Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTI-NEUROPILIN ANTIGEN-BINDING PROTEINS AND METHODS OF USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2018/119171
Kind Code:
A1
Abstract:
Provided herein are antigen-binding proteins (ABPs) that selectively bind to NRP-1 and its isoforms and homologs, and compositions comprising the ABPs. Also provided are methods of using the ABPs, such as therapeutic and diagnostic methods.

Inventors:
HICKLIN DANIEL (US)
SEIDEL-DUGAN CYNTHIA (US)
WINSTON WILLIAM (US)
SALMERON-GARCIA JOSE-ANDRES (US)
NIELSON NELS (US)
BRODKIN HEATHER (US)
Application Number:
PCT/US2017/067782
Publication Date:
June 28, 2018
Filing Date:
December 21, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
POTENZA THERAPEUTICS INC (US)
International Classes:
A61K39/395; A61P35/00; C07K16/28; G01N33/53; A61K39/00
Domestic Patent References:
WO2007056470A22007-05-18
WO2014058915A22014-04-17
WO2012006503A12012-01-12
WO2008143666A22008-11-27
WO2008077546A12008-07-03
WO2003011878A22003-02-13
WO2000061739A12000-10-19
WO2001029246A12001-04-26
WO2003085119A12003-10-16
WO2003084570A12003-10-16
WO2005035586A12005-04-21
WO2005035778A12005-04-21
WO2005053742A12005-06-16
WO2002031140A12002-04-18
WO1997030087A11997-08-21
WO1998058964A11998-12-30
WO1999022764A11999-05-14
WO2004056312A22004-07-08
WO2003085107A12003-10-16
WO1999058572A11999-11-18
WO1999051642A11999-10-14
WO1994029351A21994-12-22
WO1993016185A21993-08-19
WO1990003430A11990-04-05
WO1987000195A11987-01-15
WO2006029879A22006-03-23
WO2005100402A12005-10-27
WO2006044908A22006-04-27
WO2009036379A22009-03-19
WO2010105256A12010-09-16
WO2012009568A22012-01-19
Foreign References:
US6602684B12003-08-05
US20050123546A12005-06-09
US20030157108A12003-08-21
US20040093621A12004-05-13
US20030115614A12003-06-19
US20020164328A12002-11-07
US20040132140A12004-07-08
US20040110704A12004-06-10
US20040110282A12004-06-10
US20040109865A12004-06-10
US9150641B22015-10-06
GB98099518A
US6737056B12004-05-18
US7332581B22008-02-19
US6194551B12001-02-27
US7361740B22008-04-22
US7371826B22008-05-13
US5648260A1997-07-15
US5624821A1997-04-29
US7521541B22009-04-21
US4816567A1989-03-28
US8258082B22012-09-04
US8691730B22014-04-08
US5585089A1996-12-17
US5693761A1997-12-02
US5693762A1997-12-02
US6180370B12001-01-30
US5591669A1997-01-07
US5589369A1996-12-31
US5545807A1996-08-13
US5565332A1996-10-15
US5573905A1996-11-12
US5567610A1996-10-22
US5229275A1993-07-20
US5571894A1996-11-05
US5587458A1996-12-24
US20030215914A12003-11-20
US5204244A1993-04-20
US5534615A1996-07-09
US4767704A1988-08-30
US4657866A1987-04-14
US4927762A1990-05-22
US4560655A1985-12-24
US5122469A1992-06-16
US5500362A1996-03-19
US5821337A1998-10-13
US6267958B12001-07-31
US6171586B12001-01-09
Other References:
LIANG W C ET AL: "Function Blocking Antibodies to Neuropilin-1 Generated from a Designed Human Synthetic Antibody Phage Library", JOURNAL OF MOLECULAR BIOLOGY, ACADEMIC PRESS, UNITED KINGDOM, vol. 366, no. 3, 23 February 2007 (2007-02-23), pages 815 - 829, XP026268831, ISSN: 0022-2836, [retrieved on 20070201], DOI: 10.1016/J.JMB.2006.11.021
APPLETON BRENT A ET AL: "Structural studies of neuropilin/antibody complexes provide insights into semaphorin and VEGF binding", EMBO (EUROPEAN MOLECULAR BIOLOGY ORGANIZATION) JOURNAL, WILEY, DE, vol. 26, no. 23, 28 November 2007 (2007-11-28), pages 4902 - 4912, XP002504809, ISSN: 0261-4189, DOI: 10.1038/SJ.EMBOJ.7601906
GREG M. DELGOFFE ET AL: "Stability and function of regulatory T cells is maintained by a neuropilin-1-semaphorin-4a axis", NATURE, vol. 501, no. 7466, 4 August 2013 (2013-08-04), GB, pages 252 - 256, XP055250992, ISSN: 0028-0836, DOI: 10.1038/nature12428
T.-H. SHIN ET AL: "Enhancement of the Tumor Penetration of Monoclonal Antibody by Fusion of a Neuropilin-Targeting Peptide Improves the Antitumor Efficacy", MOLECULAR CANCER THERAPEUTICS, vol. 13, no. 3, 16 January 2014 (2014-01-16), pages 651 - 661, XP055196026, ISSN: 1535-7163, DOI: 10.1158/1535-7163.MCT-13-0748
CHAUDHARY; ELKORD, VACCINES, vol. 4, no. 3, September 2016 (2016-09-01), pages 28
BOS ET AL., JEXPMED, vol. 210, no. 11, 2013, pages 2435 - 66
TENG ET AL., CANCER RES., vol. 70, no. 20, 2010, pages 7800
APPLETON ET AL., EMBO J., vol. 26, no. 23, 28 November 2007 (2007-11-28), pages 4902 - 4912
DELGOFFE ET AL., NATURE, vol. 501, no. 7466, 12 September 2013 (2013-09-12), pages 252 - 6
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 2012, COLD SPRING HARBOR LABORATORY PRESS
PAUL: "Fundamental Immunology", 2013, LIPPINCOTT WILLIAMS & WILKINS
SCHROEDER; CAVACINI, J. ALLERGY CLIN. IMMUNOL., vol. 125, 2010, pages 41 - 52
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH
AL-LAZIKANI ET AL., J. MOL. BIOL., vol. 273, 1997, pages 927 - 948
MACCALLUM ET AL., J. MOL. BIOL., vol. 262, 1996, pages 732 - 745
LEFRANC ET AL., DEV. COMP. IMMUNOL., vol. 27, 2003, pages 55 - 77
HONEGGE; PLIICKTHUN, J. MOL. BIOL., vol. 309, 2001, pages 657 - 70
ABHINANDAN; MARTIN, IMMUNOLOGY, vol. 45, 2008, pages 3832 - 3839
"The Pharmacology of Monoclonal Antibodies", vol. 113, pages: 269 - 315
ARABI GHAHROUDI ET AL., FEBS LETTERS, vol. 414, 1998, pages 521 - 526
MUYLDERMANS ET AL., TRENDS IN BIOCHEM. SCI., vol. 26, 2001, pages 230 - 245
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 525
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 329
PRESTA, CURR. OP. STRUCT. BIOL., vol. 2, 1992, pages 593 - 596
APPLETON, THE EMBO JOURNAL, vol. 26, 2007, pages 4902 - 4912
MARKS ET AL., BIOLTECHNOLOGY, vol. 10, 1992, pages 779 - 783
BARBAS ET AL., PROC. NAT. ACAD. SCI. U.S.A., vol. 91, 1994, pages 3809 - 3813
SCHIER ET AL., GENE, vol. 169, 1995, pages 147 - 155
YELTON ET AL., J. IMMUNOL., vol. 155, 1995, pages 1994 - 2004
JACKSON ET AL., J. IMMUNOL., vol. 154, 1995, pages 3310 - 33199
HAWKINS ET AL., J. MOL. BIOL., vol. 226, 1992, pages 889 - 896
COX ET AL.: "Immunoassay Methods", ASSAY GUIDANCE MANUAL, 24 December 2014 (2014-12-24)
SILMAN ET AL., CYTOMETRY, vol. 44, 2001, pages 30 - 37
FINCO ET AL., J. PHARM. BIOMED. ANAL., vol. 54, 2011, pages 351 - 358
CREIGHTON: "Proteins: Structures and Molecular Properties", 1993, W. H. FREEMAN & CO.
SEDER; AHMED: "Nature Immunol.", vol. 4, 2003, pages: 835 - 842
NOCENTINI ET AL., BR. J. PHARMACOL., vol. 165, 2012, pages 2089 - 2099
JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 97, 2008, pages 2426 - 2447
ANALYTICAL BIOCHEMISTRY, vol. 348, 2006, pages 24 - 39
SCHROEDER; CAVACINI, J. ALLERGY CLIN. IMMUNOL., vol. 125, 2010, pages S41 - 52
WRIGHT ET AL., TIBTECH, vol. 15, 1997, pages 26 - 32
SHIELDS ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 26733 - 26740
OKAZAKI ET AL., J MOL. BIOL., vol. 336, 2004, pages 1239 - 1249
YAMANE-OHNUKI ET AL., BIOTECH. BIOENG., vol. 87, 2004, pages 614 - 622
RIPKA ET AL., ARCH. BIOCHEM. BIOPHYS., vol. 249, 1986, pages 533 - 545
KANDA ET AL., BIOTECHNOL. BIOENG., vol. 94, 2006, pages 680 - 688
AALBERSE ET AL., IMMUNOLOGY, vol. 105, 2002, pages 9 - 19
ARMOUR ET AL., MOL. IMMUNOL., vol. 40, 2003, pages 585 - 593
ARMOUR ET AL., EUR. J. IMMUNOL., vol. 29, 1999, pages 2613 - 2624
LAZAR ET AL., PROC. NATL. ACAD. SCI. USA, vol. 103, 2006, pages 4005 - 4010
IDUSOGIE ET AL., J. IMMUNOL., vol. 164, 2000, pages 4178 - 4184
HINTON ET AL., J. IMMUNOL., vol. 176, 2006, pages 346 - 356
DUNCAN; WINTER, NATURE, vol. 322, 1988, pages 738 - 740
LIU ET AL., J. BIOL. CHEM., vol. 286, 2011, pages 11211 - 11217
KOHLER ET AL., NATURE, vol. 256, 1975, pages 495 - 497
GODING J.W.: "Monoclonal Antibodies: Principles and Practice", 1986, ACADEMIC PRESS
KOZBOR, J. IMMUNOL., vol. 133, 1984, pages 3001
MORRISON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6851 - 6855
WINTER; MILSTEIN, NATURE, vol. 349, 1991, pages 293 - 299
RADER ET AL., PROC. NAT. ACAD. SCI. U.S.A., vol. 95, 1998, pages 8910 - 8915
STEINBERGER ET AL., J. BIOL. CHEM., vol. 275, 2000, pages 36073 - 36078
QUEEN ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 86, 1989, pages 10029 - 10033
JAKOBOVITS ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 90, 1993, pages 2551
JAKOBOVITS ET AL., NATURE, vol. 362, 1993, pages 255 - 258
BRUGGERMANN ET AL., YEAR IN IMMUNO., vol. 7, 1993, pages 33
HOOGENBOOM ET AL., J. MOL. BIOL., vol. 227, 1991, pages 381 - 388
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 - 597
HUDSON ET AL., NAT. MED., vol. 9, 2003, pages 129 - 134
PLIICKTHUN: "The Pharmacology of Monoclonal Antibodies", vol. 113, 1994, SPRINGER-VERLAG, pages: 269 - 315
EMANUEL ET AL., MABS, vol. 3, 2011, pages 38 - 48
VOGT; SKERRA, CHEM. BIOCHEM., vol. 5, 2004, pages 191 - 199
WAGNER ET AL., BIOCHEM. & BIOPHYS. RES. COMM., vol. 186, 1992, pages 118 - 1145
GEYER; BRENT, METH. ENZYMOL., vol. 328, 2000, pages 171 - 208
FERNANDEZ, CURR. OPINION IN BIOTECH., vol. 15, 2004, pages 364 - 373
ZAHND ET AL., J. MOL. BIOL., vol. 369, 2007, pages 1015 - 1028
EBERSBACH ET AL., J. MOL. BIOL., vol. 372, 2007, pages 172 - 185
GRAVERSEN ET AL., J. BIOL. CHEM., vol. 275, 2000, pages 37390 - 37396
SILVERMAN ET AL., NATURE BIOTECH., vol. 23, 2005, pages 1556 - 1561
SILACCI ET AL., J. BIOL. CHEM., vol. 289, 2014, pages 14392 - 14398
BINZ ET AL., NAT. BIOTECHNOL., vol. 23, 2005, pages 1257 - 1268
SKERRA, CURRENT OPIN. IN BIOTECH., vol. 18, 2007, pages 295 - 304
CHOWDHURY, METHODS MOL. BIOL., vol. 207, 2008, pages 179 - 196
HOOGENBOOM ET AL., METHODS IN MOLECULAR BIOLOGY, vol. 178, 2001, pages 1 - 37
HAM ET AL., METH. ENZ., vol. 58, 1979, pages 44
BARNES ET AL., ANAL. BIOCHEM., vol. 102, 1980, pages 255
CARTER ET AL., BIOLTECHNOLOGY, vol. 10, 1992, pages 163 - 167
YIN ET AL., MABS, vol. 4, 2012, pages 217 - 225
LINDMARK ET AL., J. IMMUNOL. METH., vol. 62, 1983, pages 1 - 13
GUSS ET AL., EMBO J., vol. 5, 1986, pages 1567 - 1575
HARLOW; LANE: "Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY
MORRIS: "Methods in Molecular Biology", vol. 66, 1996, HUMANA PRESS, article "Epitope Mapping Protocols"
JANG ET AL., ANN. CLIN. LAB. SCI., vol. 42, 2012, pages 42 - 49
RAVETCH; KINET, ANNU. REV. IMMUNOL., vol. 9, 1991, pages 457 - 492
HELLSTROM ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 83, 1986, pages 7059 - 7063
HELLSTROM ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 82, 1985, pages 1499 - 1502
BRUGGEMANN ET AL., J. EXP. MED., vol. 166, 1987, pages 1351 - 1361
CLYNES ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 95, 1998, pages 652 - 656
GAZZANO-SANTORO ET AL., J. IMMUNOL. METHODS, vol. 202, 1996, pages 163 - 171
CRAGG ET AL., BLOOD, vol. 101, 2003, pages 1045 - 1052
CRAGG ET AL., BLOOD, vol. 103, 2004, pages 2738 - 2743
PETKOVA ET AL., INT'L. IMMUNOL., vol. 18, 2006, pages 1759 - 1769
"Handbook of Pharmaceutical Excipients", 2009
"Handbook of Pharmaceutical Excipients", 2009, THE PHARMACEUTICAL PRESS
WEINMANN ET AL., CHEM MED CHEM, vol. 14, 2016, pages 1576
Y. XU ET AL.: "Addressing polyspecificity of antibodies selected from an in vitro yeast presentation system: a FACS-based, high-throughput selection and analytical tool", PEDS, vol. 26.10, 2013, pages 663 - 70, XP055233064, DOI: doi:10.1093/protein/gzt047
SIEGEL ET AL.: "High efficiency recovery and epitope-specific sorting of an scFv yeast display library", J IMMUNOL METHODS, vol. 286, no. 1-2, 2004, pages 141 - 153, XP004503454, DOI: doi:10.1016/j.jim.2004.01.005
ESTEP ET AL.: "High throughput solution-based measurement of antibody-antigen affinity and epitope binning", MABS, vol. 5, no. 2, 2013, pages 270 - 278, XP055105281, DOI: doi:10.4161/mabs.23049
ESTEP ET AL., MABS, vol. 5, 2013, pages 270 - 278
Attorney, Agent or Firm:
KABLER, Kevin et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. An isolated multivalent antigen binding protein (ABP) that specifically binds human NRP- 1 (hNRP- 1; SEQ ID NO: 130), wherein the ABP comprises the following six CDR sequences:

(a) a CDR-H3 having the sequence set forth in SEQ ID NO: 47;

(b) a CDR-H2 having the sequence X1ISGSGGX2TYYADSVX3G, wherein Xi is I or A, X2 is S or A, and X3 is K or E, as set forth in SEQ ID NO: 136;

(c) a CDR-H1 having the sequence FTFX1SX2AMV, wherein Xi is A, K, or S and X2 is Y or V, as set forth in SEQ ID NO: 137;

(d) a CDR-L3 having the sequence set forth in SEQ ID NO : 81 ;

(e) a CDR-L2 having the sequence set forth in SEQ ID NO:71 ; and

(f) a CDR-L 1 having the sequence set forth in SEQ ID NO : 63.

2. The ABP of claim 1, wherein the ABP comprises:

(a) a CDR-H3 of SEQ ID NO: 47, a CDR-H2 of SEQ ID NO: 27, a CDR- H 1 of SEQ ID NO : 12, a CDR-L3 of SEQ ID NO : 81 , a CDR-L2 of SEQ ID NO:71, and a CDR-Ll of SEQ ID NO:63;

(b) a CDR-H3 of SEQ ID NO:47, a CDR-H2 of SEQ ID NO:28, a CDR- H 1 of SEQ ID NO : 13 , a CDR-L3 of SEQ ID NO : 81 , a CDR-L2 of SEQ ID NO:71, and a CDR-Ll of SEQ ID NO:63;

(c) a CDR-H3 of SEQ ID NO: 47, a CDR-H2 of SEQ ID NO: 29, a CDR- H 1 of SEQ ID NO : 14, a CDR-L3 of SEQ ID NO : 81 , a CDR-L2 of SEQ ID NO:71, and a CDR-Ll of SEQ ID NO:63; or

(d) a CDR-H3 of SEQ ID NO: 47, a CDR-H2 of SEQ ID NO: 30, a CDR- H 1 of SEQ ID NO : 14, a CDR-L3 of SEQ ID NO : 81 , a CDR-L2 of SEQ ID NO:71, and a CDR-Ll of SEQ ID NO:63.

3. The ABP of claim 2, wherein:

(a) the ABP of claim 2(a) comprises a VH sequence of SEQ ID NO:92 and a VL sequence of SEQ ID NO: 104;

(b) the ABP of claim 2(b) comprises a VH sequence of SEQ ID NO: 93 and a VL sequence of SEQ ID NO: 104; (c) the ABP of claim 2(c) comprises a VH sequence of SEQ ID NO:94 and a VL sequence of SEQ ID NO: 104;

(d) the ABP of claim 2(d) comprises a VH sequence of SEQ ID NO: 95 and a VL sequence of SEQ ID NO: 104; or

(e) the ABP of claim 2(d) comprises a VH sequence of SEQ ID NO: 96 and a VL sequence of SEQ ID NO: 104.

4. The ABP of claim 3, wherein:

(a) the ABP of claim 2(a) comprises (i) a heavy chain of SEQ ID

NO: 1 14 and a light chain of SEQ ID NO: 126;

(b) the ABP of claim 2(b) comprises (i) a heavy chain of SEQ ID

NO: 1 15 and a light chain of SEQ ID NO: 126;

(c) the ABP of claim 2(c) comprises (i) a heavy chain of SEQ ID

NO: 1 16 and a light chain of SEQ ID NO: 126;

(d) the ABP of claim 2(d) comprises (i) a heavy chain of SEQ ID

NO: 1 17 and a light chain of SEQ ID NO: 126; or

(e) the ABP of claim 2(d) comprises (i) a heavy chain of SEQ ID

NO: 1 18 and a light chain of SEQ ID NO: 126.

5. An isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1 ; SEQ ID NO: 130), wherein the ABP comprises the following six CDR sequences:

(a) a CDR-H3 having I the sequence set forth in SEQ ID NO:41;

(b) a CDR-H2 having I the sequence set forth in SEQ ID NO: 23;

(c) a CDR-H1 having I the sequence set forth in SEQ ID NO: 8;

(d) a CDR-L3 having ; the sequence set forth in SEQ ID NO: 77;

(e) a CDR-L2 having ; the sequence set forth in SEQ ID NO: 67, and

(f) a CDR-L1 having ; the sequence set forth in SEQ ID NO: 59.

6. The ABP of claim 5, wherein:

(a) the ABP comprises a VH sequence of SEQ ID NO:85 and a VL sequence of SEQ ID NO: 100; or

(b) the ABP comprises a VH sequence of SEQ ID NO:86 and a VL sequence of SEQ ID NO: 100.

7. The ABP of claim 6, wherein:

(a) the ABP of claim 6(a) comprises a heavy chain of SEQ ID NO: 107 and a kappa light chain of SEQ ID NO: 122; and

(b) the ABP of claim 6(b) comprises a heavy chain of SEQ ID NO: 108 and a kappa light chain of SEQ ID NO: 122.

8. An isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), wherein the ABP comprises the following six CDR sequences:

(a) a CDR-H3 having the sequence ARDLGYYGSGMHX, wherein X is A or V, as set forth in SEQ ID NO: 138;

(b) a CDR-H2 having the sequence set forth in SEQ ID NO:24;

(c) a CDR-H1 having the sequence set forth in SEQ ID NO:9;

(d) a CDR-L3 having the sequence set forth in SEQ ID NO:78;

(e) a CDR-L2 having the sequence set forth in SEQ ID NO:68; and

(f) a CDR-L1 having the sequence set forth in SEQ ID NO: 60.

9. The ABP of claim 8, wherein the ABP comprises:

(a) a CDR-H3 of SEQ ID NO: 42, a CDR-H2 of SEQ ID NO: 24, a CDR- Hl of SEQ ID NO:9, a CDR-L3 of SEQ ID NO:78, a CDR-L2 of SEQ ID NO:68, and a CDR-L1 of SEQ ID NO:60; or

(b) a CDR-H3 of SEQ ID NO: 43, a CDR-H2 of SEQ ID NO: 24, a CDR- Hl of SEQ ID NO:9, a CDR-L3 of SEQ ID NO:78, a CDR-L2 of SEQ ID NO:68, and a CDR-L1 of SEQ ID NO:60.

10. The ABP of claim 8, wherein:

(a) the ABP comprises a VH sequence of SEQ ID NO:87 and a VL sequence of SEQ ID NO : 101 ; or

(b) the ABP comprises a VH sequence of SEQ ID NO : 88 and a VL sequence of SEQ ID NO : 101.

11. The ABP of claim 8, wherein:

(a) the ABP comprises a heavy chain of SEQ ID NO: 109 and a kappa light chain of SEQ ID NO: 123; or (b) the ABP comprises a heavy chain of SEQ ID NO: 110 and a kappa light chain of SEQ ID NO: 123.

12. An isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), wherein the ABP comprises the following six CDR sequences:

(a) a CDR-H3 having the sequence, ARDRGMYYASGFXP, wherein X is G or N, as set forth in (SEQ ID NO: 139);

(b) a CDR-H2 having the sequence set forth in SEQ ID NO: 25;

(c) a CDR-H1 having the sequence set forth in SEQ ID NO: 10;

(d) a CDR-L3 having the sequence set forth in SEQ ID NO:79;

(e) a CDR-L2 having the sequence set forth in SEQ ID NO:69; and

(f) a CDR-L 1 having the sequence set forth in SEQ ID NO : 61.

13. The ABP of claim 12, wherein the ABP comprises:

(a) a CDR-H3 of SEQ ID NO: 44, a CDR-H2 of SEQ ID NO: 25, a CDR- H 1 of SEQ ID NO : 10, a CDR-L3 of SEQ ID NO : 79, a CDR-L2 of SEQ ID NO:69, and a CDR-Ll of SEQ ID NO:61; or

(b) a CDR-H3 of SEQ ID NO: 45, a CDR-H2 of SEQ ID NO: 25, a CDR- H 1 of SEQ ID NO : 10, a CDR-L3 of SEQ ID NO : 79, a CDR-L2 of SEQ ID NO:69, and a CDR-Ll of SEQ ID NO:61.

14. The ABP of claim 12, wherein:

(a) the ABP comprises a VH sequence of SEQ ID NO:89 and a VL sequence of SEQ ID NO: 102; or

(b) the ABP comprises a VH sequence of SEQ ID NO:90 and a VL sequence of SEQ ID NO: 102.

15. The ABP of claim 12, wherein:

(a) the ABP comprises a heavy chain of SEQ ID NO: 111 and a kappa light chain of SEQ ID NO: 124; or

(b) the ABP comprises a heavy chain of SEQ ID NO: 112 and a kappa light chain of SEQ ID NO: 124.

16. An isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), comprising the following six CDR sequences: (a) a CDR-H3 having the sequence set forth in SEQ ID NO: 46;

(b) a CDR-H2 having the sequence set forth in SEQ ID NO: 26;

(c) a CDR-H 1 having the sequence set forth in SEQ ID NO : 11 ;

(d) a CDR-L3 having the sequence set forth in SEQ ID NO: 80;

(e) a CDR-L2 having the sequence set forth in SEQ ID NO:70; and

(f) a CDR-L 1 having the sequence set forth in SEQ ID NO : 62.

17. The ABP of claim 16, wherein the ABP comprises a VH sequence of SEQ ID NO:91 and a VL sequence of SEQ ID NO: 103.

18. The ABP of claim 16 or claim 17, wherein the ABP comprises a heavy chain of SEQ ID NO: 113 and a kappa light chain of SEQ ID NO: 125.

19. An isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), comprising the following six CDR sequences:

(a) a CDR-H3 having the sequence set forth in SEQ ID NO:48;

(b) a CDR-H2 having the sequence set forth in SEQ ID NO : 31 ;

(c) a CDR-H 1 having the sequence set forth in SEQ ID NO : 15 ;

(d) a CDR-L3 having the sequence set forth in SEQ ID NO: 82;

(e) a CDR-L2 having the sequence set forth in SEQ ID NO: 68; and

(f) a CDR-L 1 having the sequence set forth in SEQ ID NO : 64.

20. The ABP of claim 19, wherein the ABP comprises:

(a) a VH sequence of SEQ ID NO: 97 and a VL sequence of SEQ ID NO: 105, or

(b) a VH sequence of SEQ ID NO:98 and a VL sequence of SEQ ID NO: 105.

21. The ABP of claim 19 or claim 20, wherein the ABP comprises:

(a) a heavy chain of SEQ ID NO: 119 and a kappa light chain of SEQ ID NO: 127; or

(b) a heavy chain of SEQ ID NO: 120 and a kappa light chain of SEQ ID NO: 127.

22. An isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), comprising the following six CDR sequences:

(a) a CDR-H3 having the sequence set forth in SEQ ID NO: 49;

(b) a CDR-H2 having the sequence set forth in SEQ ID NO:32; (c) a CDR-Hl having the sequence set forth in SEQ ID NO: 16;

(d) a CDR-L3 having the sequence set forth in SEQ ID NO: 83;

(e) a CDR-L2 having the sequence set forth in SEQ ID NO:72; and

(f) a CDR-L 1 having the sequence set forth in SEQ ID NO : 65.

23. The ABP of claim 22, wherein the ABP comprises a VH sequence of SEQ ID NO:99 and a VL sequence of SEQ ID NO: 106.

24. The ABP of claim 16 or claim 23, wherein the ABP comprises a heavy chain of SEQ ID NO: 121 and a kappa light chain of SEQ ID NO: 128.

25. An isolated antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), comprising:

(a) a CDR-H3 having at least about 80% identity to a CDR-H3 of a VH region selected from SEQ ID NOs:41-49;

(b) a CDR-H2 having at least about 80% identity to a CDR-H2 of a VH region selected from SEQ ID NOs:23-32;

(c) a CDR-Hl having at least about 80% identity to a CDR-Hl of a VH region selected from SEQ ID NOs:8-16;

(d) a CDR-L3 having at least about 80% identity to a CDR-L3 of a VL region selected from SEQ ID NOs:77-83;

(e) a CDR-L2 having at least about 80% identity to a CDR-L2 of a VL region selected from SEQ ID NOs:67-72; and

(f) a CDR-Ll having at least about 80% identity to a CDR-Ll of a VL region selected from SEQ ID NOs:59-65.

26. The ABP of claim 25 wherein the CDR-H3, CDR-H2, CDR-Hl, CDR-L3, CDR-L2, and CDR-Ll are each identified according to a numbering scheme selected from the Kabat numbering scheme, the Chothia numbering scheme, or the IMGT numbering scheme.

27. The ABP of claim 25 or 26, the CDR-Hl is identified as defined by both the Chothia and Kabat numbering schemes, inclusive of the boundaries of both numbering schemes.

28. The ABP of claim 25, wherein:

(a) the CDR-H3 comprises a CDR-H3 selected from SEQ ID NOs:41- 49, or a variant thereof having 1, 2, or 3 amino acid substitutions; (b) the CDR-H2 comprises a CDR-H3 selected from SEQ ID NOs: 23- 32, or a variant thereof having 1, 2, or 3 amino acid substitutions;

(c) the CDR-H1 comprises a CDR-H1 selected from SEQ ID NOs:8-16, or a variant thereof having 1 or 2 amino acid substitutions;

(d) the CDR-L3 comprises a CDR-L3 selected from SEQ ID NOs:77-83, or a variant thereof having 1 or 2 amino acid substitutions;

(e) the CDR-L2 comprises a CDR-L2 selected from SEQ ID NOs: 67-72, or a variant thereof having 1 amino acid substitution; and

(f) the CDR-L1 comprises a CDR-L1 selected from SEQ ID NOs: 59-65, or a variant thereof having 1 or 2 amino acid substitutions.

29. The ABP of any one of claims 1-28, wherein the amino acid substitutions are conservative amino acid substitutions.

30. An ABP that specifically binds human NRP-1, wherein the ABP does one or more of the following:

(a) competes or cross-competes for binding to NRP-1 with an antibody selected from MAB 1, MAB2, MAB3, MAB4, MAB5, MAB6, MAB7, MAB8, MAB9, MAB10, MABl l, MAB 12, MAB 13, MAB 14, or MAB15, each as provided in Appendix A of this disclosure;

(b) is specific for cell surface NRP-1;

(c) specifically blocks NRP-1 binding to a transmembrane semaphorin polypeptide;

(d) blocks the interaction between a NRP-1 polypeptide and a vascular endothelial cell growth factor (VEGF) polypeptide;

(e) is capable of inhibiting Treg suppression in a human subject;

(f) co-stimulates an effector T cell in combination with antigen presentation from an antigen-presenting cell;

(g) inhibits the suppression of an effector T cell by a regulatory T cell;

(h) reduces the number of effector T cells in a tissue or in systemic circulation;

(i) does not substantially bind platelets;

(j) does not substantially cause thrombocytopenia when administered to a patient; (k) blocks SEMA3 binding to NRP- 1 ;

(1) does not bind to NRP- 1 -negative cells;

(m) specifically binds one or more of NRP 1 residues selected from the group consisting of Y297, T316, D320, E348, T349, K350, K351, K352, Y353, Y354, E412, T413, G414 and 1415; or

(n) is capable of any combination of (a) - (m).

31. The ABP of any one of claims 1-30, wherein the ABP antibody does not compete or cross- compete for binding with an antibody selected from MAB1, MAB2, MAB3, MAB4, MAB5, MAB6, MAB7, MAB8, MAB9, MAB 10, MABl l, MAB 12, MAB 13, MAB 14, or MAB 15, each as provided in Appendix A of this disclosure.

32. The ABP of any one of claims 1-31, wherein the NRP-1 is selected from hNRP-1 (SEQ ID NO: 130), cNRP-1 (SEQ ID NO: 132), mNRP-1 (SEQ ID NO: 134), rNRP-1 (SEQ ID NO: 135), and combinations thereof.

33. The ABP of any one of claims 1-32, wherein the ABP comprises an antibody.

34. The ABP of claim 33, wherein the antibody is a monoclonal antibody.

35. The ABP of claim 33 or 34, wherein the antibody is selected from a human antibody, a humanized antibody or a chimeric antibody.

36. The ABP of any one of claims 1-35, wherein the ABP is multivalent.

37. The ABP of any one of claims 1-35, wherein the ABP comprises an antibody fragment.

38. The ABP of any one of claims 1-37, wherein the ABP comprises an alternative scaffold.

39. The ABP of any one of claims 1-38, wherein the ABP comprises an immunoglobulin constant region.

40. The ABP claim 39, wherein the ABP comprises heavy chain constant region of a class selected from IgA, IgD, IgE, IgG, or IgM.

41. The ABP of claim 40, wherein the ABP comprises a heavy chain constant region of the class IgG and a subclass selected from IgG4, IgGl, IgG2, or IgG3.

42. The ABP of claim 41, wherein the IgG is an IgG4.

43. The ABP of claim 41, wherein the IgG is an IgGl .

44. The ABP of any one of claims 1-43, wherein the ABP comprises a common light chain antibody, an antibody with a knobs-into-holes modification, an scFv attached to an IgG, a Fab attached to an IgG, a diabody, a tetravalent bispecific antibody, a DVD-IgMAB, a DARTT M, a DuoBody®, a CovX-Body, an Fcab antibody, a TandAb®, a tandem Fab, a ZybodyMAB, or combinations thereof.

45. The ABP of any one of claims 1-43, wherein the ABP reduces binding of semaphorin 3 A to NRP-1 by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90%.

46. The ABP of any one of claims 1-43, wherein the ABP reduces binding of semaphorin 3 A to NRP-1 by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90%, and wherein the ABP does not block binding of VEGF to NRP-1.

47. The ABP of claim 45, wherein the ABP reduces binding of semaphorin 3A to NRP-1 by at least about 50%.

48. The ABP of claim 30(h), wherein the tissue is a tumor.

49. The ABP of any one of claims 1-48, wherein the NRP-1 is expressed on the surface of a target cell.

50. The ABP of any one of claims 1-49, wherein the ABP comprises a polypeptide sequence having a pyroglutamate (pE) residue at its N-terminus.

51. The ABP of any one of claims 1-50, wherein the ABP comprises a VH sequence in which an N-terminal Q is substituted with pE.

52. The ABP of any one of claims 1-51, wherein the ABP comprises a VL sequence in which an N-terminal E is substituted with pE.

53. The ABP of any one of claims 1-52, wherein the ABP comprises a heavy chain sequence in which an N-terminal Q is substituted with pE.

54. The ABP of any one of claims 1-53, wherein the ABP comprises a light chain sequence in which an N-terminal E is substituted with pE.

55. The ABP of any one of claims 1-54 for use as a medicament.

56. The ABP of any one of claims 1-53 for use in the treatment of a cancer or viral infection.

57. The ABP of claim 56 for use in the treatment of a cancer, wherein the cancer is selected from a solid tumor and a hematological tumor.

58. A kit comprising an ABP of any one of claims 1-54, and instructions for use of the ABP.

59. The kit of claim 58, wherein the ABP is lyophilized.

60. The kit of claim 59, further comprising a fluid for reconstitution of the lyophilized ABP.

61. An isolated polynucleotide encoding an ABP of any one of claims 1-54, a VH thereof, a VL thereof, a light chain thereof, a heavy chain thereof or an antigen-binding portion thereof.

62. A vector comprising the polynucleotide of claim 61.

63. A host cell comprising the polynucleotide of claim 61 or the vector of claim 62.

64. The host cell of claim 63, wherein the host cell is selected from a bacterial cell, a fungal cell, and a mammalian cell.

65. The host cell of claim 63, wherein the host cell is selected from an E. coli cell, a Saccharomyces cerevisiae cell, and a CHO cell.

66. A cell-free expression reaction comprising the polynucleotide of claim 61 or vector of claim 62.

67. A method of producing an ABP of any one of claims 1-54, comprising expressing the ABP in the host cell of claim 63 and isolating the expressed ABP.

68. A pharmaceutical composition comprising an ABP of any one of claims 1-54 and a pharmaceutically acceptable excipient.

69. The pharmaceutical composition of claim 68, wherein the ABP is present in the composition in an amount effective to locally inhibit a NRP-l:semaphorin interaction in a tumor.

70. The pharmaceutical composition of claim 68, wherein the anti-NRP-1 antibody is present in the composition in an amount effective to inhibit an interaction between NRP-1 and a transmembrane semaphorin polypeptide when administered to a human subject.

71. The pharmaceutical composition of claim 68, wherein the anti-NRP- 1 antibody specifically blocks NRP-1 binding to a transmembrane semaphorin polypeptide.

72. The pharmaceutical composition of claim 68, wherein the anti-NRP-1 antibody does not affect the interaction between a NRP- 1 polypeptide and a vascular endothelial cell growth factor (VEGF) polypeptide.

73. The pharmaceutical composition of claim 68, wherein the anti-NRP-1 antibody is capable of inhibiting Treg suppression in the human subject.

74. The pharmaceutical composition of claim 68, wherein the anti-NRP-1 antibody is capable of decreasing Treg survival and/or stability in the human subject.

75. The pharmaceutical composition of claim 68, wherein the anti-NRP-1 antibody is present in the composition in an amount effective to locally inhibit the NRP-l :semaphorin-4 interaction in a tumor.

76. The pharmaceutical composition of claim 68, wherein the anti-NRP-1 antibody is present in the composition in an amount effective to locally inhibit the NRP-l :semaphorin-3 interaction in a tumor.

77. The pharmaceutical composition of claim 68, wherein the anti-NRP-1 antibody is present in the composition in an amount effective to prevent development of an undesired autoimmune and/or inflammatory manifestation.

78. The pharmaceutical composition of claim 68, wherein the human subject is suffering from a cancer.

79. The pharmaceutical composition of claim 68, wherein the amount of the ABP in the pharmaceutical composition is sufficient to (a) reduce the suppression of effector T cells by regulatory T cells; (b) activate effector T cells; (c) reduce the number of regulatory T cells in a tissue or systemically; (d) induce or enhance proliferation of effector T cells; (e) inhibit the rate of tumor growth; (f) induce tumor regression; or (g) combinations thereof, in a subject.

80. The pharmaceutical composition of any one of claims 68-79, for use as a

medicament.

81. The pharmaceutical composition of any one of claims 68-80, for use in the treatment of a cancer or a viral infection.

82. The pharmaceutical composition of claim 81, for use in the treatment of a cancer, wherein the cancer is selected from brain, prostate, breast, colon, skin, and lung cancer.

83. A method of inhibiting a function or decreasing stability of a regulatory T cell (Treg) in a subject, comprising exposing the Treg in vivo to an inhibitor of neuropilin-1 (NRP- l):semaphorin axis in the Treg, wherein an effective amount of an ABP of any one of claims 1-54, or a pharmaceutical composition of any one of claims 67-82, is administered to the subject.

84. A method of increasing T effector cell (Tea) function or exposing the Teff in vivo to the ABP of any one of claims 1-54, comprising administering to a subject an effective amount of a pharmaceutical composition of any one of claims 68-82.

85. The method of claim 83 or 84, wherein the subject has a cancer.

86. The method of any one of claims 83-85, wherein the method induces or enhances an immune response to a cancer-associated antigen.

87. The method of any one of claims 83-86, wherein the ABP is capable of (a) decreasing Treg survival and/or stability in the human subject; (b) binding to an extracellular domain of the NRP-1 polypeptide; or (c) a combination thereof.

88. The method of any one of claims 83-87, further comprising administering one or more additional therapeutic agents.

89. The method of claim 88, wherein the additional therapeutic agent is selected from radiation, a cytotoxic agent, a chemotherapeutic agent, a cytostatic agent, an anti-hormonal agent, a VEGF inhibitor, an immunostimulatory agent, an anti-angiogenic agent, and combinations thereof.

90. The method of claim 88, wherein the additional therapeutic agent is an

immunostimulatory agent.

91. The method of claim 90, wherein the additional therapeutic agent is a chimeric antigen receptor T cell.

92. The method of claim 90, wherein the immunostimulatory agent comprises an agent that blocks signaling of an inhibitory receptor expressed by an immune cell or a ligand thereof.

93. The method of claim 92, wherein the inhibitory receptor expressed by an immune cell or ligand thereof is selected from PVRIG, VISTA, CCR4, CD27, CTLA-4, PD-1, PD-L1, LAG-3, Tim3, TIGIT, neuritin, BTLA, KIR, and combinations thereof.

94. The method of claim 90, wherein the immunostimulatory agent comprises an agonist to a stimulatory receptor expressed by an immune cell.

95. The method of claim 93, wherein the stimulatory receptor expressed by an immune cell is selected from OX40, ICOS, GITR, CD28, CD37, CD40, 4-1BB, and combinations thereof.

96. The method of claim 90, wherein the immunostimulatory agent comprises a cytokine.

97. The method of claim 90, wherein the immunostimulatory agent comprises a vaccine to a cancer-associated antigen.

98. A method of modulating an immune response in a subject in need thereof, comprising administering to the subject an effective amount of an ABP of any one of claims 1-54 or a pharmaceutical composition of any one of claims 67-82.

99. The method of any one of claims 83-98, further comprising administering one or more additional therapeutic agents to the subject.

100. The method of claim 99, wherein the additional therapeutic agent is (i) an agonist to a stimulatory receptor of an immune cell or (ii) an antagonist of an inhibitory receptor of an immune cell, wherein the receptor of an immune cell is selected from OX40, CD2, CD27, CDS, ICAM-1, LFA-1 (CDl la/CD18), ICOS (CD278), 4-1BB (CD137), CD28, CD30, CD40, BAFFR, HVEM, CD 7, LIGHT, NKG2C, GITR, SLAMF7, NKp80, CD160, B7-H3, CD83 ligand, and combinations thereof.

101. The method of claim 99, wherein the additional therapeutic agent is an oncolytic virus selected from herpes simplex virus, vesicular stomatitis virus, adenovirus, Newcastle disease virus, vaccinia virus, a maraba virus, and combinations thereof.

102. The method of any one of claims 99-101, wherein the additional therapeutic agent is formulated in the same pharmaceutical composition as the ABP.

103. The method of any one of claims 99-101, wherein the additional therapeutic agent is formulated in a different pharmaceutical composition from the ABP.

104. The method of any one of claims 99-101 or 103, wherein the additional therapeutic agent is administered prior to administering the ABP.

105. The method of any one of claims 99-101 or 103, wherein the additional therapeutic agent is administered after administering the ABP.

106. The method of any one of claims 99-105, wherein the additional therapeutic agent is administered contemporaneously with the ABP.

107. The method of any one of claims 83-106, wherein the method does not substantially cause thrombocytopenia in the subject.

108. The ABP of any one of claims 1-57, wherein the ABP specifically binds to human NRP-1 with a kD of less than 20nM, less than 10 nM, less than 5 nM, less than 2 nM, less than 1 nM, less than 0.5 nM, or less than 0.2 nM.

109. The ABP of any one of claims 1-57 or claim 108, wherein the ABP specifically binds to NRP-1 from humans, mice, and cynomolgus monkeys.

110. The ABP of any one of claims 1-57 or 108-110, wherein the ABP binds to a different epitope on NRP-1 than the epitope on NRP-1 to which SEC 10 binds.

111. The ABP of any one of claims 1-57 or 108-110, wherein the ABP binds to the bl domain of NRP-1.

112. The ABP of any one of claims 1-57 or claim 108 or claim 109, wherein the ABP specifically binds one or more residues on NRPl (SEQ ID NO: 130) chosen from the group consisting of specifically binds one or more of NRPl residues selected from the group consisting of Y297, T316, D320, E348, T349, K350, K351, K352, Y353, Y354, E412, T413, G414 and 1415.

113. An anti-human NRP-1 antibody or an antigen-binding fragment thereof, comprising: a heavy chain variable region comprising a CDR-H3 consisting of SEQ ID NO: 47, a CDR-H2 consisting of SEQ ID NO:30, and a CDR-H1 consisting of SEQ ID NO: 14; and a light chain variable region comprising a CDR-L3 consisting of SEQ ID NO: 81, a CDR- L2 consisting of SEQ ID NO:71, and a CDR-L1 consisting of SEQ ID NO:63.

114. The anti -human NRP-1 antibody or an antigen-binding fragment thereof according to claim 1, which is selected from any one of the following (1) and (2):

(1) an anti -human NRP-1 antibody or an antigen-binding fragment thereof, comprising a heavy chain variable region consisting of SEQ ID NO: 96, and a light chain variable region consisting of SEQ ID NO: 104; and

(2) an anti -human NRP- 1 antibody or the antigen-binding fragment thereof, comprising a heavy chain variable region consisting of SEQ ID NO: 96 in which E of the amino acid number 1 is modified to pyroglutamate, and a light chain variable region consisting of SEQ ID NO: 104.

115. The anti -human NRP-1 antibody or an antigen-binding fragment thereof according to claim 2, which is selected from the group consisting of (1) to (4):

(1) an anti -human NRP-1 antibody comprising a heavy chain consisting of SEQ ID NO: 118, and a light chain consisting of SEQ ID NO: 126,

(2) an anti -human NRP- 1 antibody comprising a heavy chain consisting of SEQ ID NO: 118 in which E of the amino acid number 1 is modified to pyroglutamate, and a light chain consisting of SEQ ID NO: 126,

(3) an anti -human NRP- 1 antibody comprising a heavy chain consisting of the amino acid sequence of amino acid numbers 1 to 453 of SEQ ID NO: 118, and a light chain consisting of SEQ ID NO: 126; and

(4) an anti -human NRP- 1 antibody comprising a heavy chain consisting of the amino acid sequence of amino acid numbers 1 to 453 of SEQ ID NO: 118 in which E of the amino acid number 1 is modified to pyroglutamate, and a light chain consisting of SEQ ID NO: 126.

116. A polynucleotide, which is selected from the group consisting of (1) and (2): (1) a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti-human NRP- 1 antibody or the antigen-binding fragment thereof according to claim 114(1), and

(2) a polynucleotide comprising a base sequence encoding the light chain variable region of the anti-human NRP- 1 antibody or the antigen-binding fragment thereof according to claim 114(1).

117. A polynucleotide, which is selected from the group consisting of (1) and (2):

(1) a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody according to claim 115(1), and

(2) a polynucleotide comprising a base sequence encoding the light chain of the anti-human NRP-1 antibody according to claim 115(1).

118. An expression vector comprising:

(a) a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof of claim 114(1), and/or

(b) a polynucleotide comprising a base sequence encoding the light chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof of claim 114(1).

119. An expression vector comprising:

(a) a polynucleotide comprising a base sequence encoding the heavy chain of the anti- human NRP-1 antibody of claim 115(1), and/or

(b) a polynucleotide comprising a base sequence encoding the light chain of the anti- human NRP-1 antibody of claim 115(1).

120. A host cell transformed with an expression vector selected from the group consisting of (a) to (d):

(a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP-1 antibody or the antigen-binding fragment thereof of claim 114(1), and a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen-binding fragment thereof;

(b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP- 1 antibody or the antigen-biding fragment thereof according to claim 114(1) and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen- binding fragment thereof; (c) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP- 1 antibody or the antigen-binding fragment thereof according to claim 114(1); and

(d) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the anti-human NRP-1 antibody or the antigen-binding fragment thereof according to claim 114(1).

121. A host cell transformed with an expression vector selected from the group consisting of (a) to (d):

(a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of claim 115(1) and a polynucleotide comprising a base sequence encoding the light chain of the antibody;

(b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of claim 115(1) and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the antibody;

(c) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of claim 115(1); and

(d) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the anti -human NRP-1 antibody of claim 115(1).

122. A method for producing an anti -human NRP-1 antibody or an antigen-binding

fragment thereof, comprising culturing host cell(s) selected from the group consisting of (a) to (c) below to express a tetravalent anti -human NRP-1 antibody or an antigen- binding fragment thereof:

(a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP-1 antibody or the antigen-binding fragment thereof of 114(1) and a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen-binding fragment thereof;

(b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP-1 antibody or the antigen-binding fragment thereof of claim 114(1) and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen-binding fragment thereof; and

(c) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP- 1 antibody or the antigen-binding fragment thereof of claim 114( 1) and a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen-binding fragment thereof.

123. A method for producing an anti -human NRP-1 antibody, comprising culturing host cell(s) selected from the group consisting of (a) to (c) below to express an anti-human NRP- 1 antibody:

(a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of 1 15( 1) and a polynucleotide comprising a base sequence encoding the light chain of the antibody;

(b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of claim 1 15(1) and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the antibody; and

(c) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of claim 1 15(1) and a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the antibody.

124. A pharmaceutical composition comprising the anti-human NRP-1 antibody of claim 115 and a pharmaceutically acceptable excipient.

125. A pharmaceutical composition comprising the anti-human NRP-1 antibody of claim 115( 1), the anti-human NRP-1 antibody of claim 1 15(2), the anti-human NRP- 1 antibody of claim 115(3), and/or the anti-human NRP-1 antibody of claim 1 15(4), and a

pharmaceutically acceptable excipient.

126. The pharmaceutical composition of any one of claims 124 and 125, which is a pharmaceutical composition for treating cancer.

127. The pharmaceutical composition of any one of claims 124 - 126, wherein the composition is administered in combination with radiation, a cytotoxic agent, a chemotherapeutic agent, a cytostatic agent, an anti-hormonal agent, a VEGF inhibitor, an immunostimulatory agent, an anti-angiogenic agent, and combinations thereof.

128. The anti-human NRP- 1 antibody of claim 1 15, for preventing or treating cancer.

129. Use of the anti-human NRP-1 antibody of claim 1 15 for manufacture of a pharmaceutical composition for preventing or treating cancer.

130. A method for preventing or treating cancer, comprising administering a therapeutically effective amount of the anti -human NRP-1 antibody of claim 1 15.

131. The method of claim 130, further comprising administering one or more additional therapeutic agents.

132. The method of claim 131, wherein the additional therapeutic agent is selected from the group consisting of radiation, a cytotoxic agent, a chemotherapeutic agent, a cytostatic agent, an anti-hormonal agent, a VEGF inhibitor, an immunostimulatory agent, an anti- angiogenic agent, and combinations thereof.

Description:
ANTI-NEUROPILIN ANTIGEN-BINDING PROTEINS

AND METHODS OF USE THEREOF

FIELD

[1] Provided herein are antigen-binding proteins (ABPs) with binding specificity for NRP-1 and compositions comprising such ABPs, including pharmaceutical compositions, diagnostic

compositions, and kits. Also provided are methods of making NRP-1 ABPs, and methods of using NRP-1 ABPs, for example, for therapeutic purposes, diagnostic purposes, and research purposes.

BACKGROUND

[2] Multiple studies have demonstrated that tumors are able to establish an immunosuppressive microenvironment to escape immune surveillance and promote tumor development. Regulatory T cells (Tregs) are an important component of the immunosuppressive milieu in the tumor environment and work by dampening T cell immunity to tumor associated antigens. Tregs are therefore a major obstacle in mounting an effective anti-tumor immune response. Depletion of Tregs in murine models of cancer inhibits tumor growth; however, the accompanying autoimmune and inflammatory disorders associated with a complete depletion of Tregs may limit the clinical utility of this approach. Strategies which specifically target Tregs, in the inflammatory tumor microenvironment, may be a viable alternative. Recent studies in several laboratories have identified Neuropilin 1 (NRP-1) as a candidate target for modulating Treg activity in tumors without impacting Tregs in the periphery (see, e.g., Chaudhary and Elkord, Vaccines (2016) Sep;4(3): 28; Bos et al., J Exp Med (2013) 210 (11):2435-66; Teng et al, Cancer Res. (2010) 70 (20):7800-..

[3] NRP-1 is a multifunctional 130-kDa transmembrane protein with a large extracellular domain containing two N-terminal CUB domains (al and a2), two coagulation factor V/VIII homology domains (bl and b2) and a single MAM domain (c). The cytoplasmic tail is short and does not display any catalytic activity on its own. NRP-1 is a receptor with multiple known ligands and co-receptors, including semaphorins, VEGF, P1GF and plexins, among others (Appleton et al., Embo J. (2007) Nov 28; 26(23): 4902-4912).

[4] NRP-1 is expressed on human and murine Tregs, and this expression identifies a highly- suppressive Treg subset. Within the tumor microenvironment, NRP-1 expression is required for Treg stability and function but does not impact Tregs outside the inflammatory environment of tumors. Recent studies have identified the immune cell-expressed ligand semaphorin 4A (Sema4a) as an additional ligand for NRP-1, and demonstrated that the sema4a/NRP-l interaction is an important mediator of Treg stability in vitro and in inflammatory sites in vivo. These data suggest that NRP-1 is required for Treg lineage stability and function (see, e.g., Delgoffe et al., Nature (2013) Sep

12;501(7466):252-6.).

[5] Several lines of evidence support the utility of targeting the interaction of NRP-1 and its associated proteins, in particular targeting the NRP-l/Sema axis, on Tregs as a strategy for modulating the immunosuppressive microenvironment found in tumors. For instance, mice with Treg targeted NRP-1 knock-out exhibit reduced tumor growth in several murine tumor models, without any other autoimmune phenotypes. Additionally, antagonists to NRP-1 or Sema reverse Treg suppressive activity and demonstrate anti-tumor efficacy again in the absence of autoimmune adverse events. Furthermore, the NRP-1 -VEGFA axis has been proposed as an important pathway regulating the chemotaxis of Tregs into the tumor micro-environment, and an antagonistic Ab that blocks this interaction on Tregs could inhibit the influx of these suppressive cells into the tumor.

[6] There is emerging evidence suggesting the NRP-1 is expressed on the surface of immune cells in human tumors. NRP-1+ Tregs are found in the draining lymph nodes (DLN) from cervical cancer patents, and there was a significant drop in the percentage of Tregs in DLN in patients with a pathological response to preoperative chemoradiation. In addition, NRP-1+ Tregs have been observed in tumor infiltrating lymphocytes (TILs) in patients with melanoma and head and neck squamous cell carcinoma.

[7] Thus, there is a need for therapeutics that can antagonize NRP-1 without inducing autoimmune disease. Provided herein are ABPs that fulfill this need.

SUMMARY

[8] Provided herein are ABPs that specifically bind NRP-1 and methods of using such ABPs.

[9] In one aspect, provided herein are is an isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), wherein the ABP comprises the following six CDR sequences:

(a) a CDR-H3 having the sequence set forth in SEQ ID NO: 47;

(b) a CDR-H2 having the sequence X1ISGSGGX2TYYADSVX3G, wherein Xi is I or A, X 2 is S or A, and X 3 is K or E, as set forth in SEQ ID NO: 136;

(c) a CDR-H1 having the sequence FTFX1SX2AMV, wherein Xi is A, K, or S and X 2 is Y or V, as set forth in SEQ ID NO: 137;

(d) a CDR-L3 having the sequence set forth in SEQ ID NO : 81 ;

(e) a CDR-L2 having the sequence set forth in SEQ ID NO:71; and

(f) a CDR-L 1 having the sequence set forth in SEQ ID NO : 63.

[10] In one embodiment, the ABP comprises a CDR-H3 of SEQ ID NO:47, a CDR-H2 of SEQ ID NO : 27, a CDR-H 1 of SEQ ID NO : 12, a CDR-L3 of SEQ ID NO : 81 , a CDR-L2 of SEQ ID NO : 71 , and a CDR-L1 of SEQ ID NO:63; or a CDR-H3 of SEQ ID NO:47, a CDR-H2 of SEQ ID NO:28, a CDR-Hl of SEQ ID NO: 13, a CDR-L3 of SEQ ID NO:81, a CDR-L2 of SEQ ID NO:71, and a CDR- Ll of SEQ ID NO:63; or a CDR-H3 of SEQ ID NO:47, a CDR-H2 of SEQ ID NO:29, a CDR-H1 of SEQ ID NO : 14, a CDR-L3 of SEQ ID NO : 81 , a CDR-L2 of SEQ ID NO : 71 , and a CDR-L 1 of SEQ ID NO: 63; or a CDR-H3 of SEQ ID NO: 47, a CDR-H2 of SEQ ID NO: 30, a CDR-H1 of SEQ ID NO : 14, a CDR-L3 of SEQ ID NO : 81 , a CDR-L2 of SEQ ID NO : 71 , and a CDR-L 1 of SEQ ID NO:63.

[11] In another embodiment, the ABP comprises a VH sequence of SEQ ID NO:92 and a VL sequence of SEQ ID NO: 104; a V H sequence of SEQ ID NO:93 and a V L sequence of SEQ ID NO: 104; a V H sequence of SEQ ID NO:94 and a V L sequence of SEQ ID NO: 104; a V H sequence of SEQ ID NO:95 and a V L sequence of SEQ ID NO: 104; or a V H sequence of SEQ ID NO:96 and a V L sequence of SEQ ID NO: 104.

[12] In another embodiment, the ABP comprises a heavy chain of SEQ ID NO: 1 14 and a light chain of SEQ ID NO: 126; a heavy chain of SEQ ID NO: 115 and a light chain of SEQ ID NO: 126; a heavy chain of SEQ ID NO: 1 16 and a light chain of SEQ ID NO: 126; a heavy chain of SEQ ID NO: 1 17 and a light chain of SEQ ID NO: 126; or a heavy chain of SEQ ID NO: 1 18 and a light chain of SEQ ID NO: 126.

[13] In another aspect is provided an isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1 ; SEQ ID NO: 130), wherein the ABP comprises the following six CDR sequences:

(a) a CDR-H3 having I the sequence set forth in SEQ ID NO:41;

(b) a CDR-H2 having I the sequence set forth in SEQ ID NO: 23;

(c) a CDR-H1 having I the sequence set forth in SEQ ID NO: 8;

(d) a CDR-L3 having ; the sequence set forth in SEQ ID NO: 77;

(e) a CDR-L2 having ; the sequence set forth in SEQ ID NO: 67, and

(f) a CDR-L 1 having ; the sequence set forth in SEQ ID NO: 59.

[14] In one embodiment, the ABP comprises a VH sequence of SEQ ID NO: 85 and a VL sequence of SEQ ID NO: 100; or a V H sequence of SEQ ID NO: 86 and a V L sequence of SEQ ID NO: 100. In another embodiment, the ABP comprises a heavy chain of SEQ ID NO: 107 and a kappa light chain of SEQ ID NO: 122; and a heavy chain of SEQ ID NO: 108 and a kappa light chain of SEQ ID NO: 122.

[15] In another aspect is provided an isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1 ; SEQ ID NO: 130), wherein the ABP comprises the following six CDR sequences:

(a) a CDR-H3 having the sequence ARDLGYYGSGMHX, wherein X is A or V, as set forth in SEQ ID NO: 138;

(a) a CDR-H2 having the sequence set forth in SEQ ID NO: 24;

(b) a CDR-H1 having the sequence set forth in SEQ ID NO:9;

(c) a CDR-L3 having the sequence set forth in SEQ ID NO: 78; (d) a CDR-L2 having the sequence set forth in SEQ ID NO: 68; and

(e) a CDR-Ll having the sequence set forth in SEQ ID NO: 60.

[16] In one embodiment, the ABP comprises: a CDR-H3 of SEQ ID NO:42, a CDR-H2 of SEQ ID NO:24, a CDR-Hl of SEQ ID NO:9, a CDR-L3 of SEQ ID NO:78, a CDR-L2 of SEQ ID NO:68, and a CDR-Ll of SEQ ID NO:60; or a CDR-H3 of SEQ ID NO:43, a CDR-H2 of SEQ ID NO:24, a CDR- Hl of SEQ ID NO:9, a CDR-L3 of SEQ ID NO:78, a CDR-L2 of SEQ ID NO:68, and a CDR-Ll of SEQ ID NO:60. In another embodiment, the ABP comprises a VH sequence of SEQ ID NO:87 and a V L sequence of SEQ ID NO : 101 ; or the ABP comprises a V H sequence of SEQ ID NO : 88 and a V L sequence of SEQ ID NO: 101. In another embodiment, the ABP comprises a heavy chain of SEQ ID NO: 109 and a kappa light chain of SEQ ID NO: 123; or the ABP comprises a heavy chain of SEQ ID NO: 110 and a kappa light chain of SEQ ID NO: 123.

[17] In another aspect is provided an isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), wherein the ABP comprises the following six CDR sequences:

(a) a CDR-H3 having the sequence, ARDRGMYYASGFXP, wherein X is G or N, as set forth in (SEQ ID NO: 139);

(b) a CDR-H2 having the sequence set forth in SEQ ID NO: 25;

(c) a CDR-Hl having the sequence set forth in SEQ ID NO: 10;

(d) a CDR-L3 having the sequence set forth in SEQ ID NO:79;

(e) a CDR-L2 having the sequence set forth in SEQ ID NO:69; and

(f) a CDR-Ll having the sequence set forth in SEQ ID NO:61.

[18] In one embodiment the ABP comprises a CDR-H3 of SEQ ID NO:44, a CDR-H2 of SEQ ID NO: 25, a CDR-Hl of SEQ ID NO: 10, a CDR-L3 of SEQ ID NO: 79, a CDR-L2 of SEQ ID NO: 69, and a CDR-Ll of SEQ ID NO:61; or a CDR-H3 of SEQ ID NO:45, a CDR-H2 of SEQ ID NO:25, a CDR-Hl of SEQ ID NO: 10, a CDR-L3 of SEQ ID NO: 79, a CDR-L2 of SEQ ID NO: 69, and a CDR- Ll of SEQ ID NO:61. In another embodiment, the ABP comprises a VH sequence of SEQ ID NO: 89 and a VL sequence of SEQ ID NO: 102; or a VH sequence of SEQ ID NO:90 and a VL sequence of SEQ ID NO: 102. In another embodiment, the ABP comprises a heavy chain of SEQ ID NO: 111 and a kappa light chain of SEQ ID NO: 124; or the ABP comprises a heavy chain of SEQ ID NO: 112 and a kappa light chain of SEQ ID NO: 124.

[19] In another aspect is provided an isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), comprising the following six CDR sequences: (a) a CDR-H3 having the sequence set forth in SEQ ID NO: 46;

(b) a CDR-H2 having the sequence set forth in SEQ ID NO: 26;

(c) a CDR-H1 having the sequence set forth in SEQ ID NO: 11;

(d) a CDR-L3 having the sequence set forth in SEQ ID NO: 80;

(e) a CDR-L2 having the sequence set forth in SEQ ID NO:70; and

(f) a CDR-L1 having the sequence set forth in SEQ ID NO: 62.

[20] In one embodiment, the ABP comprises a VH sequence of SEQ ID NO:91 and a VL sequence of SEQ ID NO: 103. In another embodiment, the ABP comprises a heavy chain of SEQ ID NO: 113 and a kappa light chain of SEQ ID NO: 125.

[21] In another aspect is provided an isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), comprising the following six CDR sequences:

(a) a CDR-H3 having the sequence set forth in SEQ ID NO:48;

(b) a CDR-H2 having the sequence set forth in SEQ ID NO: 31;

(c) a CDR-H1 having the sequence set forth in SEQ ID NO: 15;

(d) a CDR-L3 having the sequence set forth in SEQ ID NO: 82;

(e) a CDR-L2 having the sequence set forth in SEQ ID NO: 68; and

(f) a CDR-L1 having the sequence set forth in SEQ ID NO: 64.

[22] In one embodiment, the ABP comprises a VH sequence of SEQ ID NO:97 and a VL sequence of SEQ ID NO: 105, or a V H sequence of SEQ ID NO:98 and a V L sequence of SEQ ID NO: 105. In another embodiment, the ABP comprises: a heavy chain of SEQ ID NO: 119 and a kappa light chain of SEQ ID NO: 127; or a heavy chain of SEQ ID NO: 120 and a kappa light chain of SEQ ID NO: 127.

[23] In another aspect is provided an isolated multivalent antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), comprising the following six CDR sequences:

(a) a CDR-H3 having the sequence set forth in SEQ ID NO: 49;

(b) a CDR-H2 having the sequence set forth in SEQ ID NO:32;

(c) a CDR-H1 having the sequence set forth in SEQ ID NO: 16;

(d) a CDR-L3 having the sequence set forth in SEQ ID NO: 83;

(e) a CDR-L2 having the sequence set forth in SEQ ID NO:72; and

(f) a CDR-L1 having the sequence set forth in SEQ ID NO: 65. [24] In one embodiment, the ABP comprises a VH sequence of SEQ ID NO:99 and a VL sequence of SEQ ID NO: 106. In another embodiment, the ABP comprises a heavy chain of SEQ ID NO: 121 and a kappa light chain of SEQ ID NO: 128.

[25] In another aspect is provided an isolated antigen binding protein (ABP) that specifically binds human NRP-1 (hNRP-1; SEQ ID NO: 130), comprising a CDR-H3 having at least about 80% identity to a CDR-H3 of a V H region selected from SEQ ID NOs:41-49; a CDR-H2 having at least about 80% identity to a CDR-H2 of a VH region selected from SEQ ID NOs:23-32; a CDR-Hl having at least about 80% identity to a CDR-Hl of a V H region selected from SEQ ID NOs:8-16; a CDR-L3 having at least about 80% identity to a CDR-L3 of a V L region selected from SEQ ID NOs:77-83; a CDR-L2 having at least about 80% identity to a CDR-L2 of a VL region selected from SEQ ID NOs:67-72; and a CDR-L1 having at least about 80% identity to a CDR-L1 of a VL region selected from SEQ ID NOs:59-65. In one embodiment, the CDR-H3, CDR-H2, CDR-Hl, CDR-L3, CDR-L2, and CDR-L1 are each identified according to a numbering scheme selected from the Kabat numbering scheme, the Chothia numbering scheme, or the IMGT numbering scheme. In another embodiment, the CDR-Hl is identified as defined by both the Chothia and Kabat numbering schemes, inclusive of the boundaries of both numbering schemes. In one embodiment, the CDR-H3 comprises a CDR-H3 selected from SEQ ID NOs:41-49, or a variant thereof having 1, 2, or 3 amino acid substitutions; the CDR-H2 comprises a CDR-H3 selected from SEQ ID NOs:23-32, or a variant thereof having 1, 2, or 3 amino acid substitutions; the CDR-Hl comprises a CDR-Hl selected from SEQ ID NOs:8-16, or a variant thereof having 1 or 2 amino acid substitutions; the CDR-L3 comprises a CDR-L3 selected from SEQ ID NOs:77-83, or a variant thereof having 1 or 2 amino acid substitutions; the CDR-L2 comprises a CDR-L2 selected from SEQ ID NOs:67-72, or a variant thereof having 1 amino acid substitution; and the CDR-L1 comprises a CDR-L1 selected from SEQ ID NOs:59-65, or a variant thereof having 1 or 2 amino acid substitutions. In one embodiment, the amino acid substitutions are conservative amino acid substitutions.

[26] In another aspect is provided an ABP that specifically binds human NRP-1, wherein the ABP:

(a) competes or cross-competes for binding to NRP-1 with an antibody selected from MAB 1, MAB2, MAB3, MAB4, MAB5, MAB6, MAB7, MAB8, MAB9, MAB10, MAB11, MAB12, MAB13, MAB 14, or MAB15, each as provided in Appendix A of this disclosure;

(b) is specific for cell surface NRP-1;

(c) specifically blocks NRP-1 binding to a transmembrane semaphorin polypeptide;

(d) blocks the interaction between a NRP-1 polypeptide and a vascular endothelial cell

growth factor (VEGF) polypeptide;

(e) is capable of inhibiting Treg suppression in a human subject; (f) co-stimulates an effector T cell in combination with antigen presentation from an antigen- presenting cell;

(g) inhibits the suppression of an effector T cell by a regulatory T cell;

(h) reduces the number of effector T cells in a tissue or in systemic circulation;

(i) does not substantially bind platelets;

(j) does not substantially cause thrombocytopenia when administered to a patient;

(k) blocks SEMA3 binding to NRP-1;

(1) does not bind to NRP- 1 -negative cells; or

(m) is capable of any combination of (a) - (1).

[27] In one embodiment, the ABP antibody does not compete or cross-compete for binding with an antibody selected from MAB 1, MAB2, MAB3, MAB4, MAB5, MAB6, MAB7, MAB8, MAB9, MAB10, MAB11, MAB12, MAB 13, MAB 14, or MAB 15, each as provided in Appendix A of this disclosure. In one embodiment, the ABP is an ABP selected from MAB 1, MAB2, MAB3, MAB4, MAB5, MAB6, MAB7, MAB8, MAB9, MAB10, MAB 11, MAB 12, MAB13, MAB14, or MAB 15, each as provided in Appendix A of this disclosure. In one embodiment, the NRP-1 is selected from hNRP-1 (SEQ ID NO: 130), cNRP-1 (SEQ ID NO: 132), mNRP-1 (SEQ ID NO: 134), rNRP-1 (SEQ ID NO: 135), and combinations thereof.

[28] In one embodiment, the ABP comprises an antibody. In one embodiment, the antibody is a monoclonal antibody. In another embodiment, the antibody is selected from a human antibody, a humanized antibody or a chimeric antibody. In one embodiment, the ABP is multivalent. In another embodiment, the ABP comprises an antibody fragment. In another embodiment, the ABP comprises an alternative scaffold. In another embodiment, the ABP comprises an immunoglobulin constant region. In another embodiment, the ABP comprises heavy chain constant region of a class selected from IgA, IgD, IgE, IgG, or IgM. In another embodiment, ABP comprises a heavy chain constant region of the class IgG and a subclass selected from IgG4, IgGl, IgG2, or IgG3. In another embodiment, the IgG is an IgG4. In another embodiment, the IgG is an IgGl .

[29] In one embodiment, the ABP comprises a common light chain antibody, an antibody with a knobs-into-holes modification, an scFv attached to an IgG, a Fab attached to an IgG, a diabody, a tetravalent bispecific antibody, a DVD-IgMAB, a DARTT M, a DuoBody ® , a CovX-Body, an Fcab antibody, a TandAb ® , a tandem Fab, a Zybody^^, or combinations thereof.

[30] In one embodiment, the ABP blocks binding of semaphorin 3 A (SEMA3A) to NRP-1 by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90%. In on embodiment, the ABP reduces binding of semaphorin 3A to NRP-1 by at least about 50%. In one embodiment, the tissue is a tumor. In another embodiment, the NRP-1 is expressed on the surface of a target cell.

[31] In one embodiment, the ABP comprises a polypeptide sequence having a pyroglutamate (pE) residue at its N-terminus. In another embodiment, the ABP comprises a VH sequence in which an N- terminal Q is substituted with pE. In another embodiment, the ABP comprises a VH sequence in which an N-terminal E is substituted with pE. In another embodiment, the ABP comprises a VL sequence in which an N-terminal E is substituted with pE. In another embodiment, the ABP comprises a heavy chain sequence in which an N-terminal Q is substituted with pE. In another embodiment, the ABP comprises a heavy chain sequence in which an N-terminal E is substituted with pE. In another embodiment, the ABP comprises a light chain sequence in which an N-terminal E is substituted with pE.

[32] In one embodiment, the ABP specifically binds to human NRP-1 with a kD of less than 20nM, less than 10 nM, less than 5 nM, less than 2 nM, less than 1 nM, less than 0.5 nM, or less than 0.2 nM. In another embodiment, the ABP specifically binds to NRP-1 from humans, mice, and cynomolgus monkeys. In one embodiment, the ABP binds to a different epitope on NRP-1 than the epitope on NRP-1 to which SEC10 binds. In one embodiment, the ABP binds to the al, a2, b l, or b2 domain of NRP-1. In another embodiment, the ABP binds to more than one domain of NRP- 1. In another embodiment, the ABP binds to the b2 domain of NRP-1. In another embodiment, the ABP binds to the b l domain of NRP- 1.

[33] In another aspect is provided any of the ABPs disclosed herein for use as a medicament. In another embodiment, the ABP is provided for use in the treatment of a cancer or viral infection. In one embodiment, the cancer is selected from a solid tumor and a hematological tumor.

[34] In another aspect is provided a kit comprising any of the ABPs disclosed herein, and instructions for use of the ABP. In one embodiment, the kit comprises a lyophilized ABP. In another embodiment, the kit comprises a fluid for reconstitution of the lyophilized ABP.

[35] In another aspect is provided an isolated polynucleotide encoding an ABP disclosed herein, a

VH thereof, a VL thereof, a light chain thereof, a heavy chain thereof or an antigen-binding portion thereof.

[36] In another aspect is provided a vector comprising the isolated polynucleotide encoding an ABP disclosed herein, a VH thereof, a VL thereof, a light chain thereof, a heavy chain thereof or an antigen-binding portion thereof.

[37] In another aspect is provided a host cell comprising any of the vectors or polynucleotides disclosed herein. In one embodiment, the host cell is selected from a bacterial cell, a fungal cell, and a mammalian cell. In another embodiment, the host cell is selected from an E. coli cell, a

Saccharomyces cerevisiae cell, and a CHO cell.

[38] In another aspect is provided a cell-free expression reaction comprising any of the vectors or polynucleotides disclosed herein.

[39] In another aspect is provided a method of producing an ABP as disclosed herein, comprising expressing the ABP in the host cell disclosed herein and isolating the expressed ABP.

[40] In another aspect is provided a pharmaceutical composition comprising any of the ABPs disclosed herein and a pharmaceutically acceptable excipient. In one embodiment, the ABP is present in the composition in an amount effective to locally inhibit the NRP-l :semaphorin-4 interaction in a tumor. In one embodiment, the anti-NRP-1 antibody is present in the composition in an amount effective to inhibit an interaction between NRP- 1 and a transmembrane semaphorin polypeptide when administered to a human subject. In another embodiment, the anti-NRP-1 antibody specifically blocks NRP-1 binding to a transmembrane semaphorin polypeptide. In another embodiment, the anti- NRP-1 antibody blocks the interaction between a NRP-1 polypeptide and a vascular endothelial cell growth factor (VEGF) polypeptide. In another embodiment, the anti-NRP-1 antibody blocks binding of a semaphorin polypeptide. In one embodiment, the anti-NRPl antibody blocks SEMA3 binding. In another embodiment, the anti-NRP-1 antibody blocks SEMA4 binding. In another embodiment, the antibody blocks interaction between a NRP-1 polypeptide and SEMA3. In another embodiment, the antibody blocks interaction between a NRP-1 polypeptide and VEGF. In one embodiment, the antibody blocks a semaphorin polypeptide binding but does not block VEGF binding. In another embodiment, the anti-NRP-1 antibody is capable of inhibiting Treg suppression in the human subject. In another embodiment, the anti-NRP-1 antibody is capable of decreasing Treg survival and/or stability in the human subject. In one embodiment, the anti-NRP-1 antibody is present in the composition in an amount effective to locally inhibit the NRP-1 :semaphorin-4 interaction in a tumor. In another embodiment, the anti-NRP-1 antibody is present in the composition in an amount effective to prevent development of an undesired autoimmune and/or inflammatory manifestation. In one embodiment, human subject is suffering from a cancer. In one embodiment, the amount of the ABP in the pharmaceutical composition is sufficient to (a) reduce the suppression of effector T cells by regulatory T cells; (b) activate effector T cells; (c) reduce the number of regulatory T cells in a tissue or systemically; (d) induce or enhance proliferation of effector T cells; (e) inhibit the rate of tumor growth; (f) induce tumor regression; or (g) combinations thereof, in a subject.

[41] In one embodiment, the pharmaceutical composition is for use as a medicament. In one embodiment, the pharmaceutical composition is for use in the treatment of a cancer or a viral infection. In one embodiment, the pharmaceutical composition is for use in the treatment of a cancer, wherein the cancer is selected from brain, prostate, breast, colon, skin, and lung cancer. In one embodiment, the pharmaceutical composition comprises a pharmaceutically acceptable excipient. In one embodiment, the ABP in the pharmaceutical composition is sufficient to (a) reduce the suppression of effector T cells by regulatory T cells; (b) activate effector T cells; (c) reduce the number of regulatory T cells in a tissue or systemically; (d) induce or enhance proliferation of effector T cells; (e) inhibit the rate of tumor growth; (f) induce tumor regression; or (g) combinations thereof, in a subject.

[42] In another aspect is provided a method of inhibiting a function or decreasing stability of a regulatory T cell (Treg) in a subject, comprising exposing the Treg in vivo to an inhibitor of neuropilin-1 (NRP-l):semaphorin-4A axis in the Treg, wherein an effective amount of an ABP provided herein or a pharmaceutical composition provided herein is administered to the subject. In one embodiment, the method comprises increasing T effector cell (T eff ) function or exposing the T eff in vivo to an ABP provided herein, comprising administering to a subject an effective amount of a pharmaceutical composition provided herein. In one embodiment, the subject has a cancer. In one embodiment, the method induces or enhances an immune response to a cancer-associated antigen. In one embodiment, the ABP is capable of (a) decreasing Treg survival and/or stability in the human subject; (b) binding to an extracellular domain of the NRP- 1 polypeptide; or (c) a combination thereof.

[43] In one embodiment, the method further comprises administering one or more additional therapeutic agents. In one embodiment, the additional therapeutic agent is selected from radiation, a cytotoxic agent, a chemotherapeutic agent, a cytostatic agent, an anti-hormonal agent, a VEGF inhibitor, an immunostimulatory agent, an anti-angiogenic agent, and combinations thereof. In one embodiment, the additional therapeutic agent is an immunostimulatory agent. In one embodiment, the immunostimulatory agent comprises an agent that blocks signaling of an inhibitory receptor expressed by an immune cell or a ligand thereof. In one embodiment, the inhibitory receptor expressed by an immune cell or ligand thereof is selected from PVRIG, VISTA, CCR4, CD27, CTLA-4, PD- 1, PD- Ll, LAG-3, Tim3, TIGIT, neuritin, BTLA, KIR, and combinations thereof. In one embodiment, the immunostimulatory agent comprises an agonist to a stimulatory receptor expressed by an immune cell. In one embodiment, the stimulatory receptor expressed by an immune cell is selected from OX40, GITR, ICOS, CD28, CD37, CD40, 4- IBB, and combinations thereof. In one embodiment, the immunostimulatory agent comprises a cytokine. In another embodiment, the immunostimulatory agent comprises a vaccine to a cancer-associated antigen.

[44] In another aspect is provided a method of modulating an immune response in a subject in need thereof, comprising administering to the subject an effective amount of an ABP provided herein. In one embodiment, the method further comprises administering one or more additional therapeutic agents to the subject. In one embodiment, the additional therapeutic agent is (i) an agonist to a stimulatory receptor of an immune cell or (ii) an antagonist of an inhibitory receptor of an immune cell, wherein the receptor of an immune cell is selected from OX40, CD2, CD27, CDS, ICAM-1, LFA-1 (CD l la/CD 18), ICOS (CD278), 4- 1BB (CD 137), CD28, CD30, CD40, BAFFR, HVEM, CD7, LIGHT, NKG2C, GITR, SLAMF7, NKp80, CD 160, B7-H3, CD83 ligand, and combinations thereof. In another embodiment, the additional therapeutic agent is an oncolytic virus selected from herpes simplex virus, vesicular stomatitis virus, adenovirus, Newcastle disease virus, vaccinia virus, a maraba virus, and combinations thereof. In one embodiment, the additional therapeutic agent is formulated in the same pharmaceutical composition as the ABP. In another embodiment, the additional therapeutic agent is formulated in a different pharmaceutical composition from the ABP. [45] In one embodiment, the additional therapeutic agent is administered prior to administering the ABP. In another embodiment, the additional therapeutic agent is administered after administering the ABP. In another embodiment, the additional therapeutic agent is administered contemporaneously with the ABP. In one embodiment, the method does not substantially cause thrombocytopenia in the subject.

[46] In another aspect is provided an anti -human NRP- 1 antibody or an antigen-binding fragment thereof, comprising a heavy chain variable region comprising a CDR-H3 consisting of SEQ ID NO:47, a CDR-H2 consisting of SEQ ID NO:30, and a CDR-H1 consisting of SEQ ID NO: 14; and a light chain variable region comprising a CDR-L3 consisting of SEQ ID NO: 81, a CDR-L2 consisting of SEQ ID NO:71, and a CDR-L1 consisting of SEQ ID NO: 63. In one embodiment, the antibody or antigen-binding fragment is selected from any one of the following (1) and (2):

(1) an anti -human NRP-1 antibody or an antigen-binding fragment thereof, comprising a heavy chain variable region consisting of SEQ ID NO: 96, and a light chain variable region consisting of SEQ ID NO: 104; and

(2) an anti -human NRP-1 antibody or the antigen-binding fragment thereof, comprising a heavy chain variable region consisting of SEQ ID NO: 96 in which E of the amino acid number 1 is modified to pyroglutamate, and a light chain variable region consisting of SEQ ID NO: 104.

[47] In one embodiment is a method for producing an anti -human NRP-1 antibody or an antigen- binding fragment thereof, comprising culturing host cell(s) selected from the group consisting of (a) to (c) below to express a tetravalent anti -human NRP-1 antibody or an antigen-binding fragment thereof:

(a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP- 1 antibody or the antigen-binding fragment thereof of the above embodiment (1) and a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen-binding fragment thereof;

(b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP- 1 antibody or the antigen-binding fragment thereof of the above embodiment (1) and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen- binding fragment thereof; and

(c) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP- 1 antibody or the antigen-binding fragment thereof of claim the above embodiment (1) and a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen-binding fragment thereof.

[48] In another embodiment is provided (1) a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti -human NRP-1 antibody or the antigen-binding fragment thereof of the above aspect, and (2) a polynucleotide comprising a base sequence encoding the light chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof of the above aspect.

[49] In another embodiment is provided an expression vector comprising: (a) a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof of the above aspect, and/or (b) a polynucleotide comprising a base sequence encoding the light chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof of the above aspect.

[50] In another embodiment is provided a host cell transformed with an expression vector selected from the group consisting of (a) to (d):

(a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof of the above aspect, and a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen- binding fragment thereof;

(b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof of the above aspect and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen-binding fragment thereof of the above aspect;

(c) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof of the above aspect; and

(d) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the anti -human NRP-1 antibody or the antigen-binding fragment thereof of the above aspect.

[51] In another embodiment is provided an anti-human NRP-1 antibody or an antigen-binding fragment thereof according to the above aspect, which is selected from the group consisting of (1) to (1) an anti-human NRP-1 antibody comprising a heavy chain consisting of SEQ ID NO: 118, and a light chain consisting of SEQ ID NO: 126;

(2) an anti -human NRP-1 antibody comprising a heavy chain consisting of SEQ ID NO: 118 in which E of the amino acid number 1 is modified to pyroglutamate, and a light chain consisting of SEQ ID NO: 126;

(3) an anti -human NRP-1 antibody comprising a heavy chain consisting of the amino acid sequence of amino acid numbers 1 to 453 of SEQ ID NO: 118, and a light chain consisting of SEQ ID NO: 126; and

(4) an anti -human NRP-1 antibody comprising a heavy chain consisting of the amino acid sequence of amino acid numbers 1 to 453 of SEQ ID NO: 118 in which E of the amino acid number 1 is modified to pyroglutamate, and a light chain consisting of SEQ ID NO: 126.

[52] In one embodiment, the anti -human NRP-1 antibody is for use in preventing or treating cancer. In another embodiment, the anti -human NRP-1 antibody is for manufacture of a

pharmaceutical composition for preventing or treating cancer.

[53] A polynucleotide, which is selected from the group consisting of (1) and (2):

(1) a polynucleotide comprising a base sequence encoding the heavy chain of the anti- human NRP-1 antibody according to the above embodiment (1), and

(2) a polynucleotide comprising a base sequence encoding the light chain of the anti- human NRP-1 antibody according to the above embodiment (1).

[54] An expression vector comprising:

(a) a polynucleotide comprising a base sequence encoding the heavy chain of the anti-human NRP-1 antibody of the above embodiment (1), and/or

(b) a polynucleotide comprising a base sequence encoding the light chain of the anti-human NRP-1 antibody of the above embodiment (1).

[55] A host cell transformed with an expression vector selected from the group consisting of (a) to (d):

(a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of the above embodiment (1) and a polynucleotide comprising a base sequence encoding the light chain of the antibody;

(b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of the above embodiment (1) and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the antibody; (c) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of the above embodiment (1); and

(d) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the anti-human NRP-1 antibody of the above embodiment (1).

[56] A method for producing an anti -human NRP- 1 antibody, comprising culturing host cell(s) selected from the group consisting of (a) to (c) below to express an anti -human NRP-1 antibody:

(a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of the above embodiment (1) and a polynucleotide comprising a base sequence encoding the light chain of the antibody;

(b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of the above embodiment (1) and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the antibody; and

(c) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody of the above embodiment (1) and a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the antibody.

[57] In one embodiment is provided a pharmaceutical composition comprising the anti-human NRP-1 antibody of the above embodiment and a pharmaceutically acceptable excipient. In another embodiment is provided a pharmaceutical composition comprising the anti -human NRP-1 antibody of the above embodiment (1), the anti-human NRP-1 antibody of the above embodiment (2), the anti- human NRP-1 antibody of the above embodiment (3), and/or the anti-human NRP-1 antibody of the above embodiment (4), and a pharmaceutically acceptable excipient. In one embodiment the pharmaceutical composition is a pharmaceutical composition for treating cancer. In another embodiment, the composition is administered in combination with radiation, a cytotoxic agent, a chemotherapeutic agent, a cytostatic agent, an anti-hormonal agent, a VEGF inhibitor, an

immunostimulatory agent, an anti-angiogenic agent, or combinations thereof.

[58] In another embodiment is provided a method for preventing or treating cancer, comprising administering a therapeutically effective amount of the anti -human NRP-1 antibody of the above aspect. In one embodiment, the method further comprises administering one or more additional therapeutic agents. In one embodiment, the additional therapeutic agent is selected from the group consisting of radiation, a cytotoxic agent, a chemotherapeutic agent, a cytostatic agent, an anti- hormonal agent, a VEGF inhibitor, an immunostimulatory agent, an anti-angiogenic agent, and combinations thereof.

BRIEF DESCRIPTION OF THE DRAWINGS

[59] Figure 1A and IB are two graphs showing tumor growth inhibition in CT26 tumor-bearing mice treated with a murine version of MABs 2, 3, 4, 5, 7, 12, 13, 14, and 15, as well as an IgG control and the anti-NRP-1 antibody SEC 10 as a comparator. Mice were treated with MAB monotherapy (Figure 1A) or in combination with a PD-1 antibody (Figure IB). Antibody treatment times (days) are shown by arrows. Figure 1C is a graph showing tumor growth inhibition in CT26 tumor-bearing mice treated with monotherapy and combination therapy as described herein. Provided are: i) a murine version of MAB 12, ii) a PD-1 inhibitor, and iii) a combination of mMAB 12 and the PD-1 inhibitor. Antibody treatment times (days) are shown by arrows.

[60] Figure 2 is three graphs showing tumor growth inhibition in MC38 tumor-bearing mice treated with a murine version of MABs 2, 3, 4, 5, 7, 12, 13, 14, and 15, as well as an IgG control and SEC 10 as a comparator. Mice were treated with MAB monotherapy (Figure 2A) or in combination with a PD-L1 antibody (Figure 2B). Antibody treatment times (days) are shown by arrows. The antitumor efficacy of mMAB12 alone or in combination with PD-L1 antibody in the MC38 syngeneic colon mouse tumor model is shown in Figure 2C.

[61] Figure 3 is two graphs showing epitope binning data for the anti-NRP-1 antibodies MAB 12 and SEC 10. The top panel shows binning data for MAB 12 and SEC 10 with 5 μg/mL MAB 12 immobilized on anti-human Fc AHC sensors. The bottom panel shows binning data for MAB 12 and SEC 10 with 5 μg/mL SEC 10 immobilized on the sensors. NRPl protein is bound to the immobilized antibody and binding of the second antibody is evaluated. The traces show that MAB 12 and SEC 10 are able to simultaneously bind NRPl .

DETAILED DESCRIPTION

1. Definitions

[62] Unless otherwise defined, all terms of art, notations and other scientific terminology used herein are intended to have the meanings commonly understood by those of skill in the art to which this invention pertains. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not necessarily be construed to represent a difference over what is generally understood in the art. The techniques and procedures described or referenced herein are generally well understood and commonly employed using conventional methodologies by those skilled in the art, such as, for example, the widely utilized molecular cloning methodologies described in Sambrook et al., Molecular Cloning: A Laboratory Manual 4th ed. (2012) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY. As appropriate, procedures involving the use of commercially available kits and reagents are generally carried out in accordance with manufacturer-defined protocols and conditions unless otherwise noted. [63] As used herein, the singular forms "a," "an," and "the" include the plural referents unless the context clearly indicates otherwise. The terms "include," "such as," and the like are intended to convey inclusion without limitation, unless otherwise specifically indicated.

[64] As used herein, the term "comprising" also specifically includes embodiments "consisting of and "consisting essentially of the recited elements, unless specifically indicated otherwise

[65] The term "about" indicates and encompasses an indicated value and a range above and below that value. In certain embodiments, the term "about" indicates the designated value ± 10%, ± 5%, or ± 1%. In certain embodiments, where applicable, the term "about" indicates the designated value(s) ± one standard deviation of that value(s).

[66] The term "immunoglobulin" refers to a class of structurally related proteins generally comprising two pairs of polypeptide chains: one pair of light (L) chains and one pair of heavy (H) chains. In an "intact immunoglobulin," all four of these chains are interconnected by disulfide bonds. The structure of immunoglobulins has been well characterized. See, e.g., Paul, Fundamental

Immunology 7th ed., Ch. 5 (2013) Lippincott Williams & Wilkins, Philadelphia, PA. Briefly, each heavy chain typically comprises a heavy chain variable region (VH) and a heavy chain constant region (CH). The heavy chain constant region typically comprises three domains, abbreviated CHI, Cm, and CH3. Each light chain typically comprises a light chain variable region (VL) and a light chain constant region. The light chain constant region typically comprises one domain, abbreviated CL.

[67] The term "antigen-binding protein" (ABP) refers to a protein comprising one or more antigen- binding domains that specifically bind to an antigen or epitope. In some embodiments, the antigen- binding domain binds the antigen or epitope with specificity and affinity similar to that of naturally occurring antibodies. In some embodiments, the ABP comprises an antibody. In some embodiments, the ABP consists of an antibody. In some embodiments, the ABP consists essentially of an antibody. In some embodiments, the ABP comprises an alternative scaffold. In some embodiments, the ABP consists of an alternative scaffold. In some embodiments, the ABP consists essentially of an alternative scaffold. In some embodiments, the ABP comprises an antibody fragment. In some embodiments, the ABP consists of an antibody fragment. In some embodiments, the ABP consists essentially of an antibody fragment. A "NRP-1 ABP," "anti-NRP-1 ABP," or "NRP-1 -specific ABP" is an ABP, as provided herein, which specifically binds to the antigen NRP- 1. In some embodiments, the ABP binds the extracellular domain of NRP- 1. In certain embodiments, a NRP- 1 ABP provided herein binds to an epitope of NRP-1 that is conserved between or among NRP-1 proteins from different species.

[68] The term "antibody" is used herein in its broadest sense and includes certain types of immunoglobulin molecules comprising one or more antigen-binding domains that specifically bind to an antigen or epitope. An antibody specifically includes intact antibodies (e.g., intact

immunoglobulins), antibody fragments, and multi-specific antibodies. An antibody is one type of ABP. [69] The term "antigen-binding domain" means the portion of an ABP that is capable of specifically binding to an antigen or epitope. One example of an antigen-binding domain is an antigen-binding domain formed by a VH -VL dimer of an antibody. Another example of an antigen- binding domain is an antigen-binding domain formed by diversification of certain loops from the tenth fibronectin type III domain of an adnectin.

[70] The terms "full length antibody," "intact antibody," and "whole antibody" are used herein interchangeably to refer to an antibody having a structure substantially similar to a naturally occurring antibody structure and having heavy chains that comprise an Fc region. For example, when used to refer to an IgG molecule, a "full length antibody" is an antibody that comprises two heavy chains and two light chains. An "anti -human NRP-1 antibody" is the intact antibody, as provided herein, which specifically binds to the human NRP- 1.

[71] The term "Fc region" means the C-terminal region of an immunoglobulin heavy chain that, in naturally occurring antibodies, interacts with Fc receptors and certain proteins of the complement system. The structures of the Fc regions of various immunoglobulins, and the glycosylation sites contained therein, are known in the art. See Schroeder and Cavacini, J. Allergy Clin. Immunol, 2010, 125:S41-52, incorporated by reference in its entirety. The Fc region may be a naturally occurring Fc region, or an Fc region modified as described in the art or elsewhere in this disclosure.

[72] The VH and VL regions may be further subdivided into regions of hypervariability

("hypervariable regions (HVRs);" also called "complementarity determining regions" (CDRs)) interspersed with regions that are more conserved. The more conserved regions are called framework regions (FRs). Each VH and VL generally comprises three CDRs and four FRs, arranged in the following order (from N-terminus to C-terminus): FR1 - CDR1 - FR2 - CDR2 - FR3 - CDR3 - FR4. The CDRs are involved in antigen binding, and influence antigen specificity and binding affinity of the antibody. See Kabat et al., Sequences of Proteins of Immunological Interest 5th ed. (1991) Public Health Service, National Institutes of Health, Bethesda, MD, incorporated by reference in its entirety.

[73] The light chain from any vertebrate species can be assigned to one of two types, called kappa (K) and lambda (λ), based on the sequence of its constant domain.

[74] The heavy chain from any vertebrate species can be assigned to one of five different classes (or isotypes): IgA, IgD, IgE, IgG, and IgM. These classes are also designated α, δ, ε, γ, and μ, respectively. The IgG and IgA classes are further divided into subclasses on the basis of differences in sequence and function. Humans express the following subclasses: IgGl, IgG2, IgG3, IgG4, IgAl, and IgA2.

[75] The amino acid sequence boundaries of a CDR can be determined by one of skill in the art using any of a number of known numbering schemes, including those described by Kabat et al., supra ("Kabat" numbering scheme); Al-Lazikani et al., 1997, J. Mol. Biol, 273:927-948 ("Chothia" numbering scheme); MacCallum et al., 1996, J. Mol. Biol. 262:732-745 ("Contact" numbering scheme); Lefranc et al., Dev. Comp. Immunol, 2003, 27:55-77 ("EVIGT" numbering scheme); and Honegge and Pliickthun, J. Mol. Biol, 2001, 309:657-70 ("AHo" numbering scheme); each of which is incorporated by reference in its entirety.

[76] Table 1 provides the positions of CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2, and CDR- H3 as identified by the Kabat and Chothia schemes. For CDR-H1, residue numbering is provided using both the Kabat and Chothia numbering schemes.

[77] CDRs may be assigned, for example, using antibody numbering software, such as Abnum, available at www.bioinf.org.uk/abs/abnum/, and described in Abhinandan and Martin, Immunology, 2008, 45:3832-3839, incorporated by reference in its entirety.

Table 1. Residues in CDRs according to Kabat and Chothia numbering schemes.

* The C-terminus of CDR-H1, when numbered using the Kabat numbering convention, varies between H32 and H34, depending on the length of the CDR.

[78] The "EU numbering scheme" is generally used when referring to a residue in an antibody heavy chain constant region (e.g., as reported in Kabat et al, supra). Unless stated otherwise, the EU numbering scheme is used to refer to residues in antibody heavy chain constant regions described herein.

[79] An "antibody fragment" or an "antigen-binding fragment" comprises a portion of an intact antibody, such as the antigen-binding or variable region of an intact antibody. Antibody fragments include, for example, Fv fragments, Fab fragments, F(ab')2 fragments, Fab' fragments, scFv (sFv) fragments, and scFv-Fc fragments.

[80] "Fv" fragments comprise a non-covalently-linked dimer of one heavy chain variable domain and one light chain variable domain.

[81] "Fab" fragments comprise, in addition to the heavy and light chain variable domains, the constant domain of the light chain and the first constant domain (CHI) of the heavy chain. Fab fragments may be generated, for example, by recombinant methods or by papain digestion of a full- length antibody.

[82] "F(ab')2" fragments contain two Fab' fragments joined, near the hinge region, by disulfide bonds. F(ab')2 fragments may be generated, for example, by recombinant methods or by pepsin digestion of an intact antibody. The F(ab') fragments can be dissociated, for example, by treatment with β-mercaptoethanol.

[83] "Single-chain Fv" or "sFv" or "scFv" antibody fragments comprise a VH domain and a VL domain in a single polypeptide chain. The VH and VL are generally linked by a peptide linker. See Pluckthun A. (1994). Any suitable linker may be used. In some embodiments, the linker is a

(GGGGS)n (SEQ ID NO: 140). In some embodiments, n = 1, 2, 3, 4, 5, or 6. See Antibodies from Escherichia coli. In Rosenberg M. & Moore G.P. (Eds.), The Pharmacology of Monoclonal

Antibodies vol. 113 (pp. 269-315). Springer- Verlag, New York, incorporated by reference in its entirety.

[84] "scFv-Fc" fragments comprise an scFv attached to an Fc domain. For example, an Fc domain may be attached to the C-terminal of the scFv. The Fc domain may follow the VH or VL, depending on the orientation of the variable domains in the scFv (i.e., VH -VL or VL -VH ). Any suitable Fc domain known in the art or described herein may be used. In some cases, the Fc domain comprises an IgG4 Fc domain.

[85] The term "single domain antibody" refers to a molecule in which one variable domain of an antibody specifically binds to an antigen without the presence of the other variable domain. Single domain antibodies, and fragments thereof, are described in Arabi Ghahroudi et al., FEBS Letters, 1998, 414:521-526 and Muyldermans et al., Trends in Biochem. Set , 2001, 26:230-245, each of which is incorporated by reference in its entirety. Single domain antibodies are also known as sdAbs or nanobodies.

[86] The term "monoclonal antibody" refers to an antibody from a population of substantially homogeneous antibodies. A population of substantially homogeneous antibodies comprises antibodies that are substantially similar and that bind the same epitope(s), except for variants that may normally arise during production of the monoclonal antibody. Such variants are generally present in only minor amounts. A monoclonal antibody is typically obtained by a process that includes the selection of a single antibody from a plurality of antibodies. For example, the selection process can be the selection of a unique clone from a plurality of clones, such as a pool of hybridoma clones, phage clones, yeast clones, bacterial clones, or other recombinant DNA clones. The selected antibody can be further altered, for example, to improve affinity for the target ("affinity maturation"), to humanize the antibody, to improve its production in cell culture, and/or to reduce its immunogenicity in a subject.

[87] The term "chimeric antibody" refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.

[88] "Humanized" forms of non-human antibodies are chimeric antibodies that contain minimal sequence derived from the non-human antibody. A humanized antibody is generally a human antibody (recipient antibody) in which residues from one or more CDRs are replaced by residues from one or more CDRs of a non-human antibody (donor antibody). The donor antibody can be any suitable non-human antibody, such as a mouse, rat, rabbit, chicken, or non-human primate antibody having a desired specificity, affinity, or biological effect. In some instances, selected framework region residues of the recipient antibody are replaced by the corresponding framework region residues from the donor antibody. Humanized antibodies may also comprise residues that are not found in either the recipient antibody or the donor antibody. Such modifications may be made to further refine antibody function. For further details, see Jones et al., Nature, 1986, 321 :522-525; Riechmann et al., Nature, 1988, 332:323-329; and Presta, Curr. Op. Struct. Biol, 1992, 2:593-596, each of which is incorporated by reference in its entirety.

[89] A "human antibody" is one which possesses an amino acid sequence corresponding to that of an antibody produced by a human or a human cell, or derived from a non-human source that utilizes a human antibody repertoire or human antibody-encoding sequences (e.g., obtained from human sources or designed de novo). Human antibodies specifically exclude humanized antibodies.

[90] By "SEC 10" is meant an anti-NRP-1 antibody previously in clinical trials for treatment of solid tumors, with and without bevacizumab. See, e.g., "A Study of MNRP1685A in Patients with

Locally Advanced or Metastatic Solid Tumors," clinicaltrials.gov Identifier NCT00747734.

[91] By "SEC3" is meant the pan-anti-NRP-1 antibody set forth in SEQ ID NO: 144, also described, e.g., in Appleton, et. al, The EMBO Journal (2007) 26, 4902-4912.

[92] By "MAB59941" is meant an anti-mouse Neuropilin-1 antibody available from R&D

Systems, Clone # 761704.

[93] An "isolated ABP" or "isolated nucleic acid" is an ABP or nucleic acid that has been separated and/or recovered from a component of its natural environment. Components of the natural environment may include enzymes, hormones, and other proteinaceous or nonproteinaceous materials. In some embodiments, an isolated ABP is purified to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence, for example by use of a spinning cup sequenator. In some embodiments, an isolated ABP is purified to homogeneity by gel electrophoresis (e.g., SDS- PAGE) under reducing or nonreducing conditions, with detection by Coomassie blue or silver stain. In some embodiments, an isolated ABP may include an ABP in situ within recombinant cells, since at least one component of the ABP's natural environment is not present. In some aspects, an isolated ABP or isolated nucleic acid is prepared by at least one purification step. In some embodiments, an isolated ABP or isolated nucleic acid is purified to at least 80%, 85%, 90%, 95%, or 99% by weight. In some embodiments, an isolated ABP or isolated nucleic acid is purified to at least 80%, 85%, 90%, 95%, or 99% by volume. In some embodiments, an isolated ABP or isolated nucleic acid is provided as a solution comprising at least 85%, 90%, 95%, 98%, 99% to 100% ABP or nucleic acid by weight. In some embodiments, an isolated ABP or isolated nucleic acid is provided as a solution comprising at least 85%, 90%, 95%, 98%, 99% to 100% ABP or nucleic acid by volume.

[94] "Affinity" refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., an ABP) and its binding partner (e.g., an antigen or epitope). Unless indicated otherwise, as used herein, "affinity" refers to intrinsic binding affinity, which reflects a 1 : 1 interaction between members of a binding pair (e.g., ABP and antigen or epitope). The affinity of a molecule X for its partner Y can be represented by the dissociation equilibrium constant (KD). The kinetic components that contribute to the dissociation equilibrium constant are described in more detail below. Affinity can be measured by common methods known in the art, including those described herein, such as surface plasmon resonance (SPR) technology (e.g., BIACORE ® ) or biolayer interferometry (e.g., FORTEBIO ® ).

[95] With regard to the binding of an ABP to a target molecule, the terms "bind," "specific binding," "specifically binds to," "specific for," "selectively binds," and "selective for" a particular antigen (e.g., a polypeptide target) or an epitope on a particular antigen mean binding that is measurably different from a non-specific or non-selective interaction (e.g., with a non-target molecule). Specific binding can be measured, for example, by measuring binding to a target molecule and comparing it to binding to a non-target molecule. Specific binding can also be determined by competition with a control molecule that mimics the epitope recognized on the target molecule. In that case, specific binding is indicated if the binding of the ABP to the target molecule is competitively inhibited by the control molecule. In some aspects, the affinity of a NRP-1 ABP for a non-target molecule is less than about 50% of the affinity for NRP-1. In some aspects, the affinity of a NRP-1 ABP for a non-target molecule is less than about 40% of the affinity for NRP- 1. In some aspects, the affinity of a NRP- 1 ABP for a non-target molecule is less than about 30% of the affinity for NRP- 1. In some aspects, the affinity of a NRP- 1 ABP for a non-target molecule is less than about 20% of the affinity for NRP-1. In some aspects, the affinity of a NRP-1 ABP for a non-target molecule is less than about 10% of the affinity for NRP-1. In some aspects, the affinity of a NRP-1 ABP for a non- target molecule is less than about 1% of the affinity for NRP- 1. In some aspects, the affinity of a NRP-1 ABP for a non-target molecule is less than about 0.1% of the affinity for NRP-1.

[96] The term '¾" (sec 1 ), as used herein, refers to the dissociation rate constant of a particular ABP- antigen interaction. This value is also referred to as the k 0 ff value.

[97] The term "k a " (M^xsec "1 ), as used herein, refers to the association rate constant of a particular ABP-antigen interaction. This value is also referred to as the k on value.

[98] The term "KD" (M), as used herein, refers to the dissociation equilibrium constant of a particular ABP-antigen interaction. KD = ka/k a . In some embodiments, the affinity of an ABP is described in terms of the KD for an interaction between such ABP and its antigen. For clarity, as known in the art, a smaller KD value indicates a higher affinity interaction, while a larger KD value indicates a lower affinity interaction.

[99] The term "K A " (M 1 ), as used herein, refers to the association equilibrium constant of a particular ABP-antigen interaction. KA = k a /kd.

[100] An "affinity matured" ABP is an ABP with one or more alterations (e.g., in one or more CDRs or FRs) relative to a parent ABP (i.e., an ABP from which the altered ABP is derived or designed) that result in an improvement in the affinity of the ABP for its antigen, compared to the parent ABP which does not possess the alteration(s). In some embodiments, an affinity matured ABP has nanomolar or picomolar affinity for the target antigen. Affinity matured ABPs may be produced using a variety of methods known in the art. For example, Marks et al. (Bio/Technology, 1992, 10:779-783, incorporated by reference in its entirety) describes affinity maturation by VH and VL domain shuffling. Random mutagenesis of CDR and/or framework residues is described by, for example, Barbas et al. (Proc. Nat. Acad. Sci. U.S.A. , 1994, 91:3809-3813); Schier et al., Gene, 1995, 169: 147-155; Yelton et al, J. Immunol , 1995, 155: 1994-2004; Jackson et al., J. Immunol , 1995, 154:3310-33199; and Hawkins et al, J. Mol. Biol , 1992, 226:889-896; each of which is incorporated by reference in its entirety.

[101] An "immunoconjugate" is an ABP conjugated to one or more heterologous molecule(s), such as a therapeutic or diagnostic agent.

[102] "Effector functions" refer to those biological activities mediated by the Fc region of an antibody, which activities may vary depending on the antibody isotype. Examples of antibody effector functions include Clq binding to activate complement dependent cytotoxicity (CDC), Fc receptor binding to activate antibody-dependent cellular cytotoxicity (ADCC), and antibody dependent cellular phagocytosis (ADCP).

[103] When used herein in the context of two or more ABPs, the term "competes with" or "cross- competes with" indicates that the two or more ABPs compete for binding to an antigen (e.g., NRP-1). In one exemplary assay, NRP-1 is coated on a surface and contacted with a first NRP-1 ABP, after which a second NRP-1 ABP is added. In another exemplary assay, a first NRP-1 ABP is coated on a surface and contacted with NRP-1, and then a second NRP-1 ABP is added. If the presence of the first NRP-1 ABP reduces binding of the second NRP-1 ABP, in either assay, then the ABPs compete with each other. The term "competes with" also includes combinations of ABPs where one ABP reduces binding of another ABP, but where no competition is observed when the ABPs are added in the reverse order. However, in some embodiments, the first and second ABPs inhibit binding of each other, regardless of the order in which they are added. In some embodiments, one ABP reduces binding of another ABP to its antigen by at least 25%, at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, or at least 95%. A skilled artisan can select the concentrations of the antibodies used in the competition assays based on the affinities of the ABPs for NRP-1 and the valency of the ABPs. The assays described in this definition are illustrative, and a skilled artisan can utilize any suitable assay to determine if antibodies compete with each other. Suitable assays are described, for example, in Cox et al., "Immunoassay Methods," in Assay Guidance Manual [Internet], Updated December 24, 2014 (www.ncbi.nlm.nih.gov/books/NBK92434/; accessed September 29, 2015); Silman et al., Cytometry, 2001, 44:30-37; and Finco et al., J. Pharm. Biomed. Anal , 2011, 54:351-358; each of which is incorporated by reference in its entirety.

[104] The term "epitope" means a portion of an antigen that specifically binds to an ABP. Epitopes frequently consist of surface-accessible amino acid residues and/or sugar side chains and may have specific three-dimensional structural characteristics, as well as specific charge characteristics.

Conformational and non-conformational epitopes are distinguished in that the binding to the former but not the latter may be lost in the presence of denaturing solvents. An epitope may comprise amino acid residues that are directly involved in the binding, and other amino acid residues, which are not directly involved in the binding. The epitope to which an ABP binds can be determined using known techniques for epitope determination such as, for example, testing for ABP binding to NRP-1 variants with different point-mutations, or to chimeric NRP-1 variants.

[105] Percent "identity" between a polypeptide sequence and a reference sequence, is defined as the percentage of amino acid residues in the polypeptide sequence that are identical to the amino acid residues in the reference sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN,

MEGALIGN (DNASTAR), CLUSTALW, CLUSTAL OMEGA, or MUSCLE software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.

[106] A "conservative substitution" or a "conservative amino acid substitution," refers to the substitution an amino acid with a chemically or functionally similar amino acid. Conservative substitution tables providing similar amino acids are well known in the art. By way of example, the groups of amino acids provided in Tables 2-4 are, in some embodiments, considered conservative substitutions for one another.

Table 2. Selected groups of amino acids that are considered conservative substitutions for one another, in certain embodiments.

Table 3. Additional selected groups of amino acids that are considered conservative substitutions for one another, in certain embodiments.

Table 4. Further selected groups of amino acids that are considered conservative substitutions for one another, in certain embodiments.

YJroup A Group B D and E

Group C and Q

Group D ¾ K, and H

Group E

Group F F, Y, and W

Group G S and T

Group H C and M

[107] Additional conservative substitutions may be found, for example, in Creighton, Proteins: Structures and Molecular Properties 2nd ed. (1993) W. H. Freeman & Co., New York, NY. An ABP generated by making one or more conservative substitutions of amino acid residues in a parent ABP is referred to as a "conservatively modified variant."

[108] The term "amino acid" refers to the twenty common naturally occurring amino acids.

Naturally occurring amino acids include alanine (Ala; A), arginine (Arg; R), asparagine (Asn; N), aspartic acid (Asp; D), cysteine (Cys; C); glutamic acid (Glu; E), glutamine (Gin; Q), Glycine (Gly; G); histidine (His; H), isoleucine (lie; I), leucine (Leu; L), lysine (Lys; K), methionine (Met; M), phenylalanine (Phe; F), proline (Pro; P), serine (Ser; S), threonine (Thr; T), tryptophan (Trp; W), tyrosine (Tyr; Y), and valine (Val; V).

[109] The term "vector," as used herein, refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. The term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors."

[110] The terms "host cell," "host cell line," and "host cell culture" are used interchangeably and refer to cells into which an exogenous nucleic acid has been introduced, and the progeny of such cells. Host cells include "transformants" (or "transformed cells") and "transfectants" (or "transfected cells"), which each include the primary transformed or transfected cell and progeny derived therefrom. Such progeny may not be completely identical in nucleic acid content to a parent cell, and may contain mutations.

[Ill] The term "treating" (and variations thereof such as "treat" or "treatment") refers to clinical intervention in an attempt to alter the natural course of a disease or condition in a subject in need thereof. Treatment can be performed both for prophylaxis and during the course of clinical pathology. Desirable effects of treatment include preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.

[112] As used herein, the term "therapeutically effective amount" or "effective amount" refers to an amount of an ABP or pharmaceutical composition provided herein that, when administered to a subject, is effective to treat a disease or disorder. [113] As used herein, the term "subject" means a mammalian subject. Exemplary subjects include humans, monkeys, dogs, cats, mice, rats, cows, horses, camels, goats, rabbits, and sheep. In certain embodiments, the subject is a human. In some embodiments, the subject has a disease or condition that can be treated with an ABP provided herein. In some aspects, the disease or condition is a cancer. In some aspects, the disease or condition is a viral infection.

[114] The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic or diagnostic products (e.g., kits) that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic or diagnostic products.

[115] The term "cytotoxic agent," as used herein, refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction.

[116] A "chemotherapeutic agent" refers to a chemical compound useful in the treatment of cancer. Chemotherapeutic agents include "anti-hormonal agents" or "endocrine therapeutics" which act to regulate, reduce, block, or inhibit the effects of hormones that can promote the growth of cancer.

[117] The term "cytostatic agent" refers to a compound or composition which arrests growth of a cell either in vitro or in vivo. In some embodiments, a cytostatic agent is an agent that reduces the percentage of cells in S phase. In some embodiments, a cytostatic agent reduces the percentage of cells in S phase by at least about 20%, at least about 40%, at least about 60%, or at least about 80%.

[118] The term "tumor" refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues. The terms "cancer," "cancerous," "cell proliferative disorder," "proliferative disorder" and "tumor" are not mutually exclusive as referred to herein. The terms "cell proliferative disorder" and "proliferative disorder" refer to disorders that are associated with some degree of abnormal cell proliferation. In some embodiments, the cell proliferative disorder is a cancer. In some aspects, the tumor is a solid tumor. In some aspects, the tumor is a hematologic malignancy.

[119] The term "pharmaceutical composition" refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective in treating a subject, and which contains no additional components which are unacceptably toxic to the subject in the amounts provided in the pharmaceutical composition.

[120] The terms "modulate" and "modulation" refer to reducing or inhibiting or, alternatively, activating or increasing, a recited variable.

[121] The terms "increase" and "activate" refer to an increase of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100- fold, or greater in a recited variable.

[122] The terms "reduce" and "inhibit" refer to a decrease of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold, or greater in a recited variable. [123] The term "agonize" refers to the activation of receptor signaling to induce a biological response associated with activation of the receptor. An "agonist" is an entity that binds to and agonizes a receptor.

[124] The term "antagonize" refers to the inhibition of receptor signaling to inhibit a biological response associated with activation of the receptor. An "antagonist" is an entity that binds to and antagonizes a receptor. An antagonist in one embodiment blocks 100% of binding of a ligand to its receptor; in other embodiments, an antagonist may reduce binding by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% of binding of a ligand to its receptor.

[125] The term "a semaphorin molecule" as used herein in connection with agonists of the NRP- l :semaphorin axis of Tregs encompasses transmembrane semaphorin molecules involved in interaction with NRP-1 on Tregs (e.g., Sema3a, Sema4a), various surface- and bead-immobilized versions of such molecules, as well as multimers, derivatives, mutants, analogs, and fragments of such molecules which can be used to enhance a function or increase stability of Tregs. Non-limiting examples of such agonist semaphorin molecules include, for example, IgM-derived semaphorin fusion proteins that assemble multimeric complexes incapable of fixing complement, that crosslink NRP-1.

[126] The term "neuropilin-1 (NRP-1): semaphorin axis of a regulatory T cell (Treg)" as used herein refers to the signaling pathway initiated by semaphorin (e.g., a semaphorin expressed by a cell such as, e.g., a conventional T cell, or a recombinant semaphorin), ligation of NRP-1, and the subsequent downstream signaling.

[127] The term "effector T cell" includes T helper (i.e., CD4+) cells and cytotoxic (i.e., CD8+) T cells. CD4+ effector T cells contribute to the development of several immunologic processes, including maturation of B cells into plasma cells and memory B cells, and activation of cytotoxic T cells and macrophages. CD8+ effector T cells destroy virus-infected cells and tumor cells. See Seder and Ahmed, Nature Immunol , 2003, 4:835-842, incorporated by reference in its entirety, for additional information on effector T cells.

[128] The term "regulatory T cell" includes cells that regulate immunological tolerance, for example, by suppressing effector T cells. In some aspects, the regulatory T cell has a

CD4+CD25+Foxp3+ phenotype. In some aspects, the regulatory T cell has a CD8+CD25+ phenotype. See Nocentini et al., Br. J. Pharmacol , 2012, 165:2089-2099, incorporated by reference in its entirety, for additional information on regulatory T cells expressing NRP-1.

[129] The term "dendritic cell" refers to a professional antigen-presenting cell capable of activating a naive T cell and stimulating growth and differentiation of a B cell.

2. NRP-1 Antigen-Binding Proteins

2.1 NRP-1 Binding and Target Cells

[130] Provided herein are ABPs that specifically bind to NRP-1. In some aspects, the NRP-1 is hNRP-1 (SEQ ID NO: 130). In some aspects, the NRP- 1 is cNRP-1 (SEQ ID NO: 132). In some aspects, the NRP-1 is mNRP- 1 with the sequence provided in SEQ ID NO: 134. In some aspects, the NRP-1 is rNRP- 1 with the sequence provided in SEQ ID NO: 135.

[131] In some embodiments, the ABPs provided herein specifically bind to the extracellular domain of NRP-1.

[132] In some embodiments, the ABPs provided herein specifically bind to the extracellular domain of NRP-1 and the extracellular domain of PD-1, PD-L1, or PD-L2, i.e., are bispecific antibodies.

[133] In some embodiments, an ABP provided herein is an antibody. In some embodiments, an ABP provided herein is an antibody fragment. In some embodiments, an ABP provided herein is an alternative scaffold.

[134] The NRP-1 may be expressed on the surface of any suitable target cell. In some embodiments, the target cell is a T cell. In some embodiments, the target cell is an effector T cell. In some embodiments, the target cell is a regulatory T cell. In some embodiments, the target cell is a natural killer (NK) cell. In some embodiments, the target cell is a natural killer T (NKT) cell. In some embodiments, the target cell is a macrophage. In other embodiments, the target cell is a dendritic cell. In one embodiment, the dendritic cell is a plasmacytoid dendritic cell.

[135] In some embodiments, the NRP-1 is associated with another receptor on the surface of the cell. In some embodiments, the NRP-1 is part of a co-receptor complex. In one embodiment, the NRP-1 is associated with a plexin. In some embodiments, the NRP- 1 is associated with a VEGF receptor.

[136] In some embodiments, the ABPs provided herein comprise an immunoglobulin molecule. In some embodiments, the ABPs provided herein consist of an immunoglobulin molecule. In some embodiments, the ABPs provided herein consist essentially of an immunoglobulin molecule. In some aspects, the immunoglobulin molecule comprises an antibody. In some aspects, the immunoglobulin molecule consists of an antibody. In some aspects, the immunoglobulin molecule consists essentially of an antibody.

[137] In some embodiments, the ABPs provided herein comprise a light chain. In some aspects, the light chain is a kappa light chain. In some aspects, the light chain is a lambda light chain.

[138] In some embodiments, the ABPs provided herein comprise a heavy chain. In some aspects, the heavy chain is an IgA. In some aspects, the heavy chain is an IgD. In some aspects, the heavy chain is an IgE. In some aspects, the heavy chain is an IgG. In some aspects, the heavy chain is an

IgM. In some aspects, the heavy chain is an IgGl . In some aspects, the heavy chain is an IgG2. In some aspects, the heavy chain is an IgG3. In some aspects, the heavy chain is an IgG4. In some aspects, the heavy chain is an IgAl . In some aspects, the heavy chain is an IgA2.

[139] In some embodiments, the ABPs provided herein comprise an antibody fragment. In some embodiments, the ABPs provided herein consist of an antibody fragment. In some embodiments, the

ABPs provided herein consist essentially of an antibody fragment. In some aspects, the antibody fragment is an Fv fragment. In some aspects, the antibody fragment is a Fab fragment. In some aspects, the antibody fragment is a F(ab')2 fragment. In some aspects, the antibody fragment is a Fab' fragment. In some aspects, the antibody fragment is an scFv (sFv) fragment. In some aspects, the antibody fragment is an scFv-Fc fragment. In some aspects, the antibody fragment is a fragment of a single domain antibody.

[140] In some embodiments, an antibody fragment provided herein is derived from an illustrative antibody provided herein. In some embodiments, an antibody fragments provided herein is not derived from an illustrative antibody provided herein and may, for example, be isolated de novo according to the methods provided herein for obtaining antibody fragments.

[141] In some embodiments, an antibody fragment provided specifically binds hNRP-1. In some embodiments, an antibody fragment provided herein specifically binds cNRP-1. In some

embodiments, an antibody fragment provided herein specifically binds mNRP-1. In some

embodiments, an antibody fragment provided herein specifically binds hNRP-1 and cNRP-1. In some embodiments, an antibody fragment provided herein specifically binds hNRP-1 and mNRP-1. In some embodiments, an antibody fragment provided herein specifically binds cNRP-1 and mNRP-1. In some embodiments, an antibody fragment provided herein specifically binds hNRP-1, cNRP-1 and mNRP-1.

[142] In some embodiments, an antibody fragment provided herein retains the ability to antagonize NRP-1, as measured by one or more assays or biological effects described herein. In some embodiments, an antibody fragment provided herein retains the ability to prevent NRP-1 from interacting with one or more of its ligands, as described herein.

[143] In some embodiments, an antibody fragment provided herein competes for binding to NRP-1 with an antibody selected from MAB1, MAB2, MAB3, MAB4, MAB5, MAB6, MAB7, MAB8, MAB9, MAB 10, MAB 11, MAB 12, MAB 13, MAB14, or MAB15, each as provided in Appendix A of this disclosure.

[144] In some embodiments, the ABPs provided herein are specific for cell surface NRP-1.

[145] In some embodiments, the ABPs provided herein are specifically block NRP-1 binding to a transmembrane semaphorin polypeptide.

[146] In some embodiments, the ABPs provided herein block the interaction between a NRP-1 polypeptide and a vascular endothelial cell growth factor (VEGF) polypeptide. In one embodiment, the VEGF polypeptide is VEGFA.

[147] In some embodiments, the anti-NRP-1 antibody blocks SEMA3 binding.

[148] In some embodiments, the anti-NRP-1 antibody blocks SEMA4 binding.

[149] In some embodiments, the antibody blocks interaction between a NRP-1 polypeptide and

SEMA3.

[150] In some embodiments, the antibody blocks interaction between a NRP-1 polypeptide and VEGF. [151] In some embodiments, the ABPs provided herein are capable of inhibiting Treg suppression in a human subject.

[152] In some embodiments, the ABPs provided herein co-stimulate an effector T cell in combination with antigen presentation from an antigen-presenting cell.

[153] In some embodiments, the ABPs provided herein inhibit the suppression of an effector T cell by a regulatory T cell.

[154] In some embodiments, the ABPs provided herein reduce the number of effector T cells in a tissue or in systemic circulation.

[155] In some embodiments, a fragment of an antibody provided herein binds the same epitope of NRP-1 as such antibody.

[156] In some embodiments, the ABPs provided herein are monoclonal antibodies. In some embodiments, the ABPs provided herein are polyclonal antibodies.

[157] In some embodiments, the ABPs provided herein comprise a chimeric antibody. In some embodiments, the ABPs provided herein consist of a chimeric antibody. In some embodiments, the ABPs provided herein consist essentially of a chimeric antibody. In some embodiments, the ABPs provided herein comprise a humanized antibody. In some embodiments, the ABPs provided herein consist of a humanized antibody. In some embodiments, the ABPs provided herein consist essentially of a humanized antibody. In some embodiments, the ABPs provided herein comprise a human antibody. In some embodiments, the ABPs provided herein consist of a human antibody. In some embodiments, the ABPs provided herein consist essentially of a human antibody.

[158] In some embodiments, the ABPs provided herein are affinity matured. In some aspects, the affinity matured ABPs are affinity matured ABPs derived from an illustrative ABP provided herein.

[159] In some embodiments, the ABPs provided herein comprise an alternative scaffold. In some embodiments, the ABPs provided herein consist of an alternative scaffold. In some embodiments, the ABPs provided herein consist essentially of an alternative scaffold. Any suitable alternative scaffold may be used. In some aspects, the alternative scaffold is selected from an Adnectin™, an iMab, an Anticalin ® , an EETI-II/AGRP, a Kunitz domain, a thioredoxin peptide aptamer, an Affibody ® , a DARPin, an Affilin, a Tetranectin, a Fynomer, and an Avimer.

[160] In some embodiments, an ABP provided herein specifically blocks binding of NRP-1 to a transmembrane semaphorin polypeptide. In some aspects, the ABP inhibits binding of NRP-1 to a transmembrane semaphorin polypeptide by at least about 50%. In some aspects, the ABP inhibits binding of NRP-1 to a transmembrane semaphorin polypeptide by at least about 75%. In some aspects, the ABP inhibits binding of NRP-1 to a transmembrane semaphorin polypeptide by at least about 90%. In some aspects, the ABP inhibits binding of NRP-1 to a transmembrane semaphorin polypeptide by at least about 95%. In some embodiments, the semaphorin polypeptide is a SEMA3 polypeptide. In other embodiments, the semaphorin polypeptide is a SEMA4 polypeptide.

[161] In some embodiments, an ABP of the invention is an ABP that competes with an illustrative ABP provided herein. In some aspects, the ABP that competes with the illustrative ABP provided herein binds the same epitope as an illustrative ABP provided herein.

[162] It is known that when an antibody is expressed in cells, the antibody is modified after translation. Examples of the posttranslational modification include cleavage of lysine at the C terminal of the heavy chain by a carboxypeptidase; modification of glutamine or glutamic acid at the N terminal of the heavy chain and the light chain to pyroglutamic acid by pyroglutamylation;

glycosylation; oxidation; deamidation; and glycation, and it is known that such posttranslational modifications occur in various antibodies (See Journal of Pharmaceutical Sciences, 2008, Vol. 97, p. 2426-2447, incorporated by reference in its entirety). In some embodiments, an ABP of the invention is an antibody or antigen-binding fragment thereof which has undergone posttranslational modification. Examples of an antibody or antigen-binding fragment thereof which have undergone posttranslational modification include an antibody or antigen-binding fragments thereof which have undergone pyroglutamylation at the N terminal of the heavy chain variable region, pyroglutamylation at the N terminal of the light chain variable region, and/or deletion of lysine at the C terminal of the heavy chain. It is known in the art that such posttranslational modification due to pyroglutamylation at the N terminal and deletion of lysine at the C terminal does not have any influence on the activity of the antibody or fragment thereof (Analytical Biochemistry, 2006, Vol. 348, p. 24-39, incorporated by reference in its entirety).

[163] In some embodiments, an ABP of the invention is an anti -human NRP-1 antibody or an antigen-binding fragment thereof, comprising a heavy chain variable region comprising a CDR-H3 consisting of SEQ ID NO:47, a CDR-H2 consisting of SEQ ID NO:30, and a CDR-H1 consisting of SEQ ID NO: 14; and a light chain variable region comprising a CDR-L3 consisting of SEQ ID NO:81, a CDR-L2 consisting of SEQ ID NO: 71, and a CDR-L1 consisting of SEQ ID NO: 63

[164] In one embodiment, the anti-human NRP-1 antibody or an antigen-binding fragment thereof, comprising a heavy chain variable region consisting of SEQ ID NO: 96, and a light chain variable region consisting of SEQ ID NO: 104.

[165] In one embodiment, the anti-human NRP-1 antibody or the antigen-binding fragment thereof, comprising a heavy chain variable region consisting of SEQ ID NO: 96 in which E of the amino acid number 1 is modified to pyroglutamate, and a light chain variable region consisting of SEQ ID NO: 104.

[166] In one embodiment, the anti -human NRP-1 antibody comprising a heavy chain consisting of SEQ ID NO: 118, and a light chain consisting of SEQ ID NO: 126.

[167] In one embodiment, the anti -human NRP-1 antibody comprising a heavy chain consisting of SEQ ID NO: 118 in which E of the amino acid number 1 is modified to pyroglutamate, and a light chain consisting of SEQ ID NO: 126.

[168] In one embodiment, the anti -human NRP-1 antibody comprising a heavy chain consisting of the amino acid sequence of amino acid numbers 1 to 453 of SEQ ID NO: 118, and a light chain consisting of SEQ ID NO: 126.

[169] In one embodiment, the anti -human NRP-1 antibody comprising a heavy chain consisting of the amino acid sequence of amino acid numbers 1 to 453 of SEQ ID NO: 118 in which E of the amino acid number 1 is modified to pyroglutamate, and a light chain consisting of SEQ ID NO: 126.

2.2 NRP-1 Antagonism

[170] In some embodiments, the ABPs provided herein antagonize NRP-1 upon binding.

[171] In some embodiments, antagonism of NRP-1 by an ABP provided herein results in activation of an effector T cell. In some aspects, the effector T cell is a CD8+ T cell. In some aspects, the effector T cell is a CD4+ T cell.

[172] In some embodiments, antagonism of NRP-1 by an ABP provided herein results in activation of an NK cell. In some embodiments, antagonism of NRP-1 by an ABP provided herein results in activation of an NKT cell. In some embodiments, the NKT cell is an IL-17-secreting cell.

[173] In some embodiments, antagonism of NRP-1 by an ABP provided herein results in a reduction of the inhibitory activity of a regulatory T cell toward an effector T cell.

[174] In some embodiments, antagonism of NRP-1 by an ABP provided herein results in increased secretion of IL-2, IL-6, GM-CSF, TNF, LT-a, and/or IFN-γ by a target cell.

[175] In some embodiments, antagonism of NRP-1 by an ABP provided herein increases the proliferation, survival, and/or function of an effector T cell. In some aspects, the effector T cell is a

CD4+ effector T cell. In some aspects, the effector T cell is a CD8+ effector T cell.

[176] In some embodiments, antagonism of NRP-1 by an ABP provided herein abrogates suppression of an effector T cell by a regulatory T cell. In some aspects, the regulatory T cell is a

CD4+CD25+Foxp3+ regulatory T cell. In some aspects, the regulatory T cell is a CD8+CD25+ regulatory T cell.

[177] In some embodiments, antagonism of NRP-1 by an ABP provided herein results in an enhancement of an immune response.

[178] In some embodiments, antagonism of NRP-1 by an ABP provided herein results in the prevention of a tumor. In some embodiments, antagonism of NRP-1 by an ABP provided herein results in the delay of onset of a tumor. In some embodiments, antagonism of NRP-1 by an ABP provided herein results in a reduction of the size of a tumor. In some embodiments, antagonism of NRP-1 by an ABP provided herein results in elimination of a tumor. In some embodiments, antagonism of NRP-1 by an ABP provided herein results in a reduction in the number of metastases.

[179] In some embodiments, antagonism of NRP-1 by an ABP provided herein results in the prevention of a viral disease. In some embodiments, antagonism of NRP-1 by an ABP provided herein results in the delay of onset of a viral disease. In some embodiments, antagonism of NRP-1 by an ABP provided herein results in a reduction of the viral load in a subject. In some embodiments, antagonism of NRP-1 by an ABP provided herein results in the elimination of a viral infection. 2.3 Affinity and Kinetics of Antigen-Binding Proteins for NRP-1; Potency

[180] In some embodiments, the affinity of an ABP provided herein for NRP-1 as indicated by KD, is less than about 10 "5 M, less than about 10 "6 M, less than about 10 "7 M, less than about 10 "8 M, less than about 10 "9 M, less than about 10 "10 M, less than about 10 "11 M, or less than about 10 "12 M. In some embodiments, the affinity of the ABP is between about 10 "7 M and 10 "12 M. In some embodiments, the affinity of the ABP is between about 10 "7 M and 10 "11 M. In some embodiments, the affinity of the ABP is between about 10 "7 M and 10 "10 M. In some embodiments, the affinity of the ABP is between about 10 "7 M and 10 "9 M. In some embodiments, the affinity of the ABP is between about 10 "7 M and 10 "8 M. In some embodiments, the affinity of the ABP is between about 10 "8 M and 10 "12 M. In some embodiments, the affinity of the ABP is between about 10 "8 M and 10 "11 M. In some embodiments, the affinity of the ABP is between about 10 "9 M and 10 "11 M. In some embodiments, the affinity of the ABP is between about 10 "10 M and 10 "11 M.

2.3.1 Glycosylation Variants

[181] In certain embodiments, an ABP provided herein may be altered to increase, decrease or eliminate the extent to which it is glycosylated. Glycosylation of polypeptides is typically either "N- linked" or "O-linked."

[182] "N-linked" glycosylation refers to the attachment of a carbohydrate moiety to the side chain of an asparagine residue. The tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain. Thus, the presence of either of these tripeptide sequences in a polypeptide creates a potential glycosylation site.

[183] "O-linked" glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5- hydroxyproline or 5 -hydroxy lysine may also be used.

[184] Addition or deletion of N-linked glycosylation sites to or from an ABP provided herein may be accomplished by altering the amino acid sequence such that one or more of the above-described tripeptide sequences is created or removed. Addition or deletion of O-linked glycosylation sites may be accomplished by addition, deletion, or substitution of one or more serine or threonine residues in or to (as the case may be) the sequence of an ABP.

[185] In some embodiments, an ABP provided herein comprises a glycosylation motif that is different from a naturally occurring ABP. Any suitable naturally occurring glycosylation motif can be modified in the ABPs provided herein. The structural and glycosylation properties of

immunoglobulins, for example, are known in the art and summarized, for example, in Schroeder and Cavacini, J. Allergy Clin. Immunol , 2010, 125:S41-52, incorporated by reference in its entirety. [186] In some embodiments, an ABP provided herein comprises an IgGl Fc region with modification to the oligosaccharide attached to asparagine 297 (Asn 297). Naturally occurring IgGl antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn 297 of the Cm domain of the Fc region. See Wright et al., TIBTECH, 1997, 15:26-32, incorporated by reference in its entirety. The oligosaccharide attached to Asn 297 may include various carbohydrates such as mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the "stem" of the biantennary oligosaccharide structure.

[187] In some embodiments, the oligosaccharide attached to Asn 297 is modified to create ABPs having altered ADCC. In some embodiments, the oligosaccharide is altered to improve ADCC. In some embodiments, the oligosaccharide is altered to reduce ADCC.

[188] In some aspects, an ABP provided herein comprises an IgGl domain with reduced fucose content at position Asn 297 compared to a naturally occurring IgGl domain. Such Fc domains are known to have improved ADCC. See Shields et al., J. Biol. Chem., 2002, 277:26733-26740, incorporated by reference in its entirety. In some aspects, such ABPs do not comprise any fucose at position Asn 297. The amount of fucose may be determined using any suitable method, for example as described in WO 2008/077546, incorporated by reference in its entirety.

[189] In some embodiments, an ABP provided herein comprises a bisected oligosaccharide, such as a biantennary oligosaccharide attached to the Fc region of the ABP that is bisected by GlcNAc. Such ABP variants may have reduced fucosylation and/or improved ADCC function. Examples of such ABP variants are described, for example, in WO 2003/011878; U.S. Pat. No. 6,602,684; and U.S. Pat. Pub. No. 2005/0123546; each of which is incorporated by reference in its entirety.

[190] Other illustrative glycosylation variants which may be incorporated into the ABPs provided herein are described, for example, in U.S. Pat. Pub. Nos. 2003/0157108, 2004/0093621,

2003/0157108, 2003/0115614, 2002/0164328, 2004/0093621, 2004/0132140, 2004/0110704, 2004/0110282, 2004/0109865; International Pat. Pub. Nos. 2000/61739, 2001/29246, 2003/085119, 2003/084570, 2005/035586, 2005/035778; 2005/053742, 2002/031140; Okazaki et al., J Mol. Biol, 2004, 336: 1239-1249; and Yamane-Ohnuki et al, Biotech. Bioeng., 2004, 87: 614-622; each of which is incorporated by reference in its entirety.

[191] In some embodiments, an ABP provided herein comprises an Fc region with at least one galactose residue in the oligosaccharide attached to the Fc region. Such ABP variants may have improved CDC function. Examples of such ABP variants are described, for example, in WO

1997/30087; WO 1998/58964; and WO 1999/22764; each of which his incorporated by reference in its entirety.

[192] Examples of cell lines capable of producing defucosylated ABPs include Led 3 CHO cells, which are deficient in protein fucosylation {see Ripka et al., Arch. Biochem. Biophys. , 1986, 249:533- 545; U.S. Pat. Pub. No. 2003/0157108; WO 2004/056312; each of which is incorporated by reference in its entirety), and knockout cell lines, such as alpha- 1,6-fucosyltransferase gene or FUT8 knockout CHO cells (see Yamane-Ohnuki et al., Biotech. Bioeng., 2004, 87: 614-622; Kanda et al., Biotechnol. Bioeng. , 2006, 94:680-688; and WO 2003/085107; each of which is incorporated by reference in its entirety).

[193] In some embodiments, an ABP provided herein is an aglycosylated ABP. An aglycosylated ABP can be produced using any method known in the art or described herein. In some aspects, an aglycosylated ABP is produced by modifying the ABP to remove all glycosylation sites. In some aspects, the glycosylation sites are removed only from the Fc region of the ABP. In some aspects, an aglycosylated ABP is produced by expressing the ABP in an organism that is not capable of glycosylation, such as E. coli, or by expressing the ABP in a cell -free reaction mixture.

[194] In some embodiments, an ABP provided herein has a constant region with reduced effector function compared to a native IgGl antibody. In some embodiments, the affinity of a constant region of an Fc region of an ABP provided herein for Fc receptor is less than the affinity of a native IgGl constant region for such Fc receptor.

2.4 NRP-1 domains

[195] NRP-1 has both a transmembrane and a truncated form. The transmembrane form is as follows. Following a short stretch of secretion signal, NRP-1 consists of four different domains: two repeats of CUB domain (al/a2), two repeats of FV/VIII domain (bl/b2), a MAM (c) domain, and a fourth domain (d) that contains transmembrane and relatively short 40 to 43 amino acid cytoplasmic region. The first CUB domains have significant homology with complement factor Cls/Clr, Bone Morphogenetic Protein 1(BMP1), and Tolloid proteins. The second FV/VIII domain shares the homology with coagulation factor FV/VIII, one of the receptor type tyrosine kinase DDR, and discoidin-1. The third domain MAM is the abbreviation of meprin, A5 (former name of NRP), and receptor protein-tyrosine phosphatase mu and kappa. In one embodiment, an ABP provided herein binds to the al domain. In another embodiment, an ABP provided herein binds to the a2 domain. In another embodiment, an ABP provided herein binds to the bl domain. In another embodiment, an ABP provided herein binds to the b2 domain. In one embodiment, an ABP provided herein binds to more than one domain.

1.1. Fc Region Amino Acid Sequence Variants

[196] In certain embodiments, an ABP provided herein comprises an Fc region with one or more amino acid substitutions, insertions, or deletions in comparison to a naturally occurring Fc region. In some aspects, such substitutions, insertions, or deletions yield ABPs with altered stability, glycosylation, or other characteristics. In some aspects, such substitutions, insertions, or deletions yield aglycosylated ABPs.

[197] In some aspects, the Fc region of an ABP provided herein is modified to yield an ABP with altered affinity for an Fc receptor, or an ABP that is more immunologically inert. In some embodiments, the ABP variants provided herein possess some, but not all, effector functions. Such ABPs may be useful, for example, when the half-life of the ABP is important in vivo, but when certain effector functions (e.g., complement activation and ADCC) are unnecessary or deleterious.

[198] In some embodiments, the Fc region of an ABP provided herein is a human IgG4 Fc region comprising one or more of the hinge stabilizing mutations S228P and L235E. See Aalberse et al., Immunology, 2002, 105:9-19, incorporated by reference in its entirety. In some embodiments, the Fc region of an ABP provided herein is a human IgG4 Fc region comprising the hinge stabilizing mutations S228P. In some embodiments, the IgG4 Fc region comprises one or more of the following mutations: E233P, F234V, and L235A. See Armour et al., Mol. Immunol , 2003, 40:585-593, incorporated by reference in its entirety. In some embodiments, the IgG4 Fc region comprises a deletion at position G236.

[199] In some embodiments, the Fc region of an ABP provided herein is a human IgGl Fc region comprising one or more mutations to reduce Fc receptor binding. In some aspects, the one or more mutations are in residues selected from S228 (e.g., S228A), L234 (e.g., L234A), L235 (e.g., L235A), D265 (e.g., D265A), and N297 (e.g., N297A). In some aspects, the ABP comprises a PVA236 mutation. PVA236 means that the amino acid sequence ELLG, from amino acid position 233 to 236 of IgGl or EFLG of IgG4, is replaced by PVA. See U.S. Pat. No. 9,150,641, incorporated by reference in its entirety.

[200] In some embodiments, the Fc region of an ABP provided herein is modified as described in Armour et al., Eur. J. Immunol, 1999, 29:2613-2624; WO 1999/058572; and/or U.K. Pat. App. No. 98099518; each of which is incorporated by reference in its entirety.

[201] In some embodiments, the Fc region of an ABP provided herein is a human IgG2 Fc region comprising one or more of mutations A33 OS and P33 IS.

[202] In some embodiments, the Fc region of an ABP provided herein has an amino acid substitution at one or more positions selected from 238, 265, 269, 270, 297, 327 and 329. See U.S. Pat. No. 6,737,056, incorporated by reference in its entirety. Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so- called "DANA" Fc mutant with substitution of residues 265 and 297 with alanine. See U.S. Pat. No. 7,332,581, incorporated by reference in its entirety. In some embodiments, the ABP comprises an alanine at amino acid position 265. In some embodiments, the ABP comprises an alanine at amino acid position 297.

[203] In certain embodiments, an ABP provided herein comprises an Fc region with one or more amino acid substitutions which improve ADCC, such as a substitution at one or more of positions 298, 333, and 334 of the Fc region. In some embodiments, an ABP provided herein comprises an Fc region with one or more amino acid substitutions at positions 239, 332, and 330, as described in Lazar et al., Proc. Natl. Acad. Sci. USA, 2006, 103:4005-4010, incorporated by reference in its entirety.

[204] In some embodiments, an ABP provided herein comprises one or more alterations that improves or diminishes Clq binding and/or CDC. See U.S. Pat. No. 6, 194,551; WO 99/51642; and Idusogie et al., J. Immunol, 2000, 164:4178-4184; each of which is incorporated by reference in its entirety.

[205] In some embodiments, an ABP provided herein comprises one or more alterations to increase half-life. ABPs with increased half-lives and improved binding to the neonatal Fc receptor (FcRn) are described, for example, in Hinton et al., J. Immunol, 2006, 176:346-356; and U.S. Pat. No. 7,361,740; each of which is incorporated by reference in its entirety. Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 250, 256, 265, 272, 286, 303, 305, 307, 311, 312, 314, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424, 428, and 434 of an IgG.

[206] In some embodiments, an ABP provided herein comprises one or more Fc region variants as described in U.S. Pat. Nos. 7,371,826 5,648,260, and 5,624,821; Duncan and Winter, Nature, 1988, 322:738-740; and WO 94/29351; each of which is incorporated by reference in its entirety.

1.2. Pyroglutamate

[207] As is known in the art, both glutamate (E) and glutamine (Q) at the N-termini of recombinant proteins can cyclize spontaneously to form pyroglutamate (pE) in vitro and in vivo. See Liu et al., J. Biol. Chem. , 2011, 286: 11211-11217, incorporated by reference in its entirety.

[208] In some embodiments, provided herein are ABPs comprising a polypeptide sequence having a pE residue at the N-terminal position. In some embodiments, provided herein are ABPs comprising a polypeptide sequence in which the N-terminal residue has been converted from Q to pE. In some embodiments, provided herein are ABPs comprising a polypeptide sequence in which the N-terminal residue has been converted from E to pE.

[209] In some embodiments, provided herein are ABPs comprising VH sequences having a pE residue at the N-terminal position. In some embodiments, provided herein are ABPs comprising a VH sequence in which the N-terminal residue has been converted from Q to pE. In some embodiments, provided herein is an ABP comprising a VH sequence selected from SEQ ID Nos: 85-90, 97-99, wherein the N-terminal Q residue has been converted to pE. In some embodiments, provided herein is a composition comprising an ABP, wherein the ABP comprises a VH selected from SEQ ID NOs: 85-90, 97-99, in which at least about 20%, at least about 40%, at least about 60%, at least about 80%, at least about 90%, at least about 95%, or at least about 99% of the N-terminal residues of such VH in such composition have been converted from Q to pE.

[210] In some embodiments, provided herein are ABPs comprising VH sequences having a pE residue at the N-terminal position. In some embodiments, provided herein are ABPs comprising a VH sequence in which the N-terminal residue has been converted from E to pE. In some embodiments, provided herein is an ABP comprising a VH sequence selected from SEQ ID Nos: 91-96, wherein the N-terminal E residue has been converted to pE. In some embodiments, provided herein is a composition comprising an ABP, wherein the ABP comprises a VH selected from SEQ ID NOs:91-96, in which at least about 20%, at least about 40%, at least about 60%, at least about 80%, at least about 90%, at least about 95%, or at least about 99% of the N-terminal residues of such VH in such composition have been converted from E to pE.

[211] In some embodiments, provided herein are ABPs comprising VL sequences having a pE residue at the N-terminal position. In some embodiments, provided herein are ABPs comprising a VL sequence in which the N-terminal residue has been converted from E to pE. In some embodiments, provided herein is an ABP comprising a VL sequence set forth in SEQ ID No: 120, wherein the N- terminal E residue has been converted to pE. In some embodiments, provided herein is a composition comprising an ABP, wherein the ABP comprises a VL set forth in SEQ ID NO: 120, in which at least about 20%, at least about 40%, at least about 60%, at least about 80%, at least about 90%, at least about 95%, or at least about 99% of the N-terminal residues of such VL in such composition have been converted from E to pE.

[212] In some embodiments, provided herein are ABPs comprising heavy chain sequences having a pE residue at the N-terminal position. In some embodiments, provided herein are ABPs comprising a heavy chain sequence in which the N-terminal residue has been converted from Q to pE. In some embodiments, provided herein is an ABP comprising a heavy chain sequence selected from SEQ ID Nos: 107- 1 12, 1 19-121, wherein the N-terminal Q residue has been converted to pE. In some embodiments, provided herein is a composition comprising an ABP, wherein the ABP comprises a heavy chain selected from SEQ ID NOs: 107-1 12, 1 19- 121, in which at least about 20%, at least about 40%, at least about 60%, at least about 80%, at least about 90%, at least about 95%, or at least about 99% of the N-terminal residues of such heavy chain in such composition have been converted from Q to pE.

[213] In some embodiments, provided herein are ABPs comprising heavy chain sequences having a pE residue at the N-terminal position. In some embodiments, provided herein are ABPs comprising a heavy chain sequence in which the N-terminal residue has been converted from E to pE. In some embodiments, provided herein is an ABP comprising a heavy chain sequence selected from SEQ ID Nos: 1 13- 1 18, wherein the N-terminal E residue has been converted to pE. In some embodiments, provided herein is a composition comprising an ABP, wherein the ABP comprises a heavy chain selected from SEQ ID NOs: 1 13-1 18, in which at least about 20%, at least about 40%, at least about 60%, at least about 80%, at least about 90%, at least about 95%, or at least about 99% of the N- terminal residues of such heavy chain in such composition have been converted from E to pE.

[214] In some embodiments, provided herein are ABPs comprising light chain sequences having a pE residue at the N-terminal position. In some embodiments, provided herein are ABPs comprising a light chain sequence in which the N-terminal residue has been converted from E to pE. In some embodiments, provided herein is an ABP comprising a kappa light chain sequence selected from SEQ ID NOs: 124- 125, wherein the N-terminal E residue has been converted to pE. In some embodiments, provided herein is a composition comprising an ABP, wherein the ABP comprises a kappa light chain selected from SEQ ID NOs: 124-125, in which at least about 20%, at least about 40%, at least about 60%, at least about 80%, at least about 90%, at least about 95%, or at least about 99% of the N- terminal residues of such light chain in such composition have been converted from E to pE.

1.3. Cysteine Engineered Antigen-Binding Protein Variants

[215] In certain embodiments, provided herein are cysteine engineered ABPs, also known as "thioMAbs," in which one or more residues of the ABP are substituted with cysteine residues. In particular embodiments, the substituted residues occur at solvent accessible sites of the ABP. By substituting such residues with cysteine, reactive thiol groups are introduced at solvent accessible sites of the ABP and may be used to conjugate the ABP to other moieties, such as drug moieties or linker- drug moieties, for example, to create an immunoconjugate.

[216] In certain embodiments, any one or more of the following residues may be substituted with cysteine: V205 of the light chain; Al 18 of the heavy chain Fc region; and S400 of the heavy chain Fc region. Cysteine engineered ABPs may be generated as described, for example, in U.S. Pat. No. 7,521,541, which is incorporated by reference in its entirety.

2. Methods of Making NRP-1 Antigen-Binding Proteins

2.1. NRP-1 Antigen Preparation

[217] The NRP-1 antigen used for isolation of the ABPs provided herein may be intact NRP-1 or a fragment of NRP-1. The NRP-1 antigen may be, for example, in the form of an isolated protein or a protein expressed on the surface of a cell.

[218] In some embodiments, the NRP- 1 antigen is a non-naturally occurring variant of NRP-1, such as a NRP-1 protein having an amino acid sequence or post-translational modification that does not occur in nature.

[219] In some embodiments, the NRP- 1 antigen is truncated by removal of, for example, intracellular or membrane -spanning sequences, or signal sequences. In some embodiments, the NRP- 1 antigen is fused at its C-terminus to a human IgGl Fc domain or a polyhistidine tag.

2.2. Methods of Making Monoclonal Antibodies

[220] Monoclonal antibodies may be obtained, for example, using the hybridoma method first described by Kohler et al., Nature, 1975, 256:495-497 (incorporated by reference in its entirety), and/or by recombinant DNA methods (see e.g., U.S. Patent No. 4,816,567, incorporated by reference in its entirety). Monoclonal antibodies may also be obtained, for example, using phage or yeast-based libraries. See e.g., U.S. Patent Nos. 8,258,082 and 8,691,730, each of which is incorporated by reference in its entirety.

[221] In the hybridoma method, a mouse or other appropriate host animal is immunized to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization. Alternatively, lymphocytes may be immunized in vitro. Lymphocytes are then fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell. See Goding J.W., Monoclonal Antibodies: Principles and Practice 3 rd ed. (1986) Academic Press, San Diego, CA, incorporated by reference in its entirety.

[222] The hybridoma cells are seeded and grown in a suitable culture medium that contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells. For example, if the parental myeloma cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.

[223] Useful myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive media conditions, such as the presence or absence of HAT medium. Among these, preferred myeloma cell lines are murine myeloma lines, such as those derived from MOP-21 and MC-11 mouse tumors (available from the Salk Institute Cell Distribution Center, San Diego, CA), and SP-2 or X63-Ag8-653 cells (available from the American Type Culture Collection, Rockville, MD). Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies. See e.g., Kozbor, J. Immunol, 1984, 133:3001, incorporated by reference in its entirety.

[224] After the identification of hybridoma cells that produce antibodies of the desired specificity, affinity, and/or biological activity, selected clones may be subcloned by limiting dilution procedures and grown by standard methods. See Goding, supra. Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium. In addition, the hybridoma cells may be grown in vivo as ascites tumors in an animal.

[225] DNA encoding the monoclonal antibodies may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies). Thus, the hybridoma cells can serve as a useful source of DNA encoding antibodies with the desired properties. Once isolated, the DNA may be placed into expression vectors, which are then transfected into host cells such as bacteria (e.g., E. coli), yeast (e.g., Saccharomyces or Pichia sp.), COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce antibody, to produce the monoclonal antibodies.

[226] In another aspect is provided a method for producing an anti -human NRP-1 antibody or an antigen-binding fragment thereof, comprising culturing host cell(s) selected from the group consisting of (a) to (c) below to express an anti -human NRP-1 antibody or an antigen-binding fragment thereof: (a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti -human NRP- 1 antibody or the antigen- binding fragment thereof provided herein and a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen-binding fragment thereof; (b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti-human NRP-1 antibody or the antigen-binding fragment thereof provided herein and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen-binding fragment thereof; and (c) a host cell transformed with an expression vector comprising a

polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP-1 antibody or the antigen-binding fragment thereof provided herein and a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen-binding fragment thereof.

[227] In another aspect is provided a method for producing an anti -human NRP-1 antibody, comprising culturing host cell(s) selected from the group consisting of (a) to (c) below to express an anti -human NRP-1 antibody: (a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP- 1 antibody provided herein and a polynucleotide comprising a base sequence encoding the light chain of the antibody; (b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody provided herein and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the antibody; and (c) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP- 1 antibody provided herein and a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the antibody.

2.3. Methods of Making Chimeric Antibodies

[228] Illustrative methods of making chimeric antibodies are described, for example, in U.S. Pat. No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA, 1984, 81:6851-6855; each of which is incorporated by reference in its entirety. In some embodiments, a chimeric antibody is made by using recombinant techniques to combine a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) with a human constant region.

2.4. Methods of Making Humanized Antibodies

[229] Humanized antibodies may be generated by replacing most, or all, of the structural portions of a non-human monoclonal antibody with corresponding human antibody sequences. Consequently, a hybrid molecule is generated in which only the antigen-specific variable, or CDR, is composed of non-human sequence. Methods to obtain humanized antibodies include those described in, for example, Winter and Milstein, Nature, 1991, 349:293-299; Rader et al., Proc. Nat. Acad. Sci. U.S.A. , 1998, 95:8910-8915; Steinberger et al, J. Biol. Chem. , 2000, 275:36073-36078; Queen et al., Proc. Natl. Acad. Sci. U.S.A., 1989, 86: 10029-10033; and U.S. Patent Nos. 5,585,089, 5,693,761,

5,693,762, and 6,180,370; each of which is incorporated by reference in its entirety.

2.5. Methods of making Human Antibodies

[230] Human antibodies can be generated by a variety of techniques known in the art, for example by using transgenic animals (e.g., humanized mice). See, e.g., Jakobovits et al., Proc. Natl. Acad. Sci. U.S.A., 1993, 90:2551; Jakobovits et al., Nature, 1993, 362:255-258; Bruggermann et al., Year in Immuno. , 1993, 7:33; and U.S. Patent Nos. 5,591,669, 5,589,369 and 5,545,807; each of which is incorporated by reference in its entirety. Human antibodies can also be derived from phage-display libraries (see e.g., Hoogenboom et al, J. Mol. Biol , 1991, 227:381-388; Marks et al., J. Mol. Biol, 1991, 222:581-597; and U.S. Pat. Nos. 5,565,332 and 5,573,905; each of which is incorporated by reference in its entirety). Human antibodies may also be generated by in vitro activated B cells (see e.g., U.S. Patent. Nos. 5,567,610 and 5,229,275, each of which is incorporated by reference in its entirety). Human antibodies may also be derived from yeast-based libraries (see e.g., U.S. Patent No. 8,691,730, incorporated by reference in its entirety).

2.6. Methods of Making Antibody Fragments

[231] The antibody fragments provided herein may be made by any suitable method, including the illustrative methods described herein or those known in the art. Suitable methods include recombinant techniques and proteolytic digestion of whole antibodies. Illustrative methods of making antibody fragments are described, for example, in Hudson et al., Nat. Med., 2003, 9: 129-134, incorporated by reference in its entirety. Methods of making scFv antibodies are described, for example, in Pliickthun, in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer- Verlag, New York, pp. 269-315 (1994); WO 93/16185; and U.S. Pat. Nos. 5,571,894 and 5,587,458; each of which is incorporated by reference in its entirety.

2.7. Methods of Making Alternative Scaffolds

[232] The alternative scaffolds provided herein may be made by any suitable method, including the illustrative methods described herein or those known in the art. For example, methods of preparing Adnectins™ are described in Emanuel et al., mAbs, 2011, 3:38-48, incorporated by reference in its entirety. Methods of preparing iMabs are described in U.S. Pat. Pub. No. 2003/0215914, incorporated by reference in its entirety. Methods of preparing Anticalins ® are described in Vogt and Skerra, Chem. Biochem. , 2004, 5: 191-199, incorporated by reference in its entirety. Methods of preparing Kunitz domains are described in Wagner et al., Biochem. & Biophys. Res. Comm. , 1992, 186: 118-1145, incorporated by reference in its entirety. Methods of preparing thioredoxin peptide aptamers are provided in Geyer and Brent, Meth. Enzymol , 2000, 328: 171-208, incorporated by reference in its entirety. Methods of preparing Affibodies are provided in Fernandez, Curr. Opinion in Biotech , 2004, 15:364-373, incorporated by reference in its entirety. Methods of preparing DARPins are provided in Zahnd et al., J. Mol. Biol , 2007, 369: 1015-1028, incorporated by reference in its entirety. Methods of preparing Affilins are provided in Ebersbach et al., J. Mol. Biol, 2007, 372: 172-185, incorporated by reference in its entirety. Methods of preparing Tetranectins are provided in Graversen et al, J. Biol. Chem., 2000, 275:37390-37396, incorporated by reference in its entirety. Methods of preparing Avimers are provided in Silverman et al., Nature Biotech., 2005, 23: 1556-1561, incorporated by reference in its entirety. Methods of preparing Fynomers are provided in Silacci et al., J. Biol. Chem. , 2014, 289: 14392-14398, incorporated by reference in its entirety.

[233] Further information on alternative scaffolds is provided in Binz et al., Nat. Biotechnol , 2005 23: 1257-1268; and Skerra, Current Opin. in Biotech., 2007 18:295-304, each of which is incorporated by reference in its entirety.

2.8. Methods of Making Variants

[234] In some embodiments, an ABP provided herein is an affinity matured variant of a parent ABP, which may be generated, for example, using phage display-based affinity maturation techniques. Briefly, one or more CDR residues may be mutated and the variant ABPs, or portions thereof, displayed on phage and screened for affinity. Such alterations may be made in CDR

"hotspots," or residues encoded by codons that undergo mutation at high frequency during the somatic maturation process {see Chowdhury, Methods Mol. Biol , 2008, 207: 179-196, incorporated by reference in its entirety), and/or residues that contact the antigen.

[235] Any suitable method can be used to introduce variability into a polynucleotide sequence(s) encoding an ABP, including error-prone PCR, chain shuffling, and oligonucleotide-directed mutagenesis such as trinucleotide-directed mutagenesis (TRIM). In some aspects, several CDR residues (e.g., 4-6 residues at a time) are randomized. CDR residues involved in antigen binding may be specifically identified, for example, using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted for mutation.

[236] The introduction of diversity into the variable regions and/or CDRs can be used to produce a secondary library. The secondary library is then screened to identify ABP variants with improved affinity. Affinity maturation by constructing and reselecting from secondary libraries has been described, for example, in Hoogenboom et al., Methods in Molecular Biology, 2001, 178: 1-37, incorporated by reference in its entirety.

2.9. Vectors, Host Cells, and Recombinant Methods

[237] Also provided are isolated nucleic acids encoding NRP-1 ABPs, vectors comprising the nucleic acids, and host cells comprising the vectors and nucleic acids, as well as recombinant techniques for the production of the ABPs. [238] In another aspect is provided a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti -human NRP-1 antibody or the antigen-binding fragment thereof provided herein. In another aspect is provided a polynucleotide comprising a base sequence encoding the light chain variable region of the anti -human NRP-1 antibody or the antigen-binding fragment thereof provided herein.

[239] In another aspect is provided a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody provided herein. In another aspect is provided a polynucleotide comprising a base sequence encoding the light chain of the anti -human NRP-1 antibody provided herein.

[240] For recombinant production of an ABP, the nucleic acid(s) encoding it may be isolated and inserted into a replicable vector for further cloning (i.e., amplification of the DNA) or expression. In some aspects, the nucleic acid may be produced by homologous recombination, for example as described in U.S. Patent No. 5,204,244, incorporated by reference herein in its entirety.

[241] In another aspect is provided an expression vector comprising (a) a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof provided herein and/or (b) a polynucleotide comprising a base sequence encoding the light chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof.

[242] In another aspect is provided an expression vector comprising (a) a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody provided herein and/or (b) a polynucleotide comprising a base sequence encoding the light chain of the anti- human NRP-1 antibody.

[243] Many different vectors are known in the art. The vector components generally include one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence, for example as described in U.S. Patent No. 5,534,615, incorporated by reference in its entirety.

[244] Illustrative examples of suitable host cells are provided below. These host cells are not meant to be limiting, and any suitable host cell may be used to produce the ABPs provided herein.

[245] In another aspect is provided a host cell transformed with an expression vector selected from the group consisting of (a) to (d): (a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP-1 antibody or the antigen-binding fragment thereof provided herein, and a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen- binding fragment thereof; (b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti- human NRP-1 antibody or the antigen-biding fragment thereof provided herein and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the antibody or the antigen-binding fragment thereof; (c) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof provided herein; and (d) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain variable region of the anti -human NRP- 1 antibody or the antigen-binding fragment thereof provided herein.

[246] In another aspect is provided host cell transformed with an expression vector selected from the group consisting of (a) to (d): (a) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP- 1 antibody provided herein and a polynucleotide comprising a base sequence encoding the light chain of the antibody; (b) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP-1 antibody provided herein and an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the antibody; (c) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the heavy chain of the anti -human NRP- 1 antibody provided herein; and (d) a host cell transformed with an expression vector comprising a polynucleotide comprising a base sequence encoding the light chain of the anti -human NRP-1 antibody provided herein.

[247] Suitable host cells include any prokaryotic (e.g., bacterial), lower eukaryotic (e.g., yeast), or higher eukaryotic (e.g., mammalian) cells. Suitable prokaryotes include eubacteria, such as Gram- negative or Gram-positive organisms, for example, Enterobacteriaceae such as Escherichia (E. coli), Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella (S. typhimurium), Serratia (S. marcescans), Shigella, Bacilli (B. subtilis and B. licheniformis), Pseudomonas (P. aeruginosa), and Streptomyces . One useful E. coli cloning host is E. coli 294, although other strains such as E. coli B, E. coli XI 776, and E. coli W3110 are also suitable.

[248] In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are also suitable cloning or expression hosts for NRP-1 ABP-encoding vectors. Saccharomyces cerevisiae, or common baker's yeast, is a commonly used lower eukaryotic host microorganism. However, a number of other genera, species, and strains are available and useful, such as Schizosaccharomyces pombe, Kluyveromyces (K. lactis, K. fragilis, K. bulgaricus K. wickeramii, K. waltii, K.

drosophilarum, K. thermotolerans , and K. marxianus), Yarrowia, Pichia pastoris, Candida (C.

albicans), Trichoderma reesia, Neurospora crassa, Schwanniomyces (S. occidentalis), and filamentous fungi such as, for example Penicillium, Tolypocladium, and Aspergillus {A. nidulans and A. niger).

[249] The host cells used to produce the NRP- 1 ABPs disclosed herein may be cultured in a variety of media. Commercially available media such as, for example, Ham's F10, Minimal Essential Medium (MEM), RPMI-1640, and Dulbecco's Modified Eagle's Medium (DMEM) are suitable for culturing the host cells. In addition, any of the media described in Ham et al., Meth. Enz. , 1979, 58:44; Barnes et al., Anal. Biochem. , 1980, 102:255; and U.S. Patent Nos. 4,767,704, 4,657,866, 4,927,762, 4,560,655, and 5,122,469; or WO 90/03430 and WO 87/00195 may be used. Each of the foregoing references is incorporated herein by reference in its entirety.

[250] Any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics, trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art.

[251] The culture conditions, such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.

[252] When using recombinant techniques, the ABP can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the ABP is produced intracellularly, as a first step, the particulate debris, either host cells or lysed fragments, is removed, for example, by centrifugation or ultrafiltration. For example, Carter et al. (Bio/Technology, 1992, 10: 163-167, incorporated by reference in its entirety) describes a procedure for isolating ABPs which are secreted to the periplasmic space of E. coli. Briefly, cell paste is thawed in the presence of sodium acetate (pH 3.5), EDTA, and phenylmethylsulfonylfluoride (PMSF) over about 30 min. Cell debris can be removed by centrifugation.

[253] In some embodiments, the ABP is produced in a cell-free system. In some aspects, the cell- free system is an in vitro transcription and translation system as described in Yin et al., mAbs, 2012, 4:217-225, incorporated by reference in its entirety. In some aspects, the cell-free system utilizes a cell -free extract from a eukaryotic cell or from a prokaryotic cell. In some aspects, the prokaryotic cell is E. coli. Cell-free expression of the ABP may be useful, for example, where the ABP accumulates in a cell as an insoluble aggregate, or where yields from periplasmic expression are low.

[254] Where the ABP is secreted into the medium, supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon ® or Millipore ® Pellcon ® ultrafiltration unit. A protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.

[255] The ABP composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being a particularly useful purification technique. The suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc domain that is present in the ABP. Protein A can be used to purify ABPs that comprise human γΐ, γ2, or γ4 heavy chains (Lindmark et al., J. Immunol. Meth , 1983, 62: 1-13, incorporated by reference in its entirety). Protein G is useful for all mouse isotypes and for human γ3 (Guss et al, EMBO J., 1986, 5: 1567- 1575, incorporated by reference in its entirety).

[256] The matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or

poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose. Where the ABP comprises a Cm domain, the BakerBond ABX ® resin is useful for purification.

[257] Other techniques for protein purification, such as fractionation on an ion-exchange column, ethanol precipitation, Reverse Phase HPLC, chromatography on silica, chromatography on heparin Sepharose ® , chromatofocusing, SDS-PAGE, and ammonium sulfate precipitation are also available, and can be applied by one of skill in the art.

[258] Following any preliminary purification step(s), the mixture comprising the ABP of interest and contaminants may be subjected to low pH hydrophobic interaction chromatography using an elution buffer at a pH between about 2.5 to about 4.5, generally performed at low salt concentrations (e.g., from about 0 to about 0.25 M salt).

3. Assays

[259] A variety of assays known in the art may be used to identify and characterize the NRP-1 ABPs provided herein.

3.1. Binding, Competition, and Epitope Mapping Assays

[260] Specific antigen-binding activity of the ABPs provided herein may be evaluated by any suitable method, including using SPR, BLI, RIA, KinExA, flow cytometry, and MSD-SET.

Additionally, antigen-binding activity may be evaluated by ELISA assays and western blot assays.

[261] Assays for measuring competition between two ABPs, or an ABP and another molecule (e.g., one or more ligands of NRP-1) are described elsewhere in this disclosure and, for example, in Harlow and Lane, Antibodies: A Laboratory Manual ch.14, 1988, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y, incorporated by reference in its entirety.

[262] Assays for mapping the epitopes to which the ABPs provided herein bind are described, for example, in Morris "Epitope Mapping Protocols," mMethods in Molecular Biology vol. 66, 1996, Humana Press, Totowa, N.J., incorporated by reference in its entirety. In some embodiments, the epitope is determined by peptide competition. In some embodiments, the epitope is determined by mass spectrometry. In some embodiments, the epitope is determined by crystallography.

3.2. NRP-1 Antagonism Assays

[263] In some embodiments, the ABPs provided herein are screened to identify or characterize ABPs with antagonistic activity against NRP-1. Any suitable assay may be used to identify or characterize such ABPs. In some aspects, the assay measures the amount of a cytokine secreted by an effector T cell after contacting the effector T cell with an ABP provided herein. In some aspects, the cytokine is selected from IL-2, IL-6, LT-a, TNF, GM-CSF, IFNy, and combinations thereof. In some aspects, the cytokine is selected from sCD40L, VEGF, TGF-a, RANTES, PDGF-AB/BB, PDGF-AA, ΜΙΡ-Ιβ, MIP-la, MDC (CCL22), MCP-3, MCP-1, IP-10, IL-17A, IL-2Ra, IL-15, IL-13, IL-12 (p70), IL-12 (p40), IL-10, IL-9, IL-8, IL-7, IL-5, IL-4, IL-3, IL-2, IL-2Ra, IL-1RA, IL-Ιβ, IL-la, IFNy, IFNa2, GRO, GM-CSF, G-CSF, fractalkine, Flt-3 ligand, FGF-2, eotaxin, EGF, and combinations thereof.

[264] In some embodiments, the effector cells are co-stimulated with an agonist of CD3, to promote the secretion of cytokines by the effector cell. In some aspects, the CD3 agonist is provided at a submaximal level.

[265] In some aspects, such assays may measure the proliferation of an effector T cell after contacting the effector T cell with an ABP provided herein. In some aspects, proliferation of the effector T cell is measured by dilution of a dye (e.g., carboxyfluorescein diacetate succinimidyl ester; CFSE), by tritiated thymidine uptake, by luminescent cell viability assays, or by other assays known in the art.

[266] In some aspects, such assays may measure the differentiation, cytokine production, viability (e.g., survival), proliferation, or suppressive activity of a regulatory T cell after contacting the regulatory T cell with an ABP provided herein.

[267] In some aspects, such assays may measure the cytotoxic activity of an NK cell after contacting the NK cell with an ABP provided herein. In some aspects, the cytotoxic activity of the NK cell is measured using a cytotoxicity assay that quantifies NK-mediated killing of target cells (e.g., a K562 cell line). See Jang et al., Ann. Clin. Lab. Sci. , 2012, 42:42-49, incorporated by reference in its entirety.

[268] In some aspects, such assays may measure the amount of granzyme B. In some aspects, such assays may measure the amount of perforin.

3.3. Assays for Effector Functions

[269] Effector function following treatment with the ABPs provided herein may be evaluated using a variety of in vitro and in vivo assays known in the art, including those described in Ravetch and Kinet, Annu. Rev. Immunol , 1991, 9:457-492; U.S. Pat. Nos. 5,500,362, 5,821,337; Hellstrom et al, Proc. Nat'lAcad. Sci. USA, 1986, 83:7059-7063; Hellstrom et al., Proc. Nat 'lAcad. Sci. USA, 1985, 82: 1499-1502; Bruggemann et al, J. Exp. Med. , 1987, 166: 1351-1361; Clynes et al., Proc. Nat'l Acad. Sci. USA, 1998, 95:652-656; WO 2006/029879; WO 2005/100402; Gazzano-Santoro et al, J. Immunol. Methods, 1996, 202: 163-171; Cragg et al., Blood, 2003, 101 : 1045-1052; Cragg et al. Blood, 2004, 103:2738-2743; and Petkova et al., Int 'l. Immunol , 2006, 18: 1759-1769; each of which is incorporated by reference in its entirety.

4. Pharmaceutical Compositions

[270] The ABPs provided herein can be formulated in any appropriate pharmaceutical composition and administered by any suitable route of administration. Suitable routes of administration include, but are not limited to, the intraarterial, intradermal, intramuscular, intraperitoneal, intravenous, nasal, parenteral, pulmonary, and subcutaneous routes.

[271] In another aspect is provided a pharmaceutical composition comprising an anti-human NRP - 1 antibody or an antigen-binding fragment thereof provided herein and pharmaceutically acceptable excipients.

[272] In another aspect is provided a pharmaceutical composition comprising plural kinds of anti- human NRP-1 antibodies or antigen-binding fragments thereof provided herein. For example, the pharmaceutical composition comprises an antibody or an antigen-binding fragment thereof, which does not undergo posttranslational modification and an antibody or an antigen-binding fragment thereof derived from posttranslational modification of the antibody or the antigen-binding fragment thereof.

[273] In one embodiment, the pharmaceutical composition comprises at least two kinds of anti- human NRP-1 antibodies selected from (1) to (4): (1) an anti-human NRP-1 antibody comprising a heavy chain consisting of SEQ ID NO: 118, and a light chain consisting of SEQ ID NO: 126, (2) an anti -human NRP-1 antibody comprising a heavy chain consisting of SEQ ID NO: 118 in which E of the amino acid number 1 is modified to pyroglutamate, and a light chain consisting of SEQ ID NO: 126, (3) an anti-human NRP-1 antibody comprising a heavy chain consisting of the amino acid sequence of amino acid numbers 1 to 453 of SEQ ID NO: 118, and a light chain consisting of SEQ ID NO: 126; and (4) an anti-human NRP-1 antibody comprising a heavy chain consisting of the amino acid sequence of amino acid numbers 1 to 453 of SEQ ID NO: 118 in which E of the amino acid number 1 is modified to pyroglutamate, and a light chain consisting of SEQ ID NO: 126.

[274] In one embodiment, the pharmaceutical composition comprises an anti -human NRP-1 antibody comprising a heavy chain consisting of SEQ ID NO: 118, and a light chain consisting of SEQ ID NO: 126, an anti -human NRP-1 antibody comprising a heavy chain consisting of the amino acid sequence of amino acid numbers 1 to 453 of SEQ ID NO: 118, and a light chain consisting of SEQ ID NO: 126, and a pharmaceutically acceptable excipient.

[275] The pharmaceutical composition may comprise one or more pharmaceutical excipients. Any suitable pharmaceutical excipient may be used, and one of ordinary skill in the art is capable of selecting suitable pharmaceutical excipients. Accordingly, the pharmaceutical excipients provided below are intended to be illustrative, and not limiting. Additional pharmaceutical excipients include, for example, those described in the Handbook of Pharmaceutical Excipients, Rowe et al. (Eds.) 6th Ed. (2009), incorporated by reference in its entirety. [276] In some embodiments, the pharmaceutical composition comprises an anti-foaming agent. Any suitable anti-foaming agent may be used. In some aspects, the anti-foaming agent is selected from an alcohol, an ether, an oil, a wax, a silicone, a surfactant, and combinations thereof. In some aspects, the anti-foaming agent is selected from a mineral oil, a vegetable oil, ethylene bis stearamide, a paraffin wax, an ester wax, a fatty alcohol wax, a long chain fatty alcohol, a fatty acid soap, a fatty acid ester, a silicon glycol, a fluorosilicone, a polyethylene glycol-polypropylene glycol copolymer,

polydimethylsiloxane-silicon dioxide, ether, octyl alcohol, capryl alcohol, sorbitan trioleate, ethyl alcohol, 2-ethyl-hexanol, dimethicone, oleyl alcohol, simethicone, and combinations thereof.

[277] In some embodiments, the pharmaceutical composition comprises a cosolvent. Illustrative examples of cosolvents include ethanol, poly(ethylene) glycol, butylene glycol, dimethylacetamide, glycerin, propylene glycol, and combinations thereof.

[278] In some embodiments, the pharmaceutical composition comprises a buffer. Illustrative examples of buffers include acetate, borate, carbonate, lactate, malate, phosphate, citrate, hydroxide, diethanolamine, monoethanolamine, glycine, methionine, guar gum, monosodium glutamate, and combinations thereof.

[279] In some embodiments, the pharmaceutical composition comprises a carrier or filler.

Illustrative examples of carriers or fillers include lactose, maltodextrin, mannitol, sorbitol, chitosan, stearic acid, xanthan gum, guar gum, and combinations thereof.

[280] In some embodiments, the pharmaceutical composition comprises a surfactant. Illustrative examples of surfactants include -alpha tocopherol, benzalkonium chloride, benzethonium chloride, cetrimide, cetylpyridinium chloride, docusate sodium, glyceryl behenate, glyceryl monooleate, lauric acid, macrogol 15 hydroxystearate, myristyl alcohol, phospholipids, polyoxyethylene alkyl ethers, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene stearates, polyoxylglycerides, sodium lauryl sulfate, sorbitan esters, vitamin E polyethylene (glycol) succinate, and combinations thereof.

[281] In some embodiments, the pharmaceutical composition comprises an anti-caking agent. Illustrative examples of anti-caking agents include calcium phosphate (tribasic), hydroxymethyl cellulose, hydroxypropyl cellulose, magnesium oxide, and combinations thereof.

[282] Other excipients that may be used with the pharmaceutical compositions include, for example, albumin, antioxidants, antibacterial agents, antifungal agents, bioabsorbable polymers, chelating agents, controlled release agents, diluents, dispersing agents, dissolution enhancers, emulsifying agents, gelling agents, ointment bases, penetration enhancers, preservatives, solubilizing agents, solvents, stabilizing agents, sugars, and combinations thereof. Specific examples of each of these agents are described, for example, in the Handbook of Pharmaceutical Excipients, Rowe et al. (Eds.) 6th Ed. (2009), The Pharmaceutical Press, incorporated by reference in its entirety.

[283] In some embodiments, the pharmaceutical composition comprises a solvent. In some aspects, the solvent is saline solution, such as a sterile isotonic saline solution or dextrose solution. In some aspects, the solvent is water for injection. [284] In some embodiments, the pharmaceutical compositions are in a particulate form, such as a microparticle or a nanoparticle. Microparticles and nanoparticles may be formed from any suitable material, such as a polymer or a lipid. In some aspects, the microparticles or nanoparticles are micelles, liposomes, or polymersomes.

[285] Further provided herein are anhydrous pharmaceutical compositions and dosage forms comprising an ABP, since water can facilitate the degradation of some ABPs.

[286] Anhydrous pharmaceutical compositions and dosage forms provided herein can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions. Pharmaceutical compositions and dosage forms that comprise lactose and at least one active ingredient that comprises a primary or secondary amine can be anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.

[287] An anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions can be packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g. , vials), blister packs, and strip packs.

4.1. Parenteral Dosage Forms

[288] In certain embodiments, the ABPs provided herein are formulated as parenteral dosage forms. Parenteral dosage forms can be administered to subjects by various routes including, but not limited to, subcutaneous, intravenous (including infusions and bolus injections), intramuscular, and intraarterial. Because their administration typically bypasses subjects' natural defenses against contaminants, parenteral dosage forms are typically, sterile or capable of being sterilized prior to administration to a subject. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry (e.g., lyophilized) products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions.

[289] Suitable vehicles that can be used to provide parenteral dosage forms are well known to those skilled in the art. Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and nonaqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.

[290] Excipients that increase the solubility of one or more of the ABPs disclosed herein can also be incorporated into the parenteral dosage forms.

[291] In some embodiments, the parenteral dosage form is lyophilized. Exemplary lyophilized formulations are described, for example, in U.S. Pat. Nos. 6,267,958 and 6,171,586; and WO 2006/044908; each of which is incorporated by reference in its entirety.

5. Dosage and Unit Dosage Forms

[292] In human therapeutics, the doctor will determine the posology which he considers most appropriate according to a preventive or curative treatment and according to the age, weight, condition and other factors specific to the subject to be treated.

[293] In certain embodiments, a composition provided herein is a pharmaceutical composition or a single unit dosage form. Pharmaceutical compositions and single unit dosage forms provided herein comprise a prophylactically or therapeutically effective amount of one or more prophylactic or therapeutic ABPs.

[294] The amount of the ABP or composition which will be effective in the prevention or treatment of a disorder or one or more symptoms thereof will vary with the nature and severity of the disease or condition, and the route by which the ABP is administered. The frequency and dosage will also vary according to factors specific for each subject depending on the specific therapy (e.g. , therapeutic or prophylactic agents) administered, the severity of the disorder, disease, or condition, the route of administration, as well as age, body, weight, response, and the past medical history of the subject. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.

[295] In certain embodiments, exemplary doses of a composition include milligram or microgram amounts of the ABP per kilogram of subject or sample weight (e.g. , about 10 micrograms per kilogram to about 50 milligrams per kilogram, about 100 micrograms per kilogram to about 25 milligrams per kilogram, or about 100 micrograms per kilogram to about 10 milligrams per kilogram). In certain embodiment, the dosage of the ABP provided herein, based on weight of the ABP, administered to prevent, treat, manage, or ameliorate a disorder, or one or more symptoms thereof in a subject is 0.1 mg/kg, 1 mg/kg, 2 mg/kg, 3 mg/kg, 4 mg/kg, 5 mg/kg, 6 mg/kg, 10 mg/kg, 15 mg/kg, 20 mg/kg, 25 mg/kg, 30 mg/kg, 40 mg/kg, or more of a subject's body weight. It may be necessary to use dosages of the ABP outside the ranges disclosed herein in some cases, as will be apparent to those of ordinary skill in the art. Furthermore, it is noted that the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with subject response.

[296] Different therapeutically effective amounts may be applicable for different diseases and conditions, as will be readily known by those of ordinary skill in the art. Similarly, amounts sufficient to prevent, manage, treat or ameliorate such disorders, but insufficient to cause, or sufficient to reduce, adverse effects associated with the ABPs provided herein are also encompassed by the dosage amounts and dose frequency schedules provided herein. Further, when a subject is administered multiple dosages of a composition provided herein, not all of the dosages need be the same. For example, the dosage administered to the subject may be increased to improve the prophylactic or therapeutic effect of the composition or it may be decreased to reduce one or more side effects that a particular subject is experiencing.

[297] In certain embodiments, treatment or prevention can be initiated with one or more loading doses of an ABP or composition provided herein followed by one or more maintenance doses.

[298] In certain embodiments, a dose of an ABP or composition provided herein can be administered to achieve a steady-state concentration of the ABP in blood or serum of the subject. The steady-state concentration can be determined by measurement according to techniques available to those of skill or can be based on the physical characteristics of the subject such as height, weight and age.

[299] In certain embodiments, administration of the same composition may be repeated and the administrations may be separated by at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months. In other embodiments, administration of the same composition may be repeated and the composition may be given once weekly, once every two weeks, once every three weeks, or once every four weeks. In certain embodiments, the first dose administered to the patient may be a "loading dose." A loading dose may be a higher dose than subsequent doses.

[300] As discussed in more detail elsewhere in this disclosure, an ABP provided herein may optionally be administered with one or more additional agents useful to prevent or treat a disease or disorder. The effective amount of such additional agents may depend on the amount of ABP present in the formulation, the type of disorder or treatment, and the other factors known in the art or described herein.

6. Therapeutic Applications

[301] For therapeutic applications, the ABPs of the invention are administered to a mammal, generally a human, in a pharmaceutically acceptable dosage form such as those known in the art and those discussed above. For example, the ABPs of the invention may be administered to a human intravenously as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intra-cerebrospinal, subcutaneous, intra-articular, intrasynovial, intrathecal, or intratumoral routes. The ABPs also are suitably administered by peritumoral, intralesional, or perilesional routes, to exert local as well as systemic therapeutic effects. The intraperitoneal route may be particularly useful, for example, in the treatment of ovarian tumors.

[302] The ABPs provided herein may be useful for the treatment of any disease or condition involving NRP-1. In some embodiments, the disease or condition is a disease or condition that can benefit from treatment with an anti-NRP-1 ABP. In some embodiments, the disease or condition is a tumor. In some embodiments, the disease or condition is a cell proliferative disorder. In some embodiments, the disease or condition is a cancer.

[303] In some embodiments, the ABPs provided herein are provided for use as a medicament. In some embodiments, the ABPs provided herein are provided for use in the manufacture or preparation of a medicament. In some embodiments, the medicament is for the treatment of a disease or condition that can benefit from an anti-NRP-1 ABP. In some embodiments, the disease or condition is a tumor. In some embodiments, the disease or condition is a cell proliferative disorder. In some embodiments, the disease or condition is a cancer. In some embodiments, the disease or condition is a viral infection.

[304] In some embodiments, provided herein is a method of treating a disease or condition in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject. In some aspects, the disease or condition is a cancer. In some aspects, the disease or condition is a viral infection.

[305] Any suitable cancer may be treated with the ABPs provided herein. Illustrative suitable cancers include, for example, acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), adrenocortical carcinoma, anal cancer, appendix cancer, astrocytoma, basal cell carcinoma, brain tumor, bile duct cancer, bladder cancer, bone cancer, breast cancer, bronchial tumor, carcinoma of unknown primary origin, cardiac tumor, cervical cancer, chordoma, colon cancer, colorectal cancer, craniopharyngioma, ductal carcinoma, embryonal tumor, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, fibrous histiocytoma, Ewing sarcoma, eye cancer, germ cell tumor, gallbladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, gestational trophoblastic disease, glioma, head and neck cancer, hepatocellular cancer, histiocytosis, Hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumor, Kaposi sarcoma, kidney cancer, Langerhans cell histiocytosis, laryngeal cancer, lip and oral cavity cancer, liver cancer, lobular carcinoma in situ, lung cancer, macroglobulinemia, malignant fibrous histiocytoma, melanoma, Merkel cell carcinoma, mesothelioma, metastatic squamous neck cancer with occult primary, midline tract carcinoma involving NUT gene, mouth cancer, multiple endocrine neoplasia syndrome, multiple myeloma, mycosis fungoides, myelodysplastic syndrome,

myelodysplastic/myeloproliferative neoplasm, nasal cavity and par nasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-small cell lung cancer, oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytomas, pituitary tumor, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell cancer, renal pelvis and ureter cancer, retinoblastoma, rhabdoid tumor, salivary gland cancer, Sezary syndrome, skin cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, spinal cord tumor, stomach cancer, T-cell lymphoma, teratoid tumor, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, urethral cancer, uterine cancer, vaginal cancer, vulvar cancer, and Wilms tumor.

[306] In some embodiments, provided herein is a method of antagonizing NRP-1 in a target cell of a subject in need thereof by administering an effective amount of an ABP provided herein to the subject. In some aspects, antagonism of NRP-1 by an ABP provided herein results in increased secretion of IL-2, LT-a, IL-6, TNF, GM-CSF, ΙΚΝγ or combinations thereof by a target cell.

[307] In some embodiments, provided herein is a method of increasing the proliferation, survival, and/or function of an effector T cell in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject. In some aspects, the effector T cell is a CD4+ effector T cell. In some aspects, the effector T cell is a CD8+ effector T cell.

[308] In some embodiments, provided herein is a method of abrogating suppression of an effector T cell by a regulatory T cell in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject. In some aspects, the regulatory T cell is a CD4+CD25+Foxp3+ regulator T cell. In some aspects, the regulatory T cell is a CD8+CD25+ regulatory T cell.

[309] In some embodiments, provided herein is a method of increasing the activity of a natural killer (NK) cell, a natural killer T (NKT) cell, a macrophage, or a dendritic cell (e.g., a plasmacytoid dendritic cell) in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject.

[310] In some embodiments, provided herein is a method of treating a subject having a cancer without concomitant platelet reduction. In some aspects, the method does not result in a substantive amount of thrombocytopenia in the subject.

[311] In some embodiments, provided herein is a method of enhancing an immune response in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject.

[312] In some embodiments, provided herein is a method delaying the onset of a tumor in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject.

[313] In some embodiments, provided herein is a method preventing the onset of a tumor in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject.

[314] In some embodiments, provided herein is a method of delaying the onset of a cancer in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject.

[315] In some embodiments, provided herein is a method of preventing the onset of a cancer in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject.

[316] In some embodiments, provided herein is a method of reducing the size of a tumor in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject.

[317] In some embodiments, provided herein is a method of reducing the number of metastases in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject.

[318] In some embodiments, provided herein is a method of reducing viral titer a subject in need thereof by administering an effective amount of an ABP provided herein to the subject.

[319] In some embodiments, provided herein is a method for extending the period of overall survival, median survival time, or progression-free survival in a subject in need thereof by administering an effective amount of an ABP provided herein to the subject.

[320] In some embodiments, provided herein is a method for treating a subject who has become resistant to a standard of care therapeutic by administering an effective amount of an ABP provided herein to the subject. In some embodiments, the standard-of-care therapeutic to which the subject has become resistant is a PD-1 inhibitor. In other embodiments, the standard-of-care therapeutic to which the subject has become resistant is a PD-Ll inhibitor. In other embodiments, the standard-of-care therapeutic to which the subject has become resistant is a CTLA-4 inhibitor.

7. Combination Therapies

[321] In some embodiments, an ABP provided herein is administered with at least one additional therapeutic agent. Any suitable additional therapeutic agent may be administered with an ABP provided herein. In some aspects, the additional therapeutic agent is selected from radiation, a cytotoxic agent, a chemotherapeutic agent, a cytostatic agent, an anti-hormonal agent, an EGFR inhibitor, an immunostimulatory agent, an anti-angiogenic agent, and combinations thereof.

[322] In some embodiments, the additional therapeutic agent comprises an immunostimulatory agent.

[323] In some embodiments, the immunostimulatory agent is an agent that blocks signaling of an inhibitory receptor of an immune cell, or a ligand thereof. In some aspects, the inhibitory receptor or ligand is selected from PVRIG, VISTA, CCR4, CD27, CTLA-4, PD-1, PD-Ll, LAG-3, Tim3, TIGIT, neuritin, BTLA, KIR, and combinations thereof. In some aspects, the agent is selected from an anti- PD-1 antibody (e.g., pembrohzumab or nivolumab), and anti-PD-Ll antibody (e.g., atezolizumab), an anti-CTLA-4 antibody (e.g., ipilimumab), and combinations thereof. In some aspects, the agent is pembrolizumab. In some aspects, the agent is nivolumab. In some aspects, the agent is atezolizumab.

[324] In some embodiments, the additional therapeutic agent is an agent that inhibits the interaction between PD-1 and PD-Ll . In some aspects, the additional therapeutic agent that inhibits the interaction between PD-1 and PD-Ll is selected from an antibody, a peptidomimetic and a small molecule. In some aspects, the additional therapeutic agent that inhibits the interaction between PD-1 and PD-Ll is selected from pembrolizumab, nivolumab, atezolizumab, avelumab, durvalumab, BMS- 936559, sulfamonomethoxine 1, and sulfamethizole 2. In some embodiments, the additional therapeutic agent that inhibits the interaction between PD-1 and PD-Ll is any therapeutic known in the art to have such activity, for example as described in Weinmann et al., Chem Med Chem, 2016, 14: 1576 (DOI: 10.1002/cmdc.201500566), incorporated by reference in its entirety. In some embodiments, the agent that inhibits the interaction between PD-1 and PD-Ll is formulated in the same pharmaceutical composition an ABP provided herein. In some embodiments, the agent that inhibits the interaction between PD-1 and PD-Ll is formulated in a different pharmaceutical composition from an ABP provided herein. In some embodiments, the agent that inhibits the interaction between PD-1 and PD-L1 is administered prior to administration of an ABP provided herein. In some embodiments, the agent that inhibits the interaction between PD-1 and PD-L1 is administered after administration of an ABP provided herein. In some embodiments, the agent that inhibits the interaction between PD-1 and PD-L1 is administered contemporaneously with an ABP provided herein, but the agent and ABP are administered in separate pharmaceutical compositions.

[325] In some embodiments, the immunostimulatory agent is an agent that, when administered alone and at its recommended dosage, results in a certain amount of thrombocytopenia in the subject. In some aspects, such an agent may be administered in combination with an ABP provided herein at a reduced dosage. Such combination therapy may be safely administered without resulting in substantive platelet deterioration or thrombocytopenia.

[326] In some embodiments, the immunostimulatory agent is an agonist of a co-stimulatory receptor of an immune cell. In some aspects, the co-stimulatory receptor is selected from OX40, ICOS, CD28, CD37, GITR, CD40, and 4- IBB, and combinations thereof. In some embodiments, the agonist is an antibody.

[327] In some embodiments, the immunostimulatory agent is a cytokine. In some aspects, the cytokine is selected from IL-2, IL-5, IL-7, IL-12, IL-15, IL-21, and combinations thereof.

[328] In some embodiments, the immunostimulatory agent is an oncolytic virus. In some aspects, the oncolytic virus is selected from a herpes simplex virus, a vesicular stomatitis virus, an adenovirus, a Newcastle disease virus, a vaccinia virus, and a maraba virus.

[329] In some embodiments, the immunostimulatory agent is a T cell with a chimeric antigen receptor (CAR-T cell). In some embodiments, the immunostimulatory agent is a bi- or multi-specific T cell-directed antibody. In some embodiments, the immunostimulatory agent is an anti-TGF-β antibody. In some embodiments, the immunostimulatory agent is a TGF-β trap.

[330] In some embodiments, the additional therapeutic agent is a vaccine to a tumor antigen. Any suitable antigen may be targeted by the vaccine, provided that it is present in a tumor treated by the methods provided herein. In some aspects, the tumor antigen is a tumor antigen that is overexpressed in comparison its expression levels in normal tissue. In some aspects, the tumor antigen is selected from cancer testis antigen, differentiation antigen, NY-ESO-1, MAGE-A1, MART, and combinations thereof.

[331] Further examples of additional therapeutic agents include a taxane (e.g., paclitaxel or docetaxel); a platinum agent (e.g., carboplatin, oxaliplatin, and/or cisplatin); a topoisomerase inhibitor (e.g., irinotecan, topotecan, etoposide, and/or mitoxantrone); folinic acid (e.g., leucovorin); or a nucleoside metabolic inhibitor (e.g., fluorouracil, capecitabine, and/or gemcitabine). In some embodiments, the additional therapeutic agent is folinic acid, 5 -fluorouracil, and/or oxaliplatin. In some embodiments, the additional therapeutic agent is 5 -fluorouracil and irinotecan. In some embodiments, the additional therapeutic agent is a taxane and a platinum agent. In some

embodiments, the additional therapeutic agent is paclitaxel and carboplatin. In some embodiments, the additional therapeutic agent is pemetrexate. In some embodiments, the additional therapeutic agent is a targeted therapeutic such as an EGFR, RAF or MEK-targeted agent.

[332] The additional therapeutic agent may be administered by any suitable means. In some embodiments, an ABP provided herein and the additional therapeutic agent are included in the same pharmaceutical composition. In some embodiments, an ABP provided herein and the additional therapeutic agent are included in different pharmaceutical compositions.

[333] In embodiments where an ABP provided herein and the additional therapeutic agent are included in different pharmaceutical compositions, administration of the ABP can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent. In some aspects, administration of an ABP provided herein and the additional therapeutic agent occur within about one month of each other. In some aspects, administration of an ABP provided herein and the additional therapeutic agent occur within about one week of each other. In some aspects, administration of an ABP provided herein and the additional therapeutic agent occur within about one day of each other. In some aspects, administration of an ABP provided herein and the additional therapeutic agent occur within about twelve hours of each other. In some aspects, administration of an ABP provided herein and the additional therapeutic agent occur within about one hour of each other.

8. Kits

[334] Also provided are kits comprising the ABPs provided herein. The kits may be used for the treatment, prevention, and/or diagnosis of a disease or disorder, as described herein.

[335] In some embodiments, the kit comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, and IV solution bags. The containers may be formed from a variety of materials, such as glass or plastic. The container holds a composition that is by itself, or when combined with another composition, effective for treating, preventing and/or diagnosing a disease or disorder. The container may have a sterile access port. For example, if the container is an intravenous solution bag or a vial, it may have a port that can be pierced by a needle. At least one active agent in the composition is an ABP provided herein. The label or package insert indicates that the composition is used for treating the selected condition.

[336] In some embodiments, the kit comprises (a) a first container with a first composition contained therein, wherein the first composition comprises an ABP provided herein; and (b) a second container with a second composition contained therein, wherein the second composition comprises a further therapeutic agent. The kit in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.

[337] Alternatively, or additionally, the kit may further comprise a second (or third) container comprising a pharmaceutically-acceptable excipient. In some aspects, the excipient is a buffer. The kit may further include other materials desirable from a commercial and user standpoint, including filters, needles, and syringes.

EXAMPLES

[338] The following are examples of methods and compositions of the invention. It is understood that various other embodiments may be practiced, given the general description provided herein.

Example 1. Antibody Selection

Materials and methods

[339] Antigens were biotinylated using the EZ-Link Sulfo-NHS-Biotinylation Kit from Pierce. Goat F(ab') 2 anti-human kappa-FITC (LC-FITC), ExtrAvidin-PE (EA-PE) and Streptavidin-AF633 (SA- 633) were obtained from Southern Biotech, Sigma, and Molecular Probes, respectively. Streptavidin MicroBeads and MACS LC separation columns were purchased from Miltenyi Biotec. Goat anti- human IgG-PE (Human-PE) was obtained from Southern Biotech.

Naive Discovery

[340] Eight naive human synthetic yeast libraries each of ~10 9 diversity were propagated as previously described (see, e.g., Y. Xu et al, Addressing polyspecificity of antibodies selected from an in vitro yeast presentation system: a FACS-based, high-throughput selection and analytical tool. PEDS 26.10, 663-70 (2013); WO2009036379; WO2010105256; and WO2012009568.) For the first two rounds of selection, a magnetic bead sorting technique utilizing the Miltenyi MACS system was performed, as previously described (see, e.g., Siegel et al, High efficiency recovery and epitope- specific sorting of an scFv yeast display library." J Immunol Methods 286(1-2), 141-153 (2004).) Briefly, yeast cells (-10 10 cells/library) were incubated with 5 ml of 100 nM biotinylated antigen for 30 min at 30°C in wash buffer (phosphate-buffered saline (PBS)/0.1% bovine serum albumin (BSA)). After washing once with 40 ml ice-cold wash buffer, the cell pellet was resuspended in 20 mL wash buffer, and Streptavidin MicroBeads (500 μΐ) were added to the yeast and incubated for 15 min at 4°C. Next, the yeast were pelleted, resuspended in 20 mL wash buffer, and loaded onto a Miltenyi LS column. After the 20 mL were loaded, the column was washed 3 times with 3 ml wash buffer. The column was then removed from the magnetic field, and the yeast were eluted with 5 mL of growth media and then grown overnight. The following rounds of selection were performed using flow cytometry. Approximately 2x 10 7 yeast were pelleted, washed three times with wash buffer, and incubated at 30°C with either decreasing concentrations of biotinylated antigen (100 to 1 nM) under equilibrium conditions, 100 nM biotinylated antigens of different species in order to obtain species cross-reactivity, or with a poly-specificity depletion reagent (PSR) to remove non-specific antibodies from the selection. For the PSR depletion, the libraries were incubated with a 1 : 10 dilution of biotinylated PSR reagent as previously described (see, e.g., Y. Xu et al, Addressing polyspecificity of antibodies selected from an in vitro yeast presentation system: a FACS-based, high-throughput selection and analytical tool. PEDS 26.10, 663-70 (2013).) Yeast were then washed twice with wash buffer and stained with LC-FITC (diluted 1 : 100) and either SA-633 (diluted 1 :500) or EAPE (diluted 1 :50) secondary reagents for 15 min at 4°C. After washing twice with wash buffer, the cell pellets were resuspended in 0.3 mL wash buffer and transferred to strainer-capped sort tubes. Sorting was performed using a FACS ARIA sorter (BD Biosciences) and sort gates were determined to select for antibodies with desired characteristics. Selection rounds were repeated until a population with all of the desired characteristics was obtained. After the final round of sorting, yeast were plated and individual colonies were picked for characterization.

Antibody Optimization

[341] Optimization of antibodies was performed via a light chain diversification protocol, and then by introducing diversities into the heavy chain and light chain variable regions as described below. A combination of some of these approaches was used for each antibody.

[342] Light chain batch diversification protocol: Heavy chain plasmids from a naive selection output were extracted from the yeast via smash and grab, propagated in and subsequently purified from E.coli, and transformed into a light chain library with a diversity of 5 x 10 6 . Selections were performed with one round of MACS and four rounds of FACS employing the same conditions as the naive discovery.

[343] CDRH1 and CDRH2 selection: The CDRH3 of a single antibody was recombined into a premade library with CDRH1 and CDRH2 variants of a diversity of 1 x 10 8 and selections were performed with one round of MACS and four rounds of FACS as described in the naive discovery. For each FACS round the libraries were looked at for PSR binding, species cross-reactivity, and affinity pressure, and sorting was performed in order to obtain a population with the desired characteristics.

[344] VH Mutant selection: The heavy chain variable region (VH) was mutagenized via error prone PCR. The library was then created by transforming this mutagenized VH and the heavy chain expression vector into yeast already containing the light chain plasmid of the parent. Selections were performed similar to previous cycles using FACS sorting for two rounds. For each FACS round the libraries were looked at for PSR binding, species cross-reactivity, and affinity pressure, and sorting was performed in order to obtain a population with the desired characteristics.

Antibody production and purification

[345] Yeast clones were grown to saturation and then induced for 48 h at 30°C with shaking. After induction, yeast cells were pelleted and the supernatants were harvested for purification. IgGs were purified using a Protein A column and eluted with acetic acid, pH 2.0. Fab fragments were generated by papain digestion and purified over KappaSelect® (GE Healthcare Life Sciences).

ForteBio KD measurements

[346] ForteBio affinity measurements were performed on an Octet RED384 generally as previously described (see, e.g., Estep et al, High throughput solution-based measurement of antibody-antigen affinity and epitope binning. Mabs 5(2), 270-278 (2013)). Briefly, ForteBio affinity measurements were performed by loading IgGs on-line onto AHQ sensors. Sensors were equilibrated off-line in assay buffer for 30 min and then monitored on-line for 60 seconds for baseline establishment. Sensors with loaded IgGs were exposed to 100 nM antigen for 3 minutes, and afterwards were transferred to assay buffer for 3 min for off-rate measurement. For monovalent affinity assessment Fabs were used instead of IgGs. For this assessment, the unbiotinylated Fc fusion antigen was loaded on-line onto the AHQ sensors. Sensors were equilibrated off-line in assay buffer for 30 min and then monitored online for 60 seconds for baseline establishment. Sensors with loaded antigen were exposed to 200 nM Fab for 3 minutes, and afterwards they were transferred to assay buffer for 3 min for off-rate measurement. All kinetics were analyzed using the 1: 1 binding model.

ForteBio Epitope Binning/Li gand Blocking

[347] Epitope binning/ligand blocking was performed using a standard sandwich format cross- blocking assay. Control anti-target IgG was loaded onto AHQ sensors and unoccupied Fc -binding sites on the sensor were blocked with an irrelevant human IgGl antibody. The sensors were then exposed to 100 nM target antigen followed by a second anti -target antibody or ligand. Additional binding by the second antibody or ligand after antigen association indicates an unoccupied epitope (non-competitor), while no binding indicates epitope blocking (competitor or ligand blocking).

Size Exclusion Chromatography

[348] A TSKgel® SuperSW mAb HTP column (22855) was used for fast SEC analysis of mammalian produced mAbs at 0.4 mL/min with a cycle time of 6 min/run. 200 mM Sodium

Phosphate and 250 mM Sodium Chloride was used as the mobile phase.

Dynamic Scanning Fluorimetry

[349] 10 of 20x Sypro Orange is added to 20 of 0.2-lmg/mL mAb or Fab solution. A RT- PCR instrument (BioRad CFX96 RT PCR) is used to ramp the sample plate temperature from 40 to 95 C at 0.5C increment, with 2min equilibrate at each temperature. The negative of first derivative for the raw data is used to extract Tm.

Example 2. Antibody Characterization

[350] ForteBio KD Measurements : Quantitative binding of antibodies to recombinant monomelic human, mouse, or cynomolgus monkey NRP-1 was measured using biolayer interferometry (BLI) using FORTEBIO®. Affinity measurements of selected antibodies were performed generally as described in Estep et al. , Mobs, 2013, 5:270-278, incorporated by reference in its entirety.

FORTEBIO affinity measurements were performed by loading IgGs (human IgGl N297A) on-line onto AHQ sensors. Sensors were equilibrated off-line in assay buffer for 30 min and then monitored on-line for 60 seconds for baseline establishment. Sensors with loaded IgGs were exposed to a single concentration of antigen (100 nM) for 3 minutes. Afterwards they were transferred to assay buffer for 3 minutes for off-rate measurement. Kinetics were analyzed using the 1 : 1 binding model. A summary of KD measurements for antibodies binding a single concentration of human, cynomolgus monkey, and mouse NRP-1 is shown in Table 5 below. [351] Additional KD measurements were performed with eight antibodies (human IgG4 S228P) using multi-concentration kinetics. The binding affinities for human NRP-1 -His, cynomolgus monkey NRP-1 -His, and mouse NRP-1 -His were measured using an Octet QKe instrument

(ForteBio). A strategy of capturing antibodies on sensors followed by association/dissociation of monomeric NRP-1 proteins was used to avoid avidity effects in the assay. The BLI analysis was performed at 30°C using IX kinetics buffer (ForteBio) as assay buffer. Anti-Human IgG Fc Capture (AHC) biosensors (ForteBio) were first presoaked in assay buffer for greater than 5 minutes. Test antibody (5 μg/mL) was captured on the sensor for 250 seconds. Sensors were then dipped in assay buffer for 60 seconds to establish a baseline before measuring binding to each NRP-1 protein. Sensors were then dipped into varying concentrations of human NRP-l-His (93.3 to 0.7nM, 2-fold dilutions in assay buffer), cynomolgus monkey NRP-l-His (93.3 to 1.5nM, 2-fold dilutions in assay buffer), or mouse NRP-l-His (93.3 to 1.5nM, 2-fold dilutions in assay buffer) for 250 seconds to measure association. Dissociation of NRP-1 was then measured by dipping sensors into assay buffer for 600 seconds. Agitation at all steps was 1000 rpm. Kinetic parameters were generated with Octet Data Analysis Software Version 8.2.0.7 using reference subtraction (antibody "binding" to buffer), dissociation based inter-step correction, 1 to 1 binding model, and global fit (Rmax unlinked by sensor). KD values are shown in Table 6.

[352] MSD-SET KD Measurements : Solution equilibrium affinity measurements of selected antibodies binding human NRP-1 were performed generally as previously described. See Estep et al., supra, incorporated by reference in its entirety. Briefly, solution equilibrium titrations (SET) were performed in PBS + 0.1% IgG-Free BSA (PBSF) with antigen held constant at 10-100 pM and incubated with 3 -to 5 -fold serial dilutions of Fab or mAbs starting at ΙΟρΜ-ΙΟηΜ. Antibodies (20 nM in PBS) were coated onto standard bind MSD-ECL plates overnight at 4°C or at room temperature for 30 min. Plates were then blocked by BSA for 30 min with shaking at 700 rpm, followed by three washes with wash buffer (PBSF + 0.05% Tween® 20). SET samples were applied and incubated on the plates for 150s with shaking at 700 rpm followed by one wash. Antigen captured on a plate was detected with 250ng/mL sulfotag-labeled streptavidin in PBSF by incubation on the plate for 3 min. The plates were washed three times with wash buffer and then read on the MSD Sector Imager 2400 instrument using lx Read Buffer T with surfactant. The percent free antigen was plotted as a function of titrated antibody in Prism and fit to a quadratic equation to extract the KD. TO improve throughput, liquid handling robots were used throughout MSD-SET experiments, including SET sample preparation.

Table 5. Antibody Binding Affinities - Single Concentration Kinetics

2 1.86E-09 2.43E-09 1.94E-09 2.30E-10

3 1.08E-09 1.19E-09 9.90E-10 6.00E-11

4 8.51E-10 9.25E-10 7.46E-10 4.60E-11

5 3.23E-09 4.09E-09 5.06E-09 2.80E-10

6 4.72E-09 5.54E-09 6.98E-09 4.50E-10

7 1.12E-08 1.09E-08 1.47E-08 N.D.

8 6.13E-10 6.42E-10 5.52E-10 9.60E-11

9 6.45E-10 6.43E-10 5.66E-10 1.90E-11

10 8.68E-10 8.66E-10 7.46E-10 6.40E-11

11 4.85E-10 4.80E-10 4.46E-10 2.10E-11

12 4.81E-10 4.69E-10 4.40E-10 2.60E-11

13 1.41E-09 1.58E-09 7.42E-09 5.40E-10

14 1.12E-09 1.10E-09 5.00E-09 2.80E-10

15 8.51E-10 9.20E-09 5.41E-08 1.80E-10

Table 6. Antibody Binding Affinities - Multiple Concentration Kinetics

Example 3. Anti-Tumor Efficacy of Nine Anti-NRP-1 MABs Alone and in Combination with a PD-1 or PD-L1 Antibody

Nine optimized antibodies were evaluated for anti-tumor efficacy using immunocompetent mice. The assay was conducted with a panel of murine versions of MABs 2, 3, 4, 5, 7, 12, 13, 14, and 15, as well as an IgG control and SECIO (SEQ ID NOS 141-142) as a comparator. The antibodies were tested as chimeric mouse IgG2a antibodies containing the N297A mutation which abolishes ADCC and CDC effector functions. Anti-tumor efficacy was measured using the mouse colon CT26 syngeneic tumor model grown in female BALB/c mice. 3xl0 5 mouse CT26 cells were implanted subcutaneously on Day 1. The mice were randomized based on body weight and antibodies were administered intraperitoneally at the indicated dose on the same day as tumor cell implantation. The anti-NRP-1 antibodies were administered as a monotherapy at 500 μg/dose or in combination with an anti-PD-1 immune checkpoint inhibitor which was used at 200 μg/dose. Figure 1A shows the monotherapy effect of antibodies in the CT26 model, and Figure IB shows the effect of combination of anti-NRP-1 antibodies with anti-PD-1. The black arrows along the horizontal axis indicate the treatment days of the antibodies. The average tumor volume from 10 mice per group is shown for each treatment group.

[353] Figure 1C shows a subset of data from Figures 1A and IB comparing mMAB12 alone and in combination with an anti-PD-1 checkpoint antibody in the mouse colon CT26 syngeneic tumor model. mMAB 12 at 500μg/animal inhibited tumor growth by 61.6% TGI (tumor growth inhibition) compared to control antibody-treated mice. This effect was statistically significant by Student's t test (p<0.05). The anti-PD-1 checkpoint antibody administered at 200μg/animal was less efficacious than mMAB 12 (37.8% TGI, p<0.05). However, the combination of mMAB 12 with the PD-1 antibody resulted in additive anti-tumor efficacy (79.0% TGI, p<0.001) compared to the monotherapy treatments. The effect of the combination was statistically significant when compared to PD-1 and mMAB 12 (p<0.05 in both cases). There was no untoward toxicity exhibited by the treated mice which all gained weight over the course of the treatment, except for one non-treatment-related expired mouse in the mMAB 12 group.

[354] The same nine antibodies were evaluated in a second tumor model, the mouse colon MC38 syngeneic model. 5xl0 5 mouse MC38 cells were implanted subcutaneously into female C57B1/6 mice. The mice were randomized into treatment groups when the tumors reached an average tumor volume of 60 mm 3 to 90 mm 3 followed by initiation of treatment on Day 1. The anti-NRP-1 antibodies were administered as a monotherapy at 500 μg/dose or in combination with an anti-PD-Ll immune checkpoint inhibitor which was used at 250 μg/dose. The anti-PD-Ll antibody works in the same immune checkpoint pathway as the PD-1 antibody. Figure 2A shows the monotherapy effect of antibodies in the MC38 model, and Figure 2B shows the effect of combination of anti-NRP-1 antibodies with anti-PD-Ll. The black arrows along the horizontal axis indicate the treatment days. The average tumor volume from 10 mice per group is shown for each treatment group.

[355] The anti-tumor efficacy of mMAB12 in the MC38 syngeneic colon mouse tumor model is shown in Figure 2C. The mMAB 12 at 500μg/animal inhibited tumor growth by 77.3% TGI (p<0.05) compared to control antibody-treated mice. The MC38 model is very sensitive to PD-1 antibody blockade. Therefore, an antibody against PD-Ll at 250μg/animal which works in the same immune checkpoint pathway as the PD-1 antibody was used to demonstrate potential combination benefits. As expected, PD-Ll monotherapy blocked tumor growth at 77.5% TGI (p<0.05). However, the combination of mMAB 12 with the PD-Ll antibody did not demonstrate additional anti -tumor benefits (76.2% TGI). As with the CT26 model there was no untoward toxicity exhibited by the treated mice which all gained weight over the course of the treatment. Four antibodies (MABs 2, 5, 12, and 13) were selected based on their efficacies in the CT26 and MC38 studies and retested in the MC38 model under the same conditions (alone and in combination with anti-PD-Ll). The findings in the repeat MC38 study confirmed the above findings for efficacy and tolerability.

Example 4. Evaluation of Blockade of NRP-1 Ligands

[356] Quantitative ligand blocking studies, measuring the ability of antibodies to block the binding of recombinant human SEMA3A and human VEGFA to recombinant human NRP-1, was measured by a blocking ELISA. To measure the ability of antibody to block the SEMA3A/NRP-1 interaction, the assay plate was coated with human SEMA3A at 2^g/mL in PBS, overnight at 4°C. Biotinylated human NRP-1 (500ng/mL in 1% BSA/PBS) was incubated with test antibody (30-0.0(^g/mL, 4-fold dilution in 1% BSA/PBS) prior to addition to the assay plate, and then HRP conjugated streptavidin (1 :200 in 1% BSA/PBS) was used for detection of NRP-1 bound to SEMA3A. Briefly, to measure the ability of antibody to block the VEGFA/NRP- 1 interaction, the assay plate was coated with human NRP-1 at 2^g/mL in PBS, overnight at 4°C. Test antibody (30-0.002μg/mL, 4-fold dilution in 1% BSA/PBS) was incubated with VEGFA (125ng/mL) prior to addition to the assay plate, biotinylated anti-VEGFA antibody (O^g/mL in 1% BSA/PBS) was added, and then HRP conjugated streptavidin (1 :200 in 1% BSA/PBS) was used for detection of VEGFA bound to NRP-1. The IC 50 values for 15 IgGl format test antibodies blocking SEMA3A/VEGFA binding are shown in Table 7.

Table 7. IC50 values for Blocking Assays with IgGl Format Antibodies

[357] Eight MABs were converted to IgG4 S228P format and the assay was repeated. A summary of the averages is shown in Table 8.

Table 8. Averages for Blocking Assays with IgG4 Format Antibodies

humanizing site-directed mutation

Example 5. Epitope Binning of MAB 12 vs SEC10

[358] Epitope binning for MAB 12 and SEC 10 was measured using BioLayer Interferometry (BLI) using an Octet® QKe instrument (ForteBio®). MAB 12 or SEC 10 at 5μg/mL was immobilized on anti -human Fc AHC sensors for 300 seconds. Sensors were then dipped in kinetics buffer for baseline determination. Next, sensors were dipped in human IgG at 200μg/ml for 400 seconds to saturate all the IgG Fc binding sites on the sensors. After baseline determination, the sensors were exposed to ΙΟΟηΜ human NRP-l-HIS for 300 seconds to allow for antigen binding. Finally, sensors were transferred to wells containing 20μg/mL of either MAB 12 or SEC10 for 300 seconds to analyze antibody binding. If the test antibody showed clear binding in the last step, it was considered a non- competitor (different epitope bin), and if the test antibody did not show clear binding, it was considered a competitor (same epitope bin).

[359] Results are shown in Figure 3. Capturing MAB 12 and then binding NRP- 1 does not prevent SEC 10 from also binding NRP-1 (top panel). Similarly, capturing SEC 10 and then binding NRP-1 does not prevent MAB 12 from also binding NRP-1 (bottom panel). Self-binning (e.g., capture MAB12, bind NRP-1, test binding of MAB12) served as a positive control for binning. These data show that MAB 12 and SEC 10 can simultaneously bind NRP- 1 , and must therefore bind to different epitopes.

Example 6. Binding of anti-NRP-1 antibodies to NRP-1 domains

[360] In order to understand the approximate binding domain for antibodies binding to human NRP- 1, the ability of antibodies to bind fragments of NRP-1 that contained different domains of NRP-1 extracellular region was measured by BLI using an Octet® QKe instrument (ForteBio®).

Recombinant human NRP-l-Fc fusion proteins consisted of al, ala2, ala2bl, a2blb2, or ala2blb2 domains, and the differences in antibody binding to each protein led to determination of which primary domain the antibody binds. The BLI analysis was performed at 29°C or 30°C using IX kinetics buffer (ForteBio) as the assay buffer. Briefly, antibodies ^g/mL) were captured onto anti- human IgG Fc (AHC) biosensors for 250 seconds. Then sensors were dipped into assay buffer (100 seconds) to achieve a baseline prior to measuring binding to each NRP-1 protein. A quenching step using human IgG Fc (150nM, 250nM or 500nM, depending on the experiment) for 250 seconds was performed next. Sensors were then dipped into each NRP-1 protein at 500nM for 300 seconds, followed by dissociation of each NRP-1 protein in assay buffer for 900 or 1000 seconds. Agitation was performed at 900 rpm or 1000 rpm for all steps, depending on the experiment.

[361] Table 9 shows the results of the assays described above. The binding domain for each antibody is shown in the far-right column.

Table 9. NRP1 Domain Binding Specificity

*SEQ ID Nos 141-142

** Described in Appleton, et. al., The EMBO Journal (2007) 26, 4902-4912.

***Described in Delgoffe GM, Woo S-R, Turnis ME, Gravano DM, Guy C, Overacre AE, et al. Stability and function of regulatory T cells is maintained by a neuropilin-l-semaphorin-4a axis. Nature 501(7466):252-6. Available from R&D Systems.

Example 7: Mutational analysis for epitope determination

[362] To identify the epitope for MAB 12 binding to the bl domain of human NRPl, single point mutations were made within the human NRPl bl domain. Either alanine substitutions or NRP2 specific residues were used (MAB12 does not bind NRP2). Proteins were expressed in HEK293 cells, secreted as soluble protein, purified on Ni-NTA resin, and characterized by SDS-PAGE. Binding was assessed by Bio-Layer Interferometry (BLI) using the Octet platform. MAB12 was captured on anti- human Fc sensors, washed, and exposed to either monomelic wild type human NRPl bl domain or to monomeric mutant NRPl bl . Residues considered part of the binding epitope demonstrated reduced binding (e.g., a KD more than 5-fold poorer than that of binding to wild type human NRPl bl) or no binding. Single point mutants P317A, D320A, T349A, K352G, Y353A, Y354A, and T413A resulted in reduced binding, whereas K35 IN and E412H resulted in no binding.

Example 8: Structure determination of MAB12 complexed with NRPl

[363] The binding epitope was also identified through crystallographic studies. MAB 12 Fab was complexed with human NRPl bl, purified by size exclusion chromatography and concentrated to lOmg/ml. Crystals were grown out of 42% PEG200, HEPES pH 7. X-ray data was collected at Argonne National Laboratories (GM/CA CAT 23ID-D) and processed using CCP4 and Phenix. NRPl bl residues within a contact distance of 3.8A from the heavy and light chain were considered part of the binding epitope and include Y297, T316, D320, E348, T349, K350, K351, K352, Y353, Y354, E412, T413, G414 and 1415.

Example 9: Analysis of amino acid modifications of MAB12

[364] Analysis of the amino acid modifications of purified MAB 12 suggested that the deletion of lysine at the C terminal of the heavy chain occurred in most of the purified antibodies and that the pyroglutamylation of glutamic acid at the N terminal of the light chain occurred in some of the purified antibodies.

INCORPORATION BY REFERENCE

[365] The entire disclosures of all patent and non-patent publications cited herein are each incorporated by reference in their entireties for all purposes.

OTHER EMBODIMENTS

[366] The disclosure set forth above may encompass multiple distinct inventions with independent utility. Although each of these inventions has been disclosed in its preferred form(s), the specific embodiments thereof as disclosed and illustrated herein are not to be considered in a limiting sense, because numerous variations are possible. The subject matter of the inventions includes all novel and nonobvious combinations and subcombinations of the various elements, features, functions, and/or properties disclosed herein. The following claims particularly point out certain combinations and subcombinations regarded as novel and nonobvious. Inventions embodied in other combinations and subcombinations of features, functions, elements, and/or properties may be claimed in this application, in applications claiming priority from this application, or in related applications. Such claims, whether directed to a different invention or to the same invention, and whether broader, narrower, equal, or different in scope in comparison to the original claims, also are regarded as included within the subject matter of the inventions of the present disclosure.

APPENDIX A: SEQUENCE REFERENCE TABLE

MAB4 VHCDR2 IINPLGGSTLYAQKFQG

MAB5 VHCDR2 IINPQGGDTSYAQKFQG

MAB6 VHCDR2 IINPQGGDTSYAQKFQG

MAB7 VHCDR2 RIKRDGSEKYYVDSVKG

MAB8 VHCDR2 IISGSGGSTYYADSVKG

MAB9 VHCDR2 IISGSGGATYYADSVKG

MAB10 VHCDR2 AISGSGGATYYADSVKG

MAB11 VHCDR2 AISGSGGATYYADSVEG

MAB12 VHCDR2 AISGSGGATYYADSVEG

MAB13 VHCDR2 EISHSGSTNYNPSLKS

MAB14 VHCDR2 EISHSGSTNYNPSLKS

MAB15 VHCDR2 DIWHSGMTNYNPSLKS

MAB1 VHFR3 RVTMTRDTPTSTVYMELS SLRSEDTAVYYC

MAB2 VH FR3 RVTMTRDASTSTVYMELS SLRSEDTAVYYC

MAB3 VHFR3 RVTMTRDTSTSTVYMELS SLRSEDTAVYYC

MAB4 VHFR3 RVTMTRDTSTSTVYMELS SLRSEDTAVYYC

MAB5 VHFR3 RVTMTRDTSTSTVYMELS SLRSEDTAVYYC

MAB6 VHFR3 RVTMTRDTSTSTVYMELS SLRSEDTAVYYC

MAB7 VHFR3 RFTISRDNAKNSLYLQMNSLRAEDTAVYYC

MAB8 VHFR3 RFTISRDNSKNTLYLQMNSLRAEDTAVYYC

MAB9 VHFR3 RFTISRDNSKNTLYLQMNSLRAEDTAVYYC

MAB10 VHFR3 RFTISRDNSKNTLYLQMNSLRAEDTAVYYC

MAB11 VHFR3 RFTISRDNSKNTLYLQMSSLRAEDTAVYYC

MAB12 VHFR3 RFTISRDNSKNTLYLQMNSLRAEDTAVYYC

MAB13 VHFR3 RVTISVDTSKNQFSLKLSSVTAADTAVYYC

MAB14 VHFR3 RVTISVDTSKNQFSLKLSPVTAADTAVYYC

MAB15 VHFR3 RVTISVDTSKNQFSLKLSSVTAADTAVYYC

MAB1 VHCDR3 ARGARRITGYGMDV

MAB2 VHCDR3 ARGARRITGYGMDV

MAB3 VHCDR3 ARDLGYYGSGMHA

MAB4 VHCDR3 ARDLGYYGSGMHV

MAB5 VHCDR3 ARDRGMYYASGFGP

MAB6 VHCDR3 ARDRGMYYASGFNP

MAB7 VHCDR3 ARDQGYKTPTDFDL

MAB8 VHCDR3 AKDPGYDSSRYYYSNYGMDV

MAB9 VHCDR3 AKDPGYDSSRYYYSNYGMDV

MAB10 VH CDR3 AKDPGYDSSRYYYSNYGMDV

MAB11 VH CDR3 AKDPGYDSSRYYYSNYGMDV

MAB12 VH CDR3 AKDPGYDSSRYYYSNYGMDV

MAB13 VHCDR3 ARARPYREPYGMDV

MAB14 VHCDR3 ARARPYREPYGMDV

MAB15 VHCDR3 ARGPGYDSSGYSRRFDP

MAB1 VHFR4 WGQGTTVTVSS

MAB2 VHFR4 WGQGTTVIVSS

MAB3 VHFR4 WGQGTLVTVSS

MAB4 VHFR4 WGQGTLVTVSS

MAB5 VHFR4 WGQGTLVTVSS

MAB6 VHFR4 WGQGTLVTVSS

MAB7 VHFR4 WGRGTLVTVSS

MAB8 VHFR4 WGQGTTVTVSS

MAB9 VHFR4 WGQGTTVTVSS

MAB10 VHFR4 WGQGTTVTVSS

MAB11 VHFR4 WGQGTTVTVSS MAB12 VH FR4 WGQGTTVTVSS

MAB13 VH FR4 WGQGTTVTVSS

MAB14 VH FR4 WGQGTTVTVSS

MAB15 VH FR4 WGQGTLVTVSS

MAB1 VL FR1 DIQMTQSPSSVSASVGDRVTITC

MAB2 VL FR1 DIQMTQSPSSVSASVGDRVTITC

MAB3 VL FR1 DIQMTQSPSSVSASVGDRVTITC

MAB4 VL FR1 DIQMTQSPSSVSASVGDRVTITC

MAB5 VL FR1 EIVMTQSPGTLSLSPGERATLSC

MAB6 VL FR1 EIVMTQSPGTLSLSPGERATLSC

MAB7 VL FR1 DIQMTQSPSSLSASVGDRVTITC

MAB8 VL FR1 DIQMTQSPSSLSASVGDRVTITC

MAB9 VL FR1 DIQMTQSPSSLSASVGDRVTITC

MAB10 VL FR1 DIQMTQSPSSLSASVGDRVTITC

MAB11 VL FR1 DIQMTQSPSSLSASVGDRVTITC

MAB12 VL FR1 DIQMTQSPSSLSASVGDRVTITC

MAB13 VL FR1 DIQLTQSPSSVSASVGDRVTITC

MAB14 VL FR1 DIQLTQSPSSVSASVGDRVTITC

MAB15 VL FR1 DIQMTQSPSTLSASVGDRVTITC

MAB1 VL CDR1 RASQGISSWLA

MAB2 VL CDR1 RASQGISSWLA

MAB3 VL CDR1 RASQGISRWLA

MAB4 VL CDR1 RASQGISRWLA

MAB5 VL CDR1 RASQSVSSSYLA

MAB6 VL CDR1 RASQSVSSSYLA

MAB7 VL CDR1 QASQDITNYLN

MAB8 VL CDR1 RASQSISSYLN

MAB9 VL CDR1 RASQSISSYLN

MAB10 VL CDR1 RASQSISSYLN

MAB11 VL CDR1 RASQSISSYLN

MAB12 VL CDR1 RASQSISSYLN

MAB13 VL CDR1 RASQDISSWLA

MAB14 VL CDR1 RASQDISSWLA

MAB15 VL CDR1 RASQSISSWLA

MAB1 VL FR2 WYQQKPGKAPKLLIY

MAB2 VL FR2 WYQQKPGKAPKLLIY

MAB3 VL FR2 WYQQKPGKAPKLLIY

MAB4 VL FR2 WYQQKPGKAPKLLIY

MAB5 VL FR2 WYQQKPGQAPRLLIY

MAB6 VL FR2 WYQQKPGQAPRLLIY

MAB7 VL FR2 WYQQKPGKAPKLLIY

MAB8 VL FR2 WYQQKPGKAPKLLIY

MAB9 VL FR2 WYQQKPGKAPKLLIY

MAB10 VL FR2 WYQQKPGKAPKLLIY

MAB11 VL FR2 WYQQKPGKAPKLLIY

MAB12 VL FR2 WYQQKPGKAPKLLIY

MAB13 VL FR2 WYQQKPGKAPKLLIY

MAB14 VL FR2 WYQQKPGKAPKLLIY

MAB15 VL FR2 WYQQKPGKAPKLLIY

MAB1 VL CDR2 AASNLQS

MAB2 VL CDR2 AASNLQS

MAB3 VL CDR2 AASSLQS

MAB4 VL CDR2 AASSLQS MAB5 VL CDR2 GASNRAT

MAB6 VL CDR2 GASNRAT

MAB7 VL CDR2 DASNLET

MAB8 VL CDR2 GASSLQS

MAB9 VL CDR2 GASSLQS

MAB10 VL CDR2 GASSLQS

MAB11 VL CDR2 GASSLQS

MAB12 VL CDR2 GASSLQS

MAB13 VL CDR2 AASSLQS

MAB14 VL CDR2 AASSLQS

MAB15 VL CDR2 KASSLES

MAB1 VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC

MAB2 VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC

MAB3 VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC

MAB4 VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC

MAB5 VL FR3 GIPDRFSGSGSGTDFTLTISRLEPEDFAVYYC

MAB6 VL FR3 GIPDRFSGSGSGTDFTLTISRLEPEDFAVYYC

MAB7 VL FR3 GVPSRFSGSGSGTDFTFTISSLQPEDIATYYC

MAB8 VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC

MAB9 VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC

MAB10 VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC

MAB11 VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC

MAB12 VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC

MAB13 VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC

MAB14 VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC

MAB15 VL FR3 GVPSRFSGSGSGTEFTLTISSLQPDDFATYYC

MAB1 VL CDR3 QQASVFPFT

MAB2 VL CDR3 QQASVFPFT

MAB3 VL CDR3 QQANLLPFT

MAB4 VL CDR3 QQANLLPFT

MAB5 VL CDR3 QQLSSFPIT

MAB6 VL CDR3 QQLSSFPIT

MAB7 VL CDR3 QQSDVLPIT

MAB8 VL CDR3 QQTYSLYT

MAB9 VL CDR3 QQTYSLYT

MAB10 VL CDR3 QQTYSLYT

MAB11 VL CDR3 QQTYSLYT

MAB12 VL CDR3 QQTYSLYT

MAB13 VL CDR3 QQELAFPRT

MAB14 VL CDR3 QQELAFPRT

MAB15 VL CDR3 QQLNSYPPT

MAB1 VL FR4 FGGGTKVEIK

MAB2 VL FR4 FGGGTKVEIK

MAB3 VL FR4 FGGGTKVEIK

MAB4 VL FR4 FGGGTKVEIK

MAB5 VL FR4 FGGGTKVEIK

MAB6 VL FR4 FGGGTKVEIK

MAB7 VL FR4 FGGGTKVEIK

MAB8 VL FR4 FGGGTKVEIK

MAB9 VL FR4 FGGGTKVEIK

MAB10 VL FR4 FGGGTKVEIK

MAB11 VL FR4 FGGGTKVEIK

MAB12 VL FR4 FGGGTKVEIK 84 MAB13 VL FR4 FGGGTKVEIK

84 MAB14 VL FR4 FGGGTKVEIK

84 MAB15 VL FR4 FGGGTKVEIK

85 MAB1 VH Full QVQLVQSGAGVKKPGASVKVSCKASGYTFRSYYMLWV

RQAPGQGLEWMGIIDPSDGSTSYAQKFQGRVTMTRDTPT STVYMELS SLRSEDTA VYYCARGARRITGYGMD VWGQG TTVTVSS

86 MAB2 VH Full QVQLVQSGAEVKKPGASVKVSCKASGYTFRSYYMLWVR

QAPGQGLEWMGIIDPSDGSTSYAQKFQGRVTMTRDASTS TVYMELS SLRSEDTA VYYCARGARRITGYGMD VWGQGT TVIVSS

87 MAB3 VH Full QAQLVQSGAEVKKPGASVKVSCKASGYTFSRYYMHWV

RQAPGQGLEWMGIINPLGGSTLYAQKFQGRVTMTRDTST STVYMELS SLRSEDTA VYYCARDLGYYGSGMHAWGQG TLVTVSS

88 MAB4 VH Full QVQLVQSGAEVKKPGASVKVSCKASGYTFSRYYMHWV

RQAPGQGLEWMGIINPLGGSTLYAQKFQGRVTMTRDTST STVYMELS SLRSEDTA VYYCARDLGYYGSGMHVWGQG TLVTVSS

89 MAB5 VH Full QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWV

RQAPGQGLEWMGIINPQGGDTSYAQKFQGRVTMTRDTS TSTVYMELSSLRSEDTAVYYCARDRGMYYASGFGPWGQ GTLVTVSS

90 MAB6 VH Full QVQLVQSGAKVKKPGASVKVSCKASGYTFTSYYMHWV

RQVPGQGLEWMGIINPQGGDTSYAQKFQGRVTMTRDTS TSTVYMELSSLRSEDTAVYYCARDRGMYYASGFNPWGQ GTLVTVSS

91 MAB7 VH Full EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMEWVR

Q APGKGLEWVARIKRDGSEKYYVD S VKGRFΉ SRDNAK NSLYLQMNSLRAEDTAVYYCARDQGYKTPTDFDLWGRG TLVTVSS

92 MAB8 VH Full EVQLLESGGGLVQPGGSLRLSCAASGFTFASYAMVWVR

QAPGKGLEWVSIISGSGGSTYYADSVKGRFTISRDNSKNT LYLQMNSLRAEDTAVYYCAKDPGYDSSRYYYSNYGMD VWGQGTTVTVSS

93 MAB9 VH Full EVQLLESGGGLVQPGGSLRLSCAASGFTFKSYAMVWVR

QAPGKGLEWVSIISGSGGATYYADSVKGRFTISRDNSKNT LYLQMNSLRAEDTAVYYCAKDPGYDSSRYYYSNYGMD VWGQGTTVTVSS

94 MAB10 VH Full EVQLLESGGGLVQPGGSLRLSCAASGFTFSSVAMVWVRQ

APGKGLEWVSAISGSGGATYYADSVKGRFTISRDNSKNT LYLQMNSLRAEDTAVYYCAKDPGYDSSRYYYSNYGMD VWGQGTTVTVSS

95 MAB11 VH Full EVQLLESGGGLVQPGGSLRLSCAASGFTFSSVAMVWVRQ

APGKGLEWVSAISGSGGATYYADSVEGRFTISRDNSKNT LYLQMSSLRAEDTAVYYCAKDPGYDSSRYYYSNYGMDV WGQGTTVTVSS

96 MAB12 VH Full EVQLLESGGGLVQPGGSLRLSCAASGFTFSSVAMVWVRQ

APGKGLEWVSAISGSGGATYYADSVEGRFTISRDNSKNT LYLQMNSLRAEDTAVYYCAKDPGYDSSRYYYSNYGMD VWGQGTTVTVSS

97 MAB13 VH Full QVQLQQWGAGLLKPSETLSLTCAVYGGSFRGYYWEWIR

QPPGKGLEWIGEISHSGSTNYNPSLKSRVTISVDTSKNQFS LKLSSVTAADTAVYYCARARPYREPYGMDVWGQGTTVT vss

98 MAB14 VH Full QVQLQQWGAGLLKPSETLSLTCAVYGGSFPvGYYWEWSPv

QPPGKGLEWIGEISHSGSTNYNPSLKSRVTISVDTSKNQFS LKLSPVTAADTAVYYCARARPYREPYGMDVWGQGTTVT VSS

99 MAB15 VH Full QVQLQQWGAGLLKPSETLSLTCAVYGGSFVKYYWSWIR

QPPGKGLEWIGDIWHSGMTNYNPSLKSRVΉSVDTSKNQ FSLKLSSVTAADTAVYYCARGPGYDSSGYSRRFDPWGQG TLVTVSS

100 MAB1 VL Full DIQMTQSPSSVSASVGDRVTITCRASQGISSWLAWYQQKP

GKAPKLLIYAASNLQSGVPSRFSGSGSGTDFTLTISSLQPE

DFATYYCQQASVFPFTFGGGTKVEIK

100 MAB2 VL Full DIQMTQSPSSVSASVGDRVTITCRASQGISSWLAWYQQKP

GKAPKLLIYAASNLQSGVPSRFSGSGSGTDFTLTISSLQPE

DFATYYCQQASVFPFTFGGGTKVEIK

101 MAB3 VL Full DIQMTQSPSSVSASVGDRVTITCRASQGISRWLAWYQQK

PGKAPKLLIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPE

DFATYYCQQANLLPFTFGGGTKVEIK

101 MAB4 VL Full DIQMTQSPSSVSASVGDRVTITCRASQGISRWLAWYQQK

PGKAPKLLIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPE

DFATYYCQQANLLPFTFGGGTKVEIK

102 MAB5 VL Full EIVMTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQK

PGQAPRLLIYGASNRATGIPDRFSGSGSGTDFTLTISRLEPE DFAVYYCQQLSSFPITFGGGTKVEIK

102 MAB6 VL Full EIVMTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQK

PGQAPRLLIYGASNRATGIPDRFSGSGSGTDFTLTISRLEPE DFAVYYCQQLSSFPITFGGGTKVEIK

103 MAB7 VL Full DIQMTQSPSSLSASVGDRVTITCQASQDITNYLNWYQQKP

GKAPKLLIYDASNLETGVPSRFSGSGSGTDFTFTISSLQPE

DIATYYCQQSDVLPITFGGGTKVEIK

104 MAB8 VL Full DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKP

GKAPKLLIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPE DFATYYCQQTYSLYTFGGGTKVEIK

104 MAB9 VL Full DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKP

GKAPKLLIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPE DFATYYCQQTYSLYTFGGGTKVEIK

104 MAB10 VL Full DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKP

GKAPKLLIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPE DFATYYCQQTYSLYTFGGGTKVEIK

104 MAB11 VL Full DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKP

GKAPKLLIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPE DFATYYCQQTYSLYTFGGGTKVEIK

104 MAB12 VL Full DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKP

GKAPKLLIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPE DFATYYCQQTYSLYTFGGGTKVEIK

105 MAB13 VL Full DIQLTQSPSSVSASVGDRVTITCRASQDISSWLAWYQQKP

GKAPKLLIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPE DF ATYYCQ QELAFPRTFGGGTKVEIK

105 MAB14 VL Full DIQLTQSPSSVSASVGDRVTITCRASQDISSWLAWYQQKP

GKAPKLLIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPE DF ATYYCQ QELAFPRTFGGGTKVEIK 106 MAB15 VL Full DIQMTQSPSTLSASVGDRVTITCRASQSISSWLAWYQQKP

GKAPKLLIYKASSLESGVPSRFSGSGSGTEFTLTISSLQPDD FATYYCQQLNSYPPTFGGGTKVEIK

107 MAB1 HC Full- QVQLVQSGAGVKKPGASVKVSCKASGYTFRSYYMLWV length RQAPGQGLEWMGIIDPSDGSTSYAQKFQGRVTMTRDTPT IgG4 STVYMELS SLRSEDTA VYYCARGARRITGYGMDVWGQG S228P TTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFP

EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSS

SLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEF

LGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQ

FNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQD

WLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLP

PSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPEN Y

KTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHE

ALHNHYTQKSLSLSLGK

108 MAB2 HC Full- QVQLVQSGAEVKKPGASVKVSCKASGYTFRSYYMLWVR length QAPGQGLEWMGIIDPSDGSTSYAQKFQGRVTMTRDASTS IgG4 TVYMELSSLRSEDTAVYYCARGARRITGYGMDVWGQGT S228P TVIVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPE

PVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSS

LGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEF

LGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQ

FNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQD

WLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLP

PSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPEN Y

KTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHE

ALHNHYTQKSLSLSLGK

109 MAB3 HC Full- QAQLVQSGAEVKKPGASVKVSCKASGYTFSRYYMHWV length RQAPGQGLEWMGIINPLGGSTLYAQKFQGRVTMTRDTST IgG4 STVYMELS SLRSEDTA VYYCARDLGYYGSGMHAWGQG S228P TLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFP

EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSS

SLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEF

LGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQ

FNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQD

WLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLP

PSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPEN Y

KTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHE

ALHNHYTQKSLSLSLGK

110 MAB4 HC Full- QVQLVQSGAEVKKPGASVKVSCKASGYTFSRYYMHWV length RQAPGQGLEWMGIINPLGGSTLYAQKFQGRVTMTRDTST IgG4 STVYMELS SLRSEDTA VYYCARDLGYYGSGMHVWGQG S228P TLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFP

EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSS

SLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEF

LGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQ

FNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQD

WLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLP

PSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY

KTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHE

ALHNHYTQKSLSLSLGK

111 MAB5 HC Full- QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWV length RQAPGQGLEWMGIINPQGGDTSYAQKFQGRVTMTRDTS

TSTVYMELSSLRSEDTAVYYCARDRGMYYASGFGPWGQ IgG4 GTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDY S228P FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVP

SSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAP

EFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPE

VQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLH

QDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVY

TLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPE

N YKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSV

MHEALHNHYTQKSLSLSLGK

112 MAB6 HC Full- QVQLVQSGAKVKKPGASVKVSCKASGYTFTSYYMHWV length RQVPGQGLEWMGIINPQGGDTSYAQKFQGRVTMTRDTS IgG4 TSTVYMELSSLRSEDTAVYYCARDRGMYYASGFNPWGQ S228P GTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDY

FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVP

SSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAP

EFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPE

VQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLH

QDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVY

TLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPE

N YKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSV

MHEALHNHYTQKSLSLSLGK

113 MAB7 HC Full- EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMEWVR length QAPGKGLEWVARIKRDGSEKYYVDSVKGRFTISRDNAK IgG4 NSLYLQMNSLRAEDTAVYYCARDQGYKTPTDFDLWGRG S228P TLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFP

EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSS

SLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEF

LGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQ

FNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQD

WLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLP

PSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPEN Y

KTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHE

ALHNHYTQKSLSLSLGK

114 MAB8 HC Full- EVQLLESGGGLVQPGGSLRLSCAASGFTFASYAMVWVR length QAPGKGLEWVSIISGSGGSTYYADSVKGRFTISRDNSKNT IgG4 LYLQMNSLRAEDTAVYYCAKDPGYDSSRYYYSNYGMD S228P VWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCL

VKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSV

VTVP S S SLGTKTYTCNVDHKP SNTKVDKRVESKYGPPCP

PCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQ

EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVL

TVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPRE

PQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESN

GQPEN YKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVF

SCSVMHEALHNHYTQKSLSLSLGK

115 MAB9 HC Full- EVQLLESGGGLVQPGGSLRLSCAASGFTFKSYAMVWVR length QAPGKGLEWVSIISGSGGATYYADSVKGRFTISRDNSKNT IgG4 LYLQMNSLRAEDTAVYYCAKDPGYDSSRYYYSNYGMD S228P VWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCL

VKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSV

VTVP S S SLGTKTYTCNVDHKP SNTKVDKRVESKYGPPCP

PCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQ

EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVL

TVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPRE PQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESN

GQPEN YKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVF SCSVMHEALHNHYTQKSLSLSLGK

116 MAB10 HC Full- EVQLLESGGGLVQPGGSLRLSCAASGFTFSSVAMVWVPvQ

length APGKGLEWVSAISGSGGATYYADSVKGRFTISRDNSKNT IgG4 LYLQMNSLRAEDTAVYYCAKDPGYDSSRYYYSNYGMD S228P VWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCL

VKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSV

VTVP S S SLGTKTYTCNVDHKP SNTKVDKRVESKYGPPCP

PCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQ

EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVL

TVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPRE

PQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESN

GQPEN YKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVF

SCSVMHEALHNHYTQKSLSLSLGK

117 MAB11 HC Full- EVQLLESGGGLVQPGGSLRLSCAASGFTFSSVAMVWVRQ

length APGKGLEWVSAISGSGGATYYADSVEGRFTISRDNSKNT IgG4 LYLQMSSLRAEDTAVYYCAKDPGYDSSRYYYSNYGMDV S228P WGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLV

KDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVV

TVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPC

PAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQE

DPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLT

VLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREP

QVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNG

QPEN YKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFS

CSVMHEALHNHYTQKSLSLSLGK

118 MAB12 HC Full- EVQLLESGGGLVQPGGSLRLSCAASGFTFSSVAMVWVRQ

length APGKGLEWVSAISGSGGATYYADSVEGRFTISRDNSKNT IgG4 LYLQMNSLRAEDTAVYYCAKDPGYDSSRYYYSNYGMD S228P VWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCL

VKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSV

VTVP S S SLGTKTYTCNVDHKP SNTKVDKRVESKYGPPCP

PCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQ

EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVL

TVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPRE

PQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESN

GQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVF

SCSVMHEALHNHYTQKSLSLSLGK

119 MAB13 HC Full- QVQLQQWGAGLLKPSETLSLTCAVYGGSFRGYYWEWIR

length QPPGKGLEWIGEISHSGSTNYNPSLKSRVTISVDTSKNQFS IgG4 LKLSSVTAADTAVYYCARARPYREPYGMDVWGQGTTVT S228P VSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVT

VSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGT

KTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGG

P S VFLFPPKPKDTLMI SRTPEVTCVVVD VS QEDPEVQFNW

YVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLN

GKEYKCKVSNKGLPSSIEKΉSKAKGQPREPQVYTLPPSQ

EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT

PPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALH

NHYTQKSLSLSLGK

120 MAB14 HC Full- QVQLQQWGAGLLKPSETLSLTCAVYGGSFRGYYWEWSR

length QPPGKGLEWIGEISHSGSTNYNPSLKSRVTISVDTSKNQFS

LKLSPVTAADTAVYYCARARPYREPYGMDVWGQGTTVT IgG4 VSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVT S228P VSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGT

KTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGG

P S VFLFPPKPKDTLMI SRTPEVTCVVVD VS QEDPEVQFNW

YVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLN

GKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQ

EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPEN YKTT

PPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALH

NHYTQKSLSLSLGK

121 MAB15 HC Full- QVQLQQWGAGLLKPSETLSLTCAVYGGSFVKYYWSWIR length QPPGKGLEWIGDIWHSGMTNYNPSLKSRVΉSVDTSKNQ IgG4 FSLKLSSVTAADTAVYYCARGPGYDSSGYSRRFDPWGQG S228P TLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFP

EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSS

SLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEF

LGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQ

FNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQD

WLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLP

PSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPEN Y

KTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHE

ALHNHYTQKSLSLSLGK

122 MAB1 LC Full- DIQMTQSPSSVSASVGDRVTITCRASQGISSWLAWYQQKP length, GKAPKLLIYAASNLQSGVPSRFSGSGSGTDFTLTISSLQPE human DFATYYCQQASVFPFTFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLN FYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

122 MAB2 LC Full- DIQMTQSPSSVSASVGDRVTITCRASQGISSWLAWYQQKP length, GKAPKLLIYAASNLQSGVPSRFSGSGSGTDFTLTISSLQPE human DFATYYCQQASVFPFTFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLN FYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

123 MAB3 LC Full- DIQMTQSPSSVSASVGDRVTITCRASQGISRWLAWYQQK length, PGKAPKLLIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPE human DFATYYCQQANLLPFTFGGGTKVEIKRTVAAPSVFIFPPS kappa DEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNS constant QESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTH

QGLSSPVTKSFNRGEC

123 MAB4 LC Full- DIQMTQSPSSVSASVGDRVTITCRASQGISRWLAWYQQK length, PGKAPKLLIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPE human DFATYYCQQANLLPFTFGGGTKVEIKRTVAAPSVFIFPPS kappa DEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNS constant QESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTH

QGLSSPVTKSFNRGEC

124 MAB5 LC Full- EIVMTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQK length, PGQAPRLLIYGASNRATGIPDRFSGSGSGTDFTLTISRLEPE human DFAVYYCQQLSSFPITFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

124 MAB6 LC Full- EIVMTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQK length, PGQAPRLLIYGASNRATGIPDRFSGSGSGTDFTLTISRLEPE human DFAVYYCQQLSSFPITFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLN FYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

125 MAB7 LC Full- DIQMTQSPSSLSASVGDRVTITCQASQDITNYLNWYQQKP length, GKAPKLLIYDASNLETGVPSRFSGSGSGTDFTFTISSLQPE human DIATYYCQQSDVLPITFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLN FYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

126 MAB8 LC Full- DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKP length, GKAPKLLIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPE human DFATYYCQQTYSLYTFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

126 MAB9 LC Full- DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKP length, GKAPKLLIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPE human DFATYYCQQTYSLYTFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

126 MAB10 LC Full- DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKP length, GKAPKLLIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPE human DFATYYCQQTYSLYTFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

126 MAB11 LC Full- DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKP length, GKAPKLLIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPE human DFATYYCQQTYSLYTFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

126 MAB12 LC Full- DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKP length, GKAPKLLIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPE human DFATYYCQQTYSLYTFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

127 MAB13 LC Full- DIQLTQSPSSVSASVGDRVTITCRASQDISSWLAWYQQKP length, GKAPKLLIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPE human DFATYYCQQELAFPRTFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

127 MAB14 LC Full- DIQLTQSPSSVSASVGDRVTITCRASQDISSWLAWYQQKP length, GKAPKLLIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPE human DFATYYCQQELAFPRTFGGGTKVEIKRTVAAPSVFIFPPSD kappa EQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQ constant ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

128 MAB15 LC Full- DIQMTQSPSTLSASVGDRVTITCRASQSISSWLAWYQQKP length GKAPKLLIYKASSLESGVPSRFSGSGSGTEFTLTISSLQPDD

FATYYCQQLNSYPPTFGGGTKVEIKRTVAAPSVFIFPPSDE QLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQE SVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG LSSPVTKSFNRGEC

GenBank ATGGAGAGGGGGCTGCCGCTCCTCTGCGCCGTGCTCGC

Accession CCTCGTCCTCGCCCCGGCCGGCGCTTTTCGCAACGATA

No. AATGTGGCGATACTATAAAAATTGAAAGCCCCGGGTA

NM 0038 CCTTACATCTCCTGGTTATCCTCATTCTTATCACCCAAG 73.5 TGAAAAATGCGAATGGCTGATTCAGGCTCCGGACCCAT

(correspon ACCAGAGAATTATGATCAACTTCAACCCTCACTTCGAT ds to TTGGAGGACAGAGACTGCAAGTATGACTACGTGGAAG

NP 00386 TCTTCGATGGAGAAAATGAAAATGGACATTTTAGGGG 4.4). AAAGTTCTGTGGAAAGATAGCCCCTCCTCCTGTTGTGT

ACGAAACACATGGTGCAGGATTTTCCATACGTTATGAA

ATTTTCAAGAGAGGTCCTGAATGTTCCCAGAACTACAC

AACACCTAGTGGAGTGATAAAGTCCCCCGGATTCCCTG

AAAAATATCCCAACAGCCTTGAATGCACTTATATTGTC

TTTGCGCCAAAGATGTCAGAGATTATCCTGGAATTTGA

AAGCTTTGACCTGGAGCCTGACTCAAATCCTCCAGGGG

GGATGTTCTGTCGCTACGACCGGCTAGAAATCTGGGAT

GGATTCCCTGATGTTGGCCCTCACATTGGGCGTTACTG

TGGACAGAAAACACCAGGTCGAATCCGATCCTCATCG

AGCAAAAGAAGGTTTCTCAGCAAACTACAGTGTCTTGC

AGAGCAGTGTCTCAGAAGATTTCAAATGTATGGAAGCT

CTGGGCATGGAATCAGGAGAAATTCATTCTGACCAGAT

CACAGCTTCTTCCCAGTATAGCACCAACTGGTCTGCAG

AGCGCTCCCGCCTGAACTACCCTGAGAATGGGTGGACT

CCCGGAGAGGATTCCTACCGAGAGTGGATACAGGTAG

ACTTGGGCCTTCTGCGCTTTGTCACGGCTGTCGGGACA

CAGGGCGCCATTTCAAAAGAAACCAAGAAGAAATATT

ATGTCAAGACTTACAAGATCGACGTTAGCTCCAACGGG

GAAGACTGGATCACCATAAAAGAAGGAAACAAACCTG

TTCTCTTTCAGGGAAACACCAACCCCACAGATGTTGTG

GTTGCAGTATTCCCCAAACCACTGATAACTCGATTTGT

CCGAATCAAGCCTGCAACTTGGGAAACTGGCATATCTA

TGAGATTTGAAGTATACGGTTGCAAGATAACAGATTAT

CCTTGCTCTGGAATGTTGGGTATGGTGTCTGGACTTATT

TCTGACTCCCAGATCACATCATCCAACCAAGGGGACAG

AAACTGGATGCCTGAAAACATCCGCCTGGTAACCAGTC

GCTCTGGCTGGGCACTTCCACCCGCACCTCATTCCTAC

ATCAATGAGTGGCTCCAAATAGACCTGGGGGAGGAGA

AGATCGTGAGGGGCATCATCATTCAGGGTGGGAAGCA

CCGAGAGAACAAGGTGTTCATGAGGAAGTTCAAGATC

GGGTACAGCAACAACGGCTCGGACTGGAAGATGATCA

TGGATGACAGCAAACGCAAGGCGAAGTCTTTTGAGGG

CAACAACAACTATGATACACCTGAGCTGCGGACTTTTC

CAGCTCTCTCCACGCGATTCATCAGGATCTACCCCGAG

AGAGCCACTCATGGCGGACTGGGGCTCAGAATGGAGC

TGCTGGGCTGTGAAGTGGAAGCCCCTACAGCTGGACC

GACCACTCCCAACGGGAACTTGGTGGATGAATGTGAT

GACGACCAGGCCAACTGCCACAGTGGAACAGGTGATG

ACTTCCAGCTCACAGGTGGCACCACTGTGCTGGCCACA

GAAAAGCCCACGGTCATAGACAGCACCATACAATCAG

AGTTTCCAACATATGGTTTTAACTGTGAATTTGGCTGG GGCTCTCACAAGACCTTCTGCCACTGGGAACATGACAA

TCACGTGCAGCTCAAGTGGAGTGTGTTGACCAGCAAG

ACGGGACCCATTCAGGATCACACAGGAGATGGCAACT

TCATCTATTCCCAAGCTGACGAAAATCAGAAGGGCAA

AGTGGCTCGCCTGGTGAGCCCTGTGGTTTATTCCCAGA

ACTCTGCCCACTGCATGACCTTCTGGTATCACATGTCT

GGGTCCCACGTCGGCACACTCAGGGTCAAACTGCGCTA

CCAGAAGCCAGAGGAGTACGATCAGCTGGTCTGGATG

GCCATTGGACACCAAGGTGACCACTGGAAGGAAGGGC

GTGTCTTGCTCCACAAGTCTCTGAAACTTTATCAGGTG

ATTTTCGAGGGCGAAATCGGAAAAGGAAACCTTGGTG

GGATTGCTGTGGATGACATTAGTATTAATAACCACATT

TCACAAGAAGATTGTGCAAAACCAGCAGACCTGGATA

AAAAGAACCCAGAAATTAAAATTGATGAAACAGGGAG

CACGCCAGGATACGAAGGTGAAGGAGAAGGTGACAAG

AACATCTCCAGGAAGCCAGGCAATGTGTTGAAGACCTT

AGACCCCATCCTCATCACCATCATAGCCATGAGTGCCC

TGGGGGTCCTCCTGGGGGCTGTCTGTGGGGTCGTGCTG

TACTGTGCCTGTTGGCATAATGGGATGTCAGAAAGAAA

CTTGTCTGCCCTGGAGAACTATAACTTTGAACTTGTGG

ATGGTGTGAAGTTGAAAAAAGACAAACTGAATACACA

GAGTACTTATTCGGAGGCATGA

130 hNRP-1 Genbank MERGLPLLCAVLALVLAPAGAFRNDKCGDTIKIESPGYLT Protein NP 00386 SPGYPHSYHPSEKCEWLIQAPDPYQRIMINFNPHFDLEDR

4.4. DCKYDYVEVFDGENENGHFRGKFCGKIAPPPVVSSGPFLF

IKFVSDYETHGAGFSIRYEIFKRGPECSQNYTTPSGVIKSP

GFPEKYPNSLECTYIVFAPKMSEIILEFESFDLEPDSNPPGG

MFCRYDRLEIWDGFPDVGPHIGRYCGQKTPGRIRSSSGIL

SMVFYTDSAIAKEGFSANYSVLQSSVSEDFKCMEALGME

SGEIHSDQITASSQYSTNWSAERSRLNYPENGWTPGEDSY

REWIQVDLGLLRFVTAVGTQGAISKETKKKYYVKTYKID

VSSNGEDWITIKEGNKPVLFQGNTNPTDVVVAVFPKPLIT

RFVRIKPATWETGISMRFEVYGCKITDYPCSGMLGMVSG

LISDSQITSSNQGDRNWMPENIRLVTSRSGWALPPAPHSYI

NEWLQIDLGEEKIVRGIIIQGGKHRENKVFMRKFKIGYSN

NGSDWKMIMDDSKRKAKSFEGNNNYDTPELRTFPALSTR

FIRIYPERATHGGLGLRMELLGCEVEAPTAGPTTPNGNLV

DECDDDQANCHSGTGDDFQLTGGTTVLATEKPTVIDSTI

QSEFPTYGFNCEFGWGSHKTFCHWEHDNHVQLKWSVLT

SKTGPIQDHTGDGNFIYSQADENQKGKVARLVSPVVYSQ

NSAHCMTFWYHMSGSHVGTLRVKLRYQKPEEYDQLVW

MAIGHQGDHWKEGRVLLHKSLKLYQVIFEGEIGKGNLG

GIAVDDISINNHISQEDCAKPADLDKKNPEIKIDETGSTPG

YEGEGEGDKNISRKPGNVLKTLDPILITIIAMSALGVLLGA

VCGVVLYCACWHNGMSERNLSALENYNFELVDGVKLK

KDKLNTQSTYSEA

131 cNRP-1 DNA: ATGGAGAAGGGGTTGCCGCTCCTCTGCGCCGCGCTCGC

Genbank CCTCGCCCTCGCCCCGGCCGGCGCTTTTCGCAACGATA Acc No. AATGTGGCGATACTATAAAAATTGAAAGCCCCGGGTA XM 0055 CCTTACATCTCCTGGTTATCCTCATTCTTATCACCCAAG 64935.2 TGAAAAATGTGAATGGCTGATTCAGGCTCCGGACCCAT

ACCAGAGAATTATGATCAACTTCAACCCTCACTTCGAT

TTGGAGGACAGAGATTGCAAGTATGACTACGTGGAAG

TCTTCGATGGAGAAAATGAAAATGGACGTTTATGGGG

AAAGTTCTGTGGAAAGATAGCCCCTCCTCCTGTTGTGT ACGAAACACACGGTGCAGGATTTTCCATACGTTATGAA

ATTTTCAAGAGAGGTCCTGAATGTTCCCAGAACTACAC

AACACCTAGTGGAGTGATAAAGTCCCCCGGATTCCCTG

AAAAATATCCCAACAGCCTTGAATGCACTTATATTGTC

TTTGCACCAAAGATGTCAGAGATTATCCTGGAATTTGA

AAGCTTTGACCTGGAGCCTGACTCAAATCCTCCAGGGG

GGATGTTCTGTCGCTACGACCGGCTGGAAATCTGGGAT

GGATTCCCTGACGTTGGCCCTCACATTGGGCGTTACTG

TGGACAGAAAACACCAGGTCGAATCCGATCCTCATCG

AGCAAAAGAAGGTTTCTCAGCAAACTACAGTGTCTTGC

AGAGCAGTGTCTCAGAAGATTTCAAATGTATGGAAGCT

GTGGGCATGGAATCAGGAGAAATTCATTCTGACCAGA

TCACAGCTTCTTCCCAGTACAGCACCAACTGGTCTGCA

GAGCGCTCCCGCCTGAACTATCCTGAGAATGGGTGGAC

TCCCGGAGAAGATTCCTACCGAGAGTGGATACAGGTG

GACTTGGGCCTTCTACGCTTCGTTACGGCTGTCGGGAC

ACAGGGCGCCATTTCAAAAGAAACCAAGAAGAAATAT

TATGTCAAGACTTACAAAATTGACATTAGCTCCAACGG

GGAAGACTGGATCACCATAAAAGAAGGAAACAAACCT

GTTCTCTTTCAGGGAAACACCAACCCCACAGACGTTGT

GGTTGCAGTATTCCCCAAGCCACTGATAACTCGATTTG

TCCGAATCAAGCCTGCAACTTGGGAAACTGGCATATCT

CTGAGATTTGAAGTATATGGTTGCAAGATAACAGATTA

TCCTTGCTCCGGAATGTTGGGTATGGTGTCTGGACTTA

TTTCTGACTCCCAGATCACATCATCCAACCAAGGGGAC

AGAAACTGGATGCCTGAAAACATCCGCCTGGTAACCA

GTCGCTCCGGCTGGGCACTGCCACCCGCACCTCATTCC

TACGTCAATGAGTGGCTCCAAATAGACCTGGGGGAGG

AGAAGATCGTGAGGGGCATCATCATTCAGGGTGGGAA

GCACCGAGAGAACAAGGTATTCATGAGGAAGTTCAAG

ATCGGGTACAGCAACAACGGCTCCGACTGGAAGATGA

TCATGGACGACAGCAAACGCAAGGCAAAGTCTTTTGA

GGGCAACAACAACTATGACACACCTGAGCTGCGGACT

TTTCCAGCTCTCTCCACGCGATTCATCAGGATCTACCCC

GAGAGAGCCACTCATGGCGGACTGGGGCTCCGAATGG

AGCTGCTGGGCTGTGAAGTGGAAGCCCCTACAGCTGG

ACCGACCACTCCCAACGGGAACCCGGTGGATGAATGT

GATGACGACCAGGCCAACTGCCACAGTGGAACAGGTG

ATGACTTCCAGCTCACAGGTGGCACCACTGTGCTGGCC

ACAGAAAAGCCCACGGTCATAGACAGCACCATACAAT

CAGAGTTTCCTACATATGGTTTTAACTGTGAATTTGGCT

GGGGCTCTCACAAGACCTTCTGCCACTGGGAACATGAC

AATCACGTGCAGCTCAAGTGGAGTGTGTTGACCAGCA

AGACGGGACCCATTCAGGATCACACAGGAGATGGCAA

CTTCATCTATTCCCAAGCTGATGAAAATCAGAAGGGCA

AAGTGGCTCGCCTGGTGAGCCCTGTGGTTTATTCCCAG

AACTCTGCCCACTGCATGACCTTCTGGTATCACATGTC

TGGGTCCCACGTCGGCACACTCAGGGTCAAACTGCGCT

ACCAGAAGCCAGAGGAGTACGATCAGCTGGTCTGGAT

GGCCATTGGACACCAAGGTGACCACTGGAAGGAAGGG

CGTGTCTTGCTTCACAAGTCTCTGAAACTTTATCAGGT

GATTTTCGAGGGCGAAATCGGAAAAGGAAACCTTGGT

GGGATTGCTGTGGATGACATTAGTATCAATAACCACAT TTCACAAGAAGATTGTGCAAAACCAGCAGACCTGGAT

AAAAAGAACCCAGAAATTAAAATTGATGAAACAGGGA

GCACACCAGGATATGAAGGTGAAGGAGAAGGTGACAA

GAACATCTCCAGGAAACCAGGCAATGTGTTGAAGACC

TTAGACCCCATCCTCATCACCATCATAGCCATGAGCGC

CCTGGGGGTCCTCCTGGGGGCTGTGTGCGGGGTCGTGC

TGTACTGTGCCTGTTGGCATAATGGGATGTCAGAAAGA

AACTTGTCTGCCCTGGAGAACTATAACTTTGAACTTGT

GGACGGTGTGAAGTTGAAAAAAGACAAACTGAATACA

CAGAGTACTTATTCGGAGGCATGA

132 cNRP-1 Protein: MEKGLPLLCAALALALAPAGAFRNDKCGDTIKIESPGYLT

UniProtK SPGYPHSYHPSEKCEWLIQAPDPYQRIMINFNPHFDLEDR B - DCKYDYVEVFDGENENGRLWGKFCGKIAPPPVVSSGQFL

G7PEQ1 FIKFVSDYETHGAGFSIRYEIFKRGPECSQNYTTPSGVIKSP

GFPEKYPNSLECTYIVFAPKMSEIILEFESFDLEPDSNPPGG

MFCRYDRLEIWDGFPDVGPHIGRYCGQKTPGRIRSSSGIL

SMVFYTDSAIAKEGFSANYSVLQSSVSEDFKCMEAVGME

SGEIHSDQITASSQYSTNWSAERSRLNYPENGWTPGEDSY

REWIQVDLGLLRFVTAVGTQGAISKETKKKYYVKTYKID

ISSNGEDWITIKEGNKPVLFQGNTNPTDVVVAVFPKPLITR

F VRIKPATWETGI SLRFEVYGCKITDYPC SGMLGMVSGLI

SDSQITSSNQGDRNWMPENIRLVTSRSGWALPPAPHSYV

NEWLQIDLGEEKIVRGIIIQGGKHRENKVFMRKFKIGYSN

NGSDWKMIMDDSKRKAKSFEGNNNYDTPELRTFPALSTR

FIRIYPERATHGGLGLRMELLGCEVEAPTAGPTTPNGNPV

DECDDDQANCHSGTGDDFQLTGGTTVLATEKPTVIDSTI

QSEFPTYGFNCEFGWGSHKTFCHWEHDNHVQLKWSVLT

SKTGPIQDHTGDGNFIYSQADENQKGKVARLVSPVVYSQ

NSAHCMTFWYHMSGSHVGTLRVKLRYQKPEEYDQLVW

MAIGHQGDHWKEGRVLLHKSLKLYQVIFEGEIGKGNLG

GIAVDDISINNHISQEDCAKPADLDKKNPEIKIDETGSTPG

YEGEGEGDKNISRKPGNVLKTLDPILITIIAMSALGVLLGA

VCGVVLYCACWHNGMSERNLSALENYNFELVDGVKLK

KDKLNTQSTYSEA

133 niNRP- 1 GenBank ATGGAGAGGGGGCTGCCGTTGCTGTGCGCCACGCTCGC

Acc. No. CCTTGCCCTCGCCCTGGCGGGCGCTTTCCGCAGCGACA NM00873 AATGTGGCGGGACCATAAAAATCGAAAACCCAGGGTA 7 CCTCACATCTCCCGGTTACCCTCATTCTTACCATCCAAG

TGAGAAGTGTGAATGGCTAATCCAAGCTCCGGAACCCT

ACCAGAGAATCATGATCAACTTCAACCCACATTTCGAT

TTGGAGGACAGAGACTGCAAGTATGACTACGTGGAAG

TAATCGATGGGGAGAATGAAGGCGGCCGCCTGTGGGG

GAAGTTCTGTGGGAAGATTGCACCTTCTCCTGTGGTGT

CTTCAGGGCCCTTTCTCTTCATCAAATTTGTCTCTGACT

ATGAGACACATGGGGCAGGGTTTTCCATCCGCTATGAA

ATCTTCAAGAGAGGGCCCGAATGTTCTCAGAACTATAC

AGCACCTACTGGAGTGATAAAGTCCCCTGGGTTCCCTG

AAAAATACCCCAACAGCTTGGAGTGCACCTACATCATC

TTTGCACCAAAGATGTCTGAGATAATCCTGGAGTTTGA

AAGTTTTGACCTGGAGCAAGACTCGAATCCTCCCGGAG

GAATGTTCTGTCGCTATGACCGGCTGGAGATCTGGGAT

GGATTCCCTGAAGTTGGCCCTCACATTGGGCGTTATTG

TGGGCAGAAAACTCCTGGCCGGATCCGCTCCTCTTCAG

GCGTTCTATCCATGGTCTTTTACACTGACAGCGCAATA

GCAAAAGAAGGTTTCTCAGCCAACTACAGTGTGCTACA GAGCAGCATCTCTGAAGATTTTAAGTGTATGGAGGCTC

TGGGCATGGAATCTGGAGAGATCCATTCTGATCAGATC

ACTGCATCTTCACAGTATGGTACCAACTGGTCTGTAGA

GCGCTCCCGCCTGAACTACCCTGAAAATGGGTGGACTC

CAGGAGAAGACTCCTACAAGGAGTGGATCCAGGTGGA

CTTGGGCCTCCTGCGATTCGTTACTGCTGTAGGGACAC

AGGGTGCCATTTCCAAGGAAACCAAGAAGAAATATTA

TGTCAAGACTTACAGAGTAGACATCAGCTCCAACGGA

GAGGACTGGATCTCCCTGAAAGAGGGAAATAAAGCCA

TTATCTTTCAGGGAAACACCAACCCCACAGATGTTGTC

TTAGGAGTTTTCTCCAAACCACTGATAACTCGATTTGT

CCGAATCAAACCTGTATCCTGGGAAACTGGTATATCTA

TGAGATTTGAAGTTTATGGCTGCAAGATAACAGATTAT

CCTTGCTCTGGAATGTTGGGCATGGTGTCTGGACTTAT

TTCAGACTCCCAGATTACAGCATCCAATCAAGCCGACA

GGAATTGGATGCCAGAAAACATCCGTCTGGTGACCAG

TCGTACCGGCTGGGCACTGCCACCCTCACCCCACCCAT

ACACCAATGAATGGCTCCAAGTGGACCTGGGAGATGA

GAAGATAGTAAGAGGTGTCATCATTCAGGGTGGGAAG

CACCGAGAAAACAAGGTGTTCATGAGGAAGTTCAAGA

TCGCCTATAGTAACAATGGCTCTGACTGGAAAACTATC

ATGGATGACAGCAAGCGCAAGGCTAAGTCGTTCGAAG

GCAACAACAACTATGACACACCTGAGCTTCGGACGTTT

TCACCTCTCTCCACAAGGTTCATCAGGATCTACCCTGA

GAGAGCCACACACAGTGGGCTTGGGCTGAGGATGGAG

CTACTGGGCTGTGAAGTGGAAGCACCTACAGCTGGAC

CAACCACACCCAATGGGAACCCAGTGGATGAGTGTGA

CGACGACCAGGCCAACTGCCACAGTGGCACAGGTGAT

GACTTCCAGCTCACAGGAGGCACCACTGTCCTGGCCAC

AGAGAAGCCAACCATTATAGACAGCACCATCCAATCA

GAGTTCCCGACATACGGTTTTAACTGCGAGTTTGGCTG

GGGCTCTCACAAGACATTCTGCCACTGGGAGCATGACA

GCCATGCACAGCTCAGGTGGAGTGTGCTGACCAGCAA

GACAGGGCCGATTCAGGACCATACAGGAGATGGCAAC

TTCATCTATTCCCAAGCTGATGAAAATCAGAAAGGCAA

AGTAGCCCGCCTGGTGAGCCCTGTGGTCTATTCCCAGA

GCTCTGCCCACTGTATGACCTTCTGGTATCACATGTCC

GGCTCTCATGTGGGTACACTGAGGGTCAAACTACGCTA

CCAGAAGCCAGAGGAATATGATCAACTGGTCTGGATG

GTGGTTGGGCACCAAGGAGACCACTGGAAAGAAGGAC

GTGTCTTGCTGCACAAATCTCTGAAACTATATCAGGTT

GAATTGCTGTGGATGATATCAGTATTAACAACCATATT

TCTCAGGAAGACTGTGCAAAACCAACAGACCTAGATA

AAAAGAACACAGAAATTAAAATTGATGAAACAGGGAG

CACTCCAGGATATGAAGGAGAAGGGGAAGGTGACAAG

AACATCTCCAGGAAGCCAGGCAATGTGCTTAAGACCCT

GGATCCCATCCTGATCACCATCATAGCCATGAGTGCCC

TGGGAGTACTCCTGGGTGCAGTCTGTGGAGTTGTGCTG

TACTGTGCCTGTTGGCACAATGGGATGTCAGAAAGGA

ACCTATCTGCCCTGGAGAACTATAACTTTGAACTTGTG

GATGGTGTAAAGTTGAAAAAAGATAAACTGAACCCAC

AGAGTAATTACTCAGAGGCGTGA 134 niNRP- 1 UniProtK MERGLPLLCATLALALALAGAFRSDKCGGTIKIENPGYLT B - SPGYPHSYHPSEKCEWLIQAPEPYQRIMINFNPHFDLEDR

P97333 DCKYDYVEVIDGENEGGRLWGKFCGKIAPSPVVSSGPFL

FIKFVSDYETHGAGFSIRYEIFKRGPECSQNYTAPTGVIKS

PGFPEKYPNSLECTYIIFAPKMSEIILEFESFDLEQDSNPPG

GMFCRYDRLEIWDGFPEVGPHIGRYCGQKTPGRIRSSSGV

LSMVFYTDSAIAKEGFSANYSVLQSSISEDFKCMEALGME

SGEIHSDQITASSQYGTNWSVERSRLNYPENGWTPGEDSY

KEWIQVDLGLLRFVTAVGTQGAISKETKKKYYVKTYRV

DI S SNGEDWISLKEGNKAIIFQGNTNPTD VVLGVF SKPLIT

RFVRIKPVSWETGISMRFEVYGCKITDYPCSGMLGMVSG

LISDSQITASNQADRNWMPENIRLVTSRTGWALPPSPHPY

TNEWLQVDLGDEKIVRGVIIQGGKHRENKVFMRKFKIAY

SN GSDWKTIMDDSKRKAKSFEGN YDTPELRTFSPLS

TRFIRIYPERATHSGLGLRMELLGCEVEAPTAGPTTPNGN

PVDECDDDQANCHSGTGDDFQLTGGTTVLATEKPTIIDST

IQSEFPTYGFNCEFGWGSHKTFCHWEHDSHAQLRWSVLT

SKTGPIQDHTGDGNFIYSQADENQKGKVARLVSPVVYSQ

SSAHCMTFWYHMSGSHVGTLRVKLRYQKPEEYDQLVW

MVVGHQGDHWKEGRVLLHKSLKLYQVIFEGEIGKGNLG

GIAVDDISIN HISQEDCAKPTDLDKKNTEIKIDETGSTPG

YEGEGEGDKNISRKPGNVLKTLDPILITIIAMSALGVLLGA

VCGVVLYCACWHNGMSERNLSALENYNFELVDGVKLK

KDKLNPQSNYSEA

135 rNRP-1 UniProtK MERGLPLLCATLALALALAGAFRSDKCGGTIKIENPGYLT

B - SPGYPHSYHPSEKCEWLIQAPEPYQRIMINFNPHFDLEDR

Q9QWJ9 DCKYDYVEVIDGENEGGRLWGKFCGKIAPSPVVSSGPFL

FIKFVSDYETHGAGFSIRYEIFKRGPECSQNYTAPTGVIKS

PGFPEKYPNSLECTYIIFAPKMSEIILEFESFDLEQDSNPPG

GVFCRYDRLEIWDGFPEVGPHIGRYCGQKTPGRIRSSSGIL

SMVFYTD S AIAKEGF S ANYS VLQ SSI SEDFKCMEALGMES

GEIHSDQITASSQYGTNWSVERSRLNYPENGWTPGEDSY

REWIQVDLGLLRFVTAVGTQGAISKETKKKYYVKTYRV

DISSNGEDWITLKEGNKAIIFQGNTNPTDVVFGVFPKPLIT

RFVRIKPASWETGISMRFEVYGCKITDYPCSGMLGMVSG

LISDSQITASNQGDRNWMPENIRLVTSRTGWALPPSPHPYI

NEWLQVDLGDEKIVRGVIIQGGKHRENKVFMRKFKIAYS

N GSDWKMIMDDSKRKAKSFEGN YDTPELRAFTPLS

TRFIRIYPERATHSGLGLRMELLGCEVEVPTAGPTTPNGN

PVDECDDDQANCHSGTGDDFQLTGGTTVLATEKPTIIDST

IQSEFPTYGFNCEFGWGSHKTFCHWEHDSHAQLRWRVLT

SKTGPIQDHTGDGNFIYSQADENQKGKVARLVSPVVYSQ

SSAHCMTFWYHMSGSHVGTLRVKLHYQKPEEYDQLVW

MVVGHQGDHWKEGRVLLHKSLKLYQVIFEGEIGKGNLG

GIAVDDISIN HIPQEDCAKPTDLDKKNTEIKIDETGSTPG

YEEGKGDKNISRKPGNVLKTLDPILITIIAMSALGVLLGAV

CGVVLYCACWHNGMSERNLSALENYNFELVDGVKLKK

DKLNPQSNYSEA

136 MABs VHCDR2 X1ISGSGGX2TYYADSVX3G, wherein Xi is I or A, X 2 is S or 8-12 Consensus A, and X3 is K or E

137 MABs VHCDR1 FTFX1SX2AMV, wherein Xi is A, K, or S, X 2 is Y or V

8-12 Consensus

138 MABs VHCDR3 ARDLGYYGSGMHX, wherein X is A or V

3-4 Consensus 139 MABs VHCDR3 ARDRGMYYASGFXP, wherein X is G or N 5-6 Consensus

140 Linker (GGGGS)n, wherein n is an integer

consensus

141 anti-NRP IgGl EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQ Antibody APGKGLEWVSQISPAGGYTNYADSVKGRFTISADTSKNT SEC 10 AYLQMNSLRAEDTAVYYCARGELPYYRMSKVMDVWGQ

GTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY

FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVP

SSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPC

PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE

DPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLT

VLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREP

QVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNG

QPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFS

CSVMHEALHNHYTQKSLSLSPGK

142 anti-NRP Kappa DIQMTQSPSSLSASVGDRVTITCRASQYFSSYLAWYQQKP Antibody light chain GKAPKLLIYGASSRASGVPSRFSGSGSGTDFTLTISSLQPE SEC 10 DFATYYCQQYLGSPPTFGQGTKVEIKRTVAAPSVFIFPPSD

EQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQ

ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQ

GLSSPVTKSFNRGEC

143 Human UniProt MERGLPLLCAVLALVLAPAGAFRNDKCGDTIKIESPGYLT NRP-1 014786. SPGYPHSYHPSEKCEWLIQAPDPYQRIMINFNPHFDLEDR

Has minor DCKYDYVEVFDGENENGHFRGKFCGKIAPPPVVSSGPFLF SNP, IKFVSDYETHGAGFSIRYEIFKRGPECSQNYTTPSGVIKSP V179 GFPEKYPNSLECTYIVFVPKMSEIILEFESFDLEPDSNPPGG

MFCRYDRLEIWDGFPDVGPHIGRYCGQKTPGRIRSSSGIL

SMVFYTDSAIAKEGFSANYSVLQSSVSEDFKCMEALGME

SGEIHSDQITASSQYSTNWSAERSRLNYPENGWTPGEDSY

REWIQVDLGLLRFVTAVGTQGAISKETKKKYYVKTYKID

VSSNGEDWITIKEGNKPVLFQGNTNPTDVVVAVFPKPLIT

RFVRIKPATWETGISMRFEVYGCKITDYPCSGMLGMVSG

LISDSQITSSNQGDRNWMPENIRLVTSRSGWALPPAPHSYI

NEWLQIDLGEEKIVRGIIIQGGKHRENKVFMRKFKIGYSN

NGSDWKMIMDDSKRKAKSFEGNNNYDTPELRTFPALSTR

FIRIYPERATHGGLGLRMELLGCEVEAPTAGPTTPNGNLV

DECDDDQANCHSGTGDDFQLTGGTTVLATEKPTVIDSTI

QSEFPTYGFNCEFGWGSHKTFCHWEHDNHVQLKWSVLT

SKTGPIQDHTGDGNFIYSQADENQKGKVARLVSPVVYSQ

NSAHCMTFWYHMSGSHVGTLRVKLRYQKPEEYDQLVW

MAIGHQGDHWKEGRVLLHKSLKLYQVIFEGEIGKGNLG

GIAVDDISINNHISQEDCAKPADLDKKNPEIKIDETGSTPG

YEGEGEGDKNISRKPGNVLKTLDPILITIIAMSALGVLLGA

VCGVVLYCACWHNGMSERNLSALENYNFELVDGVKLK

KDKLNTQSTYSEA

144 SEC3 DIQMTQSPSSLSASVGDRVTITCRASQDVSTAVAWYQQK light PGKAPKLLIYSASFLYSGVPSRFSGSGSGTDFTLTISSLQPE chain DFATYYCQQAWAYLPTFGQGTKVEIKRTVAAPSVFIFPP

SDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGN

SQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVT

HQGLSSPVTKSFNRGEC SEC3 EVQLVESGGGLVQPGGSLRLSCAASGFTISGYGIHWVRQ

Heavy APGKGLEWVAYIYPDSGYTDYADSVKGRFTISADTSKNT

Chain A Y LQ M N S L R A E DT A V Y YC A R E D F RN R R R LWYV M D YW

GQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVK

DYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVT

VP S S SLGTQTYICNVNHKPSNTKVDKKVEPKS CDKTHTCP

PCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH

EDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVL

TVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE

PQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESN

GQPEN YKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVF

SCSVMHEALHNHYTQKSLSLSPGK