Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTI-IL1RAP ANTIBODY COMPOSITIONS
Document Type and Number:
WIPO Patent Application WO/2020/035577
Kind Code:
A1
Abstract:
The present invention relates toantibody compositionsdirected to Interleukin 1 Receptor Accessory Protein (IL1RAP) and their use in the treatment and diagnosis of diseases associated with IL1RAP, such as inflammatory, autoimmune, autoinflammatory and neoplastic disorders.

Inventors:
FORSBERG GÖRAN (SE)
LIBERG DAVID (SE)
SJÖSTRÖM KJELL (SE)
VON WACHENFELDT KARIN (SE)
Application Number:
PCT/EP2019/071974
Publication Date:
February 20, 2020
Filing Date:
August 15, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CANTARGIA AB (SE)
International Classes:
C07K16/28; A61K39/395; A61P35/00
Domestic Patent References:
WO2016020502A12016-02-11
WO2015132602A12015-09-11
WO2015132602A12015-09-11
WO2016020502A12016-02-11
WO1993008829A11993-05-13
WO1996027011A11996-09-06
WO2002043478A22002-06-06
WO2013074569A12013-05-23
Foreign References:
US20170121420A12017-05-04
US38935806A2006-03-24
US39158406A2006-03-28
US47802106A2006-06-29
US63372906A2006-12-05
US6180370B12001-01-30
US5225539A1993-07-06
Other References:
H AGERSTAM ET. AL.: "Antibodies targeting human IL1RAP (IL1R3) show therapeutic effects in xenograft models of acute myeloid leukemia", PNAS, vol. 112, no. 34, 25 August 2015 (2015-08-25), pages 10786 - 10791, XP055308490, DOI: 10.1073/pnas.1422749112
H. ARGERSTAM ET. AL.: "IL1RAP antibodies block IL-1 induced expansion of candidate CML stem cells and mediate cell killing in xenograft models", BLOOD, vol. 128, 12 September 2016 (2016-09-12), pages 26832693, XP055494589, DOI: 10.1182/blood-2015-11-679985
GARLANDA ET AL., IMMUNITY, vol. 39, no. 6, 12 December 2013 (2013-12-12), pages 1003 - 18
WANG, D. ET AL., NAT IMMUNOL, vol. 11, no. 10, 2010, pages 905 - 11
THOMAS, C. ET AL., NAT STRUCT MOL BIOL, vol. 19, no. 4, 2012, pages 455 - 7
LIU, X. ET AL., PNAS, vol. 110, no. 37, 2013, pages 14918 - 23
GUNTHER, S. ET AL., J. IMMUNOL., vol. 193, 2014, pages 921 - 30
JARAS, M. ET AL., PNAS, vol. 107, no. 37, 2010, pages 16280 - 5
AGERSTAM, H. ET AL., PNAS, vol. 112, no. 34, 2015, pages 10786 - 91
DINARELLO, CA, BLOOD, vol. 87, no. 6, 1996, pages 2095 - 147
DINARELLO CA, CHEST, vol. 118, 2000, pages 503 - 508
DINARELLO, CA, CLIN EXP RHEUMATOL, vol. 20, no. 27, 2002, pages 1 - 13
DINARELLO ET AL., NATURE REVIEWS, vol. 11, 2012, pages 633 - 652
DINARELLO, MOL. MED., vol. 20, no. 1, 2014, pages S43 - S58
SCHMITZ, J. ET AL., IMMUNITY, vol. 23, 2005, pages 479 - 90
YEW LIEW, F. ET AL., NAT REV IMMUNOL, vol. 16, 2016, pages 676 - 89
FOSTER, A.M. ET AL., J IMMUNOL, vol. 192, 2014, pages 6053 - 61
MUTAMBA, S. ET AL., EUR J IMMUNOL, vol. 42, 2012, pages 607 - 17
GABAY C.OWNE, E., J LEUKOCYTE BIOL, vol. 97, 2015, pages 645 - 52
BLUMBERG. H. ET AL., J IMMUNOL, vol. 185, 2010, pages 4354 - 62
MILSTEINCUELLO, NATURE, vol. 305, 1983, pages 537 - 539
LINDHOFER ET AL., IMMUNOL., vol. 155, 1995, pages 219 - 225
TRAUNECKER ET AL., EMBO J., vol. 10, 1991, pages 3655 - 3659
SURESH ET AL., METHODS IN ENZYMOLOGY, vol. 121, 1986, pages 210
BRENNAN ET AL., SCIENCE, vol. 229, 1985, pages 81
SHALABY ET AL., J. EXP. MED., vol. 175, 1992, pages 217 - 225
KOSTELNY ET AL., J. IMMUNOL., vol. 148, no. 5, 1992, pages 1547 - 1553
HOLLINGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
GRUBER ET AL., J. IMMUNOL., vol. 152, 1994, pages 5368
TUTT ET AL., J. IMMUNOL., vol. 147, 1991, pages 60
KONTERMANNBRINKMANN, DRUG DISCOVERY TODAY, vol. 20, no. 7, 2015
WU C. ET AL., NAT, BIOTECHNOL., vol. 25, 2007, pages 1290 - 1297
WU C. ET AL., MABS, vol. 1, 2009, pages 339 - 347
WANG ET AL., NATURE IMMUNOLOGY, vol. 11, 2010, pages 905 - 912
HIGGINS D.THOMPSON J.GIBSON T.THOMPSON J.D.HIGGINS D.G.GIBSON T.J.: "CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice", NUCLEIC ACIDS RES., vol. 22, 1994, pages 4673 - 4680, XP002956304
LONBERG, N. ET AL., NATURE, vol. 368, no. 6474, 1994, pages 856 - 859
HUANG ET AL., PROC. NATL. ACAD. SCI. USA., vol. 94, no. 24, 1997, pages 12829 - 12832
WANG ET AL., NATURE IMMUNOLOGY, vol. 11, pages 905 - 912
XU ET AL., MABS, vol. 7, no. 1, 2015, pages 231 - 242
"Remington: The Science and Practice of Pharmacy", 2005, LIPPINCOTT, WILLIAMS & WILKINS
THIE, VOEDISCH ET AL., METHODS MOL BIOL, vol. 525, 2009, pages 309 - 322
KOLKMANSTEMMER, NAT BIOTECHNOL., vol. 19, no. 5, May 2001 (2001-05-01), pages 423 - 8
JOHNSONHAWKINS: "Affinity Maturation of Antibodies Using Phage Display", 1996, OXFORD UNIVERSITY PRESS
PELED, KUANG ET AL., ANNU REV IMMUNOL., vol. 26, 2008, pages 481 - 511
KOPSIDAS, ROBERTS ET AL., IMMUNOL LETT., vol. 107, no. 2, 15 November 2006 (2006-11-15), pages 163 - 8
BARBAS ET AL., PROC. NAT. ACAD. SCI. USA, vol. 91, 1994, pages 3809 - 13
SCHIER ET AL., GENE, vol. 169, 1995, pages 147 - 55
JACKSON ET AL., J. IMMUNOL., vol. 1 55, no. 7, 1995, pages 331 0 - 2004
HAWKINS ET AL., J. MOL. BIOL., vol. 226, 1992, pages 889 - 96
BENHAR, EXPERT OPIN BIOL THER., vol. 7, no. 5, May 2007 (2007-05-01), pages 763 - 79
QUEEN, SCHNEIDER ET AL., PNAS, vol. 86, no. 24, 1989, pages 10029 - 33
MARKS ET AL., BIO/TECHNOLOGY, vol. 10, 1992, pages 779 - 83
LABRIJN ET AL., PROC NATL ACAD SCI USA, vol. 110, no. 13, 26 March 2013 (2013-03-26), pages 5145 - 5150
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 525
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 327
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1536
PRESTA, CURR. OP. STRUCT. BIOL., vol. 2, 1992, pages 593 - 596
GUDJONSSON ET AL., J INVEST DER, vol. 127, 2007, pages 1292 - 1308
KUNDU-RAYCHAUDHURI ET AL., INDIAN J DERMATOLOGY, VENEREOLOGY, AND LEPROLOGY, vol. 80, no. 3, 2014, pages 204 - 213
Attorney, Agent or Firm:
SMITH, Stephen Edward (GB)
Download PDF:
Claims:
CLAIMS

1. A composition comprising at least a first binding agent with specificity for interleukin-1 receptor accessory protein (IL1 RAP) and a second binding agent with specificity for IL1 RAP, wherein the first and second binding agents bind to at least two different extracellular domains of IL1 RAP.

2. The composition according to claim 1 , wherein each of the first and second

binding agents is an anti-l L1 RAP antibody or an antigen-binding fragment.

3. The composition according to any one of the preceding claims, a) wherein a first binding agent is an anti-l L1 RAP antibody or antigen-binding fragment thereof, selected from the group consisting of:

i) the reference antibody“CAN04”;

ii) an antibody comprising the variable light chain (Vi_) amino acid sequence:

DIQMTQSPSSLSASVGDRVTITCQASQGINNYLNWYQQKPGKA PKLLIHYTSGLHAGVPSRFSGSGSGTDYTLTISSLEPEDVATYYC QQYSILPWTFGGGTKVEIKR (SEQ ID NO:1 ) and the variable heavy chain (VH) amino acid sequence:

QVQLVQSGAEVKKPGSSVKVSCKASGYAFTSSWMNWVRQAPG QGLEWMGRIYPGDGNTHYAQKFQGRVTLTADKSTSTAYMELSS LRSEDTAVYYCGEGYLDPMDYWGQGTLVTVSS (SEQ ID NO:4); iii) an antibody comprising at least one of the following six

complementary determining regions (CDRs):

Light chain CDR1 : SASQGINNYLN (SEQ ID NO: 8) or ASQGINNYLN (SEQ ID NO: 29)

Light chain CDR2: YTSGLHAGV (SEQ ID NO: 9) or

YTSGLHA (SEQ ID NO: 22)

Light chain CDR3: QQYSILPWT (SEQ ID NO: 10) or

QYSILPWT (SEQ ID NO: 23)

Heavy chain CDR1 : GYAFTSSSWMN (SEQ ID NO: 11 ) or GYAFTSS (SEQ ID NO: 24) Heavy chain CDR2: RIYPGDGNTHYAQKFQG (SEQ ID NO: 12) or YPGDGN (SEQ ID NO: 25)

Heavy chain CDR3: GYLDPMDY (SEQ ID NO: 13); and iv) an antibody binding to the same epitope as the antibody of part (i) above;

v) an antibody capable of inhibiting the binding of the antibody of part (i) above to human IL1 RAP;

vi) an antibody capable of binding to extracellular domain 2 of IL1 RAP; and

vii) an antigen-binding fragment of an antibody of (i) to (vi) above; and b) wherein a second binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof selected from the group consisting of:

i) the reference antibody“CAN03”;

ii) an antibody comprising the variable light chain (Vi_) amino acid sequence:

DILLTQSPAILSVSPGERVSFSCRASQSIGTSIHWYQRRTNGSPR LLIKSASESISGIPSRFSGSGSGTDFTLSINSVESEDIADYYCQQS NSWPTTFGAGTKLELKR (SEQ ID NO: 14), and the variable heavy chain (VH) amino acid sequence: DVKLVESGGGLVKPGGSLKLSCAASGFTFSIYTMSWVRQTPEK RLEWVATISIGGSYINYPDSVKGRFTISRDNAKNTLYLQMSSLKS EDTAIYYCSREVDGSYAMDYWGQGTSVTVSS (SEQ ID NO: 15); iii) an antibody comprising at least one of the following six

complementary determining regions (CDRs):

Light chain CDR1 : RASQSIGTSIH

(SEQ ID NO: 16) or ASQSIGTSIH (SEQ ID NO: 30)

Light chain CDR2: SASESIS (SEQ ID NO: 17)

Light chain CDR3: QQSNSWPTT (SEQ ID NO: 18) or QSNSWPTT (SEQ ID NO: 26)

Heavy chain CDR1 : GFTFSIYTMS (SEQ ID NO: 19) or GFTFSIY (SEQ ID NO: 27) Heavy chain CDR2: TISIGGSYINYPDSVKG (SEQ ID NO: 20) or SIGGSY (SEQ ID NO: 28)

Heavy chain CDR3: EVDGSYAMDY (SEQ ID NO: 21 ); iv) an antibody binding to the same epitope as antibody of part (i) above;

v) an antibody capable of inhibiting the binding of antibody of part (i) above to human IL1 RAP; and

vi) an antibody capable of binding to extracellular domain 3 of

IL1 RAP; and

vii) an antigen-binding fragment of an antibody of (i) to (vi) above.

4. The composition according to claim 3, wherein the antibody of (a) (iii) comprises at least one of the CDRs, such as at least two, such as at least three, such as at least four, such as at least five, such as all six of the CDRs.

5. The composition according to claim 4, wherein the antibody comprises all three light chain CDRs and/or all three heavy chain CDRs.

6. The composition according to claim 3, wherein the antibody of (b) (iii) comprises at least one of the CDRs, such as at least two, such as at least three, such as at least four, such as at least five, such as all six of the CDRs.

7. The composition according to claim 6, wherein the antibody comprises all three light chain CDRs and/or all three heavy chain CDRs.

8. The composition according to any one of the preceding claims, wherein the first binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a variable heavy chain (VH), and a variable light chain (Vi_) selected from the group consisting of: a) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; b) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; c) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; d) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; e) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; f) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; g) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; h) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; i) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; j) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; k) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; and

I) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3.

9. The composition according to any one of the preceding claims, wherein: a) the first binding agent is an anti-l L1 RAP antibody or antigen-binding fragment thereof comprising: a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NOs: 1 , 2, or 3; and a heavy chain variable domain (VH) comprising or consisting of the amino acid sequence of SEQ ID NOs: 4, 5, 6, or 7; and b) the second binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising: a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14; and a heavy chain variable domain (VH) comprising or consisting of the amino acids of SEQ ID NO: 15.

10. The composition according to any one of the preceding claims, wherein: a) the first binding agent is an anti-l L1 RAP antibody or antigen-binding

fragment thereof comprising: a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 1 ; and a heavy chain variable domain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 4; and b) the second binding agent is an anti-l L1 RAP antibody or antigen-binding fragment thereof comprising: a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14; and a heavy chain variable domain (VH) comprising or consisting of the amino acids of SEQ ID NO: 15.

1 1. The composition according to any one of the preceding claims, wherein the first binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 1 , or an amino acid sequence having at least 60% sequence identity, such as at least 70% sequence identity, such as at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 1.

12. The composition according to any one of the preceding claims, wherein the first binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 4, or an amino acid sequence having at least 60% sequence identity, such as at least 70% sequence identity, such as at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 4.

13. The composition according to any one of the preceding claims, wherein the first binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a light chain CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 8 or SEQ ID NO: 29, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ

ID NO: 8 or SEQ ID NO: 29, respectively.

14. The composition according to any one of the preceding claims, wherein the first binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a light chain CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 22, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 9 or SEQ ID NO: 22, respectively.

15. The composition according to any one of the preceding claims, wherein the first binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a light chain CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 23, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 10 or SEQ ID NO: 23, respectively.

16. The composition according to any one of the preceding claims, wherein the first binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a heavy chain CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 11 or SEQ ID NO: 24, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 1 1 or SEQ ID NO: 24, respectively.

17. The composition according to any one of the preceding claims, wherein the first binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a heavy chain CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 12 or SEQ ID NO: 25, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 12 or SEQ ID NO: 25, respectively.

18. The composition according to any one of the preceding claims, wherein the first binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a heavy chain CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 13, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 13.

19. The composition according to any one of the preceding claims, wherein the second binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 14.

20. The composition according to any one of the preceding claims, wherein the second binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 15, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 15.

21. The composition according to any one of the preceding claims, wherein the second binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a light chain CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 16 or SEQ ID NO: 30, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 16 or SEQ ID NO: 30, respectively.

22. The composition according to any one of the preceding claims, wherein the second binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a light chain CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 17, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 17.

23. The composition according to any one of the preceding claims, wherein the second binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a light chain CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 18 or SEQ ID NO: 26, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 18 or SEQ ID NO: 26, respectively.

24. The composition according to any one of the preceding claims, wherein the second binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a heavy chain CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 19 or SEQ ID NO: 27, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 19 or SEQ ID NO: 27, respectively.

25. The composition according to any one of the preceding claims, wherein the second binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a heavy chain CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 20 or SEQ ID NO: 28, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 20 or SEQ ID NO: 28, respectively.

26. The composition according to any one of the preceding claims, wherein the second binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a heavy chain CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 21 , or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 21.

27. The composition according to any one of the preceding claims, wherein the proportion of the first binding agent relative to the second binding agent in the composition is between 10:1 and 1 :10, such as between 5:1 and 1 :5, such as between 2:1 and 1 :2, such as between 3:2 and 2:3, such as between 1 1 :9 and 9:1 1 , such as 1 :1.

28. The composition according to any one of the preceding claims, wherein the first and second binding agents are antibody molecules of isotype subtype lgG1 , lgG2, lgG3 or lgG4.

29. The composition according to any one of the preceding claims, wherein the composition is capable of inducing internalisation of cell membrane-bound IL1 RAP.

30. The composition according to any one of the preceding claims, wherein the first and/or the second binding agent(s) lack the capacity to induce antibody dependent cell mediated cytotoxicity (ADCC).

31. A bi-epitopic binding agent comprising: a first antigen-binding region, and

a second antigen-binding region, wherein the first antigen binding region and the second antigen binding region bind to different extracellular domains of human interleukin-receptor accessory protein (IL1 RAP).

32. The bi-epitopic binding agent according to claim 31 , wherein the bi-epitopic binding agent is a polypeptide.

33. The bi-epitopic binding agent according to any one of claims 31 to 32, wherein the bi-epitopic binding agent is an antibody.

34. The bi-epitopic binding agent according to any one of claims 31 to 33, a) wherein the first antigen-binding region is selected from the group consisting of:

i) an antigen-binding region which binds to the same antigen as the

reference antibody“CAN04”;

ii) an antigen-binding region which binds to the same epitope as the

antibody CAN04;

iii) an antigen-binding region which is capable of inhibiting the binding of the antibody CAN04 to human IL1 RAP;

iv) an antigen-binding region which is capable of binding to domain 2 of IL1 RAP;

v) an amino acid sequence comprising or consisting of:

the variable light chain (Vi_) amino acid sequence:

DIQMTQSPSSLSASVGDRVTITCQASQGINNYLNWYQQKPGKAPKLLI HYTSGLHAGVPSRFSGSGSGTDYTLTISSLEPEDVATYYCQQYSILP WTFGGGTKVEIKR (SEQ ID NO:1 ) and

the variable heavy chain (VH) amino acid sequence:

DVKLVESGGGLVKPGGSLKLSCAASGFTFSIYTMSWVRQTPEKRLE WVATISIGGSYINYPDSVKGRFTISRDNAKNTLYLQMSSLKSEDTAIYY CSREVDGSYAMDYWGQGTSVTVSS (SEQ ID NO:4) of the antibody CAN04,

vi) an amino acid sequence comprising or consisting of :

the heavy chain amino acid sequence SEQ ID NO: 35 and the light chain amino acid sequence SEQ ID NO: 36,

vii) an amino acid sequence comprising or consisting of at least one of the following six complementary determining regions (CDRs): Light chain CDR1 : SASQGINNYLN (SEQ ID NO: 8) or ASQGINNYLN (SEQ ID NO: 29)

Light chain CDR2: YTSGLHAGV (SEQ ID NO: 9) or YTSGLHA (SEQ ID NO: 22)

Light chain CDR3: QQYSILPWT (SEQ ID NO: 10) or QYSILPWT (SEQ ID NO: 23)

Heavy chain CDR1 : GYAFTSSSWMN (SEQ ID NO: 11 ) or GYAFTSS (SEQ ID NO: 24)

Heavy chain CDR2: RIYPGDGNTHYAQKFQG (SEQ ID NO: 12) or YPGDGN (SEQ ID NO: 25)

Heavy chain CDR3: GYLDPMDY (SEQ ID NO: 13);

and

viii) an antigen-binding fragment of an antibody of (i) to (vi) above; and b) wherein the second antigen-binding region is selected from the group consisting of:

i) an antigen-binding region which binds to the same antigen as the

reference antibody“CAN03”;

ii) an antigen-binding region which binds to the same epitope as the

antibody CAN03;

iii) an antigen-binding region which is capable of inhibiting the binding of the antibody CAN03 to human IL1 RAP;

iv) an antigen-binding region which is capable of binding to the

extracellular domain 3 of IL1 RAP;

v) an amino acid sequence comprising or consisting of the variable light chain (Vi_) amino acid sequence:

DILLTQSPAI LSVSPGERVSFSCRASQSIGTSIHWYQRRTNGSPRLLIK

SASESISGIPSRFSGSGSGTDFTLSINSVESEDIADYYCQQSNSWPTT

FGAGTKLELKR (SEQ ID NO: 14),

and the variable heavy chain (VH) amino acid sequence:

DVKLVESGGGLVKPGGSLKLSCAASGFTFSIYTMSWVRQTPEKRLE

WVATISIGGSYINYPDSVKGRFTISRDNAKNTLYLQMSSLKSEDTAIYY

CSREVDGSYAMDYWGQGTSVTVSS (SEQ ID NO: 15) of the antibody

CAN03,

vi) an amino acid sequence comprising or consisting of : the heavy chain amino acid sequence SEQ ID NO: 37 and the light chain amino acid sequence SEQ ID NO: 38,

vii) an antigen binding region comprising at least one of the following six complementary determining regions (CDRs):

Light chain CDR1 : RASQSIGTSIH

(SEQ ID NO: 16) or ASQSIGTSIH (SEQ ID NO: 30)

Light chain CDR2: SASESIS (SEQ ID NO: 17)

Light chain CDR3: QQSNSWPTT (SEQ ID NO: 18) or QSNSWPTT (SEQ ID NO: 26)

Heavy chain CDR1 : GFTFSIYTMS (SEQ ID NO: 19) or GFTFSIY (SEQ ID NO: 27)

Heavy chain CDR2: TISIGGSYINYPDSVKG (SEQ ID NO: 20) or SIGGSY (SEQ ID NO: 28)

Heavy chain CDR3: EVDGSYAMDY (SEQ ID NO: 21 ); and

viii) an antigen-binding fragment of an antibody of (i) to (vi) above.

35. The bi-epitopic binding agent according to any one of claims 31 to 33, wherein the first antigen-binding region comprises an amino acid sequence comprising or consisting of the heavy chain amino acid sequence SEQ ID NO: 35 and the light chain amino acid sequence SEQ ID NO: 36 and wherein the second antigen- binding region comprises the heavy chain amino acid sequence SEQ ID NO: 37 and the light chain amino acid sequence SEQ ID NO: 38.

36. The bi-epitopic binding agent according to any one of claims 31 and 34, wherein the first antigen-binding region comprises a variable heavy chain (VH), and a variable light chain (Vi_) selected from the group consisting of: a) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; b) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; c) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; d) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; e) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; f) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; g) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; h) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; i) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; j) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; k) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO:

6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; and

I) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3.

37. The bi-epitopic binding agent according to any one of claims 31 to 36, wherein: a) the first antigen-binding region comprises: a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NOs: 1 , 2, or 3; and a heavy chain variable domain (VH) comprising or consisting of the amino acid sequence of SEQ ID NOs: 4, 5, 6, or 7; and b) the second antigen-binding region comprises: a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14; and a heavy chain variable domain (VH) comprising or consisting of the amino acids of SEQ ID NO: 15.

38. The bi-epitopic binding agent according to any one of claims 31 to 37, wherein: a) the first antigen-binding region comprises: a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 1 ; and a heavy chain variable domain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 4; and b) the second antigen- binding region comprises: a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14; and a heavy chain variable domain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 15.

39. The bi-epitopic binding agent according to any one of claims 31 to 38, wherein the first antigen-binding region has a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 1 , or an amino acid sequence having at least 60% sequence identity, such as at least 70% sequence identity, such as at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 1.

40. The bi-epitopic binding agent according to any one of claims 31 to 39, wherein the first antigen-binding region has a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 4, or an amino acid sequence having at least 60% sequence identity, such as at least 70% sequence identity, such as at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, , such as at least 99% sequence identity to SEQ ID NO: 4.

41. The bi-epitopic binding agent according to any one of claims 31 to 40, wherein the first antigen-binding region has a light chain CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 8 or SEQ ID NO: 29, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 8 or SEQ ID NO: 29, respectively.

42. The bi-epitopic binding agent according to any one of claims 31 to 41 , wherein the first antigen-binding region has a light chain CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 22, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 9 or SEQ ID NO: 22, respectively.

43. The bi-epitopic binding agent according to any one of claims 31 to 42, wherein the first antigen-binding region has a light chain CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 23, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 10 or SEQ ID NO: 23, respectively.

44. The bi-epitopic binding agent according to any one of claims 31 to 43, wherein the first antigen-binding region has a heavy chain CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 11 or SEQ ID NO: 24, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 1 1 or SEQ ID NO: 24, respectively.

45. The bi-epitopic binding agent according to any one of claims 31 to 44, wherein the first antigen-binding region has a heavy chain CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 12 or SEQ ID NO: 25, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 12 or SEQ ID NO: 25, respectively.

46. The bi-epitopic binding agent according to any one of claims 31 to 45, wherein the first antigen-binding region has a heavy chain CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 13, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 13.

47. The bi-epitopic binding agent according to any one of claims 31 to 46, wherein the second antigen-binding region has a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 14.

48. The bi-epitopic binding agent according to any one of claims 31 to 47, wherein the second antigen-binding region has a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 15, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 15.

49. The bi-epitopic binding agent according to any one of claims 31 to 48, wherein the second antigen-binding region has a light chain CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 16 or SEQ ID NO: 30, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 16 or SEQ ID NO: 30, respectively.

50. The bi-epitopic binding agent according to any one of claims 31 to 49, wherein the second antigen-binding region has a light chain CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 17, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 17.

51. The bi-epitopic binding agent according to any one of claims 31 to 50, wherein the second antigen-binding region has a light chain CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 18 or SEQ ID NO: 26, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 18 or SEQ ID NO: 26, respectively

52. The bi-epitopic binding agent according to any one of claims 31 to 51 , wherein the second antigen-binding region has a heavy chain CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 19 or SEQ ID NO: 27, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 19 or SEQ ID NO: 27, respectively

53. The bi-epitopic binding agent according to any one of claims 31 to 52, wherein the second antigen-binding region has a heavy chain CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 20 or SEQ ID NO: 28, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 20 or SEQ ID NO: 28, respectively

54. The bi-epitopic binding agent according to any one of claims 31 to 53, wherein the second antigen-binding region has a heavy chain CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 21 , or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 21.

55. The bi-epitopic binding agent according to any one of claims 31 to 54, wherein the bi-epitopic binding agent is a dual-variable-domain antibody, a bi-epitopic Fab-fragment, a bi-epitopic scFv, a bivalent bispecific antibody, a monovalent bispecific antibody, a‘knob-in-hole’ bispecific antibody, a scFv2_Fc bispecific antibody, a BiTE/scFv2 bispecific antibody, a DVD-lg bispecific antibody, an IgG- Fab bispecific antibody, a FAb-lgG bispecific antibody, a DART-based bispecific antibody, a DNL-Fab3 bispecific antibody or a scFv-HAS-scFv bispecific antibody.

56. A composition comprising one or more polynucleotides which, collectively or individually, encode either (i) first and second binding agents as defined in claim 1 or (ii) a bi-epitopic binding agent according to claim 31.

57. An isolated polynucleotide encoding either (i) first and second binding agents as defined in claim 1 or (ii) a bi-epitopic binding agent according to claim 31.

58. An expression vector comprising one or more polynucleotides which, collectively or individually, encode either (i) first and second binding agents as defined in claim 1 or (ii) a bi-epitopic binding agent according to claim 31.

59. A host cell comprising one or more polynucleotides which, collectively or individually, encode either (i) first and second binding agents as defined in claim 1 or (ii) a bi-epitopic binding agent according to claim 31.

60. A host cell comprising one or more expression vectors according to claim 58.

61. The host cell according to any one of claims 59 to 60, wherein said host cell is a eukaryotic or prokaryotic host cell.

62. A pharmaceutical composition comprising the composition according to any one of claims 1 to 30; the bi-epitopic binding agent according to any one of claims 31 to 55; a polynucleotide according to any one of claims 56 to 57 ; a vector according to claim 58; or a host cell according to any one of claims 59 to 61 ; and a pharmaceutically acceptable excipient.

63. A composition according to any one of claims 1 to 30; the bi-epitopic binding agent according to any one of claims 31 to 55; a polynucleotide according to any one of claims 56 to 57 ; a vector according to claim 58; a host cell according to any one of claims 59 to 61 ; or a pharmaceutical composition according to claim 62, for use as a medicament.

64. A composition according to any one of claims 1 to 30; the bi-epitopic binding agent according to any one of claims 31 to 55; a polynucleotide according to any one of claims 56 to 57 ; a vector according to claim 58; a host cell according to any one of claims 59 to 61 ; or a pharmaceutical composition according to claim 62, for use as a diagnostic and/or prognostic agent.

65. A composition according to any one of claims 1 to 30; the bi-epitopic binding agent according to any one of claims 31 to 55; a polynucleotide according to any one of claims 56 to 57 ; a vector according to claim 58; a host cell according to any one of claims 59 to 61 ; a pharmaceutical composition according to claim 62, for use in the treatment, amelioration, prevention, diagnosis or prognosis of an IL1 RAP- associated disease or disorder in a mammal.

66. The composition according to claim 65, wherein the IL1 RAP-associated disease or disorder is selected from the group consisting of proliferative disorders, autoimmune disorders, and inflammatory disorders, such as auto inflammatory disorders.

67. The composition according to any one of claims 1 to 30 or claims 65 to 66; the bi- epitopic binding agent according to any one of claims 31 to 55; the polynucleotide according to any one of claims 56 to 57 ; the vector according to claim 58; the host cell according to any one of claims 59 to 61 ; or the pharmaceutical composition according to claim 62, for use in the treatment of a disease or disorder selected from the group consisting of proliferative disorders, autoimmune disorders, and inflammatory disorders, such as auto inflammatory disorders.

68. The composition, bi-epitopic binding agent, polynucleotide, vector, host cell, or pharmaceutical composition according to any one of claims 66 and 68, wherein the disease or disorder is selected from the group consisting of rheumatoid arthritis, osteoarthritis, multiple sclerosis, artherosclerosis, scleroderma (systemic sclerosis), lupus, systemic lupus erythematosus (SLE), (acute) glomerulonephritis, asthma, chronic obstructive pulmonary diseases (COPD), respiratory distress- syndrome (ARDS), inflammatory bowel disease, colitis, vasculitis, uveitis, dermatitis, atopic dermatitis, alopecia, rhinitis (allergica), allergic conjunctivitis, myasthenia gravis, sclerodermitis, sarcoidosis, psoriatic arthritis, psoriasis, ankylosingspondylitis, juvenile idiopathic arthritis, Graves’ disease, Sjogren’s syndrome, endometriosis, Crohns disease, Behget disease, celiac disease, diabetes mellitus type 1 , Graves’ disease, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis, systemic lupus erythematosus, familial Mediterranean fever (FMF), hyperimmunoglobulinemia D with recurrent fever (HIDS), TNF receptor-associated periodic syndrome (TRAPS), cryopyrin-associated periodic syndromes (CAPS, such as Muckle- Wells syndrome, familial cold urticaria, and neonatal onset), multisystem inflammatory disease (NOMID), periodic fever, aphthous stomatitis, pharyngitis and adenitis (PFAPA syndrome), Blau syndrome, pyogenic sterile arthritis, pyoderma gangrenosum, acne (PAPA), deficiency of the interleukin-1 -receptor antagonist (DIRA), adult-onset Still's disease and systemic-onset juvenile idiopathic arthritis, cardiovascular diseases.

69. The composition, bi-epitopic binding agent, polynucleotide, vector, host cell, or pharmaceutical composition according to any one of claims 66 and 68, wherein the disease or disorder is a neoplastic disorder.

70. The composition, bi-epitopic binding agent, polynucleotide, vector, host cell, or pharmaceutical composition according to claim 69, wherein the neoplastic disorder is selected from the group consisting of prostate cancer, breast cancer, lung cancer, colorectal cancer, melanomas, bladder cancer, brain/CNS cancer, cervical cancer, oesophageal cancer, gastric cancer, head/neck cancer, kidney cancer, liver cancer, lymphomas, ovarian cancer, pancreatic cancer, sarcomas, chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML).

71. Use of a composition according to any one of claims 1 to 30; or the bi-epitopic binding agent according to any one of claims 31 to 55; or a polynucleotide according to any one of claims 56 to 57 ; or a vector according to claim 58; or a host cell according to any one of claims 59 to 61 ; or a pharmaceutical composition according to claim 62, in the manufacture of a medicament for the treatment, amelioration, prevention, diagnosis or prognosis of an I L1 RAP-associated disease or disorder in a mammal.

72. A method of treatment, amelioration, prevention, diagnosis or prognosis of an I L1 RAP-associated disease or disorder, comprising administration of a composition according to any one of claims 1 to 30; or the bi-epitopic binding agent according to any one of claims 31 to 55; or a polynucleotide according to any one of claims 56 to 57 ; or a vector according to claim 58; or a host cell according to any one of claims 59 to 61 ; or a pharmaceutical composition according to claim 62, to a subject in need thereof.

73. A first binding agent with specificity for IL1 RAP as defined in any one of Claim 1 to 30 for use in the treatment, amelioration, prevention, diagnosis or prognosis of an IL1 RAP-associated disease or disorder in a mammal, wherein the first binding agent is for use in combination with one or more further binding agents with specificity for IL1 RAP, wherein the first and further binding agents bind to at least two different extracellular domains of IL1 RAP.

Description:
ANTI-IL1 RAP ANTIBODY COMPOSITIONS

Field of invention

The present invention relates to antibodies directed to Interleukin 1 Receptor Accessory Protein (IL1 RAP) and their use in the treatment and diagnosis of diseases associated with IL1 RAP, such as inflammatory disorders, autoimmune disorders and neoplastic disorders.

Background of invention

Inflammatory and autoimmune disorders, and to a certain degree also neoplastic disorders, all have an aetiology related to cytokine mediated inflammation. The Interleukin- 1 family of cytokines is believed to be of particular relevance for the aetiology of these indications.

Interleukin-1 Receptor Associated Protein in lnterleukin-1 family signal transduction

The lnterleukin-1 (IL-1 ) family of cytokines include IL-1 a, IL-1 b, IL-18, IL-33, IL-36a, IL- 36b and II_-36g. These cytokines have structural relations and mediate pro-inflammatory effects. Each cytokine binds a specific receptor that in turn dimerizes with a co-receptor leading to signal transduction. Signaling through these receptor complexes involve the MyD88 adaptor protein and subsequent NFKB activation. In addition to the pro- inflammatory cytokines above, the IL-1 family also include two receptor antagonists, IL- 1 Ra and IL-36Ra, that compete with IL-1 and IL-36 respectively for binding to their receptors. Binding of the receptor antagonists is non-productive and does not induce a signal.

IL1 RAP is the co-receptor for the interleukin-1 receptor (IL1 R1 ), the IL-33 receptor (ST2) and the IL-36 receptor (IL1 Rrp2) and is critical for for mediating the effects of these cytokines (Garlanda et al, Immunity. 2013 Dec 12;39(6):1003-18). The structure of the IL- 1 receptor with IL1 RAP has been solved (Wang, D., et al., Nat Immunol 2010 1 1 (10):905- 1 1 , Thomas, C., et al., Nat Struct Mol Biol 2012 19(4):455-7) and the interaction between IL1 RAP and ST2 or IL1 Rrp2 are most likely very similar to the IL1 R1 -IL1 RAP interaction (Liu, X. et al., PNAS 2013 1 10(37): 14918-23, Gunther, S. et al., J. Immunol. 2014 193: 921 -30).

Antibodies generated towards IL1 RAP have the ability to block IL1 RAP-mediated signaling (Jaras, M., et al., PNAS 2010 107(37): 16280-5, figure 3). The binding epitope is crucial for the effect, since not all antibodies have the ability to block signaling even though they have the ability to bind IL1 RAP and mediate efficient ADCC (Agerstam, H. et al., PNAS 2015 1 12(34): 10786-91 ).

Anti-IL1 RAP antibodies that bind to different domains of IL1 RAP are known from WO 2015/132602 and WO 2016/020502. Of these, CAN 03 and CAN04 bind to domain 3 and 2 respectively and both show complete inhibition of IL-1 signaling and partial inhibition of IL-33 signaling.

Interleukin-1 biology lnterleukin-1 (IL-1 ) is a potent pro-inflammatory cytokine that can be produced by a variety of cell types, including mononuclear phagocytes, in response to infection and inflammation. The IL-1 family consists of seven agonists, including IL-1 a and IL-1 b, and three naturally occurring receptor antagonists, including the IL-1 receptor antagonist (IL- 1 Ra) (Dinarello, CA, Blood 1996, 87(6): 2095-147). Two IL-1 receptors, IL-1 R type I and IL-1 R type II, have been identified. Both receptors can interact with all three forms of the IL-1 family molecules. IL-1 RI is responsible for mediating IL-1 -induced cellular activation. However, the IL-1 / IL-1 RI complex cannot signal by itself, but is dependent on association with a second receptor chain, IL-1 R Accessory Protein (IL1 RAP) (Dinarello, CA, Blood 1996, 87(6): 2095-147). In contrast to IL-1 Rl, IL-1 RII does not induce cellular activation upon binding to IL-1 and thus IL-1 RII functions as regulatory decoy receptor, leading to a net decrease in IL-1 available to bind to IL-1 Rl.

IL-1 is a potent pro-inflammatory cytokine, which is induced at sites of local infection or inflammation and is involved in the regulation of a variety of physiological and cellular events (summarised in Dinarello CA, CHEST, 2000, 118: 503-508 and Dinarello, CA, Clin Exp Rheumatol, 2002, 20(5 Suppl 27): S1 -13). It is capable of activating several cell types including leukocytes and endothelial cells. IL-1 induces and amplifies immunological responses by promoting the production and expression of adhesion molecules, cytokines, chemokines and other inflammatory mediators such as prostaglandin E 2 and nitric oxide (NO). As a consequence, local inflammation is amplified and sustained. In addition, the IL- 1 -induced production of inflammatory mediators results in fever, headache, hypotension and weight loss. Furthermore, IL-1 is a hematopoietic growth factor and has been shown to reduce the nadir of leukocytes and platelets in patients during bone marrow transplantation. IL-1 has also been shown to promote angiogenesis by inducing the production of vascular endothelial growth factor, thereby promoting pannus formation and blood supply in rheumatic joints. Finally, IL-1 has been shown to promote the bone and cartilage degradation in rheumatic diseases.

IL-1 is implicated in a wide range of diseases and conditions ranging from gout to cancer (for reviews, see Dinarello ef a/., 2012, Nature Reviews 11 :633-652 and Dinarello, 2014, Mol. Med. 20(suppl. 1 ):S43-S58; the disclosures of which are incorporated herein by reference), including:

• Joint, bone and muscle diseases, such as rheumatoid arthritis and osteoarthritis;

• Hereditary systemic autoinflammatory diseases, such as familial Mediterranean fever;

• Systemic autoinflammatory diseases, such as systemic juvenile idiopathic arthritis and adult-onset Still’s disease;

• Common inflammatory diseases, such as gout and type 2 diabetes;

• Acute-onset ischemic diseases, such as myocardial infarction; and

• Cancer.

A number of therapies for blocking IL-1 activity are approved and in development. Targeting IL- 1 began in 1993 with the introduction of anakinra ( Kineret Amgen), a recombinant form of the naturally occurring IL- 1 receptor antagonist (IL- 1 Ra), which blocks the activity of both IL- 1 a and IL- 1 b; this therapeutic has since been used to demonstrate a role for IL- 1 in numerous diseases (see above). Anakinra currently dominates the field of IL- 1 therapeutics owing to its good safety record, short half-life and multiple routes of administration. Neutralising IL- 1 with antibodies or soluble receptors has also proved to be effective, and the soluble decoy receptor rilonacept ( Arcaiys , Regeneron) and the anti- IL- 1 b neutralizing monoclonal antibody canakinumab (llaris Novartis) have been approved. Other therapeutic approaches, including IL- 1 a neutralisation, a therapeutic vaccine targeting IL- 1 b and a chimeric IL- 1 Ra, are in early clinical trials. In addition, orally active small-molecule inhibitors of IL- 1 production, such as caspase 1 inhibitors, have been developed and are being tested Interleukin-33 biology

Interleukin-33 (IL-33) was identified as an IL-1 family member that induced type 2 immune responses (Schmitz, J., et al., Immunity 2005 23:479-90) by activating cells like T helper 2 (T H 2) cells and mast cells. Subsequent studies have however expanded the roles of IL- 33 and it is now considered to have other pro-inflammatory effects as well (Yew Liew, F., et al., Nat Rev Immunol 2016 16:676-89). IL-33 is released by damaged or necrotic cells as a stress signal, a so called alarmin, and exerts its effects by binding to its receptor ST2. Binding of IL-33 to ST2 recruits IL1 RAP and signaling through MYD88 is induced. A soluble form of ST2 exist that has been suggested to act as a decoy receptor. Interestingly, IL-33 has potent pro-inflammatory roles at the same time as it induces cells that limit the immune response and induce tissue repair, such as T reg and M2 macrophages.

IL-33 is involved in inflammatory diseases both as an inflammatory mediator released through stress or cell death or from failing of inducing regulatory responses (such as induction of T reg and M2 macrophages). IL-33 has been coupled to a number of diseases (for review see Yew Liew, F., et al., Nat Rev Immunol 2016 16:676-89) such as:

• Asthma

• Allergic diseases such as allergic rhinitis and atopic dermatitis

· Cardiovascular diseases

• Rheumatoid arthritis

• IBD

• Diabetes and obesity

• COPD

· Cancer

Development of IL-33 inhibitory antibodies are ongoing (Pfizer, Johnson&Johnson) and clinical trials targeting IL-33 function has been initiated in asthma and atopic dermatitis. Interleukin-36 biology

IL1 Rrp2 is expressed on human monocytes and dendritic cells which are induced by IL- 36 cytokines to produce a number of inflammatory cytokines (Foster, A.M., et al., J Immunol 2014 192:6053-61 , Mutamba, S., et al., Eur J Immunol 2012 42:607-17). IL-36 has received special attention in skin diseases such as psoriasis where there is evidence that IL-36 act upstream to induce a number of other pathological cytokines (for review see Gabay C. and Owne, E., J Leukocyte Biol 2015 97:645-52). Treatment of mice with antibodies that block IL1 Rrp2 function lead to reduction of skin pathology in immunodeficient mice transplanted with human psoriatic skin (Blumberg. H., et al., J Immunol 2010 185:4354-62) and a severe life-threatening form of psoriasis (GPP) is caused by a mutation in a natural IL-36 receptor antagonist (IL-36Ra). Reports also support a function of IL-36 cytokines in lung pathology, including COPD and asthma, but the role of these cytokines in these diseases is less clear.

Current treatments of inflammatory and autoimmune diseases are not able to inhibit the key cytokines described above, in a concerted manner.

Summary of invention

The present inventors have developed novel antibodies and antibody compositions that have been found to block I L1 RAP-mediated signalling via key inflammatory cytokines, in a concerted and synergistic manner. This unexpected property makes the agents of the present invention particularly suitable for treatment of inflammatory and autoimmune diseases and other conditions associated with IL1 RAP and/or responsive to inhibition of IL-1 signalling, IL-33 signalling and/or IL-36 signalling.

In one aspect, the present invention concerns a composition comprising at least a first binding agent with specificity for anti-lnterleukin-1 Receptor Accessory Protein (IL1 RAP) and a second binding agent with specificity for IL1 RAP, wherein the first and second binding agents bind to at least two different extracellular domains of IL1 RAP. In another aspect, the present invention concerns a bi-epitopic binding agent comprising: a first antigen-binding region, and

a second antigen binding region, wherein the first antigen binding region and the second antigen binding region bind to different extracellular domains of human interleukin-receptor accessory protein (IL1 RAP).

In one aspect, the present invention concerns a composition comprising one or more polynucleotides, which, collectively or individually, encode either (i) first and second binding agents or (ii) a bi-epitopic binding agent as defined herein. In another aspect, the present invention concerns an isolated polynucleotide encoding either (i) first and second binding agents or (ii) a bi-epitopic binding agent as defined herein. In yet another aspect, the present invention concerns an expression vector comprising one or more polynucleotides which, collectively or individually, encode either (i) first and second binding agents or (ii) a bi-epitopic binding agent as defined herein. In yet another aspect, the current invention concerns a host cell comprising one or more polynucleotides which, collectively or individually, encode either (i) first and second binding agents or (ii) a bi- epitopic antibody as defined herein. In yet another aspect, the current invention concerns a host cell comprising one or more expression vectors as described herein.

In one aspect, the current invention concerns a composition comprising at least two binding agents; or a bi-epitopic binding agent; or a polynucleotide; or a vector; or a host cell; or a pharmaceutical composition as described herein for use as a medicament.

In one aspect, the current invention concerns a composition comprising at least two binding agents; or a bi-epitopic binding agent; or a polynucleotide; or a vector; or a host cell; or a pharmaceutical composition as described herein for use as a diagnostic and/or prognostic agent.

In one aspect, the current invention concerns a composition comprising at least two binding agents; or a bi-epitopic binding agent; or a polynucleotide; or a vector; or a host cell; or a pharmaceutical composition as described herein for use in the treatment, amelioration, prevention, diagnosis or prognosis of a I L1 RAP-associated disease or disorder. Description of drawings

Figure 1. Binding of exemplary antibodies in an indirect ELISA to human IL-1 RAP. The exemplary antibodies of the invention, CAN03 and CAN04, was found to possess the highest affinities for human IL-1 RAP.

Figure 2. Binding of exemplary antibodies to human ILI RAP-expressing KU812 CML cells. The graph shows the mean fluorescence intensity (MFI) value as measured by flow cytometry of KU812 cells stained with I L1 RAP-targeting monoclonal antibodies at a concentration of 0.1 pg/mL. The exemplary antibodies of the invention, CAN03 and CAN04, has the highest MFI of the compared antibodies.

Figure 3. Ability of the exemplary antibodies CAN03 and CAN04 and their combination to block IL-1 b or IL-33 signalling in a IL-1/IL-33-responsive HEK reporter system. (A) Both CAN03 and CAN04 blocks IL-1 b signalling completely although CAN04 has superior potency. The combination of CAN03 and CAN04 (1 :1 ) has the same efficacy and potency as CAN04 on its own. (B) CAN03 and CAN04 inhibits IL-33 signalling with 70% (CAN03) or 50% (CAN04) efficacy. The combination of CAN03 and CAN04 (1 :1 ) shows a synergistic increase in both potency and efficacy compared to the antibodies alone.

Figure 4. Ability of the exemplary antibodies CAN03 and CAN04 and their combination to block IL-36a signalling in a IL-36-responsive modified HEK reporter system. CAN04 at the maximum tested concentration blocks IL-36a signalling to 45% of the signal at 1 ng/ml IL- 36a (A) and to 75% at 10 ng/ml (B), CAN03 has very little effect at both concentrations of IL-36a. The combination of CAN03 and CAN04 (1 :1 ) however shows a synergistic increase in both potency and efficacy compared to the antibodies alone.

Figure 5. Ability of the exemplary antibodies CAN03 and CAN04 and their combination to block IL-1 b induced IL-6 production in the Hs578T (HTB-126) breast cancer cell line. Cells were stimulated with IL1 b in the presence or absence of the indicated antibodies. CAN03 and CAN04 at 10 pg/ml inhibits IL-6 production to 70% (CAN03) or 50% (CAN04) of the control while the combination of CAN03 and CAN04 (at 5 + 5 pg/ml) shows a synergistic increase in inhibition to 10% of control. Figure 6. Ability of the exemplary bi-epitopic antibody bsACAN03xCAN04 (containing one CAN 04 VH/VL domain and one CAN03 VH/VL domain) and CAN04 to block IL-1 b or IL- 33 signalling in a IL-1/IL-33-responsive HEK reporter system. (A) CAN04 as well bsACAN03xCAN04 blocks I L-1 b signalling completely. (B) CAN04 inhibits IL-33 signalling although with partial efficacy. Inclusion of the CAN03 domain in the bi-epitopic bsACAN03xCAN04 antibody however allows for complete inhibition of IL-33 signalling.

Detailed description of the invention

The invention is as defined in the claims.

Definitions

The term“agent” as herein refers to a compound or substance capable of interacting with interleukin-1 accessory protein (IL1 RAP). The agent is as defined in the claims, preferably an antibody, an antibody-mimetic or a composition of antibodies.

The term“affinity-matured antibody” refers to an antibody with one or more alterations in one or more HVRs thereof which result in an improvement in the affinity of the antibody for antigen, compared to a parent antibody which does not possess those alteration(s). Preferred affinity-matured antibodies will have nanomolar or even picomolar affinities for the target antigen. The technique of affinity maturation provides a method for enhancing antigen-neutralizing ability of an antibody and may increase the antigen-binding activity by introducing mutation(s) into amino acid residue(s) in the CDRs and/or framework regions of an antibody variable domain. Improving the antigen-binding properties of an antibody may improve the biological activity of the antibody in vitro or reduce the dosage, and may further improve the efficacy in vivo (in the body).

The term“amino acid” as used herein includes the standard twenty genetically-encoded amino acids and their corresponding stereoisomers in the ‘D’ form (as compared to the natural ‘L’ form), omega-amino acids and other naturally-occurring amino acids, unconventional amino acids (e.g. a,a-disubstituted amino acids, N-alkyl amino acids, etc.) and chemically derivatised amino acids (see below).

When an amino acid is being specifically enumerated, such as“alanine” or“Ala” or“A”, the term refers to both L-alanine and D-alanine unless explicitly stated otherwise. Other unconventional amino acids may also be suitable components for polypeptides of the present invention, as long as the desired functional property is retained by the polypeptide. For the peptides shown, each encoded amino acid residue, where appropriate, is represented by a single letter designation, corresponding to the trivial name of the conventional amino acid.

In one embodiment, the antibody polypeptides as defined herein comprise or consist of L- amino acids.

The term“bi-epitopic antibody” is an antibody with specificity for two different epitopes of the same antigen, such as wherein the two epitopes are different domains of IL1 RAP. Bi-epitopic antibodies can be prepared as full-length antibodies or low molecular weight forms thereof (e.g. F(ab') 2 bi-epitopic antibodies, sc(Fv)2 bi-epitopic antibodies, diabody bi-epitopic antibodies, tri-bodies and miniantibodies).

Methods for making bi-epitopic antibodies are known in the art. Traditionally, the recombinant production of bi-epitopic antibodies is based on the co-expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chains have different specificities [Milstein and Cuello, Nature, 305:537-539 (1983)]. Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of ten different antibody molecules, of which only one has the correct bispecific structure. Further improvements of the quadroma technique has been made through creation of chimeric quadroma which facilitates enrichment of antibody with correct bi-epitopic structure (Lindhofer et al. Immunol. 155:219-225 (1995). The purification of the correct molecule is usually accomplished by affinity chromatography steps. Similar procedures are disclosed in WO 93/08829, published 13 May 1993, and in Traunecker et al., EMBO J., 10:3655-3659 (1991 ).

Antibody variable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CHI) containing the site necessary for light-chain binding present in at least one of the fusions. DNAs encoding the immunoglobulin heavy-chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host organism. For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121 :210 (1986).

According to another approach described in WO 96/2701 1 , the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture. The preferred interface comprises at least a part of the CH3 region of an antibody constant domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.

Bi-epitopic antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab' )2 bi-epitopic antibodies).

Techniques for generating bispecific antibodies from antibody fragments have been described in the literature. For example, bi-epitopic antibodies can be prepared using chemical linkage. Brennan et al., Science 229:81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab' )2 fragments. Bi-epitopic antibodies can in one embodiment be considered as a type of bispecific antibody, and can thus be prepared in the same manner as bispecific antibodies. The fragments referred to herein above are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal di thiols and prevent intermolecular disulfide formation. The Fab' fragments generated are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab' - TNB derivatives is then reconverted to the Fab '-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab' -TNB derivative to form the bi-epitopic antibody. The bi-epitopic antibodies produced can be used as agents for the selective immobilization of enzymes.

Fab' fragments may be directly recovered from E. coli and chemically coupled to form bi- epitopic antibodies. Shalaby et al., J. Exp. Med. 175:217-225 (1992) describe the production of a fully humanized bispecific antibody F(ab' )2 molecule. Each Fab' fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bi-epitopic antibody. The bi-epitopic antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets.

Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, bi-epitopic antibodies have been produced using leucine zippers. Kostelny et al., J. Immunol. 148(5): 1547- 1553 (1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The "diabody" technology described by Hollinger et al., Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (Vi_) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and Vi_ domains of one fragment are forced to pair with the complementary Vi_ and VH domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See, Gruber et al., J. Immunol. 152:5368 (1994). Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al., J. Immunol. 147:60 (1991 ). A more recent approach for manufacturing bispecific antibodies is described in Kontermann & Brinkmann, (2015) Drug Discovery Today 20(7). The methods described therein can directly be applied by those of skill in the art for producing bi-epitopic antibodies.

Another method of making and using bispecific or multispecific constructs is known as the dock-and-lock technology (U.S. patent application Ser. Nos. 1 1/389,358, filed Mar. 24, 2006; 1 1/391 ,584, filed Mar. 28, 2006; 1 1/478,021 , filed Jun. 29, 2006; 1 1/633,729, filed Dec. 5, 2006). The DNL bispecific antibodies are designed by utilizing the specific interaction between the regulatory (R) subunits of cAMP-dependent protein kinase (PKA) and the anchoring domains of A kinase anchor proteins (AKAPs), the mechanism of which has been applied in generation of bioactive conjugates of distinct protein and nonprotein molecules. The generated bispecific antibody is without a Fc region, and has a rather short half-life suitable for pre-targeting approaches. Another approach is the generation of dual-variable-domain Immunoglobulin (DVD-lg) antibodies which combines the variable regions from two different antibodies in tandem by naturally occurring linkers. The binding affinity of both variable regions can be preserved and function independently without significant steric hindrance (Wu C. et al., Nat, Biotechnol. 25; 1290-1297 (2007). This approach has been used to generate antibodies which simultaneously bind IL-1 a and I L-1 b (Wu C. et al. MAbs 1 :339-347 (2009).

The term“extracellular domain” as used herein refers to a domain of IL1 RAP which is located extracellularly, in whole or in part, when IL1 RAP forms a cell membrane-bound complex with an IL1 receptor (e.g. IL1 R type I or type II). In human IL1 RAP, three extracellular domains are known to exist:

Domain 1 : Amino acids 21 to 134

Domain 2: Amino acids 135 to 234

Domain 3: Amino acids 235 to 367

(wherein the amino acid numbering is according to Accession No. Q9NPH3 within UniProtKB/Swiss-Prot).

The structure of IL1 RAP is further defined in Wang et al., 2010, Nature Immunology, 11 :905-912 (the disclosures of which are incorporated herein by reference; see also Example C below).

Thus, the first or second binding agent of the compositions of the invention, or the first or second antigen-binding region of the bi-epitopic antibodies of the invention, may have specificity for domain 1 of IL1 RAP. For example, the epitope to which the antibody or antigen-binding fragment binds may be located within amino acids 21 to 39, 40 to 59, 60, to 79, 80, to 99, 100 to 1 19, or between amino acids 120 to 134 of IL1 RAP. However, it will be appreciated that the epitope may be non-linear.

In addition, or alternatively, the first or second binding agent of the compositions of the invention, or the first or second antigen-binding region of the bi-epitopic antibodies of the invention, may have specificity for domain 2 of IL1 RAP. For example, the epitope to which the antibody or antigen-binding fragment binds may be located within amino acids 135 to 154, 155 to 174, 175 to 194, 195 to 214 or between amino acids 215 to 234 of IL1 RAP. However, it will be appreciated that the epitope may be non-linear. In addition, or alternatively, the first or second binding agent of the compositions of the invention, or the first or second antigen-binding region of the bi-epitopic antibodies of the invention, may have specificity for domain 3 of IL1 RAP. For example, the epitope to which the antibody or antigen-binding fragment binds may be located within amino acids 235 to 249, 250 to 269, 270 to 289, 290, to 309, 310, to 329, 330, to 349 or between amino acids 350 to 367 of IL1 RAP. However, it will be appreciated that the epitope may be non-linear.

The term“sequence identity” as used herein refers to a comparative measure of two amino acid or polynucleotide sequences. The level of sequence identity indicates the likelihood that a first sequence is derived from a second sequence. Amino acid sequence identity requires identical amino acid sequences between two aligned sequences. Thus, a candidate sequence sharing 70% amino acid identity with a reference sequence, requires that, following alignment, 70% of the amino acids in the candidate sequence are identical to the corresponding amino acids in the reference sequence. Identity may be determined by aid of computer analysis, such as, without limitations, the ClustalW computer alignment program (Higgins D., Thompson J., Gibson T., Thompson J.D., Higgins D.G., Gibson T.J., 1994. CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic Acids Res. 22:4673-4680), and the default parameters suggested therein. The ClustalW software is available as a ClustalW WWW Service at the European Bioinformatics Institute from http://www.ebi.ac.uk/clustalw. Using this program with its default settings, the mature (bioactive) part of a query and a reference polypeptide are aligned. The number of fully conserved residues are counted and divided by the length of the reference polypeptide.

The ClustalW algorithm may similarly be used to align nucleotide sequences. Sequence identities may be calculated in a similar way as indicated for amino acid sequences.

Antibodies and antigen binding fragments

In one embodiment, one or both of said binding agents are individually an antibody or an antigen binding fragment.

By“an antibody or an antigen-binding fragment” we include substantially intact antibody molecules, as well as chimeric antibodies, humanised antibodies, isolated human antibodies, single chain antibodies, bi-epitopic antibodies, bispecific antibodies, antibody heavy chains, antibody light chains, homodimers and heterodimers of antibody heavy and/or light chains, and antigen-binding fragments and derivatives of the same. Suitable antigen-binding fragments and derivatives include, but are not necessarily limited to, Fv fragments (e.g. single chain Fv and disulphide-bonded Fv), Fab-like fragments (e.g. Fab fragments, Fab’ fragments and F(ab) 2 fragments), single variable domains (e.g. VH and Vi_ domains) and domain antibodies (dAbs, including single and dual formats [i.e. dAb-linker-dAb]). The potential advantages of using antibody fragments, rather than whole antibodies, are several-fold. The smaller size of the fragments may lead to improved pharmacological properties, such as better penetration of solid tissue. Moreover, antigen- binding fragments such as Fab, Fv, ScFv and dAb antibody fragments can be expressed in and secreted from E. coli, thus allowing the facile production of large amounts of the said fragments.

The term "antibody" refers to an immunoglobulin molecule, a fragment of an immunoglobulin molecule, or a derivative of either thereof. The term “immunoglobulin molecule” in turn refers to a class of structurally related glycoproteins consisting of two pairs of polypeptide chains, one pair of light (L) low molecular weight chains and one pair of heavy (H) chains, all four inter-connected by disulfide bonds. Each heavy chain typically is comprised of a heavy chain variable region (VH) and a heavy chain constant region (CH). The heavy chain constant region typically is comprised of three domains, CH1 , CH2, and CH3. Each light chain typically is comprised of a light chain variable region (Vi_) and a light chain constant region (CL). The light chain constant region typically is comprised of one domain, CL. The VH and VL regions may be further subdivided into regions of hypervariability (or hypervariable regions which may be hypervariable in sequence and/or form of structurally defined loops), also termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FRs). Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4.

The term“antigen” refers to a molecule comprising at least one epitope. The antigen may for example be a polypeptide, polysaccharide, protein, lipoprotein or glycoprotein.

The term“antigen-binding fragment” refers to fragments of antibodies retaining the ability to specifically bind to an antigen. A“bi-epitopic antibody” as used herein refers to an antibody capable of binding to two different epitopes simultaneously. A bi-epitopic antibody may be a bispecific antibody. A “bispecific antibody” as used herein refers to an antibody capable of binding to two different epitopes on two different molecules.

The term“chimeric antibody” refers to antibodies comprising regions derived from different species. The chimeric antibody may for example comprise variable regions from one animal species and constant regions from another animal species. For example, a chimeric antibody can be an antibody having variable regions which derive from a mouse monoclonal antibody and constant regions which are human. Such antibodies may also be referred to as humanised antibodies.

The antigen binding fragment of an antibody may also be a“diabody”, which are small antibody fragments with two antigen-binding sites. Diabodies preferably comprises a heavy chain variable domain (VH) connected to a light chain variable domain (Vi_) in the same polypeptide chain (VH-VL).

The term “domain antibodies” (dAbs) refers to antigen-binding fragments of antibodies, preferably ranging from 1 1 kDa to 15 kDa.

The term“dual-variable-domain antibody” refers to antibody molecules with two different binding specificities. Each light and heavy chain contains two different variable regions joined by short linker sequences. The N-terminal variable regions of the heavy and light chains are of one binding specificity, and the adjacent variable regions of the same heavy and light chains are of a different specificity. These extended heavy and light chains are synthesized and assembled into covalent molecules containing two heavy chains and two light chains.

The term“epitope” refers to a determinant capable of specific binding to an antibody. Epitopes may for example be comprised within polypeptides, polysaccharide, proteins, lipoproteins or glycoproteins. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. Epitopes may be conformational or non-conformational, wherein binding to the former but not the latter is lost in the presence of denaturing solvents. Epitopes may be continuous or discontinuous, wherein a discontinuous epitope is a conformational epitope on a protein antigen which is formed from at least two separate regions in the primary sequence of the protein.

The term“Fab fragment” refers to an antigen-binding fragment of an antibody, consisting of one constant and one variable domain of each of the heavy and the light chain.

The term“humanized antibodies” refers to antibodies from non-human species whose protein sequences have been modified to increase their similarity to antibody variants produced naturally in humans.

The antibody according to the invention may be a human or a humanized antibody. A human antibody as used herein is an antibody, which is obtained from a system using human immunoglobulin sequences. Human antibodies may for example be antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom. Human antibodies may also be isolated from a host cell transformed to express the antibody, e.g., from a transfectoma. Human antibodies may also be isolated from a recombinant, combinatorial human antibody library.

Human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies can be subjected to in vitro mutagenesis or in vivo somatic mutagenesis and thus the amino acid sequences of the VH and Vi_ regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and Vi_ sequences, may not naturally exist within the human antibody germline repertoire in vivo.

A human antibody is preferably at least 90%, more preferably at least 95%, even more preferably at least 96%, 97%, 98%, or 99% identical in amino acid sequence to the amino acid sequence encoded by a wild type human immunoglobulin gene.

Said transgenic of transchromosomal animal may contain a human immunoglobulin gene miniloci that encodes unrearranged human heavy (m and/or y) and k light chain immunoglobulin sequences. Furthermore, the animal may contain one or more mutations that inactivate the endogenous heavy and light chain loci. Examples of such animals are described in Lonberg, N. et al. (1994) Nature 368 (6474):856-859 and WO 02/43478. The antibody according to the invention may be a chimeric antibody, i.e. an antibody comprising regions derived from different species. The chimeric antibody may for example comprise variable regions from one species of animal and constant regions from another species of animal. For example, a chimeric antibody can be an antibody having variable regions which derive from a mouse monoclonal antibody and constant regions which are human. Such antibodies may also be referred to as humanized antibodies.

Thus, the antibody according to the invention may also be a humanized antibody, which is encoded partly by sequences obtained from human germline immunoglobulin sequences and partly from other sequences. Said other sequences are preferably germline immunoglobulins from other species, more preferably from other mammalian species. In particular a humanized antibody may be an antibody in which the antigen binding site is derived from an immunoglobulin from a non-human species, preferably from a non-human mammal, e.g. from a mouse or a rat, whereas some or all of the remaining immunoglobulin- derived parts of the molecule is derived from a human immunoglobulin. The antigen binding site from said non-human species may for example consist of a complete Vi_ or VH or both or one or more CDRs grafted onto appropriate human framework regions in Vi_ or VH or both. Thus, in a humanized antibody, the CDRs can be from a mouse or rat monoclonal antibody and the other regions of the antibody are of human origin.

By“reference antibody‘CAN03’” we include an intact IgG antibody comprising a heavy chain variable regions having the amino acid sequence of SEQ ID NO: 14; a light chain variable regions having the amino acid sequence of SEQ ID NO:15; a heavy chain constant region having the amino acid sequence of SEQ ID NO: 33, and a light chain constant region having the amino acid sequence of SEQ ID NO: 34 . Alternatively, a humanised version of CAN03 may be used as the reference antibody. The reference antibody CAN03 is described in WO 2016/020502.

By“reference antibody‘CAN04’” we include an intact IgG antibody comprising a heavy chain variable region selected from the group consisting of SEQ ID NO: 4, SEQ ID NO: 5, SEQ I D NO: 6, and SEQ ID NO: 7; a light chain variable region selected from the group consisting of SEQ ID NO: 1 , SEQ ID NO: 2, and SEQ ID NO: 3; a heavy chain constant region having the amino acid sequence of SEQ ID NO: 31 ; and a light chain constant region having the amino acid sequence of SEQ ID NO: 32. In a preferred embodiment, the heavy chain variable region is SEQ ID NO: 4, and the light chain variable region selected from the group consisting of SEQ ID NO: 1 . Alternatively, a humanised version of CAN04 may be used as the reference antibody. The reference antibody CAN04 is described in WO 2015/132602.

The term“scFv” refers to single-chain Fv. A Fv fragment is a fragment wherein the variable regions of the heavy (VH) and the light (Vi_) chain are associated to form the binding site of the antibody. Flexible linkers can be used to covalently join a VH and a Vi_ chain, forming a scFv that retains the binding specificity of the original antibody.

The term “single-chain antibodies” refers to antibodies that are single polypeptides comprising one or more antigen binding sites. Single chain antibodies may comprise the two domains of the Fv fragment, Vi_ and VH.

Binding to extracellular domains of I L1 RAP

In one aspect, the present invention concerns a composition comprising at least a first binding agent with specificity for anti-lnterleukin-1 Receptor Accessory Protein (IL1 RAP) and a second binding agent with specificity for IL1 RAP, wherein the first and second binding agents bind to at least two different extracellular domains of IL1 RAP. For example, the first and second binding agents may bind, respectively, to the following extracellular domains od IL1 RAP:

(a) domain 1 and domain 2;

(b) domain 1 and domain 3; or

(c) domain 2 and domain 3.

In one embodiment, the composition comprises binding agents that collectively bind to all three extracellular domains of IL1 RAP, i.e. domains 1 , 2 and 3.

By“interleukin-1 receptor accessory protein”, “IL1 RAP” and “IL1 -RAP” we specifically include the human IL1 RAP protein, for example as described in GenBank Accession NO: AAB84059, NCBI Reference Sequence: NP_002173.1 and UniProtKB/Swiss-Prot Accession NO: Q9NPH3-1 (see also Huang et al., 1997, Proc. Natl. Acad. Sci. USA. 94 (24), 12829-12832, the disclosures of which are incorporated herein by reference). IL1 RAP is also known in the scientific literature as IL1 R3, C3orf13, FLJ37788, IL-1 RAcP and EG3556. Thus, the binding agents of the invention have specificity for IL1 RAP. By“specificity” we mean that the binding agents are capable of binding to IL1 RAP in vivo, i.e. under the physiological conditions in which IL1 RAP exists within the human body. Preferably, the binding agent does not bind to any other protein in vivo. Such binding specificity may be determined by methods well known in the art, such as ELISA, immunohistochemistry, immunoprecipitation, Western blots and flow cytometry using transfected cells expressing IL1 RAP. Advantageously, the binding agent is capable of binding selectively to IL1 RAP, i.e. it bind at least 10-fold more strongly to IL1 RAP than to any other proteins.

The reference antibody‘CAN04’ binds to domain 2 of IL1 RAP (see Wang et al., 2010, Nature Immunology, 11 :905-912, the disclosures of which are incorporated herein by reference), i.e. within amino acids 135 to 234 of IL1 RAP (see Accession No. Q9NPH3 within UniProtKB/Swiss-Prot). For example, the epitope to which the antibody or antigen- binding fragment may be located within amino acids 135 to 154, 155 to 174, 175 to 194, 195 to 214 or between amino acids 215 to 234 of IL1 RAP. However, it will be appreciated that the epitope may be non-linear.

In one embodiment, the antibody or antigen-binding fragment in the compositions of the invention is capable of binding to an epitope on the extracellular domain of IL1 RAP which overlaps, at least in part, with the epitope on IL1 RAP to which reference antibody CAN04 is capable of binding. Thus, the antibody or antigen-binding fragment may be capable of binding to an epitope located at/within domain 2 of IL1 RAP.

The reference antibody‘CAN03’ binds to domain 3 of IL1 RAP. Thus, it will be appreciated that the antibody or an antigen-binding fragment in the compositions of the invention also binds to domain 3 of IL1 RAP.

By“domain 3” of IL1 RAP we include the structural region defined by amino acids 235 to 369 of IL1 RAP according to numbering used in Accession No. Q9NPH3 of UniProtKB/Swiss-Prot (see also Wang et al., 2010, Nature Immunology, 11 :905-912, the disclosures of which are incorporated herein by reference). For example, the epitope to which the antibody or antigen-binding fragment binds may be located within amino acids 235 to 239, 240 to 249, 250 to 259, 260 to 269, 270 to 279, 280 to 289, 290 to 299, 300 to 309, 310 to 319, 320 to 329, 330 to 229, 240 to 349, 350 to 359 or between amino acids 360 to 369 of I L1 RAP. The expression “capable of inhibiting the binding of reference antibody ‘CAN04’ (or ‘CAN03’) to human IL1 RAP” refers to the presence of the antibody polypeptides which inhibit, in whole or in part, the binding of ‘CAN04’ (or‘CAN03’) to human IL1 RAP. An antibody capable of inhibiting the binding of antibody CAN04 (or CAN03) is to be understood as an antibody that is competing with CAN04 (or CAN03) for binding to IL1 RAP. Such competitive binding inhibition can be determined using assays and methods well known in the art, for example using Surface Plasmon Resonance, or flow cytometry, or an ELISA.

Composition comprising at least two binding agents that bind to at least two different extracellular domains of IL1RAP

In one embodiment, the present invention concerns a composition wherein: a) a first binding agent is an anti-l L1 RAP antibody or antigen-binding fragment thereof, selected from the group consisting of:

i) the reference antibody“CAN04”,

ii) an antibody comprising the variable light chain (Vi_) amino acid sequence:

DIQMTQSPSSLSASVGDRVTITCQASQGINNYLNWYQQKP GKAPKLLIHYTSGLHAGVPSRFSGSGSGTDYTLTISSLEPED VATYYCQQYSILPWTFGGGTKVEIKR (SEQ ID NO: 1 ) and the variable heavy chain (VH) amino acid sequence:

QVQLVQSGAEVKKPGSSVKVSCKASGYAFTSSWMNWVRQ APGQGLEWMGRIYPGDGNTHYAQKFQGRVTLTADKSTSTA YMELSSLRSEDTAVYYCGEGYLDPMDYWGQGTLVTVSS

(SEQ ID NO: 4),

iii) an antibody comprising at least one of the following six complementary determining regions (CDRs):

Light chain CDR1 : SASQGINNYLN (SEQ ID NO: 8) or ASQGINNYLN (SEQ ID NO: 29)

Light chain CDR2: YTSGLHAGV (SEQ ID NO: 9) or YTSGLHA (SEQ ID NO: 22)

Light chain CDR3: QQYSILPWT (SEQ ID NO: 10) or QYSILPWT (SEQ ID NO: 23) Heavy chain CDR1 : GYAFTSSSWMN (SEQ ID NO: 1 1 ) or GYAFTSS (SEQ ID NO: 24)

Heavy chain CDR2: RIYPGDGNTHYAQKFQG (SEQ ID NO: 12) or YPGDGN (SEQ ID NO: 25)

Heavy chain CDR3: GYLDPMDY (SEQ ID NO: 13); and iv) an antibody binding to the same epitope as the antibody of part (i) above,

v) an antibody capable of inhibiting the binding of the antibody of part (i) above to human IL1 RAP;

vi) an antibody capable of binding to extracellular domain 2 of IL1 RAP; and

vii) an antigen-binding fragment of an antibody of (i) to (vi) above; and b) wherein a second binding agent is an anti-l L1 RAP antibody or antigen- binding fragment thereof selected from the group consisting of:

i) the reference antibody“CAN03”;

ii) an antibody comprising the variable light chain (Vi_) amino acid sequence:

DILLTQSPAILSVSPGERVSFSCRASQSIGTSIHWYQRRTNG SPRLLIKSASESISGIPSRFSGSGSGTDFTLSINSVESEDIAD YYCQQSNSWPTTFGAGTKLELKR (SEQ ID NO: 14), and the variable heavy chain (V H ) amino acid sequence:

DVKLVESGGGLVKPGGSLKLSCAASGFTFSIYTMSWVRQT PEKRLEWVATISIGGSYINYPDSVKGRFTISRDNAKNTLYLQ MSSLKSEDTAIYYCSREVDGSYAMDYWGQGTSVTVSS

(SEQ ID NO: 15);

iii) an antibody comprising at least one of the following six complementary determining regions (CDRs):

Light chain CDR1 : RASQSIGTSIH

(SEQ ID NO: 16) or ASQSIGTSIH (SEQ ID NO: 30)

Light chain CDR2: SASESIS (SEQ ID NO: 17) Light chain CDR3: QQSNSWPTT (SEQ ID NO: 18) or

QSNSWPTT (SEQ ID NO: 26)

Heavy chain CDR1 : GFTFSIYTMS (SEQ ID NO: 19) or GFTFSIY (SEQ ID NO: 27)

Heavy chain CDR2: TISIGGSYINYPDSVKG (SEQ ID NO: 20) or SIGGSY (SEQ ID NO: 28)

Heavy chain CDR3: EVDGSYAMDY (SEQ ID NO: 21 ); iv) an antibody binding to the same epitope as antibody of part (i) above;

v) an antibody capable of inhibiting the binding of antibody of part (i) above to human IL1 RAP;

vi) an antibody capable of binding to extracellular domain 3 of IL1 RAP; and

vii) an antigen-binding fragment of an antibody of (i) to (vi) above.

In one embodiment, the antibody of (a) (iii) above comprises at least one of the CDRs, such as at least two, such as at least three, such as at least four, such as at least five, such as all six of the CDRs. In another embodiment, said antibody comprises all three light chain CDRs and/or all three heavy chain CDRs.

In one embodiment, the antibody of (b) (iii) comprises at least one of the CDRs, such as at least two, such as at least three, such as at least four, such as at least five, such as all six of the CDRs. In another embodiment, said antibody comprises all three light chain CDRs and/or all three heavy chain CDRs.

In one embodiment, said first binding agent is an anti-l L1 RAP antibody or antigen-binding fragment thereof comprising a variable heavy chain (VH), and a variable light chain (Vi_) selected from the group consisting of: a) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; b) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; c) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; d) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; e) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; f) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; g) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; h) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; i) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; j) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; k) a variable heavy chain (V H ) comprising the amino acid sequence SEQ ID NO: 6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; and

I) a variable heavy chain (V H ) comprising the amino acid sequence SEQ ID NO: 7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3.

In one embodiment, said first binding agent is an anti-l L1 RAP antibody or antigen-binding fragment thereof comprising: a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NOs: 1 , 2, or 3; and a heavy chain variable domain (V H ) comprising or consisting of the amino acid sequence of SEQ ID NOs: 4, 5, 6, or 7; and said second binding agent is an anti-l L1 RAP antibody or antigen-binding fragment thereof comprising: a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14; and a heavy chain variable domain (V H ) comprising or consisting of the amino acids of SEQ ID NO: 15.

Preferably, said first binding agent is an anti-l L1 RAP antibody or antigen-binding fragment thereof comprising a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 1 and a heavy chain variable domain (V H ) comprising or consisting of the amino acid sequence of SEQ ID NO: 4, and said second binding agent is an anti-IL1 RAP antibody or antigen-binding fragment thereof comprising a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14 and a heavy chain variable domain (V H ) comprising or consisting of the amino acids of SEQ ID NO: 15.

In some embodiments, said first binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 1 , or an amino acid sequence having at least 60% sequence identity, such as at least 70% sequence identity, such as at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 1 .

In some embodiments, said first binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 1 , or an amino acid sequence wherein any one amino acid of SEQ ID NO: 1 , has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said first binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 4, or an amino acid sequence having at least 60% sequence identity, such as at least 70% sequence identity, such as at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 4.

In some embodiments, said first binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 4, or an amino acid sequence wherein any one amino acid of SEQ ID NO: 4, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said first binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a light chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO:10, SEQ ID NO:22, SEQ ID NO: 23, and SEQ ID NO: 29, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO:10, SEQ ID NO:22, SEQ ID NO: 23, and SEQ ID NO: 29, respectively.

In some embodiments, said first binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a light chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 29, or an amino acid sequence wherein any one amino acid of said amino acid sequence, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said first binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a heavy chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 1 1 , SEQ ID NO: 12, SEQ ID NO:13, SEQ ID NO:24, and SEQ ID NO: 25, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 1 1 , SEQ ID NO: 12, SEQ ID NO:13, SEQ ID NO:24, and SEQ ID NO: 25, respectively.

In some embodiments, said first binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a light chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 1 1 , SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 24, and SEQ ID NO: 25, or an amino acid sequence wherein any one amino acid of said amino acid sequence, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said second binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 14. In some embodiments, said second binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14, or an amino acid sequence wherein any one amino acid of SEQ ID NO: 14, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said second binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 15, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 15.

In some embodiments, said second binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 15, or an amino acid sequence wherein any one amino acid of SEQ ID NO: 15, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said second binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a light chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 26, and SEQ ID NO: 30, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO:18, SEQ ID NO: 26, and SEQ ID NO: 30, respectively.

In some embodiments, said second binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a light chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 26, and SEQ ID NO: 30, or an amino acid sequence wherein any one amino acid of said amino acid sequence, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered. In some embodiments, said second binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a heavy chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21 , SEQ ID NO: 27, and SEQ ID NO: 28, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO:21 , SEQ ID NO: 27, and SEQ ID NO: 28, respectively.

In some embodiments, said second binding agent is an anti-IL1 RAP antibody or antigen- binding fragment thereof comprising a heavy chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21 , SEQ ID NO: 27, and SEQ ID NO: 28, or an amino acid sequence wherein any one amino acid of said amino acid sequence, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, the proportion of said first binding agent relative to said second binding agent in said composition is between 10:1 and 1 :10, such as between 5:1 and 1 :5, such as between 2:1 and 1 :2, such as between 3:2 and 2:3, such as between 11 :9 and 9:1 1 , such as 1 :1.

In some embodiments, said first and second binding agents are antibody molecules of isotype subtype lgG1 , lgG2, lgG3 or lgG4.

In some embodiments, said composition is capable of inducing internalisation of cell membrane-bound IL1 RAP. By internalisation, we mean that upon binding of the binding agents within the compositions of the invention to IL1 RAP located within the membrane of a cell, the binding agent-IL1 RAP complex is internalised within the cell.

In some embodiments, said first and/or second binding agent(s) lack the capacity to induce antibody dependent cell mediated cytotoxicity (ADCC). Bi-epitopic binding agents that bind to at least two different extracellular domains of IL1RAP

In one aspect, the present invention concerns a bi-epitopic binding agent comprising: a first antigen-binding region, and

a second antigen-binding region, wherein the first distinct antigen binding region and the second distinct antigen binding region bind to two different extracellular domains of human interleukin-receptor accessory protein (IL1 RAP).

A“bi-epitopic binding agent” as used herein refers to a molecule capable of binding to two different epitopes simultaneously. A bi-epitopic binding agent may be a bispecific agent. A “bispecific agent” as used herein refers to a molecule capable of binding to two different epitopes on two different molecules.

In some embodiments, said bi-epitopic binding agent is a dual-variable-domain antibody, a bi-epitopic Fab-fragment, a bi-epitopic scFv, a bivalent bispecific antibody (such as IgG- scFv bispecific antibodies), a monovalent bispecific antibody (such as a DuoBody®, Genmab AS, Copenhagen, Denmark), a‘knob-in-hole’ bispecific antibody (for example, an scFv-KIH, scFv-KIHr, a BiTE-KIH or a BiTE- KIHr; see Xu et al., 2015, mAbs 7(1 ):231 - 242), a scFv2-Fc bispecific antibody, a BiTE/scFv2 bispecific antibody, a DVD-lg bispecific antibody, an IgG-Fab bispecific antibody, a FAb-lgG bispecific antibody, a DART-based bispecific antibody (such as DART2-Fc, DART2-Fc or DART), a DNL-Fab3 bispecific antibody, or a scFv-HSA-scFv bispecific antibody.

In one embodiment, said bi-epitopic binding agent is a polypeptide, such as an antibody. In one embodiment, the present invention concerns said bi-epitopic binding agent wherein: a) the first antigen-binding region is selected from the group consisting of:

i) an antigen-binding region which binds to the same antigen as the reference antibody“CAN04”;

ii) an antigen-binding region which binds to the same epitope as the antibody CAN04; iii) an antigen-binding region which is capable of inhibiting the binding of the antibody CAN04 to human IL1 RAP; iv) an antigen-binding region which is capable of binding to domain 2 of IL1 RAP;

v) an amino acid sequence comprising or consisting of: the variable light chain (Vi_) amino acid sequence: DIQMTQSPSSLSASVGDRVTITCQASQGINNYLNWYQQKP GKAPKLLIHYTSGLHAGVPSRFSGSGSGTDYTLTISSLEPED VATYYCQQYSILPWTFGGGTKVEIKR (SEQ ID NO:1 ) and the variable heavy chain (V H ) amino acid sequence:

DVKLVESGGGLVKPGGSLKLSCAASGFTFSIYTMSWVRQT PEKRLEWVATISIGGSYINYPDSVKGRFTISRDNAKNTLYLQ MSSLKSEDTAIYYCSREVDGSYAMDYWGQGTSVTVSS

(SEQ ID NO:4) of the antibody CAN04,

vi) an amino acid sequence comprising or consisting of at least one of the following six complementary determining regions (CDRs):

Light chain CDR1 : SASQGINNYLN (SEQ ID NO: 8) or ASQGINNYLN (SEQ ID NO: 29)

Light chain CDR2: YTSGLHAGV (SEQ ID NO: 9) or

YTSGLHA (SEQ ID NO: 22)

Light chain CDR3: QQYSILPWT (SEQ ID NO: 10) or QYSILPWT (SEQ ID NO: 23)

Heavy chain CDR1 : GYAFTSSSWMN (SEQ ID NO: 1 1 ) or GYAFTSS (SEQ ID NO: 24)

Heavy chain CDR2: RIYPGDGNTHYAQKFQG (SEQ ID NO: 12) or YPGDGN (SEQ ID NO: 25)

Heavy chain CDR3: GYLDPMDY (SEQ ID NO: 13); vii) an antigen-binding fragment of an antibody of (i) to (vi) above;

b) the second antigen-binding region is selected from the group consisting of: i) an antigen-binding region which binds to the same antigen as the reference antibody“CAN03”;

ii) an antigen-binding region which binds to the same epitope as the antibody CAN03;

iii) an antigen-binding region which is capable of inhibiting the binding of the antibody CAN03 to human IL1 RAP;

iv) an antigen-binding region which is capable of binding to the extracellular domain 3 of IL1 RAP;

v) an amino acid sequence comprising or consisting of the variable light chain (Vi_) amino acid sequence:

DILLTQSPAILSVSPGERVSFSCRASQSIGTSIHWYQRRTNG SPRLLIKSASESISGIPSRFSGSGSGTDFTLSINSVESEDIAD YYCQQSNSWPTTFGAGTKLELKR (SEQ ID NO: 14), and the variable heavy chain (VH) amino acid sequence: DVKLVESGGGLVKPGGSLKLSCAASGFTFSIYTMSWVRQT PEKRLEWVATISIGGSYINYPDSVKGRFTISRDNAKNTLYLQ MSSLKSEDTAIYYCSREVDGSYAMDYWGQGTSVTVSS

(SEQ ID NO: 15) of the antibody CAN03,

vi) an antigen binding region comprising at least one of the following six complementary determining regions (CDRs):

Light chain CDR1 : RASQSIGTSIH

(SEQ ID NO: 16) or ASQSIGTSIH (SEQ ID NO: 30)

Light chain CDR2: SASESIS (SEQ ID NO: 17)

Light chain CDR3: QQSNSWPTT (SEQ ID NO: 18) or QSNSWPTT (SEQ ID NO: 26)

Heavy chain CDR1 : GFTFSIYTMS (SEQ ID NO: 19) or GFTFSIY (SEQ ID NO: 27)

Heavy chain CDR2: TISIGGSYINYPDSVKG (SEQ ID NO: 20) or SIGGSY (SEQ ID NO: 28)

Heavy chain CDR3: EVDGSYAMDY (SEQ ID NO: 21 ); and vii) an antigen-binding fragment of an antibody of (i) to (vi) above.

In one embodiment, said first antigen-binding region comprises an amino acid sequence comprising or consisting of the heavy chain amino acid sequence SEQ ID NO: 35 and the light chain amino acid sequence SEQ ID NO: 36 and wherein said second antigen-binding region comprises the heavy chain amino acid sequence SEQ ID NO: 37 and the light chain amino acid sequence SEQ ID NO: 38.

In one embodiment, said first antigen binding region comprises a variable heavy chain (VH), and a variable light chain (Vi_) selected from the group consisting of: a) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; b) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; c) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; d) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 1 ; e) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; f) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; g) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; h) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 2; i) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 4, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; j) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 5, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; k) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 6, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3; and

L) a variable heavy chain (VH) comprising the amino acid sequence SEQ ID NO: 7, and a variable light chain (Vi_) comprising the amino acid sequence SEQ ID NO: 3.

In one embodiment, said first antigen-binding region comprises a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NOs: 1 , 2, or 3; and a heavy chain variable domain (VH) comprising or consisting of the amino acid sequence of SEQ ID NOs: 4, 5, 6, or 7; and said second antigen binding-region comprises a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14; and a heavy chain variable domain (VH) comprising or consisting of the amino acids of SEQ ID NO: 15.

Preferably, said first antigen-binding region comprises a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 1 ; and a heavy chain variable domain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 4; and said second antigen-binding region comprises a light chain variable domain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14; and a heavy chain variable domain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 15.

In some embodiments, said first antigen-binding region has a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 1 , or an amino acid sequence having at least 60% sequence identity, such as at least 70% sequence identity, such as at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 1.

In some embodiments, said first antigen-binding region has a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 1 , or an amino acid sequence wherein any one amino acid of SEQ ID NO: 1 , has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said first antigen-binding region has a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 4, or an amino acid sequence having at least 60% sequence identity, such as at least 70% sequence identity, such as at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 4.

In some embodiments, said first antigen-binding region has a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 4, or an amino acid sequence wherein any one amino acid of SEQ ID NO: 4, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said first antigen-binding region has a light chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO:10, SEQ ID NO:22, SEQ ID NO: 23, and SEQ ID NO: 29, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO:10, SEQ ID NO:22, SEQ ID NO: 23, and SEQ ID NO: 29, respectively.

In some embodiments, said first antigen-binding region has a light chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 29, or an amino acid sequence wherein any one amino acid of said amino acid sequence, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said first anti-IL1 RAP antibody has a heavy chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 1 1 , SEQ ID NO: 12, SEQ ID NO:13, SEQ ID NO:24, and SEQ ID NO: 25, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 1 1 , SEQ ID NO: 12, SEQ ID NO:13, SEQ ID NO:24, and SEQ ID NO: 25, respectively.

In some embodiments, said first antigen-binding region has a light chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 1 1 , SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 24, and SEQ ID NO: 25, or an amino acid sequence wherein any one amino acid of said amino acid sequence, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said second antigen-binding region has a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 14.

In some embodiments, said second antigen-binding region has a variable light chain (Vi_) comprising or consisting of the amino acid sequence of SEQ ID NO: 14, or an amino acid sequence wherein any one amino acid of SEQ ID NO: 14, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said second antigen-binding region has a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 15, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 15.

In some embodiments, said second antigen-binding region has a variable heavy chain (VH) comprising or consisting of the amino acid sequence of SEQ ID NO: 15, or an amino acid sequence wherein any one amino acid of SEQ ID NO: 15, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said second antigen-binding region has a light chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 26, and SEQ ID NO: 30, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO:18, SEQ ID NO: 26, and SEQ ID NO: 30, respectively.

In some embodiments, said second antigen-binding region has a light chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 26, and SEQ ID NO: 30, or an amino acid sequence wherein any one amino acid of said amino acid sequence, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In some embodiments, said second antigen-binding region has a heavy chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21 , SEQ ID NO: 27, and SEQ ID NO: 28, or an amino acid sequence having at least 80% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, such as at least 97% sequence identity, such as at least 98% sequence identity, such as at least 99% sequence identity to SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO:21 , SEQ ID NO: 27, and SEQ ID NO: 28, respectively.

In some embodiments, said second antigen-binding region has a heavy chain CDR comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21 , SEQ ID NO: 27, and SEQ ID NO: 28, or an amino acid sequence wherein any one amino acid of said amino acid sequence, has been altered for another amino acid, with the proviso that no more than 3 amino acid residues have been thus altered.

In one embodiment, the said bi-epitopic binding agent has a binding affinity (Ko) for human IL1 RAP of 200 pM or greater.

Polynucleotides, vectors and host cells

In one aspect, the present invention concerns a composition comprising one or more polynucleotides, which, collectively or individually, encode either (i) first and second binding agents or (ii) a bi-epitopic binding agent as defined herein. In another aspect, the present invention concerns an isolated polynucleotide encoding either (i) first and second binding agents or (ii) a bi-epitopic binding agent as defined herein. In yet another aspect, the present invention concerns an expression vector comprising one or more polynucleotides which, collectively or individually, encode either (i) first and second binding agents or (ii) a bi-epitopic binding agent as defined herein. In yet another aspect, the current invention concerns a host cell comprising one or more polynucleotides which, collectively or individually, encode either (i) first and second binding agents or (ii) a bi- epitopic antibody as defined herein. In yet another aspect, the current invention concerns a host cell comprising one or more expression vectors as described herein.

Pharmaceutical compositions

Whilst it is possible for the binding agents or salts of the present invention to be administered as the raw compounds as described above, it is preferred to present them in the form of a pharmaceutical formulation. Accordingly, the present invention further provides a pharmaceutical formulation, which comprises the binding agents of the present invention or a pharmaceutically acceptable salt or ester thereof, as herein defined, and a pharmaceutically acceptable carrier therefor. The pharmaceutical formulations may be prepared by conventional techniques, e.g. as described in Remington: The Science and Practice of Pharmacy 2005, Lippincott, Williams & Wilkins.

As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Preferably, the carrier is suitable for parenteral, e.g. intravenous, intramuscular or subcutaneous administration (e.g., by injection or infusion). Depending on the route of administration, the binding agents may be coated in a material to protect the polypeptide from the action of acids and other natural conditions that may inactivate or denature the polypeptide.

Optional pharmaceutically acceptable carriers comprise aqueous carriers or diluents. Examples of suitable aqueous carriers that may be employed in the compositions of the invention include water, buffered water and saline. Examples of other carriers include ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.

A composition of the invention also may include a pharmaceutically acceptable anti- oxidant. These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of presence of microorganisms may be ensured both by sterilization procedures, supra, and by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminium monostearate and gelatin.

Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration. Sterile injectable solutions can be prepared by incorporating the active agent (e.g. antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by sterilization microfiltration. Generally, dispersions are prepared by incorporating the active agent into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying (lyophilization) that yield a powder of the active agent plus any additional desired ingredient from a previously sterile-filtered solution thereof.

In one embodiment, the compositions are formulated for systemic administration or for local administration. Local administration may be at the site of a tumour or into a tumour draining lymph node. The composition may optionally be formulated for sustained release over a period of time. Thus, the composition may be provided in or as part of a matrix facilitating sustained release. Exemplary sustained release matrices may comprise a montanide or y-polyglutamic acid (PGA) nanoparticles.

It will be appreciated by persons skilled in the art that the compositions of the invention may comprise additional active ingredients, as well as one or more agents of the invention.

Routes of administration and dosages

The pharmaceutical compositions according to the invention may be administered via any suitable route known to those skilled in the art. Thus, possible routes of administration include parenteral (intravenous, subcutaneous, and intramuscular), topical, ocular, nasal, pulmonar, buccal, oral, parenteral, vaginal and rectal. Also administration from implants is possible.

As detailed above, the pharmaceutical compositions of the invention may be formulated for systemic administration or for local administration. Local administration may be at the site of a tumour or into a tumour draining lymph node. In one embodiment, the composition is formulated for sustained release over a period of time (for example, over 1 hour or more, e.g. over two, three, four, five, six, twelve, or twenty-four hours or more).

The binding agents and compositions of the invention may be used in therapy or prophylaxis. In therapeutic applications, binding agents and compositions are administered to a subject already suffering from a disorder or condition, in an amount sufficient to cure, alleviate or partially arrest the condition or one or more of its symptoms. Such therapeutic treatment may result in a decrease in severity of disease symptoms, or an increase in frequency or duration of symptom-free periods. An amount adequate to accomplish this is defined as "therapeutically effective amount". In prophylactic applications, binding agents and compositions are administered to a subject not yet exhibiting symptoms of a disorder or condition, in an amount sufficient to prevent or delay the development of symptoms. Such an amount is defined as a“prophylactically effective amount”. The subject may have been identified as being at risk of developing the disease or condition by any suitable means.

A suitable dosage of a binding agent or composition of the invention may be determined by a skilled medical practitioner. Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular agent employed, the route of administration, the time of administration, the rate of excretion of the polypeptide, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.

Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.

The binding agents and compositions may be administered in a single dose or in multiple doses. The multiple doses may be administered via the same or different routes and to the same or different locations. Alternatively, a binding agent or composition can be administered as a sustained release formulation as described above, in which case less frequent administration is required. Dosage and frequency may vary depending on the half-life of the polypeptide in the patient and the duration of treatment that is desired. The dosage and frequency of administration can also vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage may be administered at relatively infrequent intervals over a long period of time. In therapeutic applications, a relatively high dosage may be administered, for example until the patient shows partial or complete amelioration of symptoms of disease. Combined administration of two or more agents may be achieved in a number of different ways. In one embodiment, the first binding agent and the other agent(s) may be administered together in a single composition. In another embodiment, the first binding agent and the other agent(s) may be administered in separate compositions as part of a combined therapy.

Methods for treatment, amelioration, prevention, diagnosis or prognosis

In one aspect, the current invention concerns a composition comprising at least two binding agents; or a bi-epitopic binding agent; or a polynucleotide; or a vector; or a host cell; or a pharmaceutical composition as described herein for use as a medicament.

In one aspect, the current invention concerns a composition comprising at least two binding agents; or a bi-epitopic binding agent; or a polynucleotide; or a vector; or a host cell; or a pharmaceutical composition as described herein for use as a diagnostic and/or prognostic agent.

In one aspect, the current invention concerns a composition comprising at least two binding agents; or a bi-epitopic binding agent; or a polynucleotide; or a vector; or a host cell; or a pharmaceutical composition as described herein for use in the treatment, amelioration, prevention, diagnosis or prognosis of an I L1 RAP-associated disease or disorder in a mammal. In some embodiments, said IL1 RAP-associated disease or disorder is selected from the group consisting of proliferative disorders, autoimmune disorders, and inflammatory disorders, such as auto inflammatory disorders.

In one embodiment, the current invention concerns a composition comprising at least two binding agents; or a bi-epitopic binding agent; or a polynucleotide; or a vector; or a host cell; or a pharmaceutical composition as described herein for use in the treatment, amelioration, prevention, diagnosis or prognosis of a disease or disorder selected from the group consisting of rheumatoid arthritis, osteoarthritis, multiple sclerosis, artherosclerosis, scleroderma (systemic sclerosis), lupus, systemic lupus erythematosus (SLE), (acute) glomerulonephritis, asthma, chronic obstructive pulmonary diseases (COPD), respiratory distress- syndrome (ARDS), inflammatory bowel disease, colitis, vasculitis, uveitis, dermatitis, atopic dermatitis, alopecia, rhinitis (allergica), allergic conjunctivitis, myasthenia gravis, sclerodermitis, sarcoidosis, psoriatic arthritis, psoriasis, ankylosingspondylitis, juvenile idiopathic arthritis, Graves’ disease, Sjogren’s syndrome, endometriosis, Crohns disease, Behget disease, celiac disease, diabetes mellitus type 1 , Graves’ disease, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis, systemic lupus erythematosus, familial Mediterranean fever (FMF), hyperimmunoglobulinemia D with recurrent fever (HIDS), TNF receptor-associated periodic syndrome (TRAPS), cryopyrin-associated periodic syndromes (CAPS, such as Muckle-Wells syndrome, familial cold urticaria, and neonatal onset), multisystem inflammatory disease (NOMID), periodic fever, aphthous stomatitis, pharyngitis and adenitis (PFAPA syndrome), Blau syndrome, pyogenic sterile arthritis, pyoderma gangrenosum, acne (PAPA), deficiency of the interleukin-1 -receptor antagonist (DIRA), adult-onset Still's disease and systemic- onset juvenile idiopathic arthritis.

In one aspect, the current invention concerns a first binding agent with specificity for IL1 RAP as defined in herein for use in the treatment, amelioration, prevention, diagnosis or prognosis of an IL1 RAP-associated disease or disorder in a mammal, wherein the first binding agent is for use in combination with one or more further binding agents with specificity for IL1 RAP, wherein the first and further binding agents bind to at least two different extracellular domains of IL1 RAP.

Inflammatory diseases and disorders

In one embodiment, said I L1 RAP-associated disease or disorder is an inflammatory disease or disorder.

In one embodiment, said inflammatory disease or disorder is selected from the group consisting of rheumatoid arthritis, osteoarthritis, multiple sclerosis, artherosclerosis, scleroderma (systemic sclerosis), lupus, systemic lupus erythematosus (SLE), (acute) glomerulonephritis, asthma, chronic obstructive pulmonary diseases (COPD), respiratory distress- syndrome (ARDS), inflammatory bowel disease, colitis, vasculitis, uveitis, dermatitis, atopic dermatitis, alopecia, rhinitis (allergica), allergic conjunctivitis, myasthenia gravis, sclerodermitis, sarcoidosis, psoriatic arthritis, psoriasis, ankylosingspondylitis, juvenile idiopathic arthritis, Graves disease, Sjogren’s syndrome, Endometriosis, Crohns disease and Behget disease.

Autoimmune diseases and disorders

In one embodiment, said I L1 RAP-associated disease or disorder is an autoimmune disease or disorder.

In one embodiment, said autoimmune disease is selected from the group consisting of celiac disease, diabetes mellitus type 1 , Graves disease, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis, and systemic lupus erythematosus.

Autoinflammatory diseases and disorders In one embodiment, said I L1 RAP-associated disease or disorder is an autoinflammatory disease or disorder. In one embodiment, said autoinflammatory disease or disorder is selected from the group consisting of Familial Mediterranean Fever (FMF), Hyperimmunoglobulinemia D with recurrent fever (HIDS), TNF receptor associated periodic syndrome (TRAPS), cryopyrin- associated periodic syndromes (CAPS, such as Muckle-Wells syndrome, familial cold urticaria, and neonatal onset), multisystem inflammatory disease (NOMID), Periodic fever, aphthous stomatitis, pharyngitis and adenitis (PFAPA syndrome), Blau syndrome, Pyogenic sterile arthritis, pyoderma gangrenosum, acne (PAPA), Deficiency of the interleukin-1-receptor antagonist (DIRA), adult-onset Still's disease and systemic-onset juvenile idiopathic arthritis. Neoplastic disorders

In one embodiment, said I L1 RAP-associated disease or disorder is a neoplastic disorder in a mammal. In one embodiment, said neoplastic disorder is selected from the group consisting of prostate cancer, breast cancer, lung cancer, colorectal cancer, melanomas, bladder cancer, brain/CNS cancer, cervical cancer, oesophageal cancer, gastric cancer, head/neck cancer, kidney cancer, liver cancer, lymphomas, ovarian cancer, pancreatic cancer, sarcomas, chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML).

Examples

A. Binding affinity of exemplary antibodies for IL1RAP protein (i) Biacore study - anti-IL1RAP antibodies of murine origin

Materials & Methods

Goat anti-mouse IgG was immobilized on a CM5 chip according to the technical manual of capture kit and standard operation principle of BIAcore T200 (Biacore Life Sciences, GE Healthcare Europe GmbH, Uppsala, Sweden).

The binding analysis cycle consisted of three steps: (i) capture of the ligand on the chip surface by immobilized anti-mouse antibody; (ii) binding of the analyte to the captured ligand; and (iii) dissociation of bound analyte.

The capture molecule surface was regenerated after each binding cycle using the manufacturer’s recommended conditions.

All binding cycles were run at 25°C.

After five cycles of start-up, each antibody (100 nM) was injected at a flow rate of 30 mI/min, for 120 s, at the start of the cycle; then the analyte (100 nM) was injected at a flow rate of 30 mI/min, for 120 s, followed by monitoring the dissociation phase for 300 s. The exemplary antibodies of the compositions of the invention (CAN03 and CAN04) were tested along with one comparator anti-l L1 RAP antibody (CAN01 ).

Results & Conclusions

Results are shown in Table 2 below:

Table 2. Measurement of K on , K 0ff and KD.

Antibody ka (1/M-s) kd (1/s) KD (M)

CAN 01 2.34E+05 3.35E-04 1 .43E-09 CAN03 2.26E+05 7.25E-05 3.21 E-10

CAN04 4.27E+05 4.72E-05 1 .10E-10

The exemplary antibodies of the invention, CAN03 and CAN04, exhibited a high affinity for human IL1 RAP.

(ii) ELISA study - anti-IL1 RAP antibodies of murine origin

Materials & Methods

An indirect ELISA assay was performed. All samples were analysed in duplicate. Nunc- MaxiSorp 96 Micro Well™ Plates were coated with 100 ng of recombinant hILI RAP 21 - 367 (100 mI/well) diluted in 0.01 M PBS, pH 7.4, and incubated overnight at 4°C. Plates were washed with ELISA washing buffer (0.01 M PBS, 0.05% Tween 20, pH 7.4) followed by a blocking step using 150 mI/well of ELISA blocking solution (PBS, 0.5% BSA, 0.05% Tween 20, pH 7.4). After 1 h incubation at room temperature (RT) on agitation the plates were washed again using ELISA washing buffer. Samples were diluted in three-fold serial dilution (ranging from 1000 ng/ml to 0.5 ng/ml) in ELISA blocking solution and then transferred to the ELISA plate, 100 mI/well. Plates were incubated at RT for 1 h on agitation and then washed with ELISA washing solution. 100 mI/well of rabbit anti-mouse IgG conjugated to Alkaline Phosphatase (DAKO, 1 : 1000) was added and incubated 1 hour at RT on agitation. The plates were washed followed by addition of substrate (4-Nitrophenyl phosphatise disodium salt hexahydrate, SIGMA, 1 mg/ml), 100 mI/well. The plates were thereafter incubated at RT on agitation and absorbance at 405 nm measured consecutively for 30 min. Absorbance at 0 min was taken as background signal. Results & Conclusions

Results are shown in Figure 1

The exemplary antibodies, CAN03 and CAN04, were found to possess the highest binding signal for human IL1 RAP.

B. Binding of exemplary antibodies to IL1 RAP-expressing cells (i) Flow cytometry study - anti-IL 1 RAP antibodies of murine origin

Materials & Methods

Chronic myeloid leukaemia (CML) cell line KU812 cells were stained with antibodies raised against IL1 RAP or a relevant isotype control. For detection, a secondary anti-mlg- APC was used.

The two exemplary antibodies (CAN03 and CAN04) was tested along with six comparator anti-l L1 RAP antibodies (CAN01 , CAN 02, CAN05, CAN07, CAN08 and CAN09). An isotype negative control antibody was also included.

Results & Conclusions

Staining of I L1 RAP-expressing KU812 leukaemia cells reveals a higher mean

fluorescence intensity (MFI) for CAN03 and CAN04 compared to the isotype control and other comparator antibodies targeting IL1 RAP (Figure 2).

C. Epitope / domain mapping of exemplary anti-IL1RAP antibodies

Materials & Methods

In order to understand where the different antibody clones bind on IL1 RAP, a structural analysis of the protein was performed revealing that the extracellular part of the receptor could be divided into three distinct domains hereafter referred to as domains 1 , 2 and 3 (D1 , D2, D3) (see Wang et al., 2010, Nature Immunology, 11 :905-912, the disclosures of which are incorporated herein by reference). In order to determine the domain-binding pattern for the different antibody clones, a series of receptor constructs were generated and binding to these tested in an ELISA assay.

An indirect ELISA assay was performed. All samples were analysed in duplicate. Nunc- MaxiSorp 96 Micro Well™ Plates were coated with 100 ng of Rec hILI RAP Domain123 (aa21-367) (positive control), Rec hILI RAP Domain12 (aa21-234), Domainl (aa21-134) or Rec hILI RAP Domain3 (aa235-367) (100 mI/well) diluted in 0.01 M PBS, pH 7.4, and incubated overnight at 4°C. Plates were washed with ELISA washing buffer (0.01 M PBS, 0.05% Tween 20, pH 7.4) followed by a blocking step using 150 mI/well of ELISA blocking solution (PBS, 0.5% BSA, 0.05% Tween 20, pH 7.4). After 1 h incubation at room temperature (RT) on agitation the plates were washed again using ELISA washing buffer. CAN01, CAN03, CAN04, CAN05, CAN07, CAN08 and KMT-1 (positive control) were diluted in three-fold serial dilution (ranging from 1000 ng/ml to 0.5 ng/ml) in ELISA blocking solution and then transferred to the ELISA plate, 100 mI/well. Plates were incubated at RT for 1 h on agitation and then washed with ELISA washing solution. 100 mI/well of rabbit anti-mouse IgG conjugated to Alkaline Phosphatase (DAKO, 1 :1000) was added and incubated 1 hour at RT on agitation. The plates were washed followed by addition of substrate (4-Nitrophenyl phosphatise disodium salt hexahydrate, SIGMA, 1 mg/ml), 100 mI/well. The plates were thereafter incubated at RT on agitation and absorbance at 405 nm measured consecutively for 30 min. Absorbance at 0 min was taken as background signal.

The exemplary antibodies of the compositions of the invention (CAN03 and CAN04) were tested along with eight comparator anti-IL1 RAP monoclonal antibodies (CAN01 , CAN02, CAN05, CAN07, CAN08, CAN 10, and CAN1 1 , together with a polyclonal anti-IL1 RAP antibody (KMT-1 ) as a positive control.

Results & Conclusions

The majority of anti-IL1 RAP antibodies tested for target validation bind to domain 3 (D3) while the exemplary CAN04 antibody of the invention is distinct in that it binds to domain 2 (D2). Hence, the exemplary antibodies (CAN03 and CAN04) bind to different domains (D3 and D2 respectively) of IL1 RAP. The entire domain mapping data can be found summarized in the Table 4 below. Table 4. Epitope mapping of exemplary anti-IL1 RAP antibody clones.

Domainl23 Domainl2 Domainl Domain3 Suggested

Clone

(aa21-367) (aa 21-234) (aa21-134) (aa235-367) epitope

CAN03 + + D3

CAN 05 + + + D1 CAN07 + + D3 CAN 08 + + D3 CAN04 + + D2 CAN01 + + D3 CAN02 + nd*

KMT-1 + + + + polyclonal nd* = not determined as epitope mapping data could not clearly identify specific domain for these constructs, which may be attributed to binding to a structural epitope containing sequence elements from more than one domain, e.g. D2-D3 junction.

D. Specificity / cross-reactivity of exemplary anti-IL 1 RAP antibodies Materials & Methods

An important feature of good lead candidate antibodies is that they cross-react with equal or near-equal potency to the homologous protein in a relevant toxicology species. According to the general regulatory guidelines, binding to one rodent and one non-rodent would be the preferred scenario, but for antibodies this is rarely the case, and instead many labs struggle to identify any relevant toxicology species except for primates.

For the present study, cross reactivity to non-human primates like Macaca mulatta (rhesus) or Macaca fascicularis (cynomolgus) was expected since the IL1 RAP protein in these species share 99% homology to the human IL1 RAP protein.

A number of potential lead antibodies were selected and tested for binding to recombinant M. fascicularis IL1 RAP (aa21 -367) in an ELISA assay. The exemplary antibodies (CAN03 and CAN04) were tested along with seven comparator anti-l L1 RAP monoclonal antibodies (CAN01 , CAN 02, CAN07, CAN08, CAN09, Mab676 from R&D, and a polyclonal anti-IL1 RAP antibody (KMT-1 ).

Results & Conclusions

Surprisingly, several of the comparator anti-IL1 RAP antibodies tested were found not to cross-react with cynomolgus IL1 RAP, amongst them the commercial reference antibody mAb676 from R&D, Table 5. CAN03 and CAN04 do however both react with the cynomolgus protein.

Table 5. Binding to cynomolgus IL1 RAP. Values in bold denotes clones identified to cross-react with IL1 RAP from M. fascicularis.

Clone Binding to rec. M. fascicularis

IL1RAP (OD405)

CAN 01 0.324

CAN02 0.014

CAN09 0.022

CAN 03 0.870

CAN 04 0.416

CAN07 0.1 1 1

CAN08 0.375

mAb676 (R&D) 0.037

KMT-1 0.481

E. Inhibition of IL-1 \i, IL-33, IL-36a signalling by exemplary anti-IL1RAP antibodies

(i) Effect of different antibodies and antibody combinations on IL-1 and IL-33 signalling in the HEK-Blue IL-33/IL-1/3 cell line

Materials & Methods

As IL1 RAP is a functional part of both the IL-1 and the IL-33 receptor complexes, antibodies binding to IL1 RAP may also inhibit IL-1 and IL-33 signalling.

In order to test for the capability of potential lead candidate antibodies to block IL-1 and IL- 33 signalling, an IL-1 and IL-33 dependent reporter gene assay was set up. HEK-Blue IL- 33/I L-1 b cells (InvivoGen) respond to IL-1 or IL-33 signalling by the release of alkaline phosphatase that can be quantified by a colorimetric assay. To test the inhibitory capacity of the lead candidates HEK-Blue cells were plated at 50 000 cells/well and incubated with the test antibodies 45 minutes priorto stimulation with IL-1 b or IL-33 in a final concentration in assay of 0.3 ng/ml for each ligand. Final assay concentrations of antibodies were 200nM - 0.01 nM. In the control wells, the antibodies were replaced by PBS. The cells were incubated at 37°C o/n before measuring the amount of alkaline phosphatase released.

The exemplary antibodies of the invention (CAN03 and CAN04) were tested alone and in 1 :1 combination for inhibition of I L-1 b and IL-33 signalling.

Results & Conclusions

As depicted in Figure 3A, the exemplary antibody CAN04 induced a pronounced inhibition of IL-1 b signalling. Exemplary antibody CAN03 also inhibited signalling but with less potency than CAN04. When CAN04 and CAN03 were combined, an inhibition similar to that of CAN04 alone was detected.

In contrast to the inhibition of IL-1 b signalling, CAN03 and CAN04 when administered alone did not completely inhibit IL-33 signalling. Both antibodies blocked signalling with comparable potency but could inhibit only -70% (CAN03) or -50% (CAN04) of the signal (Figure 3B). Surprisingly however, the combination of CAN03 and CAN04 (1 :1 ) inhibited the signal to 100% and was more potent compared to each antibody alone. Thus, two antibodies binding different domains of IL1 RAP (CAN03 to D3, CAN04 to D2) act synergistically to inhibit IL1 RAP signalling.

(ii) Effect of different antibodies and antibody combinations on IL-36 signalling in a IL- 1 Rrp2-transfected HEK-Blue IL-33/IL-1/] cell line

Materials & Methods

As IL1 RAP also is a functional part of both the IL-36 receptor complex, antibodies binding to IL1 RAP may also inhibit IL-36 signalling.

The HEK-Blue I L-33/I L-1 b system (InvivoGen) does not respond to IL-36 since it lacks the IL-36 receptor (IL1 Rrp2). In order to test for the capability of potential lead candidate antibodies to block IL-36 signalling, the HEK-Blue IL-33/IL-1 b system was modified by transfection with IL1 Rrp2 (the IL36-receptor) which allowed IL-36-induced reporter gene expression. HEK-Blue cells were transiently transfected with an IL1 Rrp2-expressing vector using Lipofectamine LTX (ThermoFisher), cultured for 24h, plated at 50 000 cells/well and incubated with the test antibodies 45 minutes prior to stimulation with IL-36a at different concentrations. Assay concentrations of antibodies were 0.1 , 1 and 10 pg/ml. In the control wells, the antibodies were replaced by PBS. The cells were incubated at 37°C o/n before measuring the amount of alkaline phosphatase released.

The exemplary antibodies (CAN03 and CAN04) were tested alone and in 1 :1 combination for inhibition of signalling.

Results & Conclusions

As depicted in Figure 4A and 4B, the exemplary antibody CAN04 on its own induced a moderate inhibition of IL-36a signalling at 1 ng/ml IL-36a and very little inhibition at 10 ng/ml IL-36a. The exemplary antibody CAN03 on its own did not inhibit signalling at those concentrations of IL-36a. However, the combination of CAN03 and CAN04 (1 :1 ) was more potent compared to each antibody alone and resulted in a synergistic inhibition of the IL- 36a signal. Thus, also for IL-36 signalling, two antibodies binding different domains of IL1 RAP act synergistically to inhibit IL1 RAP signalling.

(iii) Effect in cell lines

Materials & Methods

In order to test the ability to inhibit IL1 RAP-dependent signalling in a cancer cell model, an IL-1 dependent cellular assay was set up. The IL1 RAP-expressing breast cancer cell line Hs578T responds to IL-1 b stimulation with IL-6 production and was used to test the inhibitory effect of the exemplary antibodies and an isotype control. Cells were plated at 20 000 cells/well in 12-well plates and incubated with 0.1 ng/ml IL1 b for 24 hours. IL-6 in the supernatant was measured by ELISA. Cells were incubated with or without the indicated antibodies and stimulated with IL-1 b. The final assay concentration of antibodies was 10 pg/ml (10 pg/ml of each antibody when tested alone, 5 pg/ml of each when tested in 1 :1 combination).

Results & Conclusions

As depicted in Figure 5, stimulation with I L-1 b in the presence of CAN04 or CAN03 at 10 pg/ml led to a 50% (CAN04) or 30% (CAN03) inhibition of IL-6 production after 24h, while the isotype control antibody did not have any effect. As in the HEK-Blue IL-33/I L-1 b system in example D, the combination of CAN04 and CAN03 (5 pg/ml CAN04 + 5 pg/ml CAN03) led to a synergistic increase in effect and 90% of the signal could be inhibited. Thus, CAN03 and CAN04 act synergistically when combined to reduce a natural response to IL- 1 b stimulation in a breast cancer cell line.

F. Screening of antibodies to domain 1, 2 and 3 of IL1 RAP

(i) Screening for binding to IL1RAP

Antibodies generated towards IL1 RAP are screened as in Example A (ii) above for binding to IL1 RAP and subsequently as in Example B (i) for binding to cells expressing high levels of cell surface IL1 RAP (such as KU812 CML cells or SK-MEL-5 melanoma cells).

(ii) Analysis of binding to different domains

Antibodies that react with IL1 RAP are tested for interaction with D1 , D2 and D3 as described as in Example C.

(Hi) Analysis of competitive binding by ELISA

To verify binding to similar regions of IL1 RAP as CAN03 (D3) or CAN04 (D4), competitive ELISAs may be performed as described below.

Protocol

• All samples are analysed in duplicate.

• Coat a Nunc-MaxiSorp 96 Micro Well™ Plate with 100 ul/well of recombinant hlL1 RAP 21-367 (1 ug/ml) diluted in 0.01 M PBS, pH 7.4.

• Incubate the plate overnight at 4°C.

• Wash the plate with ELISA washing buffer

(0.01 M PBS, 0.05% Tween 20, pH 7.4).

• Add 150 mI/well of ELISA blocking solution

(PBS, 0.5% BSA, 0.05% Tween 20, pH 7.4).

• Incubate the plate for 1 h at room temperature (RT) under agitation.

• Wash the plate with ELISA washing buffer.

• Add samples of test items (e.g. mAb 1 , mAb 2) to wells (100 ul/well, 10 ug/ml) • Incubate the plate for 1 h at RT.

• Wash the plate with ELISA washing solution.

• Add a solution of reference antibodies CAN03 or CAN04 (100 ul/well, 1 ug/ml) to all wells.

• Incubate the plate for 1 h at RT.

• Wash the plate with ELISA washing buffer.

• Add 100 mI/well of a suitable secondary antibody conjugated to Alkaline rabbit anti-mouse IgG conjugated to Alkaline Phosphatse (If the test items are human antibodies, a suitable secondary antibody would be Goat Anti-Mouse IgG (Fc specificj-Alkaline Phosphatase antibody, SIGMA, A1418)

• Incubate the plate for 1 h at RT under agitation.

• Wash the plate with washing buffer.

• Add 100 mI of pNPP substrate per well.

(4-Nitrophenyl phosphatise disodium salt hexahydrate, SIGMA, 1 mg/ml).

• Incubate the plate at RT under agitation and measure absorbance at 405 nm consecutively for 30 min. Absorbance at 0 min should be taken as background signal.

G. Affinity maturation of antibodies

Antibodies generated towards IL1 RAP and selected for binding to IL1 RAP and D2 or D3 respectively may be“affinity-matured” for increased affinity.

Affinity-matured antibodies are produced by procedures known in the art. Many of these methods are based on the general strategy of generating panels or libraries of variant proteins by mutagenesis followed by selection and/or screening for improved affinity. Mutagenesis is often performed at the DNA level, for example by error prone PCR (Thie, Voedisch et al. 2009, Methods Mol Biol 525: 309-322), by gene shuffling (Kolkman and Stemmer 2001 , Nat Biotechnol. May; 19(5):423-8), by use of mutagenic chemicals or irradiation, by use of ' mutator ' strains with error prone replication machinery (Greener 1996, In Vitro Mutagenesis Protocols. Humana press, NJ) or by somatic hypermutation approaches that harness natural affinity maturation machinery (Peled, Kuang et al. 2008, Annu Rev Immunol. 26:481-51 1 ). Mutagenesis can also be performed at the RNA level, for example by use of Q13 replicase (Kopsidas, Roberts et al. 2006, Immunol Lett. 2006 Nov. 15; 107(2):163-8). Random mutagenesis of HVR and/or framework residues is described by: Barbas et al., Proc. Nat. Acad. Sci. USA 91 :3809-13 (1994); Schier et al. Gene 169:147-55 (1995); Yelton et al. J. Immunol. 1 55:1 994-2004 (1995); Jackson et al., J. Immunol. 154(7):331 0-19 (1995); and Hawkins et al. J. Mol. Biol. 226:889-96 (1992); Johnson & Hawkins, Affinity Maturation of Antibodies Using Phage Display, Oxford University Press 1996.

Library-based methods allowing screening for improved variant proteins can be based on various display technologies such as phage, yeast, ribosome, bacterial or mammalian cells, and are well known in the art (Benhar 2007, Expert Opin Biol Ther. May; 7(5): 763- 79). Affinity maturation can be achieved by more directed/predictive methods for example by site-directed mutagenesis or gene synthesis guided by findings from 3D protein modeling (see for example Queen, Schneider et al. 1989, PNAS, 86(24): 10029-33 or U.S. Pat. NO: 6,180,370 or U.S. Pat. NO: 5,225,539).

Marks et al. Bio/Technology 10:779-83 (1992) describes affinity maturation by VH and Vi_ domain shuffling.

H. Production of bi-epitopic antibodies for binding to different domains of ILIRAP.

Materials & Methods

The bispecific antibody 2C9x3F8 has been produced by controlled Fab-arm exchange (cFAE), described by Labrijn et al (Proc Natl Acad Sci U S A. 2013 Mar 26; 1 10(13): 5145- SI 50). The process involves the expression of two separate parental antibodies, each containing a single point mutation, F405L and K409R in the respective CH3 domains (EU- numbering convention as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991 )). The parental antibodies were mixed and subjected to controlled reducing conditions in vitro that separated the antibodies into Heavy chain/Light chain half- molecules (Fab-arms). After re-oxidation, the Fab-arms will reassembly. AS explained above, the point mutations in the antibodies CH3 domains will affect the reassembly process in a way that highly pure bsAbs will be formed.

For bsab2C9x3F8, the parental antibodies were ch2C9 (representing CAN03) and hu3F8 (representing CAN04). The ch2C9 is a chimeric monoclonal antibody with mouse VL and VH and human constant parts (lgG1/kappa). hu3F8 is a humanized lgG1/kappa monoclonal antibody. p2C9H4hG1 is an expression vector containing all the genetic elements required to express ch2C9 heavy chain in mammalian cells. The point mutation F405L was introduced to p2C9H4hG1 and the new expression vector was named p2C9- HC-hlgG1 -F405L p3F8-VH-v2 is an expression vector containing all the genetic elements required to express hu3F8 heavy chain in mammalian cells. The point mutation K409R was introduced to p3F8-VH-v2 and the new expression vector was named p3F8-HC- hlgG1 -K409R. p2C9K9hK is an expression vector containing all the genetic elements required to express ch2C9 light chain in mammalian cells and p3F8-VL-v2 is an expression vector containing all the genetic elements required to express hu3F8 light chain in mammalian cells

Mutagenesis: To introduce the F405L or K409R mutations, p2C9H4hG1 and p3F8-VH-v2 were double digested with Bst Ell and Nsi I and synthetic gene strings (Geneart) containing the point mutations were cloned to the linearized vectors by In-Fusion cloning methodology (Clontech).

Antibody production: ch2C9-F405L and hu3F8-K409R monoclonal antibodies were produced by co-transfecting relevant heavy and light chain expression vectors in Freestyle 293F cells (Life Techologies), according to the manufacturer’s instructions. The following vectors were used: ch2C9-F405L; HC vector p2C9-HC-hlgG1 -F405L and LC vector p2C9K9hK; hu3F8-K409R; HC vector p3F8-HC-hlgG1 -K409R and LC vector p3F8-VL-v2.

Purification: Antibodies were purified by protein A affinity chromatography. Cell culture supernatants were applied to MabSelect SuRe columns (GE Healthcare), washed with PBS and eluted with 0.1 M Glycine, pH 3.0. The eluate was neutralized directly during fraction collection by 1 .0 M Tris-HCI, pH 9.0. Immediately after purification, the buffer was exchanged to PBS, pH 7.4, by gel filtration with PD-10 columns (GE Healthcare). Purity was determined by SDS-PAGE and concentration was measured by absorbance at 280 nm. Purified antibodies were stored at 4°C.

Controlled Fab-arm exchange: The bispecific antibodies were generated by a chemical reaction where equimolar amounts of ch2C9-F405L and hu3F8-K409R were mixed to a final concentration of 0.5 mg/mL each, in presence of 75 mM 2-mercaptoethylamin-HCL (2-MAE, Sigma) and incubated at 31 °C for 5 hours. After removal of 2-MAE by ultrafiltration (Viva-spin 6,10 kDa, Satorius) the samples were stored over night at 4°C to allow reoxidation of the disulfide bonds. Results & Conclusions

The present example demonstrates that pure antibodies directed to two different domains of IL1 RAP can be produced.

I. Humanization of monospecific and bi-epitopic antibodies

It is not always desirable to use non-human antibodies for human therapy, thus the antibody according to the invention may be a human antibody or a humanized antibody.

Humanization is performed, e.g., by following the method of Winter and co-workers (Jones et al., Nature, 321 :522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. (See also U.S. Pat. NO: 5,225,539). In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies also comprise, e.g., residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody includes substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also includes at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al., 1986; Riechmann et al., 1988; and Presta, Curr. Op. Struct. Biol., 2:593-596 (1992)).

J. In vitro and in vivo activity of bispecific or combinations of monospecific antibodies

Monospecific antibodies, combinations of monospecific antibodies or bispecific antibodies may be tested in the HEK-system described in Example E for inhibition of IL-1 , IL-33 or IL- 36 signalling. The monospecific antibodies, combinations of monospecific antibodies or bispecific antibodies can also be tested for inhibition of IL-1 , IL-33 or IL-36 signalling in cell lines (as for example in Example E) or primary cells that have the appropriate receptors. Readout in the latter systems can be activation of intracellular signalling pathways (including NFKB) or downstream cytokines or other induced proteins. The monospecific antibodies, combinations of monospecific antibodies or bispecific antibodies can also be tested for inhibition of LPS-induced inflammation of human cells ex vivo. LPS is administered to 2x10 5 human PBMCs in the absence or presence of the monospecific antibodies, combinations of monospecific antibodies or bispecific antibodies and the supernatant is analysed for IL-6 and other cytokines (such as IL-1 b, TNFa and IL- 8) after incubation at 37°C for 6, 24 and 48 hours. Blocking of IL-1 signalling in this system reduces inflammatory signalling and secretion of cytokines such as IL-6.

The monospecific antibodies, combinations of monospecific antibodies or bispecific antibodies can also be tested in IL-1 , IL-33 and/or IL-36-dependent disease models in vivo. One such example is Xenotransplant models of Psoriasis, conducted e.g. as described in WO 2013/074569; where human skin biopsies from psoriasis patients are transplanted onto immunodeficient mice (e.g. SCID mice) and allowed to engraft for at least four weeks. The transplanted grafts show histological features that are similar to psoriasis, including features such as epidermal thickening that can be used as a measure for treatment effects. In order to increase inflammation and the pathological features of psoriasis, autologous T cells derived from the donor’s psoriatic plaques can be administered intra-dermally or intravenously into the grafted mice. An alternative to injection of T cells is intradermal injection of autologous PBMCs that are stimulated with Staphylococcus enterotoxins (SEB and/or SEC) and IL-2. A few weeks after injection of cells, the grafts are analysed by histology, the epidermal thickness is measured and treatment effects are calculated. Other psoriasis models applicable to the present invention are described in Gudjonsson et al (2007) J Invest Der, 127:1292-1308 and Kundu-Raychaudhuri et al (2014) Indian J Dermatology, Venereology, and Leprology 80(3): 204-213.

Materials

The following study was conducted to determine whether the monoepitopic antibody CAN04 and the bi-epitopic antibody bsACAN03xCAN04 were able to inhibit I L-1 b and IL- 33 signalling.

Preparation of antibodies

All antibodies were diluted in PBS for testing. Working solutions for reporter gene assay, HEK-Blue I L-33/I L-1 b were made in PBS at 20 times at the final assay concentration. The final assay volume was 200 pi (containing 10 mI antibody or diluent (PBS) + 180 mI cells + 10 mI ligand). Final assay concentrations of antibodies were as follows; 10 to 0.0001 nM (by serial dilutions in 3-fold dilutions steps). Preparation of human ligands (IL-Ib and IL-33)

IL-1 b and IL-33 were diluted in PBS to a stock concentration of 10 pg/mL. Stock concentrations of 10 pg/mL of the ligands were stored in aliquots at -80°C until use. For the reporter gene assay, the ligands were used at a final concentration of 100 ng/mL for hi L-1 b and 2 ng/mL for h IL-33; these concentrations gave 70-80% of the maximal effect in the assay as determined in a pre-test experiment.

Dilution of ligands to final assay concentration

(a) hi L-1 b, 100ng/mL

• Stock of ligands 10pg/rnL was diluted 1 :5 (200 pi + 800 mI Selective medium, see below) = 2000ng/mL

• 2000ng/mL was diluted 1 :20 in the assay; 10 pi of LPS 2000ng/mL +

190 mI cells and compound/well to yield a final concentration of

100ng/mL

(b) hlL-33, 2ng/mL

• Stock of ligands 10pg/mL was diluted 1 :250 (10 pi + 2490 pi Selective medium, see below) = 40ng/mL

• 40ng/mL was diluted 1 :20 in the assay; 10 m I of LPS 200ng/mL + 190 pi cells and compound/well to yield a final concentration of 2ng/mL

Cell line

HEK-Blue I L-33/1 L-1 b cells, generated by stable transfection of HEK-Blue™ I L-1 b cells with the IL1 RL1 , were used as I L-33/I L-1 b sensor cells (Cat no. hkb-IL-33, InvivoGen, San Diego, US).

Methods

Study design

Stock solutions of the antibodies were prepared in PBS. Working solutions for reporter gene assay, HEK-Blue IL-33/I L-1 b were made in PBS at 20 times at the final assay concentration. The final assay volume was 200 pi (containing 10 mI antibody or diluent (PBS) + 180 mI cells + 10 mI ligand). Final assay concentrations of antibodies were as follows; 10 to 0.0001 nM (by serial dilutions in 3-fold dilutions steps. In control wells (stimulated/unstimulated cells) antibodies were replaced by 10 m I PBS. Culturing and stimulation of HEK-Blue Iί-33/Iί-1b cells

As IL1 RAP is a functional part of the IL-1 receptor complex, antibodies binding to IL1 RAP have the potential to inhibit IL-1 signalling. Since tumour cells have been reported to use IL1 RAP dependent ligands such as IL-1 b and IL-33 as a growth factor, blocking this signal may provide an important mechanism for mediating anti-tumour effects (either separately or combined with an ADCC effect). In order to test for the capability of antibodies to block IL-1 signalling, an IL-1 dependent reporter gene assay was set up. HEK-Blue I L-33/I L-1 b cells (InvivoGen) respond to IL-1 signalling by the release of alkaline phosphatase that can be quantified by a colorimetric assay. To test the inhibitory capacity of the lead candidates HEK-Blue cells were plated at 50 000 cells/well and incubated with the test antibodies 45 minutes prior to stimulation with human IL-1 a, IL-1 b, and IL-33 in a final concentration to give optimal stimulation. Final assay concentrations of antibodies were 10 nM - 0.0001 nM. In control wells, antibodies were replaced by PBS. The cells were incubated at 37°C o/n before measuring the amount of alkaline phosphatase released by QUANTI-Blue - Medium for detection and quantification of alkaline phosphatase.

The HEK-Blue IL-33/I L-1 b cells were thawed and cultured in DMEM, 10% FCS (HI) and PEST and Normocin for two passages. After two passages the cells were cultured with selection antibiotics (Zeocin, HygroGold and Blasticidin) added to the medium above for at least one passage before the experiments, as well as during the experiments. HygroGold is required to maintain the IL-1 b specificity to the cell line and Blasticidin and Zeocin are required to maintain the plasmids encoding IL1 RL1 and SEAP respectively. The experiments were run on cells of 70% confluency. The cells were split 2-3 times/week, or when they had reached 80-90% confluence.

The ligands were titrated in a dose range from 300 ng/ml to 0.01 ng/ml. To generate a good assay for testing the antibodies ability to affect the IL-1 signalling (stimulate or inhibit the amount of alkaline phosphatase release) a concentration that resulted in a robust signal on the linear slope of the dose-response curve is preferred. For this system a concentration of 100 ng/mL for hi L-1 b and 2 ng/mL for hlL-33 were selected.

Evaluation of results

Raw data was converted to % inhibition using equation 1 :

% inhibition = (1 -(A-B)/(C-B)) x 100

wherein: A = Ligand activity with compound dissolved in PBS added

B = Negative control, No Ligand, only PBS (vehicle)

C = Positive control, Ligand with PBS (vehicle), IC50 represents the concentration yielding 50% inhibition of the maximal response.

EC50 represents the concentration yielding an inhibition representing 50% inhibition with respect to calculated values for the top and bottom of the curve. Conclusions & Results

The present example demonstrates that the monoepitopic antibody CAN04, as well the biepitopic bsACAN03xCAN04 block IL-1 b signalling completely. CAN04 also inhibits IL-33 signalling but with partial efficacy. The bi-epitopic bsACAN03xCAN04 antibody however allows for complete inhibition of IL-33 signalling as illustrated in Figure 6.

Overview of sequences

SEQ ID NO: 1 : CAN04 variant 6 variable light chain (VL)

DIQMTQSPSSLSASVGDRVTITCQASQGINNYLNWYQQKPGKAPKLLIHYTSGLHAGVP

SRFSGSGSGTDYTLTISSLEPEDVATYYCQQYSILPWTFGGGTKVEIKR

SEQ ID NO: 2: CAN 04 variable light chain (VL) VL1

DIQMTQSPSSLSASVGDRVTITCSASQGINNYLNWYQQKPGKAPKLLIHYTSGLHAGVP

SRFSGSGSGTDYTLTISSLQPEDVATYYCQQYSILPWTFGGGTKVEIKR

SEQ ID NO: 3: CAN 04 variable light chain (VL) VL3

DIQMTQSPSSLSASVGDRVTITCQASQGINNYLNWYQQKPGKAPKLLIHYTSGLHAGVP

SRFSGSGSGTDFTLTISSLEPEDVATYYCQQYSILPWTFGGGTKVEIKR

SEQ ID NO: 4: CAN04 variant 6 variable heavy chain (VH)

QVQLVQSGAEVKKPGSSVKVSCKASGYAFTSSWMNWVRQAPGQGLEWMGRIYPGD

GNTHYAQKFQGRVTLTADKSTSTAYMELSSLRSEDTAVYYCGEGYLDPMDYWGQGTL

VTVSS

SEQ ID NO: 5: CAN 04 variable heavy chain (VH) VH1

QVQLVQSGAEVKKPGSSVKVSCKASGYAFSSSWMNWVRQAPGQGLEWMGRIYPGD

GNTHYAQKFQGRVTLTADKSTSTAYMELSSLRSEDTAVYYCGEGYLDPMDYWGQGTL

VTVSS

SEQ ID NO: 6: CAN 04 variable heavy chain (VH) VH3

QVQLVQSGAEVKKPGSSVKVSCKASGYTFTSSWMNWVRQAPGKGLEWMGRIYPGDG

QTHYAQKFQGRVTLTADKSTSTAYMELSSLRSEDTAVYYCGEGYLDPMDYWGQGTLV

TVSS

SEQ ID NO: 7: CAN 04 variable heavy chain (VH) VH4

QVQLVQSGAEVKKPGSSVKVSCKASGYTFTSSWMNWVRQAPGKGLEWMGRIYPGDG

QTHYAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCGEGYLDPMDYWGQGTLV T

VSS

SEQ ID NO: 8: CAN04 variant 6 Light chain CDR1 (according to Chotia)

SASQGINNYLN SEQ ID NO: 9: CAN04 variant 6 Light chain CDR2 (according to Chotia) YTSGLHAGV

SEQ ID NO: 10: CAN04 variant 6 Light chain CDR3 (according to Chotia)

QQYSILPWT

SEQ ID NO: 11 : CAN04 variant 6 Heavy chain CDR1 (according to Chotia)

GYAFTSSSWMN

SEQ ID NO: 12: CAN04 variant 6 Heavy chain CDR2 (according to Chotia)

RIYPGDGNTHYAQKFQG

SEQ ID NO: 13: CAN04 variant 6 Heavy chain CDR3 (according to Chotia)

GYLDPMDY

SEQ ID NO: 14: CAN03 variable light chain (VL)

DILLTQSPAI LSVSPGERVSFSCRASQSIGTSIHWYQRRTNGSPRLLIKSASESISGIPSRF SGSGSGTDFTLSINSVESEDIADYYCQQSNSWPTTFGAGTKLELKR

SEQ ID NO: 15: CAN03 variable heavy chain (VH)

DVKLVESGGGLVKPGGSLKLSCAASGFTFSIYTMSWVRQTPEKRLEWVATISIGGSYIN

YPDSVKGRFTISRDNAKNTLYLQMSSLKSEDTAIYYCSREVDGSYAMDYWGQGTSVT V

SS

SEQ ID NO: 16: CAN03 Light chain CDR1 (according to Chotia)

RASQSIGTSIH

SEQ ID NO: 17: CAN03 Light chain CDR2 (according to Chotia)

SASESIS

SEQ ID NO: 18: CAN03 Light chain CDR3 (according to Chotia)

QQSNSWPTT

SEQ ID NO: 19: CAN03 Heavy chain CDR1 (according to Chotia)

GFTFSIYTMS SEQ ID NO: 20: CAN03 Heavy chain CDR2 (according to Chotia) TISIGGSYINYPDSVKG

SEQ ID NO: 21 : CAN03 Heavy chain CDR3 (according to Chotia)

EVDGSYAMDY

SEQ ID NO: 22: CAN04 variant 6 Variable light chain CDR2 (according to Kabat) YTSGLHA SEQ ID NO: 23: CAN04 variant 6 Variable light chain CDR3 (according to Kabat) QYSILPWT

SEQ ID NO: 24: CAN04 variant 6 Variable heavy chain CDR1 (according to Kabat) GYAFTSS

SEQ ID NO: 25: CAN04 variant 6 Variable heavy chain CDR2 (according to Kabat) YPGDGN

SEQ ID NO: 26: CAN03 Variable light chain CDR3 (according to Kabat)

QSNSWPTT

SEQ ID NO: 27: CAN03 Variable heavy chain CDR1 (according to Kabat)

GFTFSIY

SEQ ID NO: 28: CAN03 Variable heavy chain CDR2 (according to Kabat)

SIGGSY

SEQ ID NO: 29: CAN04 variant 6 Light chain CDR1 (according to Kabat)

ASQGINNYLN

SEQ ID NO: 30: CAN03 Light chain CDR1 (according to Kabat)

ASQSIGTSIH SEQ ID NO: 31 : CAN04 Heavy chain constant region

ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQ

SSLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPE LL

GGPSVFLFPPKPKDTLMISRTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTKPRE

EQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTL P

PSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSK L

TVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

SEQ ID NO: 32: CAN04 Light chain constant region

TVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQ

DSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC

SEQ ID NO: 33: CAN03 Heavy chain constant region, Ig gamma-1 chain C region (Homo sapiens) (UnitProt Accession No. P01857)

ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQ

SSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAP EL

LGGPSVFLFPPKPKDTLMISRTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTKPR

EEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYT L

PPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS K

LTVDKSRWQQGNVFSCSVMHEALHNHYT

SEQ ID NO: 34: CAN03 Light chain constant region, Ig kappa chain C region (Homo sapiens) (UnitProt Accession No. P01834)

TVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGN

SQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC

SEQ ID NO: 35: >hu3F8-HC-hlgG1-K409R

QVQLVQSGAEVKKPGSSVKVSCKASGYAFTSSWMNWVRQAPGQGLEWMGRIYPGD

GNTHYAQKFQGRVTLTADKSTSTAYMELSSLRSEDTAVYYCGEGYLDPMDYWGQGTL

VTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHT F

PAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCP PC PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA

KTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPR EP

QVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS F

FLYSRLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

SEQ ID NO: 36: >hu3F8-LC-hk

DIQMTQSPSSLSASVGDRVTITCQASQGINNYLNWYQQKPGKAPKLLIHYTSGLHAGVP

SRFSGSGSGTDYTLTISSLEPEDVATYYCQQYSILPWTFGGGTKVEIKRTVAAPSVF IFP

PSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLS

STLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC

SEQ ID NO: 37: >ch2C9-HC-hlgG1-F405L

DVKLVESGGGLVKPGGSLKLSCAASGFTFSIYTMSWVRQTPEKRLEWVATISIGGSYIN

YPDSVKGRFTISRDNAKNTLYLQMSSLKSEDTAIYYCSREVDGSYAMDYWGQGTSVT V

SSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPA V

LQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCP AP

ELLGGPSVFLFPPKPKDTLMISRTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTK

PREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQ VY

TLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFLL Y

SKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

SEQ ID NO: 38: > ch2C9-LC-hk

DILLTQSPAILSVSPGERVSFSCRASQSIGTSIHWYQRRTNGSPRLLIKSASESISGIPS RF

SGSGSGTDFTLSINSVESEDIADYYCQQSNSWPTTFGAGTKLELKRTVAAPSVFIFP PS

DEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTL

TLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC