Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTI-FCRH5 ANTIBODIES
Document Type and Number:
WIPO Patent Application WO/2014/210064
Kind Code:
A1
Abstract:
The invention provides anti~FcRH5 antibodies and immunoconjugates and methods of using the same,

Inventors:
EBENS ALLEN J JR (US)
HAZEN MEREDITH C (US)
HONGO JO-ANNE (US)
JOHNSTON JENNIFER W (US)
JUNTTILA TEEMU T (US)
LI JI (US)
POLSON ANDREW G (US)
Application Number:
PCT/US2014/043952
Publication Date:
December 31, 2014
Filing Date:
June 24, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
GENENTECH INC (US)
International Classes:
C07K16/28; A61K39/395; A61K47/48
Domestic Patent References:
WO2006039238A22006-04-13
WO2010114940A12010-10-07
WO2010120561A12010-10-21
Other References:
HATZIVASSILIOU G ET AL: "IRTA1 and IRTA2, novel immunoglobulin superfamily receptors expressed in B cells and involved in chromosome 1q21 abnormalities in B cell malignancy", IMMUNITY, CELL PRESS, US, vol. 14, no. 3, 1 March 2001 (2001-03-01), pages 277 - 289, XP002331959, ISSN: 1074-7613, DOI: 10.1016/S1074-7613(01)00109-1
ISE TOMOKO ET AL: "Immunoglobulin superfamily receptor translocation associated 2 protein on lymphoma cell lines and hairy cell leukemia cells detected by novel monoclonal antibodies", CLINICAL CANCER RESEARCH, THE AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 11, no. 1, 1 January 2005 (2005-01-01), pages 87 - 96, XP002528392, ISSN: 1078-0432
POLSON ANDREW G ET AL: "Expression pattern of the human FcRH/IRTA receptors in normal tissue and in B-chronic lymphocytic leukemia", INTERNATIONAL IMMUNOLOGY, OXFORD UNIVERSITY PRESS, GB, vol. 18, no. 9, 1 September 2006 (2006-09-01), pages 1363 - 1373, XP002487226, ISSN: 0953-8178, [retrieved on 20060718], DOI: 10.1093/INTIMM/DXL069
K. ELKINS ET AL: "FcRL5 as a Target of Antibody-Drug Conjugates for the Treatment of Multiple Myeloma", MOLECULAR CANCER THERAPEUTICS, vol. 11, no. 10, 17 July 2012 (2012-07-17), pages 2222 - 2232, XP002587809, ISSN: 1535-7163, DOI: 10.1158/1535-7163.MCT-12-0087
DAVIS R S ET AL: "Identification of a family of Fc receptor homologs with preferential B cell expression", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, vol. 98, no. 17, 14 August 2001 (2001-08-14), pages 9772 - 9777, XP002971662, ISSN: 0027-8424, DOI: 10.1073/PNAS.171308498
Attorney, Agent or Firm:
SONEOKA, Yuko et al. (LLP1801 Page Mill Road, Suite 11, Palo Alto CA, US)
Download PDF:
Claims:
WHAT IS CLAMED IS:

1) An isolated anti-FcRH5 antibody that binds an isoform e-speeifie region of the extracellular domain of FcRHSc,

2) The antibody of claim 1 , wherein the isofonn c-specifie region comprises Ig-tike domain 9.

3) The antibody of any one of claims 1 -2, wherein the isofonn c~specific region comprises amino acids 743-850 of SEQ ID NO:l .

4) The antibody of any one of claims 1 -3, wherein the antibody comprises: a) a heavy chairs comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:38, HVR-H2 comprising the amino acid sequence of SEQ ID O:62, and HVK-H3 comprising the amino acid sequence of SEQ ID NO:86; and/or b) a light chain comprising a HVR-LI comprising the amino acid sequence of SEQ ID NO:2, HVR-L2 comprising the amino acid sequence of SEQ ID NO:14, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:26.

5) The antibody of claim 4, wherein the heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO: 50, HVR-H2 comprising the amino acid sequence of SEQ ID NO:74. and HVR-H3 comprising the amino acid sequence of SEQ TD O:98,

6) The antibody of any one of claims 1 -3, wherein the antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:39. HVR-H2 comprising the amino acid sequence of SEQ ID NO:63, and HVR-H3 comprising the amino acid sequence of SEQ ID NO;87; and/or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:3, HVR-L2 comprising the amino acid sequence of SEQ ID NO:l 5, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:27,

7} The antibody of claim 6, wherein the heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:51 , HVR-H2 comprising the amino acid sequence of SEQ ID NO:75, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:99.

8) The antibody of any one of claims 1-3, wherein the antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:40„ HVR-H2 comprising the amino acid sequence of SEQ ID MO:64, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:88; and/or b) a light chain comprising a HVR-LI comprising the amino acid sequence of SEQ ID NQ:4, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:28.

9) The antibody of claim 8„ wherein the heavy chain comprising a H'VR-Rl comprising the amino acid sequence of SEQ ID NO:52, HVR-H2 comprising the amino acid sequence of SEQ ID NO:76, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 100.

10) The antibody of any one of claims 1-3, wherein the antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NQ:41, HVR-H2 comprising the amino acid sequence of SEQ ID NO:65s and HVR-H3 comprising the amino acid sequence of SEQ ID NO:89: and'or b) a light chain comprising a HVR-LI comprishig the amino acid sequence of SEQ ID NO:5, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 17, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:29.

1 i ) The antibody of claim 10, wherein the heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:53, HVR-H2 comprising the amino acid sequence of SEQ ID NO;77. and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 101.

12) The antibody of any one of claims 1 -3» wherein the antibody comprises: a) a heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:42, HVR-H2 comprising the amino acid sequence of SEQ ID NO:66, and HVR-H3 comprising the amino acid seqiience of SEQ ID NO:90; and/or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:6, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18, and HVR.-L3 comprising the amino acid sequence of SEQ ID NO; 30.

13) The antibody of claim 12, wherein the heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:54, HVR-H2 comprising the amino acid sequence of SEQ ID NO:78, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 102.

14) The antibody of any one of claims 1-3, wherein the antibody comprises: a) a heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:43, HVR-H2 comprising the amino acid sequence of SEQ ID NO:67, and KVR-H3 comprising the amino acid sequence of SEQ ID NO:91 ; and or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:7, HVR-L2 comprising the amino acid sequence of SEQ ID NO:1 , and HVR-.L3 comprising the amino acid sequence of SEQ ID NO:31 ,

15) The antibody of claim 14, wherein the heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:55, HVR-H2 comprising the amino acid sequence of SEQ ID NO:79, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 103.

16) The antibody of any one of claims 1-3, wherein the antibody comprises: a) a heavy chain comprising a HVR-H1 comprismg the amino acid sequence of SEQ ID NQ:44, HVR-H2 comprising the amino acid sequence of SEQ ID NO:68, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:92; and/or b) a light chain comprising a HVR -U comprising the amino acid sequence of SEQ ID NO:8, HVR-L2 comprising the amino acid sequence of SEQ ID NO:20, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:32,

17) The antibody of claim 16, wherein the heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:56, HVR.-H2 comprising the amino acid sequence of SEQ ID NO:80, and HVR-H3 comprising the annuo acid sequence of SEQ ID NO: 104.

18) The antibody of any one of claims 1 -3, wherein the antibody comprises: a) a heavy chain comprising a HVR-H1 comprismg the amino acid sequence of SEQ ID NO:45, HVR-H2 comprising the amino acid sequence of SEQ ID NO;69, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:93; and/or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:9, HVR-L2 comprising the amino acid sequence of SEQ ID NO:2i , and HVR-L3 comprising the amino acid sequence of SEQ ID NO:33,

1 ) The antibody of claim 1 8, wherein the heavy chain comprising a HVR-H! comprising tlie amino acid sequence of SEQ ID NO:57, HVR-H2 comprising the amino acid sequence of SEQ ID NO:81 , and HVR-H3 comprising the amino acid sequence of SEQ ID NO:105,

20) The antibody of any one of claims 1 -3, wherein the antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:46, HVR-H2 comprising the amino acid sequence of SEQ ID NO:70, and HVR-H3 comprising the amino acid sequence of SEQ ID O:94; and/or b) a light chain comprising a HVR-Ll comprising the amino acid sequence of SEQ ID NO: 10, HVR-L2 comprising the amino acid sequence of SEQ ID O:22, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:34.

21) The antibody of claim 20, wherein the heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID O:58, HVR-H2 comprising the amino acid sequence of SEQ ID NO:82, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 106,

22) The antibody of any one of claims 1-3, wherein the antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID N :47, HVR-H2 comprising the ammo acid sequence of SEQ ID O:71 , and HVR-H3 comprising the amino acid sequence of SEQ ID NO:95; and/or b) a light chain comprising a HVR-Ll comprising the amino acid sequence of SEQ ID NO: I L HVR-L2 comprising the amino acid sequence of SEQ ID NO:23, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:35.

23) The antibody of claim 22, wherein the heavy chain comprising a HVR-H l comprising the amino acid sequence of SEQ ID O:59, HV -H2 comprising the amino acid sequence of SEQ ID NO: 83, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:107.

24) The antibody of any one of claims 1 -3, wherein the antibody comprises: a) a heavy chain comprising a I-FVR-Hl comprising the amino acid sequence of SEQ ID NO:48, HVR-H2 comprising the amino acid sequence of SEQ ID NO;72( and HVR-H3 comprising the amino acid sequence of SEQ ID N :96: and/or b) a light chain comprising a HVR-Ll comprising the amino acid sequence of SEQ ID NO: 12, HV -L2 comprising the amino acid sequence of SEQ ID NO:24, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:36.

25) The antibody of claim 24, wherein the heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:6G, HVR-H2 comprising the amino acid sequence of SEQ ID NO:84, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 108.

26) The antibody of any one of claims 1-3, wherein fee antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:49, HV -H2 comprising the amino acid sequence of SEQ ID NO:73, and HVR.-H3 comprising the amino acid sequence of SEQ ID NO:97; and/or b) a light chain comprising a HVR-Ll comprising the amino acid sequence of SEQ ID NO:13, HVR-L2 comprising the amino acid sequence of SEQ ID NO:25, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:37.

27) The antibody of claim 26, wherein the heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ TD NO;61 , HVR-H2 comprising tlie amino acid sequence of SEQ ID NO:85, and HVR-H3 comprising the amino acid sequence of SEQ 3D NO: 109.

28) The antibody of any one of the preceding claims, wherein the antibody comprises:

a) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: l 11 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1 10:

b) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: l 13 arsd/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1 12;

c) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 115 and''or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1 14;

d) a VH sequence having at least 95%o sequence identity to the amino acid sequence of SEQ ID NO: 117 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1 16;

e) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:1 1 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:118;

f) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 121 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 120;

g) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 123 and/or a VL sequence having at least 95% sequence identity to the. amino acid sequence of SEQ ID NO: 122;

h) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 125 and/or a VL sequence having at least 95% sequence identity to the amino acid seqiaence of SEQ ID NO: 124;

i) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 127 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 126;

j) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 129 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:I28; k) a VH sequence having at least 95% sequence identity io the amino acid sequence of SEQ ID NO:131 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 130;

1) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ 3D NO:133 and/or a VL sequence having at least 95% sequence identity to the amino acid seqiaence of SEQ ID NO: 132; or

m) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 135 and or a VL sequence having at ieast 95% sequence identity to the amino acid sequence of SEQ ID NO: 134.

29) The antibody of any of claims 1-28, wherein die antibody comprises:

a) a VH sequence of SEQ ID NO:l 1 1 and/or a VL sequence of SEQ ID NO:l 10;

b) a VH sequence of SEQ ID NO: 1 13 and/or a VL sequence of SEQ ID NO: 1 12;

c) a VH sequence of SEQ ID NO:l 15 and/or a VL sequence of SEQ ID NO: 114;

d) a VH sequence of SEQ ID NO:l 17 and/or a VL sequence of SEQ ID NG:i 16;

e) a VH sequence of SEQ ED NO:l 19 and/or a VL sequence of SEQ ID NO:l 18;

fj a VH sequence of SEQ ID NO:121 and/or a VL sequence of SEQ ID NO:120;

g) a VH sequence of SEQ ID NO:123 and/or a VL sequence of SEQ ID NO:122;

h) a VH sequence of SEQ ID NO: 125 and/or a VL sequence of SEQ ID NO:l 24;

i) a VH sequence of SEQ ID NO: 127 and/or a VL sequence of SEQ ID NO: 126;

j) a VH sequence of SEQ ID NO:129 and/or a VL sequence of SEQ ID NO: 128;

k) a VH sequence of SEQ ID NO:131 and/or a VL sequence of SEQ ID NO: 130;

1) a VH sequence of SEQ ID NO: 133 and/or a VL sequence of SEQ ID NO: 132; or m) a VH sequence of SEQ ID NO:l 35 and/or a VL sequence of SEQ ID NO: 134.

30) The antibody of any one of claims 1-29, wherein the antibody is a monoclonal antibody.

31) The antibody of any one of claims 1 -30, wherein ihe antibody is a human, humanized, or chimeric antibody.

32) The antibody of any one of claims 1 -31 , wherein ihe antibody is an antibody fragment that binds FeRHS.

33) The antibody of any one of claims 1 -32, wherein the antibody is an IgGl , IgG2a or IgG2b antibody.

34) The antibody of any one of claims 1 -33, wherein the antibody has one or more of the following characteristics: a) cross reactive with full length human and eyno FcRH5, b) does not cross react with FcRHl , FcRH2, FcRH.3, and/or FcRH4, c) binds to endogenous FcRH5, d) does not cross react with FcRHSa, and e) does not cross react with another Ig-like domain of FcRH5.

35) The antibody of any one of claims 1 -34, wherein the antibody is a bispecifie antibody.

36) The antibody of claim 35, wherein bispecifie antibody binds FcRI-15 and CD3.

37) Isolated nucleic acid encoding the antibody of any one of claims 1 -36. 38) A host cell comprising the nucleic acid of claim 37.

39) A method of producing an antibody comprising culturing the host cell of claim 38 so that antibody is produced.

40) An immunoconjngate comprising the antibody of any one of claims 1 -36 and a cytotoxic agent.

41) The immiinoeonjugate of claim 41 having the fonnuia Ab-(L~D)p, wherein:

(a) Ab is the antibody of any one of claim 1 to 36;

(b) L is a linker;

(c) D is a drug selected from a maytansinoid, an auristatin, a calicheami pyrrolobenzodiazepine, and a nemorubicin derivative; and

(d) p ranges from 1-8.

42) The immunoeonjugate of claim 41 , wherein D is an auristatin.

43)

and wherein R1 and R6 are each methyl, R3 and R4 are each isopropyl, Rs is H, R7 is sec-butyl, each Rs is independently selected from C3¾, Q-C¾, OH, and H; R ' is H; and R18 is -C(R8)rC(R8)r ryL

44) The immunoeonjugate of claim 41 , wherein the drug is MMAE.

45) The immunoeonjugate of claim 41. wherein D is a pyrrolobenzodiazepine of Formula A:

wherein the dotted lines indicate the optional presence of a double bond between CI and C2 or C2 and C3:

R2 is independently selected from. H, OH, =0, =CH2, CM, R, OR, = H-RD, =C(R )2,

0- SO..-R ,€G2R and COR, and optionally further selected froxn halo or dihalo, wherein RD is independently selected from R, CG2R, COR, CHO, C02l and halo;

R" and R9 are independently selected from H, R, OH, OR, SH, SR, NH2, NHR, NRR', 02, Me¾Srs and halo: R7 is independently selected from H, R, OH, OR, SH, SR, N¾ NHR, NRR', NOJ; Me3Sn and halo;

Q is independently selected from O, S and NH;

Rn is either H, or R or, where Q is Os SQ-iM, where M is a metal cation;

R and R' are each independently selected from optionally substituted Cj.g aikyl,

Cj.g heterocyclyl and C5.20 ar l grou s, and optionally in relation to the group NRR\ R and R* together with the nitrogen atom to which they are attached form an optionally substituted 4-,

5-, 6- or 7-memhered heterocyclic ring;

R12, Rk\ t9 and R:7 are as defined for R2, R6, R9 and R' respectively;

R" is a C 12 alkylene group, which chain may be interrupted by one or more heteroatoms and/or aromatic rings that are optionally substituted; and

X and X' are independently selected from O, S and N(H).

46)

A(K);

wherein n is 0 or 1.

wherein RE mid RE are each independently selected from H or D, wherein RD is independently selected from R, CO2 , COR, CHO, C02H, and halo;

wherein Ar! and Ar; are each independently optionally substituted 0<. aryl; and wherein n is 0 or 1.

wherein the horizontal wavy line indicates the covaient attachement site to the linker; Rv i and Rv? are independently selected from II, methyl, ethyl, phenyl, fluoro-substituted phenyl, and C5.6 heterocyclyl; and

n is 0 or 1.

The irnmunoeonjugate of claim 41 , wherein D is a nemorubicin derivative.

The irnmunoeonjugate of claim 49, wherein D has a structure selected from:

: and

„0

51 ) The immunoconjugate of any one of claims 41 to 50, wherein the linker is cieavable by a protease,

52) The immunoconjugate of claim 51 , wherein the linker comprises a val-cit dipeplide or a Fhe- homoLys dipeptide,

53) The immunoconjugate of any one of claims 41 io 50, wherein the iinker is acid-labile.

54) The immunoconjugate of claim 53, wherein the linker comprises hydrazone.

55) The immunoconjugate of claim 53 having the formula:

wherein S is a sulfur atom.

56) The immunoconjugate of claim 46 having a formula selected from:

P : and

125 59) A pharmaceutical formulation comprising the antibody of any one of claims 1 -36 and/or the immunoeonjugate of any one of claims 41 - 58 and a pharmaceutically acceptable carrier.

60) The pharmaceutical formulation of claim 59, further comprising an additional therapeutic agent.

61} A method of treating an individual having an FcRHS-positive cancer, the method comprising administering to the individual an effective amount of the antibody of any one of claims 1-36 and/or immunoeonjugate of any one of claims 41 to 59.

62) The method of claim 61 , wherein the FcRHS-positive cancer is a B-cell proliferative disorder.

63) The method of any one of claims 61-62, further comprising administering an additional therapeutic agent to the individual.

64) A method of inhibiting proliferation of an FeRHS -positive cell, the method comprising exposing the cell to the antibody of any one of claims 1 -36 and/or immunoeonjugate of any one of claims 41-59 under conditions permissive for binding of the antibody and/or immunoeonjugate to FcRHS on the surface of the cell, thereby inhibiting proliferation of the cell

65) The method of claim 64, wherein the cell is a B-cell.

66) The antibody of any one of claims 1 -36 conjugated to a label.

67) The antibody of claim 66, wherein the label is a positron emitter.

68) The antibody of claim 67, wherein the positron emitter is S9Zr,

69) A method of detecting human FcRI-15 in a biological sample comprising contacting the biological sample with the anti-FcRH5 antibody of any one of claims 1 -.36 or 66-68 under conditions permissive for binding of the anti-Fc.RH5 antibody to a naturally occurring human FcRHS, and detecting whether a complex is formed between the anti-FcRIi5 antibody and a naturally occurring human FcRHS in the biological sample,

70) The method of claim 69, wherein the anti~FcRH5 antibody is an antibody as in claim 9 or claim 15.

71 ) The method of any one of claims 69-70, wherein the biological sample is a blood sample.

72) A method for detecting an FcRHS-positive cancer comprising (i) administering a labeled anti- FcRH5 antibody to a subject having or suspected of having a FcRHS-positive cancer, wherein the labeled anti~FcRH5 antibody comprises die anti-FcRH5 antibody of any one of claims 1 to 36, and (ii) detecting the labeled anti-FcRH5 antibody in the subject, wherein detection of the labeled anti-FcRHS antibody indicates a FcRHS-positive cancer in the subject.

73) The method of 72, wherein the labeled anti~FcRH5 antibody comprises an anti-FcRH5 antibody conjugated to a positron emitter.

74) The method of any one of claims 72-73, wherein the positron emitter is 89Zr.

Description:
The present application claims benefit under 35 U.S.C, § 1 19 of U.S. Provisional Patent Application No, 61/838,534, filed on June 24, 2013, the disclosure of which is hereby incorporated by reference in its entirety.

SEQUENCE LISTING

The instant application contains a Sequence Listing in ASCII format and is hereby incorporated by reference in its entirety. The ASCII text file was created on June 9, 2014, is named GNE- 0413 WO .. .SL.txt and is 67,909 bytes in size.

FIELD OF THE INVENTION

Provided herein are anti~FcRH5 antibodies (e.g., bispecific antibodies) and immunoeonjugates

The Fc receptor-like 5 (F ' cRLS, also known as FcRHS and IRTA2) belongs to a family of 6 recently identified genes of the immunoglobulin superfamily (IgSF). This family of genes is closely related to the Fc receptors with the conserved genomic structure, extraceiiuiar Ig domain composition and the ITIM- and ITAM-like signaling motifs (Davis RS ei a!.. Ear J Immunol (2005) 35:674-80). Members of this family have also been called IFGPs (from Ig super-family, FcR, gp42) and SPAPs (SH2 domain-containing phosphatases anchor proteins), Six members of the Fc H/IRTA receptor family have been described: FcRHl/iRTAS, FcRH2 TKTA4, FcRH3/IRTA3, FcRH4/lRTAl ,

FcRH5/IRTA2 and FcRH6 (Poison AG et al., Int. Immunol (2006) 18(9): 1363-1373). All FcRH/TRTAs contain some combination of canonical imrnunoreeeptor tyrosine-based inhibitory motifs and

'imrnunoreeeptor tyrosine-based activation motifs-like" signaling motifs. The F RII cDNAs encode type 1 transmembrane glycoproteins with multiple Ig-like extraceiiuiar domains and cytoplasmic domains containing consensus imrnunoreeeptor tyrosine-based activating and/or inhibitory signaling motifs. The FeRFl genes are structurally related, and their protein products share 28-60% extracellular identity with each other. They also share 15-31 % identity with their closest FcR relatives, There is a high degree of homology between the different FcRHs.

The iigand(s) for FcRHS are unknown, but FcRHS has been implicated in enhanced proliferation and downstream isotype expression during the development of antigen-primed B~cel1s (Derne t-Brown J. el al, JLetikoe Bio! (2012) 91 :59-67). The FcRHS locus has three major mRNA isoforrns (FcRHSa, FcRHSb, and FcRHS c). The major FcRHS protein isoforms encoded by these transcripts share a common amino acid sequence until residue 560, featuring a common signal peptide and six extracellular Ig-like domains. FeRHSa represents a 759 amino acid secreted glycoprotein with eight Ig-like domains followed by 13 unique, predominantly polar amino acids at its C -terminus.

FeRHSb diverges from FcRHSa at amino acid residue 560 and extends for a short stretch of 32 additional residues, whose hydrophobicity is compatible with its docking to the plasma membranevia a 5 GPI anchor, FcRHSc is the longest isoform whose sequence deviates from FcRHSa at amino acid 746.

FcRHSc encodes a 977 aa type I transmembrane glycoprotein with nine extracellular Ig-type domains, harboring eight potential N-linked glyeosylation sites, a 23 ammo acid transmembrane, and a 104 amino acid cytoplasmic domain with three consensus SH2 binding motifs with the Π.ΊΜ consensus,

The FcRH genes are clustered together in the midst of the classical FcR genes, FeyRl, FcyRI!,

I Q FcyRlIl, and FceRJ, in the lq21 -23 region of chromosome 1 , This region contains 1 of the most

frequent secondary chromosomal abnormalities associated with malignant phenotype in hematopoietic tumors, especially in multiple myeloma (Hatzivassiliou Q.et al. Immunity (2001) 14:277-89). FcRH5 is expressed only in the B-ceil lineage, starting as early as pre~B-cells, but does not attain full expression until the mature B-cell stage. Unlike most knownother B - cell-specific surface proteins (e.g., CD20,

15 CD19, and CD22), FeRFfS continues to be expressed in plasma cells whereas other B-celi-speciflc markers are downregulated (Poison AG et αί, Jut Immunol (2006) 18:1363-73). In addition, FcRH5 mRNA is overexpressed in multiple myeloma cell lines with lq21 abnormalities as detected by oligonucleotide arrays (inoue 1. Am Pathol (2004) 165:71-81). The expression pattern indicates that FcRH5 could be a target for antibody-based therapies for the treatment of multiple myeloma. Multiple

20 myeloma is a malignancy of plasma cells characterized by skeletal lesions, renal failure, anemia, and hypercalcemia. It is essentially incurable by current therapies. Current drug treatments for multiple myeloma include combinations of the proteosome inhibitor bortezomib (V elcade), the

inimunomodulator lenalidomide (Revlimid), and the steroid dexamethasone,

FcRHSc specific antibody-based therapies and detection methods may be particularly

25 efficacious as they specifically recognize target cell, membrane-associated FcRH5 rather than

antibodies which recognize both solubie and membrane isoforms of FcRHS. However, only the last Ig- like domain of FcRFI5 (Ig-like domain 9) is unique extracellular region that differentiates between the three major isoforms of Fc IIS, and there is sigiiiiicani homology between the Ig-iike domains within FcRI-15. Further, the last Ig-like domain is highly conserved between FcRHl , FcRFI2, FeRH3, and

30 FcRHS. Any antibody-based therapy that specifically targeted FcRIIS would have to have minimal cross-reactivity with other FcRHs to avoid adverse off-target effects (e.g., Fc H3 is expressed on normal NK ceils), There is a need in the art for agents that aid in the diagnosis and treatment of cancer, such as FcRHS-associated cancer.

SUMMARY

35 Provided herein axe anti-FeRHS antibodies including bispecific antibodies, immunoconjugates. and methods of using the same. Provided herein are isolated anti-FcRH5 antibodies that binds an

7 iso form c-specific region of the extracellular domain of FcRHSc. In some embodiments, the isoform c- speciiic region comprises Ig-iike domain 9, In some embodiments, the iso form c-specific region comprises amino acids 743-850 of SEQ ID NO: I .

In some embodiments, the antibody comprises: a) a heavy chain comprising a HYR-H1 comprising the amino acid sequence of SEQ ID NO:38, HVR-H2 comprising the amino acid sequence of SEQ ID NO:62, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:86; and/o b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:2, HV -L2 comprising the amino acid sequence of SEQ ID NO:14. and HVR-L.3 comprising the amino acid sequence of SEQ ID O:26. In some embodiments, the heavy chain comprising a HVR-H1 comprising the ammo acid sequence of SEQ ID NO:50, HVR-H2 comprising the amino acid sequence of SEQ ID NO:74, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:98,

In some embodiments, the antibody comprises: a) a heavy chain comprising a H.VR-H1 comprising the amino acid sequence of SEQ ID NO:39, HVR-H2 comprising the amino acid sequence of SEQ ID NOi63. and iiVR-H3 comprising the amino acid sequence of SEQ ID NO:87; and/or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:3, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 15, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:27. In some embodiments, the heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:5I , HVR-H2 comprising the amino acid sequence of SEQ ID NO:75, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:99.

n some embodiments, the antibody comprises: a) a heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:40, HVR-H2 comprising the amino acid sequence of SEQ ID NO:64, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:88; and/or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:4, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:28. In some embodiments, the heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:52, HVR-H2 comprising the amino acid sequence of SEQ ID NO:76, and HVR-K3 comprising the amino acid sequence of SEQ ID NO: 100.

In some embodiments, the antibody comprises: a) a heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:41, HVR-H2 comprising the amino acid sequence of SEQ ID NO:65 s and HVR-H3 comprising the amino acid sequence of SEQ ID NO:89; and/or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:5, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 17, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:29, In some embodiments, the heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:53, HVR-H2 comprising the amino acid sequence of SEQ ID NQ:77, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:101 , In some embodiments, the antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:42. HVR-IT2 comprising the amino acid sequence of SEQ ID NO:66, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:90; and/or b) a light chain comprising a HVR-LI comprising the amino acid sequence of SEQ ID Q:6, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:30. In some embodiments, the heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID Q:54, HVR-H2 comprising the amino acid sequence of SEQ ID O:78, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 102.

In some embodiments, the antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID Q:43, HVR.-H2 comprising the amino acid sequence of SEQ I ' D O:67, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:9 i ; and/or b) a light chain comprising a HVR-LI comprising the amino acid sequence of SEQ ID NO:7, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 19, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:31. In some embodiments, the heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:55, HVR-H2 comprising the amino acid sequence of SEQ ID NO: 79, and HVR-H3 comprising the amino acid sequence of SEQ fD NO: 103.

In some embodiments, the antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:44, HVR-H2 comprising the amino acid sequence of SEQ ID NO:68, and HVR-H3 comprising the amino acid sequence of SEQ ID NQ:92; and/or b) a light chain comprising a HVR-LI comprising the amino acid sequence of SEQ ID NO:8, HVR-L2 comprising the amino acid sequence of SEQ ID NO:20„ and HVR-L3 comprising the amino acid sequence of SEQ ID NO:32. In some embodiments, the heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:56, HVR-H2 comprising the amino acid sequence of SEQ ID NO;80, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 104.

In some embodiments, die antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID N :45, HVR-H2 comprising the amino acid sequence of SEQ ED NO:69, and HVR-H3 comprising the amino acid sequence of SEQ ID Q:93; and/or b) a light chain comprising a HVR-LI comprising the amino acid sequence of SEQ ID NO:9, HVR-L2 comprising the amino acid sequence of SEQ ID NO:21, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:33. In some embodiments, the heavy chain comprising a HVR-Hl eomprismg the amino acid sequence of SEQ ID NO:57, HVR-H2 comprising the amino acid sequence of SEQ ID NO:81 , and HVR-H3 comprising the amino acid sequence of SEQ ED NO: 105.

In some embodiments, the antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:46, HVR-H2 comprising the amino acid sequence of SEQ ID NO:70, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:94; and/or b) a light chain comprising a HVR-LI comprising the amino acid sequence of SEQ ID NO: 10, HVR-L2 comprising the amino acid sequence of SEQ ID NO:22, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:34. In some embodiments, the heaw chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:58, HVR-H2 comprising the amino acid sequence of SEQ ID O:82, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:106.

In some embodiments, the antibody comprises: a) a heavy chain comprismg a HVR-Hl comprising the amino acid sequence of SEQ ID NG:47, HVR -H2 comprising the amino acid sequence of SEQ ID NO:71 , and HVR-H3 comprising the amino acid sequence of SEQ ID NO:95; and/or h) a light chain comprising a ITVR-L1 comprising the amino acid sequence of SEQ ID NG:11 , HVR.-L2 comprising the amino acid sequence of SEQ ID O:23, and HVR-L3 comprising the amino acid sequence of SEQ ID NO;35. In some embodiments, the heavy chain comprismg a HVR-Hl comprising the amino acid sequence of SEQ ID NO:59, HVR-H2 comprising the amino acid sequence of SEQ ID NO:83, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:l 07.

In some embodiments, the antibody comprises: a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:48 t HVR-H2 comprising the amino acid sequence of SEQ ID NO:72, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:96; and/or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:12. HVR-L2 comprising the amino acid sequence of SEQ ID O;24, and HVR-L3 comprismg the amino acid sequence of SEQ ID NO:36. hi some embodiments, the heavy chain comprismg a HVR-Hl comprising the amino acid sequence of SEQ ID NO:60, HVR-H2 comprising the amino acid sequence of SEQ ID NO:84 ; and HVR-H3 comprising the amino acid sequence of SEQ ID NO:108.

In some embodiments, the antibody comprises: a) a heavy chain comprising a HVR-Hl comprismg the amino acid sequence of SEQ ID NO:49, HVR-H2 comprising the amino acid sequence of SEQ ID NO:73. and HVR-H3 comprismg the amino acid sequence of SEQ ID NQ:97; and/or b) a light chain comprising a HVR-L1 comprismg the amino acid sequence of SEQ ID O:l 3, HVR-.L2 comprising the amino acid sequence of SEQ ID NO:25, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:37. In some embodiments, the heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID O:61 , HVR-H2 comprising the amino acid sequence of SEQ ID NO;85, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 109.

In some embodiments of any of the antibodies, the antibody comprises: a) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1 i 1 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1 10; b) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:l 13 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1 12; c) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:l 15 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NG:1 14; d) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1 17 and/or a VI. sequence having at least 95 % sequence identity to the amino acid sequence of SEQ ID NO:l 16; e) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1 19 and'Or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: l 18; £} a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 121 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 120; g) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 123 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 122; h) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 125 andbr a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 124; i) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:127 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 126; j) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 129 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:128; k) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:l 31 and/br a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 130: 1) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 133 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 132; or a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 135 and/or a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 134.

In some embodiments of any of the antibodies, the antibody comprises: a) a VH sequence of

SEQ ID NO: 1 11 and/or a VL sequence of SEQ ED NO: 1 10; b) a VH sequence of SEQ ID NO: 113 and/or a VL sequence of SEQ ID NO: 1 12; c) a VH sequence of SEQ ID NO: 1 15 and/or a VL sequence of SEQ ID NO: 1 14; d) a VH sequence of SEQ ID NO: 1 17 and/or a VL sequence of SEQ ID NO: 116; e) a VH sequence of SEQ ID NO:l 19 and/or a VL sequence of SEQ ID NO:1 18; f) a VH sequence of SEQ ID NO;121 and/or a VL sequence of SEQ ID NO: 120; g) a VH sequence of SEQ ID NO:123 and/or a VL sequence of SEQ ID NO: 122; h) a VH sequence of SEQ ID NO: 125 and'Or a VL sequence of SEQ ID NO: 124; i) a VH sequence of SEQ ID NO: 12? and/or a VL sequence of SEQ ID NO: 126; j) a VH sequence of SEQ ID NO: 129 and/br a VL sequence of SEQ ID NO: 128; k) a VH sequence of SEQ ID NO: 131 and/or a VL sequence of SEQ ID NO: 130: 1) a VH sequence of SEQ ID NO: 133 and/or a VL sequence of SEQ ID NO: 132, or m) a VH sequence of SEQ ID NO: 135 and or a VL sequence of SEQ ID NO: 134,

In some embodiments of any of the antibodies, the antibody is a monoclonal antibody. In some embodiments of any of the antibodies, the antibody is a human, humanized, or chimeric antibody. In some embodiments of any of the antibodies, the antibody is an antibody fragment that binds FcRHS. In some embodiments of any of the antibodies, the antibody is an IgGl, IgG2a or IgG2b antibody.

In some embodiments of any of the antibodies, the antibody has one or more of the following characteristics: a) cross reactive with full length human and cyno FcRHS, b) does not cross react with FcRBl, FcRFT.2, FcRH3, and/or FcRH4, c) binds to endogenous Fc I-15, d) does not cross react with Fe&H5a, and e) does not cross react with another Ig~like domain of FcRH5,

In some embodiments of any of the antibodies, the antibody is a bispecifie antibody. In some embodiments, the bispecifie antibody binds FcRHS and CD3.

In some embodiments, an isolated nucleic acid that encodes an antibody described herein is provided. In some embodiments, a host cell comprising the nucleic acid is provided. In some embodiments, a method of producing an antibody described herein is provided. In some embodiments, the method comprises culruring the host cell comprising the nucleic acid that encodes an antibody.

In some embodiments, immunoconjugates are provided. In. some embodiments, an

itnmunocomugate comprises an anti-Fc H5 antibody and a cytotoxic agent. In some embodiments, the anti-FcRH5 antibody binds an isoform c-specific region of die extracellular domain of FcRHSc. In some embodiments, the anti-FcRH5 antibodies binds Ig-like domain 9 of FcRHSc. In some

embodiments, an immunoconjugate has the formula Ab-(L~D)p, wherein: (a) Ab is an antibody described herein; (b) L is a linker; (c) D is a drug selected from a maytansinoid, an auristatin, a ealicheamiein, a pyrrolobenzodiazepine, and a nemombiein derivative; and (d) p ranges from 1-8. In some embodiments, D is an auristatin. In some such embodiments, D has formula Ό

wherein R' and R° are each methyl, R 3 and R 4 are each isopropyl, R 5 is H, R' is sec-butyl, each R s is independently selected from CH 3 , O-CH 3 , OH, and H; R" is H; and R 38 is -C(R 8 )j-C{R 8 ) 2 ~aryL In some embodiments, D is MMAE having the structure:

In some embodiments, D is a pyrrolobenzodiazepine of Fonnula A:

wherein the dotted lines indicate the optional presence of a double bond between CI and€2 or C2 and C3; R 2 is independently selected from H, OH, =0, =C¾, CN, R, OR, =CH-R°, -C(R°)2, 0-S0 2 -R, CQ>R and COR, and optionally forther selected from halo or dihalo, wherein R is independently selected from R, C0 2 R, COR, CHO, CC¼H, and halo; R 6 and R are independently selected from H, R, OH, OR, SH, SR, N¾, NHR, NRR', N0 2s Me 3 Sn and halo; R 7 is independently selected from H, R, OH, OR, SH, SR, N¾, NHR, NRR * , N0 2) Me 3 Sn and halo; Q is independently selected from O, S and NH; R : i is either H. or R or, where Q is O, SO s M, where M is a metal cation; R and R' are each independently selected from optionally substituted C t „g alkyl, CY12 alkyl,€3.8 heterocyclyi, C . 2 n heterocyclyl, and CK. 2G aryi groups, and optionally in relation to the group NRR.', R and R' together with the nitrogen atom to which they are attached form an optionally substituted 4-, 5-, 6~ or

7-membered heterocyclic ring; R , R !'~ , R 19 and R i? are as defined for ΡΛ R s , R 9 and R 7 respectively; R" is a C3.12 alkylene group, which chain may be interrupted by one or more heteroatoms and/or aromatic rings that are optionally substituted; and X and X' are independently selected from O, S and

wherein n is 0 or 1 .

In some embodiments, D is a nemorubicin derivative. In some embodiments, D has a structure selected from:

.0

; and

in some embodiments, an immunoconjugate comprises a linker that is cieavable by a protease. In some embodiments, the linker comprises a val-cit dipeptide or a Phe-homoLys dipeptide. In some embodiments, an immunoconjugate comprises a linker that is acid-labile. In some such embodiments, the linker comprises hydrazone.

In some embodiments, pharmaceutical formulations are provided. In. some such embodiments, a pharmaceutical formulation comprises an immunoconjugate comprising an antibody thai binds FcRH5, e.g., as described herein. In some embodiments, the anti-FcRHS antibody binds an isoform c- specific region of the extracellular domain of FcRHSc. In some embodiments, the anti-FcRHS antibodies binds ig-iike domain 9 of FcRHSc. hi some embodiments, a pharmaceutical formulation further comprises an additional therapeutic agent.

In some embodiments, methods of treating individuals having FcRHS {e.g.., FcRH5c)-positive cancers are provided, hi some such embodiments, a method comprises administering a pharmaceutical formulation comprising an immunoconjugate comprising an antibody thai binds FcRHS and/or an FcRHS bispecific antibody, e.g., as described herein. In some embodiments, the FcRHS bispecific antibody comprises an FcRHS binding a m and a CDS binding a m, In some embodiments, the aiiii- FcRH5 antibody binds an isoform c-specific region of the extracellular domain of FcRHSc. In some embodiments, the anti-FcRHS antibodies binds Ig-like domain 9 of FcRHSc. In some embodiments, the FcRHS-positive cancer is a B-cell proliferative disorder. In some embodiments, the FcRHS-positive cancer is plasma cell neoplasm, In some embodiments, the plasma cell neoplasm is multiple myeloma, hi some embodiments, a method comprises administering an additional therapeutic agent to the individual.

In some embodiments, methods of inhibiting proliferation of an FcRHS (e.g., FcRHS c)-positive ceil are provided. In some embodiments, the method comprising exposing the cell to an

immunoconjugaie comprising an antibody that binds FcRHS and/or an FcRHS bispecific antibody under conditions permissive for binding of the antibody to FcRIT5 on the surface of the cell, In some embodiments, the FcRHS bispecific antibody comprises an FcRHS binding arm and a CD3 binding arm. In some embodiments, the anti-FcRH5 antibody binds an isoform c-specific region of the extracellular domain of FcRH5c. h some embodiments, the anti-FcRH5 antibodies binds Ig-like domain 9 of FcRHSc. In some embodiments, die antibody that binds FcRHS is an antibody described herein. In some embodiments, the FcRHS-positive cancer is a B~cell proliferative disorder. In some embodiments, the FcRHS-positive cancer is plasma cell neoplasm. In some embodiments, the plasma cell neoplasm is multiple myeloma. In some embodiments, a method comprises administering an additional therapeutic agent to the individual.

In some embodiments, an antibody that binds FcRHS is conjugated to a label. In some embodiments, the anti-FcRH5 antibody binds an isoform c-specific region of the extracellular domain of FcRHSc, In some embodiments, the anti~FcRH5 antibodies binds Ig-li.ke domain 9 of FcRHSc. In some embodiments, the antibody that binds FcRH5 is an antibody described herein. In some embodiments, the label is a positron emitter. In some embodiments, the positron emitter is 5' 'Zr.

In some embodiments, a method of detecting human FcRHS in a biological sample is provided.

In some embodiments, a method comprises contacting the biological sample with an anti-FcRHS antibody under conditions permissive for binding of the anti-FcRHS antibody to a naturally occurring human f cRH5, and detecting whether a complex is formed between the anti-FcRHS antibody and a naturally occurring human FcRHS in the biological sample. In some embodiments, the anti-FcRIIS antibody binds an isoform c~specifie region of the extracellular domain of FcRHSc. In some embodiments, the anti-FcRHS antibodies binds ig-like domain 9 of FcRHSc. In some embodiments, the anti-FcRH5 antibody is an antibody described herein.

In some embodiments, a method for detecting an FcRHS -positive cancer is provided, in some such embodiments, a method comprises (i) administering a labeled anti-FcRHS antibody to a subject having or suspected of having an FcRHS-positive cancer, and (ii) detecting the labeled anti-FcRH5 antibody in the subject, wherein detection of the labeled anti-FcRH5 antibody indicates an FcRHS- positive cancer in. the subject, in some embodiments, the anti-FcRHS antibody binds an isoform c~ specific region of the extracellular domain of FcRHSc. in some embodiments, the anti-FcRH5 antibodies binds Ig-like domain 9 of FcRHSc. hi some embodiments, an anti-FcRHS antibody is an antibody described herein. BRIEF DESCRIPTION OF THE FIGURES

FIG. 1 (A) depicts the three major isoforms of FcRHS, FcRHS a (ERTA2a; UniProt Identifier Q96RD9-3), FcRHS h (IRTA2b; UniProt Identifier Q96 D9-4), and FcRHSc (IRTA2c; UniProt Identifier Q96RD9-1), The Ig-like domains are numbered and correspond to the amino acid sequence of UniProt Identifier Q69RD9-1 (SEQ ID O:l); Ig-like domain 1 (aa ("amino acid") 23-100), Ig-like domain 2 (aa 105-185). Ig-like domain 3 (aa 188-271). Ig-like domain 4 (287-373), Ig-like domain 5 (aa 380-466), Ig-like domain 6 (aa 490-555), Ig-like domain 7 (aa 568-652), Ig-like domain 8 (aa 658- 731), and Ig-like domain 9 (aa 754-835). FIG. 1 (B) depicts part of FcRHS (SEQ ID NO: 136) and the structure and homology of FcRHS amino acids 735 to 977 of FcRHSc (SEQ ID NO:2),

FIG. 2 shows binding of FeRH5 antibodies to SVT2 cells transfected with (A) human FcRHS and (B) cyno FcRHS, in different concentrations.

FIG. 3 shows binding of FcRHS antibodies to (A) EJM ceils or (B) OPM2 cells transfected with human FcRHS, and binding of subclone supematants (C) 5A10.1 , (D) 5F1 .1 , (E) 3G7.1 , and (F) 6D2.2 to MOLF2 cells which express FcRHS endogenousiy,

FIG. 4 shows (A) binding of FcRHS subclone supematants to 293 cells transfected with WT or (B) mutant FcRHS with deletion of 4 membrane proximal extracellular domains.

FIG. 5 shows binding of (A) the FcRHS antibodies to FcRHSa by ELISA, and (B) binding of subclone supematants to human B cells.

FIG. 6 shows binding of FcRHS subclone supematants to SVT2 ceils transfected with (A) FcRHl, (B) FcRH2, (C) FcRH3, or (D) FcRH4,

FIG. 7 shows the binding of FcRHS antibody subclone supematants to NK cells.

FIG. 8 shows (A) killing activity of FcRHS bisFabs, FcRHS -TDB (clone 10A8) and (B) HER2- TDB and (C) killing activity of FcRHS-bisFabs and (D) FcRH5-TDBs on FcRHS transfected 293 cells. FIG. 9 shows (A) killing activity and (B) T~ceii activation of Fc.RHS bisFabs and FcRH5- TDBs on MOLP-2 cells,

DETAILED DESCRIPTION

I. DEFINITIONS

The term "FcRHS," as used herein, refers to any native, mature FcRHS which results from processing of an FcRHS precursor protein in a cell. The temi includes FcRHS from any vertebrate source, including mammals such as primates (e.g. humans and cynomoigus monkeys) and rodents (e.g. , mice and rats), unless otherwise indicated. The term also includes naturally occurring variants of FcRHS, e.g.. splice variants or allelic variants. In some embodiments, the amino acid sequences human FcRHS proteins is FcRHS a (IKTA2a; UniProt Identifier Q96RD9-3; 759 aa), FcRHSb (IRTA2b;

UniProt Identifier Q96RD9-4; 592 aa), FcRHSc (IRTA2c; UniProt identifier Q96RD9-1 ; 977 aa (SEQ ID NO: 1), UniProt Identifier Q96FD9-2 (124 aa), and/or FeRHSd (IRTA2d; UniProt Identifier Q96RB9-5; 152 aa).

The term "glycosylated forms of FcRH5" refers to naturally occurring forms of FcRHS mat are post-transiationaily modified by the addition of carbohydrate residues.

"Percent (%) amino acid sequence identity" with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum, percent sequence identity, and not considering any conservative substitutions as part of the 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 or Megaiign (DNASTAR.) 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. For purposes herein, however, % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2, The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc, and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code. The ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.

In situations where ALIGN-2 is employed for amino acid sequence comparisons, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows:

100 times the fraction X/Ywhere X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of arrrino acid residues in B, It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. Unless specifically stated otherwise, ail % amino acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program,

The terms "anti-FcRHS antibody" and "an antibody that binds to FcRH5" refer to an antibody that is capable of binding FcRHS with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting FcRHS. In one embodiment, the extent of binding of an ami- FcRHS antibody to an unrelated, non~FcRH5 protein is less than about 10% of the binding of the antibody to FcRH5 as measured, e.g., by a radioimmunoassay (RIA), In certain embodiments, an antibody that binds to FcRHS has a dissociation constant (Kd) of < Ι μΜ. < 100 nM, < 10 nM, , < 5 Nrn, , < 4 nM, , < 3 nM, , < 2 nM, < 1 nM, < 0.1 nM, < 0.01 nlvi, or < 0.001 nM (e.g., 10 "? M or less, e.g. from 10 "8 M to 10 '!3 M, e.g., from 10 "9 M to 10 ": ? M). In certain embodiments, an nti-FcRHS antibody- binds to an epitope of FcRHS that is conserved among FcRHS from different species. In some embodiments, the anti~FcRH5 antibody binds an isoform c-specific region of the extracellular domain of FcRH5c. in some embodiments, the anti-FeRH5 antibodies binds Ig-like domain 9 of FcRII5c.

The term "antibody" is used herein in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispeciftc antibodies (e.g., bispeciftc antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.

An "antibody fragment" refers to a molecule other than an intact antibody that comprises a portion of an intact antibody and that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab¾; diabodies; linear antibodies; single-chain antibody molecules (e.g., scFv); and niultispeciilc antibodies formed from antibody fragments.

The term "epitope" refers to the particular site on an antigen molecule to which an antibody binds.

An "antibody that binds to the same epitope" as a reference antibody refers to an antibody that blocks binding of the reference antibody to its antigen in a competition assay by 50% or more, and conversely, the reference antibody blocks binding of the antibody to its antigen in a competition assay by 50% or more. An exemplary competition assay is provided herein.

The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts, in contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen. Thus, the modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridorna method, recombinant DNA methods, phage-display methods, and methods utilising transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein.

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 native antibody structure or having heavy chains that contain an Fc region as defined herein,

A "naked antibody" refers to an antibody that is not conjugated to a heterologous moiety {e.g., a cytotoxic moiety) or radioiabel. The naked antibody may be present in a pharmaceutical formulation.

"Native antibodies" refer to naturally occurring immunoglobulin molecules with varying structures. For example, native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide- bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CHI . CH2, and CH3). Similarly, from - to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain. The light chain of an antibody may be assigned to one of two types, called kappa (κ) and lambda (λ), based on the amino acid sequence of its constant domain.

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.

A "human antibody" is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues, A "humanized" antibody refers to a chimeric antibody comprising amino acid residues from non-human HVRs and amino acid residues from human FRs. in. certain embodiments, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g. , CDRs) correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody. A humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody. A "humanized form" of an antibody, e.g., a non-human antibody, refers to an antibody that has undergone humanization.

The "class" of an antibody refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: IgA, IgD, igE, igG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG I , IgGj, IgG , IgG, ( , IgAj, and IgA 2 , The heavy chain constant domains that correspond to the different classes of immunoglobulins are called a, 8, ε, γ, and μ, respectively.

The term "Fc region" herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions, ϊη one embodiment, a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl terminus of the heavy cham. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present. Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EIJ numbering system, also called the EU index, as described in Kabat et ai, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 19 1 ,

The term "variable region" or "variable domain" refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen. The variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariabie regions (HVRs), (See, e.g., Kindt el ai. Kuby Immunology, 6* ed., W.H. Freeman and Co., page 91 (2007),) A single VH or VL domain may be sufficient to confer antigen-binding specificity. Furthermore, antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portoiano e al., J, Immunol, 150:880-887 (1993); Ciarkson et al, Nature 352:624-628 (1991).

"Framework" or "FR" refers to variable domain residues other than hypervariabie region (HVR) residues. The FR of a variable domain generally consists of four FR domains: FR1 , FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1-H1(L1)-FR2-H2(L2)-FR3-H3(L3)-FR4.

A "human consensus framework" is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences. Generally, the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences, Generally, the subgroup of sequences is a subgroup as in Kabat et ai, Sequences of Proteins of Immunological Interest, Fifth Edition, ΝΓΗ Publication 91 -3242, Bethesda MD (1991), vols, 1-3. in one embodiment, for the VL, the subgroup is subgroup kappa I as in Kabat et ai, supra, in one embodiment, for the VH, the subgroup is subgroup III as in Kabat et al, supra.

The term "hypervariable region" or "HVR," as used herein, refers to each of the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops ("hypervariable loops"). Generally, native four-chain antibodies comprise six HVRs; three in the VH (Hi, H2, H3), and three in the VL (LI , L2, L3). HVRs generally comprise amino acid residues from the hypervariable loops and/or from the "complementarity determining regions" (CDRs), the latter being of highest sequence variability and/or involved in antigen recognition. Exemplary hypervariable loops occur at amino acid residues 26-32 (Li), 50-52 (L2) 5 91-96 (L3), 26-32 (HI), 53-55 (H2), and 96-101 (H3). (Chothia and Les.k, J. Mol Biol 196:901 -917 (1987).) Exemplary CDRs (GDR-L1, CDR-L2,

CDR-L3, CDR-H1, CDR-H2, and CDR-H3) occur at atnino acid residues 24-34 of Li , 50-56 of L2, 89- 97 of L3, 31-35B of HI , 50-65 of H2. and 95-102 of 113. (Kabat et ai, Sequences of Proteins of immunological interest, 5th Ed. Public Health Sendee, National Institutes of Health, Bethesda, MD (1 1).) With the exception of CDR1 in VH, CDRs generally comprise the amino acid residues that form the hypervariable loops. CDRs also comprise "specificity determining residues," or "SDRs," which are residues that contact antigen. SDRs are contained within regions of the CDRs called abbreviated-CDRs, or a-CDRs. Exemplary a-CDRs (a-CDR-Ll , a-CDR-L2, a-CDR-L3, a-CDR-Hl, a- CDR-H2, and a~CDR~H3) occur at amino acid residues 31-34 of LI, 50-55 of L2, 89-96 of L3, 31-.35B of HI, 50-58 of H2 S and 95-1 02 of H3. (See Almagro and Fransson, Front. Biased, 13:1619-1633 (2008).) Unless otherwise Indicated, HVR residues and other residues in the variable domain {e.g., FR residues) are numbered herein according to Kabat et ai, supra.

An "acceptor human framework" for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework deri ved from a human immunoglobulin framework or a human consensus framework, as defined below. An acceptor human framework "derived from" a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less. 3 or less, or 2 or less, in some embodiments, the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.

"Affinity" refers to the strength of the sum total of noncovalent Interactions between a single binding site of a molecule (e.g.. an antibody) and its binding partner {e.g., an antigen). Unless indicated otherwise, as used herein, "binding affinity" refers to intrinsic binding affinity which reflects a 1 :1 interaction between members of a binding pair (e.g., antibody and antigen). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following. An "affinity matured" antibody refers to an antibody with one or more aiterations in one or more hypervariable regions (HVRs), compared to a parent antibody which does not possess such alterations, such alterations resulting in an improvement in the affinity of the antibody for antigen.

An "immunoconjugate" is an antibody conjugated to one or more heterologous moleeule(s), including but not limited to a cytotoxic agent.

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. Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At 11 1 , I m , I i25 , Y 90 , Re 185 , Re' 88 , Sm 153 , Br' 2 , P 32 , Pb 2 ' 2 and radioactive isotopes of Lu); ehemotherapeutie agents or drugs (e.g., methotrexate, adriamicin. vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chloratnbue.il, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleoiyiic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof; and the varioiss antitumor or anticancer agents disclosed below.

"Effector functions" refer to those biological activities attributable to the Fc region of an antibody, which var with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell- mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B-eell receptor); and B~ceil activation,

An "isolated antibody" is one which has been separated from a component of its natural environment. In some embodiments, an antibody is purified to greater than 95% or 99% purity as determined by, for example, eiectrophoretic (e.g., S ' DS-PAGE, isoelectric focusing (lEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC). For review of methods for assessment of antibody purity, see, e.g., Flatman et al, J. C ro atogr. B 848:79-87 (2007).

An "isolated nucleic acid" refers to a nucleic acid molecule that has been separated from a component of its natural environment. An isolated nucleic acid includes a nucleic acid molecule contained in ceils that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomaliy or at a chromosomal location that is different from its natural chromosomal location.

"isolated nucleic acid encoding an anti-FcRH5 antibody" refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell

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."

The terms "host cell," "host cell line," and "host cell culture" are used interchangeably and refer to ceils into which exogenous nucleic acid has been introdiiced, including the progeny of such cells. Host cells include "transformants" and "transformed cells," which include the primary

transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.

As used herein, "treatment" (and grammatical variations thereof such as "treat" or "treating") refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, 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. In some embodiments, antibodies provided herein are used to delay development, of a disease or to slow the progression of a disease,

The terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation. Examples of cancer include, but are not limited to, carcinoma, lymphoma {e.g., Kodgkin's and non-Hodgkin's lymphoma), blastema, sarcoma, and leukemia. More particular examples of such cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, small intestine cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, leukemia sid other iymphoproliferative disorders, and various types of head and neck cancer.

A "B~cell malignancy" herein includes non-Hodgkin's lymphoma (NHL), including low grade/follicular NHL, small lymphocytic (SL) NHL, intermediate grade/foilicular NHL, intermediate grade dif&se NHL, high grade irnmunoblastie NHL, high grade lymphoblastic NHL, high grade small non-cleaved cell NHL, bulky disease NHL, mantle cell lymphoma, AIDS-related lymphoma, and Waldenstrom's Macroglobulinemia, non-Hodgkin's lymphoma (NHL), lymphocyte predominant Hodgkin's disease (LPHD), small lymphocytic lymphoma (SLL), chronic lymphocytic leukemia (CLL), indolent NHL including relapsed indolent NHL and rituximab-refractory indolent NHL; leukemia, including acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), Hairy cell leukemia, chronic myeioblastic leukemia; mantle cell lymphoma; and other hematologic malignancies, Such malignancies may be treated with antibodies directed against B-cell surface markers, such as FcRH5 (e.g., FcRH5c). Such diseases are contemplated herein to be treated by the administration of an antibody directed against a B~cell surface marker, such as FcRH5 (e.g., FeRHSc), and includes the administration of an unconjugated ("naked") antibody or an antibody conjugated to a cytotoxic agent as disclosed herein, Such diseases are also contemplated herein to be treated by combination therapy including an anti-FcRH5 antibody (including FeRH5 bispecific antibody) or anti-FcRHS antibody drug conjugate in combination with another antibody or antibody drug conjugate, another cytotoxic agent, radiation or other treatment administered simultaneously or i series.

The term "non-Hodgkin's lymphoma" or "NHL", as used herein, refers to a cancer of the lymphatic system other than Hodgkin's lymphomas. Hodgkin's lymphomas can generally be distinguished from non-Hodgkin's lymphomas by the presence of Reed-Stemberg cells in Hodgkin's lymphomas and the absence of said cells in non-Hodgkin's lymphomas. Examples of non-Hodgkin's lymphomas encompassed by the term as used herein include any that would be identified as such by one skilled in the art (e.g., an oncologist or pathologist) in accordance with classification schemes known in the art, such as the Revised European-American Lymphoma (REAL) scheme as described in Color Atlas of Clinical Hematology (3rd edition), A. Victor Hoffbrand and Joh E, Pettit (eds.) (Karcourt Publishers Ltd., 2000). See, in particular, the lists in FIGS, 1 1.57, 1 1.58 and 1 1.59. More specific examples include, but are not limited to, relapsed or refractory MIL, front line low grade NHL, Stage 11MV NHL, chemotherapy resistant NHL, precursor B lymphoblastic leukemia and/or lymphoma, small lymphocytic lymphoma, B-cell chronic lymphocytic leukemia and/or prolymphocyte leukemia and/or small lymphocytic lymphoma, B-cell prolymphocyte lymphoma, mimunocytoma and/or iymphopiasmacytic lymphoma, Iymphopiasmacytic lymphoma, marginal zone B-cell lymphoma, splenic marginal zone lymphoma, extranodal marginal zone— MALT lymphoma, nodal marginal zone lymphoma, hairy cell leukemia, plasmacytoma and/or plasma cell myeloma, low grade/follicuiar lymphoma, intermediate grade foliicular NHL. mantle cell lymphoma, follicle center lymphoma (follicular), intermediate grade diffuse NHL, diffuse large B-ceii lymphoma, aggressive NHL

(including aggressive front-line NHL and aggressive relapsed NHL), NIIL relapsing after or refractory to autologous stem cell transplantation, primary mediastinal large B~cell lymphoma, primary effusion lymphoma, high grade immunoblastic NHL, high grade lymphoblastic NHL, high grade small non- cleaved cell NHL, bulky disease NHL, Burkitt's lymphoma, precursor (peripheral) large granular lymphocytic leukemia, mycosis fungoides and/or Sezary syndrome, skin (cutaneous) lymphomas, anaplastic large cell lymphoma, angiocentric lymphoma.

Plasma cells disorders result from the uncontrolled division or multiplication of a plasma cell clone. Plasma cells arise from activated B lymphocytes (i.e., B-eells). Each B-cell produces a unique receptor, known as the B-cell receptor, arrayed on its cell surface that is specific for a foreign substance, i.e., antigen. When a B-cell receptor binds its cognate antigen, the cell expressing the receptor is activated to re-enter the cell cycle, producing many clonal copies of itself. The clones mature into plasma cells that reside principally in the hone marrow and thai are specialized to produce copies of the B-cell receptor that are released into the blood stream as antibodies, In a plasma ceil disorder, the plasma ceil or the parent B-cell suffers genetic damage resnlting in suppression of or insensitivity to the normal restraints on cell division and'Or activity. Daughter plasma cells derived from such cells are malignant in that they may divide unchecked and/or generate excess amount of the same immunoglobulin (antibody). Often the immunoglobulin produced is incomplete or has an incorrect conformation that can result m " accumulation of the protein (also known as monoclonal proteirj, M protein, paraprotein or amyloid protein, dependent on the specific disorder) in the serum, tissues or organs (especially the kidneys), leading to organ dysfunction and/or failure. Plasma ceil disorders include monoclonal gammopathies of undetermined significance (MGUS), multiple myeloma (MM), macroglobulinemia, heavy chain diseases, and systemic light-chain amyloidosis (AL), which are differentiated based on the proliferative nature of the clone, the extent of marrow involvement, and the type of M protein expressed. Additional plasma ceil disorders are solitary plasmacytoma,

extramedullary plasmacytoma, multiple solitary plasmacytomas, plasma cell leukemia, Waldenstrom's macroglobulinaemia, B-cell non~Hodgkin lymphomas, B-cell chronic lymphocytic leukemia.

Tl e term 'TcRH5 -positive cancer" refers to a cancer comprising cells that express FcRHS on their surface. For the purposes of determining whether a cell expresses FcRHS on the surface, FcRHS mRNA expression is considered to correlate to FcRHS expression on the cell surface. In some embodiments, expression of FcRHS mRNA is determined by a method selected from in situ hybridization and R ' F-FCR (including quantitative RT-PCR). Alternatively, expression of FcRHS on the ceil surface can be determined, for example, using antibodies to FcRHS in a method such as irnmunohistochemistry, FACS, etc. In some embodiments, FcRHS is one or more of FcRHS a, FcRIISb, FcRHSc, UniProt Identifier Q96RD9-2, and/or FeRHSd, In some embodiments, the FcRHS is FcRHSc.

'The term "FcRHS-positive cell" refers to a ceil that expresses FcRHS on its surface, in some embodiments, FcRHS is one or more of FeRFfSa, FcRHSb, FcRHSc, UniProt Identifier Q96RJD9-2, and/or FeRHSd. In some embodiments, tlie FcRHS is FcRHSc.

An "effective amount" of an agent, e.g. , a pharmaceutical formulation, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.

An "individual" or "subject" is a mammal. Mammals include, but are not limited to.

domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats). In certain, embodiments, the individual or subject is a human.

The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products. The term "pharmaceutical formulation" refers to a preparation which is in such form as ΐο permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unaccepiabiv toxic to a subject to which the formulation would be administered.

A "pharmaceutically acceptable carrier" refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject. A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, exeipient, stabilizer, or preservative.

"Aikyl" is C] -Ci 8 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms, Examples are methyl (Me, -CH3), ethyl (Et, -CH2CH3), 1 -propyl (n-Pr, n-propyl, -CH2CH2CH3), 2- propyl (i-Pr, i-propyl, ~CH(CH3)2), 1 -butyl (n~Bu, n-butyl, -CH2CH2CH2CH3). 2 -methyl- 1 -propyl (i- Bu, i-butyl -CH2CH(CH3)2), 2-buiyi (s-Bu, s-butyi, « CH(CH3)CH2CH3), 2-mefhyi-2-propyi (t-Bu, t- butyl, -C(CH3)3), 1 -pentyl (n-pentyl, -CH2CH2CK2.CH2CH3), 2-pentyi (-CH(CH3)CH2 Il2CH3), 3- pentyl (-CH(CH2CH3)2) S 2-methyl-2-butyl (~C(CH )2CH2CH3), 3-methyl-2 -butyl (- CH(CH3)CH(CH3)2), 3-methyl-1 -butyl (-CH2CH2CH(CH3)2), 2-methyl-l -butyl (- CH2CH(CH3)CH2CH3), 1 -hexyi (-CH2CH2CH2CH2CH2CH3), 2-hexyl (-

CH(CH3)CIl2 il2CH2CH3), 3-hexyl (-CH(CH2CH3)(CH2CI-l2CH3)), 2 » rnethyl-2-pentyl (- C(CH3)2CH2CH2CH3) > 3-methyl-2-pentyl (-CH(CH3)CH(CH3)CH2CH3), 4-methyl~2-pentyl (- CH(CH3)CH ' 2CH(CH3)2), 3~methyl-3-peniyi (~C(CH3)(CH2CH ' 3)2), 2~methyi-3~pentyl (- CH(CH2CH3)CH(CH3)2), 23-dimethyl-2-butyl (-C(CH3)2CH(CH3)2), 3,3-dixnethyl-2-butyl (- CH(CH3)C(CH3)3, The term "Ci-Q alkyi," as used herein refers to a straight chain or branched, saturated or unsaturated hydrocarbon having from 1 to 8 carbon atoms. Representative "Cj-Cs aikyl" groups include, but are not limited to, -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl, -n-hexyl, -11- heptyl, -n-octyi, -n-nonyl and -n-decyi; while branched Cj-Cg alkyis include, but are not limited to, - isopropyl, -sec-butyl, -isobutyl, -ieri-butyi, -isopentyl, 2-methylbutyl, unsaturated CrQ alkyis include, but are not limited to, -vinyl, -ally!, -1-b tenyl, -2-butenyl, -isobutylenyl, -l -penteny!, -2-pentenyl, - 3 -methyl -l-butenyl, ~2-methyl-2-butenyl, -2,3-dimethyi-2~butenyl, 1 -hexyi, 2~hexyl, 3-hexyl,- acetylenyl, -propynyl, -l -butynyl, -2-butynyl, -1 -pentynyl, -2-pentynyl, -3 -methyl- 1 butynyl. A C[-C 8 alkyi group can be unsubstituted or substituted with one or more groups including, but not limited to, - C C 8 alkyi, -0-(C r C 8 alkyi), -aryl, ~C(0)R\ -OC(0)R', -C(0)OR\ -C(0) ¾ , -C(0)NHR\ - C(0)N(R')2 -NHC(0)R -SO 3 R', -S(0) 2 R', -S(0)R% -OH, -halogen, -N 3 , -NH 2 , -NH(R'), - (R') 2 and -CN; where each R' is independently selected from H, -C[-Q alkyi and aryl,

The term "CrQ ? . alkyi," as used herein refers to a straight chain or branched, saturated or unsaturated hydrocarbon having from 1 to 12 carbon atoms. A Ci~C l2 alkyi group can be unsubstituted or substituted with one or more groups including, but not limited to, -C ; -CR alkyi, -0-(C R C G alkyi), - aryl, -€(G)R\ -OC(0)R\ -C(0)OR\ -C(0)NH 2 , -C(0) HR\ ~C(0)N(R¾ -NHC(0)R\ -SO 3 R', - S(0) 2 R\ -S(0)R\ -OH, -halogen, -N 3 , -NIL, -NH(R'), -N(R') 2 and -CN; where each R' is independently selected from H, -C C 8 alkyl and aryl

The term "Cj-Cg alkyi," as used herein refers to a straight chain or branched, saturated or unsaturated hydrocarbon having from 1 to 6 carbon atoms. Representative "C;-Q alkyl" groups include, but are not limited to, -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl, -and n-hexyl; while branched Cj-Cg alkyls include, but are not limited to, -isopropyl, -sec-butyL -isobutyl, -fcrf-butyl, ~ isopentyl; and 2-meihyibutyl; unsaturated€ t -C 6 alkyls include, hist are not limited to, -vinyl, -allyl, -

1- butenyl, -2-butenyl, and -isohuty!enyl, -1-pentenyi, -2-pentenyl, -3-methyl-l-butenyl, -

2- methyl-2-butenyl, ~2,3<lim.et.hyl-2~butenyl, 1-hexyl, 2-hexyI, and 3-hexyl. A Cr alkyl group can be unsubstituted or substituted with one or more groups, as described above for C;-Cg alkyl group.

The term "C Q alkyl," as used herein refers to a straight chain or branched, saturated or unsaturated hydrocarbon having from 1 to 4 carbon atoms. Representative "C1-C alkyl" groups include, but are not limited to, -methyl, -ethyl, -n-propyl. -n-butyi; while branched C 1 -C4 alkyls include, but are not limited to, -isopropyl, -seobutyl, -isobutyl, -ierf-butyl; unsaturated Q-Q alkyls include, but are not limited to, -vinyl, -allyl, -1 -buienyL -2-butenyl, and -isobutyienyl A Q- alkyi group can be ^substituted or substituted with one or more groups, as described above for C Q alkyl group.

"Alkoxy" is an alkyi group singly bonded to an oxygen, Exemplary alkoxy groups include, but are not limited to, methoxy (-OCH 3 ) and ethoxy (-OC¾Cj¾). A "Q-Q alkoxy" is an alkoxy group with 1 to 5 carbon atoms. Alkoxy groups may can be unsubstituted or substituted with one or more groups, as described above for alkyl groups.

"Alkenyl" is C2~C] g hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaiuration, i.e. a carbon-carbon, sp ' double bond. Examples include, but are not limited to: etliyiene or vinyl cyciopenienyl {-C5H 7 ), and 5-hexenyl (~C¾ CH,CH,CH 2 CH=CH 2 ). A "C-rQ alkenyi" is a hydrocarbon containing 2 to 8 normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaiuration, i.e. a carbon-carbon, sp " double bond.

"Alkynyl" is C2-C} hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaiuration. i.e.. a carbon-carbon, sp triple bond. Examples include, but are not limited to: acetyienic (-C=CH) and propargyi (-CH^CsCH), A "C?-C 8 alkynyl" is a hydrocarbon containing 2 to 8 normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaiuration, i.e. a carbon-carbon, sp triple bond,

"Alkylene" refers to a saturated, branched or straight chain or cyclic hydrocarbon radical of 1-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. Typical alkylene radicals include, but are not limited to: methylene (-CH r ) 1 ,2-ethyl (-CH 2 CH ), 1 ,3 -propyl {-CH 2 C¾C¾-), 1 ,4-butyl

i-CH 2 C¾C¾CH r ), and the like. A "Cj-Cjo alkylene" is a straight chain, saturated hydrocarbon group of the formula -(CH- )i„ r- Examples of a C:-Qo alkylene include methylene, ethylene, propylene, butylene, pentylene, hexylene, heptylene, ocytyiene, nonyiene and decalerte.

"Alkenyiene" refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene, Typical alkenyiene radicals include, but are not limited to: 1 ,2-ethyiene (-CH=€H-).

« Aikynylene" refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical of 2-1 S carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne. Typical alkynylene radicals include, but are not limited to: acetylene (-C^C-), propargyl {-€¾(¾€-), and 4-pentynyl (-C¾CH 2 CH 2 i €-).

"Aryl" refers to a earbocyclic aromatic group. Examples of aryl groups include, hut are not limited to, phenyl, naphthyl and anthracenyl. A earbocyclic aromatic group or a heterocyclic aromatic group can be unsubstituied or substituted with one or more groups including, but not limited to, -C:-Cs alkyl -0-(C,-C 8 alkyl), -aryl, -C(0)R\ -OC(0)R\ -C(0)OR', -C(0)NH 2 , -C(0)NHR\ -C(G)N{R') 2 - NHC(0)R', ~S(G) 2 R\ -S(0)R\ -OH, -halogen, ~N 3 , -NH 2 , - H(R * ), -N(R') 2 and ~CN; wherein each R' is independently selected from H, -C r C s alkyl and aryl

A "C5-C20 aryl" is an aryl group with 5 to 20 carbon atoms in the earbocyclic aromatic rings. Examples of C5-C20 aryl groups include, but are not limited to, phenyl, naphthyl and anthracenyl. A C$- C;o aryl group can be substituted or unsubsiiiuted as described above for aryl groups. A "Cs-C^ aryl" is an aryl group with 5 to 14 carbon atoms in the earbocyclic aromatic rings. Examples of iV€ i4 ar l groups include, but are not limited to, phenyl, naphthyl and anthracenyl. A Cs-C^ aryl group can be sisbstituted or unsubstituied as described above for aryl groups.

An "arylene" is an aryl group which has two covalent bonds and can be in the ortho. meta, or

in which the phenyl group can be unsubsiiiuted or substituted with up to four groups inciuding, but not iimiied to, -C C g alkyl, -0-(C r C 8 alkyl), -aryl, -C(0)R\ -OC(0)R', -C(0)OR\ -C(0)N¾ , - C(0)NHR\ ~C(0)N(R') 2 -NHC(0)R\ ~S(G) 2 R\ -8(G)R\ -OH, -halogen, -N 3 , -N¾, -NH(R'), -N(R 5 ) and -CN; wherein each R' is independently selected from H, -C r C* alkyl and aryl.

"Arylalkyi" refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp J carbon atom, is replaced with an aryl radical. Typical arylalkyi groups include, but are not limited to, benzyl, 2 -phenyl ethan~l~yl, 2-phenylethen-i-yl, naphthylrnethyl, 2-naphtliylethan-l -yi, 2-naphthylethen~l -yl 5 naphthohenzyl, 2~naphthophenyiethan-l ~yl and the like. The arylalkyl group comprises 6 to 20 carbon atoms, e.g. the alkyl moiety, including alkanyl, alkenyi or alkynyi groups, of the arylalkyl group is 1 to 6 carbon atoms and the aryi moiety is 5 to 14 carbon atoms.

"Heteroaryiaikyl" refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp 3 carbon atom, is re laced with a heteroaryl radical Typical heteroaryiaikyl groups include, but are not limited to, 2-benzimidazoiylmethy3, 2~iuryi ethyl, and the like. The heteroaryiaikyl group comprises 6 to 20 carbon atoms, e.g. the alkyl moiety, including alkanyl, aikenyl or alkynyi groups, of the heteroaryiaikyl group is 1 to 6 carbon atoms and the heteroaryl moiety is 5 to 14 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S. The heteroaryl moiety of the heteroaryiaikyl group may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S), for example: a bicycle [4,5], [5,5], [5,6], or [6,6] system.

"Substituted alkyl,' ' "substituted aryl," and "substituted arylalkyl" mean alky], aryl, and arylalkyl respectively, in which one or more hydrogen atoms are each independently replaced with a substituent. Typical substituents include, but are not limited to, -X, -R, -O " , -OR, -SR, -S ' , -NR 2 , -NR 3 , =NR, -CX 3 , -CM, -OCN, -SCN, -N=CO, -NCS, -NO, -NO,, -N 2> -N,, NC(=0)R, -C(===0)R, - C(-0)NRj, -SO 3 " , -SO 3 H, -S(==0) 2 R, -OS(-0) 2 OR, -S(=0) 2 NR, -S(=0)R, -OP«))(OR) 2 , -P(=0)(OR) 2! -P0 3> -P0 3 H 2 , -C(0)R, -C(=G)X, -C(=S)R, -C0 2 R, -CQ 2 , -C(=S)OR, -C(=Q)SR, -C(=S)SR,

-C(=0)NR 2 , -C(=S)NR 2 , -C(==NR)NR 2> where each X is independently a halogen: F, CI, Br, or I; and each R is independently -H, CVC sa alkyl, C 3 -C\o <& '£ > heierocycle, protecting group or prodrug moiety. Alkylene. alkenylene, and aikynylene groups as described above may also be similarly substituted,

"Heteroaryl" and "heierocycle" refer to a ring system in which one or more ring atoms is a heteroatom, e.g. nitrogen, oxygen, and sulfur. The heterocycle radical comprises 3 to 20 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S. A heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S) or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system.

Exemplary heterocycles are described, e.g., in Paquette, Leo A,, "Principles of Modem

Heterocyclic Chemistry" (W.A. Benjamin, New York. 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; "The Chemistry of Heterocyclic Compounds, A series of Monographs" (John Wiley Sc. Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and /. Am. Chern. Soc. (1960) 82:5566.

Examples of heterocycles include by way of example and not limitation pyridyl,

dihydroypyridyl, tetrahydropyridyl (piperidyl), thiazolyi, tetrahydrothiophenyl, sul&r oxidized tetrahydrothiophenyl, pyrimidinyl, turanyi, thienyl, pyrrolyi, pyrazolyl, imidazolyl, tetrazolyl, benzofurany!, thianaphthalenyl, itidolyi, indolenyi, qidnolinyl, isoquinolinyi, benzirnidazolyl, piperidinyi, 4-piperidonyl, pyrroiidinyl, 2-pyrrolidonyl, pyrrolinyl, tetrahydrofuranyl, bis- tetrahydrofuranyl. tetrahydropyranyl, bis-tetrahydropyranyl. tetrahydroquinolinyl,

tetrahydroisoqumolinyl, deeahydroquinolinyl, oetahydroisoquinoiinyl, azocmyl, triazinyl, 611-1 ,2,5- thiadiazinyl, 2H.6H-1 ,5,2-dithiazmyl, ihienyi, thianthrenyl, pyranyl, isobenzofuranyl, chroraenyl, xanthenyi, phenoxathmyl, 2H-pyrrolyl, isothiazolyl, isoxazoiyl, pyrazinyl, pyridaziiiyl, indolirinyl, isoindolyl, 3H-indoiyl, lH-indazolyl, purinyi, 4H-qumolizinyi, phthalazinyl, naphthyridinyl, quinoxalinyl, quinazoi yl, ehinolrnyl, pteridinyl, 4aH-earbazoiyL earbazoryl, β-carbolinyl,

phenandiridinyl, acridinyi, pyriniidinyl, phenandirolinyl, phenazinyi, phenomiazinyl, furazanyl, phenoxazinyi, isoehromanyl, ehrornanyl, imidazolidinyl, imidazoiinyl, pyrazolidinyi, pyrazoiinyL piperazinyl, mdolmyl, isoindolmyl, qumuclidinyl, morphoiinyl, oxazolidinyl, benzotriazolyi, benzisoxazoiyl, oxindolyl, benzoxazoimyi, and isatinoyl.

By way of example and not limitation, carbon bonded heterocycles are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine. position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazoie, imidazole or thiazoie, position 3, 4, or 5 of an isoxazole. pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6. 7, or 8 of a quinoiine or position 1 , 3, 4, 5, 6, 7, or 8 of an

isoqui.noline. Still more typically, carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5-pyridyl, 6-pyridyl, 3-pyridazinyl, 4-pyridazinyi, 5~pyridazinyl, ό-pyridazi.nyl, 2~pyrimidinyl, 4~ pyrimidinyi, 5-pyrimidinyl, 6-pyrimidinyi, 2 -pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2- thiazoiyl, 4-thiazolyi, or 5 -thiazoiyl.

By way of example and not limitation, nitrogen bonded heterocycles are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2- imidazoiine, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazohne, piperidine, piperazine, indole, indoline, 1 H-indazole, position 2 of a isoindole, or isoindoline, position 4 of a rnorphoiine, and position 9 of a earbazoSe, or β-carboline. Still more typically, nitrogen bonded heterocycles include 1 - aziridyl, 1 -azetedyl, 1 -pyrrolyi, 1 -imidazolyl, 1 -pyrazolyl, and 1 -piperidinyl

A "Cs-Cg heterocycie" refers to an. aromatic or non-aromatic C Q carboeycie i which one to four of the ring carbon atoms are independently replaced with a heteroatom from the group consisting of O, S and N. Representative examples of a C Cs heterocycie include, but are not limited to, benzofuranyl, benzothiop.hene, indolyl, benzopyrazoly!, eoumarinyl, isoquinolinyl, pyrrolyi, thiophenyl, furanyl, thiazoiyl, imidazolyl, pyrazolyl, triazolyl, qisinolinyl, pyrhnidinyl, pyridinyl, pyridonyl, pyrazinyi, pyridazinyL isothiazolyl, isoxazoiyl and tetrazolyl A C 3 -C 8 heterocycie can be unsubsiituied or substituted with up to seven groups including, but not limited to, ~C ; -Q alkyl, -0-(C C s aikyi), - aryl -C(0)R', -OC(0)R\ -C(G)GR\ -C(0)NH 2 , -C(0) HJR', ~C(0)N(R') 2 -NHC(0)R\ -S(0) 2 R\ - S(0)R\ -OH, -halogen, -N 3 , -N¾ s - H(R'), -N(R'):? and -CM; wherein each R' is independently seieeied from II, -€ r Q alkyl and aryl "CyCg heterocyclo" refers to a C : ,~C ? , heierocycie group defined above wherein one of the heierocycie group's hydrogen atoms is replaced with a bond. A C^-Cg heterocyclo can be unsubstituied or substituted with up to six groups including, but not limited to, -CpCg aikyl, -0-(C t ~ 8 a!kyl), -aryl, ~ C(0)R% ~OC(0)R', -C(0)OR', -C(0)NH 2 , -C(0)NHR', -C O R 1 ), -NHC(0)R', ~S(0) 2 R\ -S(0)R% -OH, -halogen, -N 3 , -NH 2 , -NH(R'), -N(R') 2 and -CN; wherein each R' is independently selected from H, -Cj-Cg alkyl and aryl.

A "C Cjo heierocycie" refers to an aromatic or non-aromatic CyC s carbocycie in which one to four of the ring carbon atoms are independently replaced with a heteroatom from the group consisting of O, S and N. A C3-C 0 heierocycie can be unsubatituted or substituted with up to seven groups including, but not limited to, -C Q alkyi, -0-(Ci-C 8 alkyl), -aryl. -C(0)R\ -QC(0)R\ -C(0)OR' s - C(Q)NH 2 , -C(0)NHR', -C(0)N(R') 2 -NHC(0)R\ -S(0) 2 R', -S(0)R\ -OH, -halogen, -N 3 , -NH 2 , - NH(R'), -N(R') 2 and -CN; wherein each R' is independently selected from II, -C r C s alkyl and aryl "Q-C 20 heterocyclo" refers to a CJ-C J O heierocycie group defined above wherein one of the heierocycie group's hydrogen atoms is replaced with a bond.

"Carbocycie" means a saturated or unsaturated ring having 3 to 7 carbon atoms as a monocyele or 7 to 12 carbon atoms as a bicycle. Monocyclic carboeycies have 3 to 6 ring atoms, still more typically 5 or 6 ring atoms. Bieyclie carboeycies have 7 to 12 ring atoms, e.g. arranged as a bicyelo [4,5], [5,5], [5,6] or [6,6] system, or 9 or 10 ring atoms arranged as a bicyelo [5,6] or [6,6] system.. Examples of monocyclic carboeycies include cyclopropyi, cyclobutyi, cyclopentyl, l -cyclopent-l-enyl, 1 -eyclopeni-2-enyl, 1 - eyclopent-3-enyl, cyclohexyl, 1 -cyclohex-l-enyi, l-cyciohex-2-enyl, l-eyelohex-3-enyl, cycloheplyl, and cyelooctyi.

A "C Cg carbocycie" is a 3-, 4-, 5-, 6-, 7- or 8-membered saturated or unsaturated non- aromatic carbocyclic ring. Representative C3-Q carboeycies include, but are not limited to, - cyclopropyi, -cyclobutyi, -cyclopentyl, -cyclopentadienyi, -cyclohexyl, -cyciohexenyl, -1 ,3- cyclohexadienyl, -1 ,4-eyclohexadienyl, -cycloheplyl -1 ,3-cycloheptadienyl, -1 ,3,5-cycloheptatrienyl, - cyelooctyi, and -cyclooctadienyl, A Cj-Cg carbocycie group can be unsubatituied or substituted with one or more groups including, but not limited to, -C r Cs alkyi, -Q-(C ; -Cg alkyl), -aryl, -C(0)R.\ ~ OC(0)R' ) -C(0)OR', -C(0)NH 2 , -C(0)NHR\ -C(0)N(R') 2 ~NHC(0)R', -S(Q) 2 R% -S(0)R', -OH, - halogen, -N 3 , -NH 2 , -NH(R'), -N(R') 2 and -CN; where each R' is independently selected from H, ~C ; ~ Cg alkyl and aryL

A "Cj-Cg carbocyclo" refers to a Cj-Cg carbocycie group defined above wherein one of the carbocycie groups' hydrogen atoms is replaced with a bond.

"Linker" refers to a chemical moiety comprising a covalent bond or a chain of atoms that covalenily attaches an antibody to a drug moiety. In various embodiments, linkers include a divalent radical such as an aikyldiyl, an aryidiyl, a hei.eroaryl.diy3, moieties such as: -(CR 2 ) a O(CR 2 ) f ,~, repeating units of aikyloxy (e.g. polyethyienoxy, PEG, poiyrnethyleneoxy) and aikylamino (e.g.

poiyethyleneamino, Jeffamine™); and diacid ester and amides including succinate, suceinamide, diglycolate, malonaie, and caproamide. In various embodiments, linkers can comprise one or more amino acid residues, such as valine, phenylalanine, lysine, and homoiysine.

The term "chirai" refers to molecules which have the property of non-superimposability of the mirror image partner, while the term "achiral" refers to molecules which are superimposable on their mirror image partner,

The term "stereoisomers" refers to compounds which have identical chemical constitution, hut differ with regard to the arrangement of d e atoms or groups in space.

"Diastereomer" refers to a stereoisomer with two or more centers of chira ty and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.

"Bnantiomers" refer to two stereoisomers of a compound which are non-superimposahle mirror images of one another.

Stereochemical definitions and conventions used herein generally follow S, P, Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and

Eliei, H. and Wilen, S,, Stereochemistry of Organic Compounds (1994) John Wiley & Sons, Inc., New York. Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarised light. In describing an optically active compound, the prefixes D and L, or R and S, are used to denote the absolute configuration of the molecule about its chirai center(s). The prefixes d and 1 or (+) and (~) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning thai the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of one another. A specific stereoisomer may also be referred to as an enantiomer. and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no

stereoselection or stereospecificity in a chemical reaction or process. The terms "racemic mixture" and "racemate" refer to an equirnolar mixture of two enantiomeric species, devoid of optical activity,

"Leaving group" refers to a functional group that can be substituted by another functionai group, Certain leaving groups are well known in the art, and examples include, but are not limited to, a haiide (e.g., chloride, bromide, iodide), mefhanesulfonyi (mesyl), p-toiuenesulfonyl (tosyl), trifiuoromethylsulfonyl (triflate), and trifluoromethylsulfonate.

The term "protecting group" refers to a substituent that is commonly employed to block or protect a particular functionality while reacting other functionai groups on the compound. For example, an "amino-protecting group" is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include, but are not limited to, acetyl, trifluoroacetyi, t-butoxycarbonyl (BOC), henzyioxycarbonyl (CBZ) and 9- tluorenyimethylenoxycarbonyl (Fmoc). For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991 , or a later edition.

As is understood by one skilled in the art, reference to "about" a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to "about X" includes description of "X",

It is understood that aspect and embodiments described herein include "consisting" and/or "consisting essentially of aspects and embodiments. As used herein, the singular form "a", "an", and "the" includes plural references unless indicated otherwise.

II. COMPOSITIONS AND METHODS

Provided herein are antibodies that bind to FcRH5 including bispeeifk antibodies and imrnunoconjugates comprising such antibodies. Antibodies and immunoconjugates may be useful, e.g., for the diagnosis or treatment of FeRH5-positive cancers, hi some embodiments, the anti-FcRH5 antibody binds an isoform c-specific region of the extracellular domain of FcRHSc. In some embodiments, the anti~FeRH5 antibodies bind ig-!ike domain 9 of FcRHSc.

Without being bound by theory, the selectio of the precise antigen for the antibodies of the present invention was driven by at least three important considerations. First, there was a need for little to no cross-reactivity with FcRI-15 isoforms other than FcRHSc, such as isoform a and isoform b, to avoid the resulting therapeutic from binding to non-target molecules and thus reducing its effectiveness. As illustrated in Figure 1, domain 9 of FcRH5 is an example of a unique sequence among the three isoforms, Next, there was a need for little to no cross-reactivity with FcRH family members other than FcRH5, such as FcRHl , Fc H2, FcRH3, and FcRH4. This is difficult because of the generally highly conserved nature of the last Ig-like domains in many of the FcRH family members. But because of the parallel need for FcRH5 isoform e specificity, an antibody that binds the last Ig-like domain was pursued. Finally, for antibodies to be used in therapeutic molecules that work to bring large structures in close proximity, such as T-celis and tumor cells using a bispeeific antibody format, it is known that tumor epitopes closer to the cell membrane are more effective (see. e.g., Bluemei ei l. Cancer Immunol Immune then (2010) 59: 1 197 1209). Sometimes described as the theory of kinetic segregation, the cell membrane proximal location of domain 9 of FcRHS is a desirable antigen target in this context. To meet these considerations and as described in detail below, certain embodiments of the antibodies of the present invention were developed,

Provided herein are isolated anti-FcRH5 antibodies that binds an isoform c-specific region of the extracellular domain of FcRHSc. In some embodiments, the isoform c-specific region comprises Ig- like domain 9, In some embodiments, the Ig-like domain 9 is also called Ig-like C24ype 8. In some embodiments, the isoform c-specific region comprises amino acids 754-835 of SEQ ID NO: I . In some embodiments, the isoform c-specific region comprises amino acids 752-834 of SEQ ID NO:l . In some embodiments, the isoform c-specific region comprises amino acids 743-850 of SEQ ID NO: I . in some embodiments, the isoform c-specific region comprises amino acids 745-851 of SEQ ID NO: 1. In some embodiments, the isoibrm c-specific region comprises amino acids about any of 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 34, or 15 from the N-terminal and/or Oterminai boundary. In some embodiments, the isoform c-specific region comprises amino acids from about any of 750, 751 , 752, 753, or 754 to about any of 830, 831, 832, 833, 834, 835, or 836 of SEQ ID NO:l . In some embodiments, the antibodies binds FeRHSe and/or the isoform e-speeific region with an affinity of < 5 n!VS, or < 4 nM, or < 3 nM, or < 2 nM, or < 1 nK arid optionally > 0.0003 nM, or > 0.001 nM, or > 0.01 nM.

In some embodiments of any of tire antibodies, the antibody has one or more of the following characteristics: a) cross reactive with foil length human and eyno FcRHS (i.e., binds foil length human FcRI-I5 and binds foil length eyno FcRHS), b) does not significantly cross react with FcRHL FcRI12, FcRH3, and/or FcRH4 (i.e., does not significantly bind FcRHI, FcRU2, FcRH3, and/or FcRH4), c) binds to endogenous FcRHS, d) does not cross react with FcRHSa (i.e., does not significantly bind FcRHSa), and e) does not cross react with another Ig-like domain of FcRHS (i.e., does not significantly bind another Ig-3ike domam of FcRHS), Methods of determining the ability to bind are known in the art and described below.

Provided herein, and in some embodiments, are antibodies comprising a) a heavy chain comprising a HVR-HI comprising the amino acid sequence of SEQ ID NO:38, HVR-H2 comprising the amino acid sequence of SEQ ID NO:62, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:86; and/or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:2, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 14, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:26, In some embodiments, the heavy chain comprising a HVR- HI comprising the amino acid sequence of SEQ ID NO:50, HVR-H2 comprising the ammo acid sequence of SEQ ID NO:74, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:98. In some embodiments, the antibody comprises a VH sequence having at least about any of 90%, 1%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 3 i 1 and/or a VL sequence having at 3east about any of 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 1 30. In some embodiments, the antibody comprises a VH sequence of SEQ ID NO: 111 and/or a VL sequence of SEQ ID NO: 1 10. In some embodiments of any of the antibodies, the antibody comprises six HVR.5 of IC8.1. In some embodiments, the antibody comprises VH domain and VL domain of 1C8.1. In some embodiments, the antibody binds an isoform c-specific region of the extracellular domain of FcRH5c (e.g., Ig-like domam 9). In some embodiments, the antibody is cross reactive with full length human and eyno FcRHS. In some embodiments, the antibody does not significantly cross react with FcRHI , FcRH2, FcRH3, and/or FcRH4, in some embodiments, the antibody binds to endogenous FcRHS. In some embodiments, the antibody binds B-celis. In some embodiments, the antibody does not significantly bind NK ceils and/or monocytes.

Provided herein, and in some embodiments, are antibodies comprising a) a heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:39, HVR-H2 comprising the ammo acid sequence of SEQ ID NO:63, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:87: and/or b) a light chain comprising a HVR-LI comprising the ammo acid sequence of SEQ ID NO:3, HV -.L2 comprising the ammo acid sequence of SEQ ID NO: 15, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:27. In some embodiments, the heavy chain comprising a HVR- HI comprising the amino acid sequence of SEQ ID NO:51, HVR-H2 comprising the amino acid sequence of SEQ ID O:75 s and HVR-H3 comprising the amino acid sequence of SEQ ID NO:99. In some embodiments, the antibody comprises a VH sequence having at least about any of 90%, 1%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NG: i 13 and or a VI. sequence having at least about any of 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 1 12. In some embodiments, the antibody comprises a VH sequence having at least about any of 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 135 and/or a VL sequence having at least about any of 90%, 1 , 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 134. In some embodiments, the antibody comprises a VH sequence of SEQ ID NO: 113 and/or a VL sequence of SEQ ID NO: 112. In some embodiments, the antibody comprises a VH sequence of SEQ ID NO: 135 and/or a VL sequence of SEQ ID NO: 134, In some embodiments of any of the antibodies, the antibody comprises six HVRs of 1G7.2. In some embodiments, the antibody comprises VH domain and VL domain of 1G7.2, in some embodiments of any of the antibodies, the antibody comprises six HVRs of 1G7.2', In some embodiments, the antibody comprises VH domain and VL domain of 107.2'. In some embodiments, the antibody hinds an isoform c~specifie region of the extracellular domain of FcRH5c (e.g., Ig~like domain 9). In some embodiments, the antibody is cross reactive with full length human and cyrso FcRH5. in some embodiments, the antibody does not significantly cross react with FcRHl , FcRH2, FcRID, and/or FcRJ-14. In some embodiments, the antibody binds to endogenous FcRH.5, In some embodiments, the antibody binds B-celis. In some embodiments, the antibody does not significantly bind N cells and/or monocytes. In some embodiments, the antibody does not significantly cross react with FcRH5a.

Provided herein, and in some embodiments, are antibodies comprising a) a heavy chain comprising a ITVR-Hl comprising the amino acid sequence of SEQ ID NO:40, HVR-H2 comprising the amino acid sequence of SEQ ID NO:64, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:88; and/or b) a light chain comprising a HVR-LI comprising the amino acid sequence of SEQ ID NO:4, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16, and HVR-L3 comprising the amino acid sequence of SEQ D NO:28. In some embodiments, the heavy chain comprising a HVR- Hl comprising the amino acid sequence of SEQ ID NO:52, HVR-H2 comprising the amino acid sequence of SEQ ID NO:76, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:100. In some embodiments, the antibody comprises a VH sequence having at least about any of 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%>, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: l 15 and/or a VL sequence having at least about any of 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 114. In some embodiments, the antibody comprises a VH sequence of SEQ ID NO:l 15 and/or a VL sequence of SEQ ID NO:l 14. In some embodiments of any of the antibodies, the antibody comprises six HVRs of 2H7.3 , In some embodiments, the antibody comprises VH domain and VL domain of 2H7.3. In some embodiments, the antibody binds an isoform c-speeific region of the extracellular domain of FeRHSc (e.g., Ig-like domain 9). In some embodiments, the antibody is cross reactive with Ml length human and cyno FcRH5. In some embodiments, the antibody does not significantly cross react with FcRHl , FcRI-13, and/or FeRH4. In some embodiments, the antibody binds to endogenous Fc.RH.5, In some embodiments, the antibody binds B-celis. In some embodiments, the antibody does not significantly bind NK ceils and/or monocytes.

Provided herein, and in some embodiments, are antibodies comprising a) a heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID O:41 , HVR-H2 comprising the amino acid sequence of SEQ ID NO:65, and HVR-H3 comprismg the amino acid sequence of SEQ ID NO:89: and ' or b) a light chain comprismg a HVR-L1 comprising the amino acid sequence of SEQ ID NO:5, HYR-L2 comprising the amino acid sequence of SEQ ID NO:17, and HVR-L3 comprismg the amino acid sequence of SEQ ID NO:29. In some embodiments, the heavy chain comprising a HVR- Hi comprising the amino acid sequence of SEQ ID NO:53, HVR-H2 comprising the ammo acid sequence of SEQ ID NO:77, and HVR-H3 comprising the ammo acid sequence of SEQ ID NO: 101. Iri some embodiments, the antibody comprises a VII sequence having at least about any of 90%, 91%,

92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 117 and/or a VL sequence having at least about any of 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:l 16. In some embodiments, the antibody comprises a VH sequence of SEQ ID NO: 1 17 and/or a VL sequence of SEQ ID NO: 1 16. In some embodiments of any of the antibodies, the antibody comprises six HVRs of 3A4.2. In some embodiments, the antibody comprises VH domain, and VL domain of 3A4.2. In some embodiments, the antibody binds an isoform c-specific region of the extracellular domain of FcRH5c (e.g., Ig-h ' ke domain 9). In some embodiments, the antibody is cross reactive with full length human and cyno FcRII5. In some embodiments, the antibody does not significantly cross react with FcRHl , FcRH2, EcRI-I3, and/or FcRH4. In some embodiments, the antibody binds to endogenous FcRHS, In some embodiments, the antibody binds B-celis. In some embodiments, the antibody does not significantly bind NK cells and/or monocytes.

Provided herein, and in some embodiments, are antibodies comprising a) a heavy chain comprising a HVR-Hl comprismg the amino acid sequence of SEQ ID Q:42, HVR-H2 comprising the amino acid sequence of SEQ ID ΝΟ:όβ, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:90; axid/or b) a light chain comprising a HVR-Ll comprising the amino acid sequence of SEQ ID NO:6, KVR-L2 comprising the amino acid sequence of SEQ ID NO: 18, and KVR-L3 comprising the amino acid sequence of SEQ ID NO:30. In some embodiments, the heavy chain comprising a HVR- HI comprising the amino acid sequence of SEQ ID NO:54, HVR-H2 comprising the amino acid sequence of SEQ ID NO:78, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 102. In some embodiments, the antibody comprises a VH sequence having at least about any of 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:l 19 and/or a VL- sequence having at least about any of 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 1 0% sequence identity to the amino acid sequence of SEQ ID NO: 118. In some embodiments, the antibody comprises a VH sequence of SEQ ID NO:l 19 and/or a VL sequence of SEQ ID NO: 118. In some embodiments of any of the antibodies, the antibody comprises six HVRs of 3B12.1.1. In some embodiments, the antibody comprises VH domain and VL domain of 3B 1.2.1.1. In some embodiments, the antibody binds an iscform c~specific region of the extracellular domain of FcRH5c (e.g., Ig-like domain 9). In some embodiments, the antibody is cross reactive with full length human and cyno FcRHS. In some embodiments, the antibody does not significantly cross react with FcRHl, FcRH2, FcRH3, and/or FcRH4. In some embodiments, the antibody binds to endogenous FcRHS. In some embodiments, the antibody binds B celis. In some embodiments, the antibody does not significantly bind NK cells and/or monocytes. In some embodiments, the antibody does not significantly cross react with FeRHSa.

Provided herein, and in some emboidments, are antibodies comprising a) a heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:43, HVR-H2 comprising the amino acid sequence of SEQ ID NO:67. and HVR-H3 comprising the amino acid sequence of SEQ ID NO:91 ; and/or b) a light chain comprising a IIVR-Ll comprising the amino acid sequence of SEQ ID NO:7, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 1 , and HVR-L3 comprising the amino acid sequence of SEQ ID NO:31. In some embodiments, the heavy chain comprising a HVR- HI comprising the amino acid sequence of SEQ ID NO:55. HVR-H2 comprising the amino acid sequence of SEQ ID NO:79, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:103. In some embodiments, the antibody comprises a VH sequence having at least about any of 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:121 and/or a VL sequence having at least about any of 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:120. In some embodiments, the antibody comprises a VH sequence of SEQ ID NO:12i and/or a VL sequence of SEQ ID NO: 120. in some embodiments of any of the antibodies, the antibody comprises six HVRs of 3C10. In some embodiments, the antibody comprises VH domain and VL domain of 3C10. In some embodiments, the antibody binds an isoform c-specifie region of the extracellular domain of FcRHSc (e.g., Ig-like domain 9). In some embodiments, the antibody is cross reactive with full length human and cyno FcRHS. in some embodiments, the antibody does not significantly cross react with FcRHl , FcR.H2, FcRH3, and/or FeRH4, In some embodiments, the antibody binds to endogenous FcRHS. In some embodiments, die antibody binds B-cells. in some embodiments, the antibody does not significantly bind NK cells and/or monocytes.

Provided herein, and in some emboidments, are antibodies comprising a) a heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:44, HVR-H2 comprising the amino acid sequence of SEQ ID NO:68, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:92; and/or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:S, HVR-L2 comprising the amino acid sequence of SEQ ID NO:20, and HVR-.L3 comprising the amino acid sequence of SEQ ID NO:32. In some embodiments, the heavy chain comprising a HVR~ HI comprising the amino acid sequence of SEQ ID NO:56, KVR-K2 comprising the amino acid sequence of SEQ ID NO:80, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:104. In some embodiments, the antibody comprises a VH sequence having at least about any of 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 123 and/or a VL sequence having at least about any of 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:122. in some embodiments, the antibody comprises a VH sequence of SEQ ID NO:123 and/or a VL sequence of SEQ ID NO: 122. In some embodiments of any of the antibodies, the antibody comprises six HVRs of 3F10. hi some embodiments, the antibody comprises VH domain and VL domain of 3F10. hi some embodiments, the antibody binds an isoform c-specific region of the extracellular domain of FcRHSc (e.g., Ig-like domain 9). In some embodiments, the antibody is cross reactive with full length human and cyno FcR.H5. In some embodiments, the antibody does not significantly cross react with FcRHl , FcRH2, FcRH3, and/or FcRH4. In some embodiments, the antibody binds to endogenous FcRH5, In some embodiments, the antibody binds B-cells. In some embodiments, the antibody does not significantly bind NK cells and/or monocytes. In some embodiments, the antibody does not significantly cross react with FcRHSa.

Provided herein, and in some emboidments, are antibodies comprising a) a heavy chain comprising a HVR-H1 comprising the amino acid sequence of SEQ ID NO:45, HVR-H2 comprising the amino acid sequence of SEQ ID NO:69, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:93; and/or b) a light chain comprising a HVR-Ll comprising the amino acid sequence of SEQ ID NO:9, HVR-L2 comprising the amino acid sequence of SEQ ID NO:21, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:33. In some embodiments, the heavy chain comprising a HVR- HI comprising the amino acid sequence of SEQ ID NO:57, HVR-H2 comprising the amino acid sequence of SEQ ID NO:8L and H R -H3 comprising the amino acid sequence of SEQ ID NO:105. In some embodiments, the antibody comprises a VH sequence having at least aboin any of 90%. 91 , 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:125 and/or a VL sequence having at least about any of 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%. 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:124. in some embodiments, the antibody comprises a VK sequence of SEQ ID NO: 125 and/or a VL sequence of SEQ ID NO: 124. In some embodiments of any of the antibodies, the antibody comprises six HVRs of 3G3. In some embodiments, the antibody comprises VH domain and VL domain of 3G3. In some embodiments, the antibody binds an isoform e-specific region of the extracellular domain of FcRHSc (e.g., Ig-Iike domain 9), In some embodiments, the antibody is cross reactive with full length human and cyno FcRH5. In some embodiments, the antibody does not significantly cross react with FcRHl , FcRH2, FcRID, and/or FcRH4. in some embodiments, the antibody binds to endogenous FcRH5, in some embodiments, the antibody binds B-cells, In some embodiments, the antibody does not significantly bind NK cells and/or monocytes. In some embodiments, die antibody does not significantly cross react wife FeRHSa.

Provided herein, and in some emboidments, are antibodies comprising a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:46, HVR-H2 comprising the amino acid sequence of SEQ iD NO:70, and HVR-H3 comprising the amino acid sequence of SEQ ID NO:94; and/or b) a light chain comprising a HVR-L1 comprising fee amino acid sequence of SEQ iD NO: 10, HVR-L2 comprising the amino acid sequence of SEQ ID NO:22, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:34. In some embodiments, the heavy chain comprising a HVR- Hl comprising the amino acid sequence of SEQ ID NO;58, HVR-II2 comprising die amino acid sequence of SEQ ID NO:82, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 106. In some embodiments, the antibody comprises a VH sequence having at least about any of 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:127 and/or a VL sequence having at least about any of 90%, 93 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 126. In some embodiments, fee antibody comprises a VH sequence of SEQ IT) NO: 127 and/or a VL sequence of SEQ ID NO: 126. In some embodiments of any of the antibodies, the antibody comprises six HVRs of 3G7.1.5. In some embodiments, the antibody comprises VH domain and VL domain of 3G7.1.5. In some embodiments, the antibody binds an isoform c-specitlc region of the extracellular domain of

FcRHSc (e.g., Ig~iike domain 9). In some embodiments, the antibody is cross reactive with full length hnman and cyno FcRHS. In some embodiments, the antibody does not significantly cross react with FcRHl , FcRH ' 2, FcRH3, and/or FeRH4. In. some embodiments, the antibody binds to endogenous FcRHS. In some embodiments, the antibody binds B-cells. In some embodiments, the antibody does not significantly bind NK ceils and/or monocytes.

Provided herein, and in some emboidments, are antibodies comprising a) a heavy chain comprising a HVR-Hl comprising the amino acid sequence of SEQ ID NO:47, HVR-H2 comprising the amino acid sequence of SEQ ID NO:71, and HVR-II3 comprising fee amino acid sequence of SEQ ID NO:95; anfe'or b) a light chain comprising a HVR-Li comprising the amino acid sequence of SEQ ID NO:l 1 , HVR-L2 comprising the amino acid sequence of SEQ ID NO:23, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:35. In some embodiments, the heavy chain comprising a HVR- Hl comprising the amino acid sequence of SEQ ID NO:59, HVR-H2 comprising the amino acid sequence of SEQ ID NO;83, and HVR-H3 comprising the ammo acid sequence of SEQ ID NO: 107. In some embodiments, the antibody comprises a VH sequence having at least about any of 90%, 1 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 129 and/or a VL sequence having at least about any of 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:128. In some embodiments, the antibody comprises a VH sequence of SEQ ID NO: 129 and/or a VL sequence of SEQ ID NO: 128, In some embodiments of any of the antibodies, the antibody comprises six HVRs of 5Ά10.1.3. In some embodiments, die antibody comprises Vii domain and VL domain of 5AI0.1.3, In some embodiments, the antibody binds an isoform c-specifie region of the extracellular domain of FeRHSc (e.g,, Ig~like domain 9). In some embodiments, the antibody is cross reactive with full length human and cytio FeRHS, In some embodiments, the antibody does not significantly cross react with FcRHl, FeRH2 f FeRIB, and/or FcRl . In some embodiments, the antibody binds to endogenous FeRHS. In some embodiments, the antibody binds B-eells. In some embodiments, the antibody does not significantly bind NK ceils and/or monocytes.

Provided herein, and in some emboidments, are antibodies comprising a) a heavy chain comprising a HVR-IT1 comprising the amino acid sequence of SEQ ID NO:48, HVR-H2 comprising the amino acid sequence of SEQ ID NO:?2, and HV.R.-H3 comprising the amino acid sequence of SEQ ID NO:96; and/or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ ID NO:l 2, HVR-L2 comprising the amino acid sequence of SEQ ID NO:24, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:36, In some embodiments, the heavy chain comprising a HVR- Hl comprising the amino acid sequence of SEQ ID NO:60, HVR-H2 comprising the amino acid sequence of SEQ ID NO:84, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 108, In some embodiments, the antibody comprises a VH sequence having at least about any of 90%, 1%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ FD NO: 131 and/or a VL sequence having at least about any of 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:! 30. In some embodiments, the antibody comprises a VH sequence of SEQ ID NO:131 and/or a VL sequence of SEQ ID NO: I30. In some embodiments of any of the antibodies, the antibody comprises six HVRs of 5FL1.5. In some embodiments, die antibody comprises VH domain and VL domain of 5F1.1.5. In some embodiments, the antibody binds an isofom c-specific region of the extracellular domain of

Fc.RHSc (e.g., Ig~iike domain 9), In some embodiments, the antibody is cross reactive with full length human and cyno FeRHS. In some embodiments, the antibody does not significantly cross react with FcRHl , FcRI-12, FcRIB, and/or FcRH4. In some embodiments, the antibody binds to endogenous FcRH5, In some embodiments, the antibody binds B~cells, In some embodiments, the antibody does not significantly bind NK cells and/or monocytes, In some embodiments, the antibody does not significantly cross react with FcRFISa, Provided herein, and in some emhoidments, are antibodies comprising a) a heavy chain comprising a HVR.-H1 comprising the amino acid seqisence of SEQ ID NO:49, HVR-H2 comprising the amino acid sequence of SEQ ID NO:73, and HV -H3 comprising the amino acid sequence of SEQ ID NO:97; and/or b) a light chain comprising a HVR-L1 comprising the amino acid sequence of SEQ iD NO:13, HVR-L2 comprising the amino acid sequence of SEQ ID NO:25, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:37. In some embodiments, the heavy chain comprising a HVR- Hl comprising the amino acid sequence of SEQ ID NO:61 , HVR-H2 comprising the amino acid sequence of SEQ ID NO:85, and HVR-H3 comprising the amino acid sequence of SEQ ID NO: 109. In some embodiments, the antibody comprises a VII sequence having at least about any of 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 133 and/or a VL sequence having at least about any of 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 132. In some embodiments, the antibody comprises a VH sequence of SEQ ID NO: 133 and/or a VL sequence of SEQ ID NO: 132. In some embodiments of any of the antibodies, the antibody comprises six HVRs of 6D2. In some embodiments, the antibody comprises VH domain and VL domain of 6D2. In some embodiments, the antibody binds an isoform c-specific region of the extracellular domain of FcRHSc (e.g., Ig-like domain 9). In some embodiments, the antibody is cross reactive with full length human and eyno FcRH5. In some embodiments, the antibody does not significantly cross react with FcRHl , FcRH2, FcRH3, and/or FcRH4. In some embodiments, the antibody hinds to endogenous FcRHS. In some embodiments, the antibody binds B-cells. In some embodiments, the antibody does not significantly bind NK ceils and/or monocytes.

Ei a further aspect provided herein, an anti~FcRH5 antibody according to any of the above embodiments is a monoclonal antibody, including a chimeric, humanized or human antibody. In one embodiment, an anti-FcRH5 antibody is an antibody fragment, e.g., a Fv. Fab, Fab', scFv, diabody, or F(ab') 2 fragment, in another embodiment, the antibody is a substantially full length antibody, e.g., an IgGl antibody or other antibody class or isotype as defined herein.

In a further aspect, the invention provides an antibody that binds to the same epitope as an anti- FcRHS antibody provided herein. In certai embodiments, an antibody is provided that binds an isoform c-specific region of the extracellular domain of FeRHSc from, within, or overlapping amino acids 754-835 of SEQ ID NO:l .

In some embodiments of any of the anti-FcRH5 antibodies, the FcRH5 antibody, particularly an FcRH5 bispecific (e.g., anti-CD3 anti-FcRH5 bispecific), may have features, singly or in combination, based upon HEK ceil line assays (HEK cells reconstituted with necessary signaling components for the TCR triggering as described in James and Valle, Nature 487:64-69 (2012), which is incorporated by reference in its entirety. In some embodiments, the features, singly or in combination, may include tumor cell interphase/immunological synapse, Lck-mediated TCR phosphorylation, ZAP70 activity including phosphorylation state and localization, CDS 8 activity including localization and binding, 2 Ar activity including localization and binding, CAAX activity including localization and binding CD45 activity including localization, pMHC activity including localization, and/or TCR activity and triggering features.

In a further aspect, an anti~FeRH5 antibody according to any of the above embodiments may incorporate any of the features, singly or i combination, as described in (a)-(e) and/or Sections 1 -7 below.

( ) hinds an isoform c- pecific region of the extracellular domain of FcRHSc

Methods of determining whether an anti-FcRHS antibody binds to an isoform e- specific region of the extracellular domain of FcRHSc are known in the art. in some embodiments, binding of an anti- FeRHS antibody to an isoform c-specific region of the extracellular domain of FeRHS c may be determined by expressing FcRH.5 polypeptides with N- and C-terminal deletions in 293 ceils and/or SVT2 ceils and testing by FACS as described in the Examples binding of the antibody to the truncated polypeptides. In some embodiments, a substantial reduction (> 70% reduction) or elimination of binding of the antibody to a truncated polypeptide relative to binding to mil-length FeRHS expressed i 293 cells indicates that the antibody does not bind to that truncated polypeptide.

In some embodiments, the isoform c-specific region comprises Ig-iike domain 9. hi some embodiments, the Ig-like domain 9 is also called Ig-like C2-type 8. In some embodiments, the isoform c-specific region comprises amino acids 754-835 of SEQ ID NO: I , Irs some embodiments, the isoform c-specific region comprises amino acids 752-834 of SEQ ID NO:l . in some embodiments, the isoform c-speeific region comprises amino acids 743-850 of SEQ ID NO: 1. In some embodiments, the isoform c-specific region comprises amino acids 745-851 of SEQ ID NO:l . In some embodiments, the isoform c-specific region comprises amino acids about any of 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, or 15 from the N-terminal and/or C-terminal boundary. I some embodiments, the isoform c-specific region comprises amino acids from about any of 750. 751, 752, 753, or 754 to about any of 830, 831 , 832, 833. 834, 835, or 836 of SEQ ID NO: l . In some embodiments, FcRIiS is human FcRI-15. in some embodiments, FeRHS is human FeRHS or eynornolgus monkey FeRHS.

(b) cross reacts with (binds) human and cyno FeRHS with an affinity of< 5 M, or <■/ nM, or < 3 nM, or < 2 nM or < 1 n and optionally > Q. OQQl nM, or > 0.001 nM, or > 0,01 nM Methods of determining binding affinity are known in the art. In some embodiments, the binding affinity may be determined according to a BiAcore wi assay, ELISA, Facs, and IHC, for example, as described in the Examples.

in some embodiments, the anti-FcRHS antibody binds human and/or cyno FeRHS with an affinity of about any of < 5 nM, or < 4 nM, or < 3 nM, or < 2 nM, or < I nM. In some embodiments, the anti-FcRHS antibody binds human and/or cyno FeRHS with an affinity of about < 5. in some embodiments, the anti-FcRHS antibody binds human and/or cyno FeRHS with an affmity of about < 4 nM. In some embodiments, the anti-FeRH5 antibody binds human and/or cyno FeRHS with an affmity of about < 3 nM, In some embodiments, the anti-FcRHS antibody binds human and/or cyno FeRHS with an affinity of about <2 nM in some embodiments, FcRHS is human FcRHS. In some embodiments, FcRHS is cynomolgus monkey FcRHS.

(c) does not cross react with (does not bind) FcRH FcRH2, FcRHS, and/or FcRH4

Methods of determining binding are known in the art. in some embodiments, the binding affinity may be determined according to a BIAcore* assay, Facs, ELISA, and IHC, for example, as described in the Examples.

In some embodiments, the anti~FeRH5 antibody binds FcRHS with an affinity of more than about any of 2, 5, 10, 20, 50, 100, 500, or 1000-fold greater than FcRH! , FcRH2, FcRH3, and/or FcRH4. In some embodiments, FcRH is human FcRH.

(d) does not cross read with (does not bind) FcRHSa

Methods of determining binding are known in the art. In some embodiments, the binding affinity may be determined according to a BIAeore* assay, Facs, FX1SA, and IHC, for example, as described in the Examples.

In some embodiments, the anti-FcRlI5 antibody binds FcRHSe with an affinity of more than about any of 2, 5, 10, 20, 50, 100, 500, or 1000-fold greater than FcRHSa. in some embodiments, FcRH is human FcRH.

(e) does not cross react with another Ig-iike domain (does not bind) of FcRHS

Methods of determining binding are known in the art. In some embodiments, the binding affinity may be determined according to a BIAeore assay, Facs, ELISA, and FHC, for example, as described in the Examples.

In some embodiments, the anti-FcRH5 antibody binds Ig-like domain 9 of FcRHS with an affinity of more than about any of 2, 5, 10, 20, 50, 100, 500, or 1000-fold greater than Ig-like domain 1 , 2, 3, 4, 5, 6, 7. and/or 8 of FcRHS. in some embodiments, FcRH is human FcRH, In some

embodiments, the Ig-like domain is Ig-like domain 1 (aa 23-100 of SEQ ID NO;l), Ig-like domain 2 (aa 105-1 85 of SEQ ID NG:1 ), Ig-hke domain 3 (aa 188-271 of SEQ ID NO:l ), Ig-like domain 4 (287-373 of SEQ ID NO;l ), Ig-like domain 5 (aa 380-466 of SEQ iD NO:l), Ig-like domain 6 (aa 490-555 of SEQ ID NO: 3), Ig-like domain 7 (aa 568-652 of SEQ ID NO: I), Ig-like domain 8 (aa 658-731 of SEQ ID NO: l ),

Binding Assays and Other Assays

In one aspect, an anti-FcRH5 antibody is tested for its antigen binding activity. For example, in certain embodiments, an anti-FcRHS antibody is tested for its ability to bind to FcRHS expressed on the surface of a cell. A FACS assay may be used for such testing.

In an exemplary competition assay, immobilized FcRHS is incubated in a solution comprising a first labeled antibody that binds to FcRH5 and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to FcRHS. The second antibody may be present in a hybridoma supernatant. As a control, immobilized FcRHS is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After incubation under conditions permissive for binding of the first antibody to FcRHS, excess unbound antibody is removed, and the amount of label associated with immobilized FcRHS is measured. If the amount of label associated with immobilized FcRHS is substantially reduced in the test sample relative to the control sample, then that indicates that the second antibody is competing with the first antibody for binding to FcRHS. in certain embodiments, immobilized FcRHS is present on the surface of a cell or in a membrane preparation obtained from a cell expressing FcRHS on its surface.

In one aspect, purified anti-FcRH5 antibodies can be further characterized by a series of assays including, but not limited to, N-termina! sequencing, amino acid analysis, non-denaturing size exclusion high pressure liquid chromatography (HFLC), mass spectrometry, ion exchange

cliromatography and papain digestion. In one embodiment, contemplated are an altered antibody that possesses some but not ail effector functions, which make it a desirable candidate for many applications in which the half life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious, Iri certain embodiments, the Fc activities of the antibody are measured to ensure that only the desired properties are maintained. In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities. For example, Fc receptor (FcR) binding assays can be conducted to ensure mat the antibody lacks FcyR binding (hence likely lacking ADCC activity}, bat retains Fc n binding ability. The primary cells for mediating ADCC. NK cells, express Fc(RIII only, whereas monocytes express Fc(RI, Fc(RII and Fc(RHI. FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu, Rev. Immunol, 9:457-92 (1991). An example of an in vitro assay to assess ADCC activity of a molecule of interest is described in U.S. Pat. No. 5,500.362 or 5,821 ,337. Useful effector cells for such assays include peripheral blood mononuclear ceils (PBMC) and Natural Killer (NK} cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed i vivo, e.g., in a animal model such as that disclosed in Ciynes el ah PNAS (USA) 95:652-656 (1998). CI q binding assays may also be carried out to confirm that the antibody is unable to bind Clq and hence lacks CDC activity. To assess complement activation, a CDC assay, e.g. as described in

Gazzano-Santoro el aL, J. Immunol. Methods 202:163 (1996), may be performed. FcRn binding and in vivo clearance half life determinations can also be performed using methods known in the art.

In certain embodiments, an antibody provided herein has a dissociation constant (Kd) of

< 1 μΜ, < 100 nM, < 10 nM, < 1 nM, < 0.1 nM, < 0.01 nM, or < 0.001 nM, and optionally is > 1 Q '! 3 M. (e.g. 10 "S M or less, e.g. from 10 "S M to 10 ' " M, e.g., from K. M to 10 "i3 M).

In some embodiments, Kd may be measured by a radiolabeled antigen binding assay (RIA) performed with the Fab version of an antibody of interest and its antigen as described by the following assay. Solution binding affinity of Fabs for antigen may be measured by equilibrating Fab with a minimal concentration of ( i; i)~iabeled antigen in d e presence of a titration series of unlabeled antigen, then capturing bound antigen with an anti-Fab antibody-coated plate (see, e.g., Chen et aL, J. MoL Biol. 293:865-881(1999)). To establish conditions for the assay, MlCROTTrER* multi-well plates (Thermo Scientific) may be coated overnight with 5 jig/ml of a capturing anti-Fab antibody (Cappel Labs) in 50 rnM sodium carbonate (pH 9,6), and subsequently blocked with 2% (w/v) bovine serum albumin in PBS for two to five hours at room temperature (approximately 23 Q C). In a non-adsorbent plate (Nunc #269620), 100 p.M or 26 pM [ ,?' ¾-antigen are mixed with serial dilutions of a Fab of interest (e.g. , consistent with assessment of the artti-VEGF antibody, Fab~12. in Presta et al, Cancer Res. 57:4593- 4599 (1997)), The Fab of interest is then incubated overnight; however, the incubation may continue for a longer period {e.g., about 65 hours) to ensure that equilibrium is reached. Thereafter, the mixtures may be transferred to the capture plate for incubation at room temperature {e.g., for one hour). The solution may be then removed and the plate washed eight times with 0.1 % polysorbate 20 (TWEEN- 20 ® ) in PBS. When the plates have dried, 150 pLAveH of scintillant (MICROSCiNT-20™; Packard) may be added, and the plates may be counted on a TOPCOUNT ! M gamma counter (Packard) for ten minutes. Concentrations of each Fab that give less than or equal to 20% of maximal binding may be chosen for use in competitive binding assays.

According to another embodiment, Kd is measured using surface piasmon resonance assays using a BIACO E^QOO or a BiACORE ' ^3000 (BIAcore, Inc., Piscataway, NJ) at 25 n C with immobilized antigen CMS chips at -10 response units (RU), Briefly, carboxymethylated dextran biosensor chips (CMS, BIACGRE. Inc.) may be activated wi N~ethyi-N'~ (3-dirnethylaminopropyl)- carbodiimide hydrochloride (EDC) and N~bydroxysuecinirmde (NHS) according to the supplier's instructions. Antigen may be diluted with 10 mM sodium acetate, pH 4.8, to 5 pg/rnl (-0,2 μΜ) before injection at a flow rate of 5 μί_Ληϋηυίε to achieve approximately 30 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine may be injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0,78 nM to 500 nM) may be injected in PBS with 0.05% polysorbate 20 (TWEEN-20™) surfactant (PBST) at 25 n C at a flow rate of approximately 25 \h!m\t\. Association rates {k n and dissociation rates (k ) may be calculated using a simple one-to-one Langmuir binding model (BIACORE" 0 Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (Kd) may be caicuiated as the ratio k See, e.g., Che et al, J, Mol. Biol. 293:865-881

6 ~s -I

(1999). If the on-rate exceeds 10 M s by the surface piasmon resonance assay above, then the on-rate may be determined by using a fluorescent quenching technique that measures the increase or decrease in fluorescence emission intensity (excitation - 295 urn; emission = 340 nm, 1.6 nni band-pass) at 25°C of a 20 nM anti-antigen antibody (Fab form) in PBS, pH 7.2, in the presence of increasing

concentrations of antigen as measured in a spectrometer, such as a stop-flow equipped specirophometer (Aviv Instruments) or a 8000-series SLM-AMTNCO 1 ?vi spectrophotometer ( ' i¾emioSpeetronic) with a stirred cuvette. 2, Antibody Fragments

in certain embodiments, an antibody provided herein is an antibody fragment. Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab% Fv, and scFv fragments, and other fragments described below. For a review of certain antibody fragments, see Hudson et al. Nat, Med. 9: 129-134 (2003), For a review of scFv fragments, see, e.g., Pluckthun, in The Pharmacology of Monoclonal Antibodies, vol. 113, osenburg and Moore eds., (Springer- Verlag, New York), pp. 269- 315 (1994); see also WO 93/161 85; and U.S. Patent Nos. 5,571 ,894 and 5,587,458. For discussion of Fab and F(ab¾ fragments comprising salvage receptor binding epitope residues and having increased i vivo half-life, see U.S. Patent No. 5,869,046.

Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EF 404,097; WO 1993/01161 ; Hudson et l., Nat. Med, 9:129-134 (2003); and Hoilinger et !., Proc. Natl Acad. Sci. USA 90: 6444-6448 (1993). Triabodies and tetrabodies are also described in Hudson et al., Nat. Med. 9:129-134 (2003).

Single-domain antibodies are antibody fragments comprising all or a portio of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody, hi certain embodiments, a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U.S. Patent No. 6,248,516 Bl).

Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as productio by recombinant host cells (e.g. E, coil or phage), as described herein.

5o Chimeric and Humanized Antih&dies

In certain embodiments, an antibody provided herein is a chimeric antibody. Certain chimeric antibodies are described, e.g., in U.S. Patent No. 4,816,567; and Morrison et al, Proc. Na . Acad, Sci, USA, 81 ;6851-6855 (1984)). In one example, a chimeric antibody comprises 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) and a human constant region. In a further example, a chimeric antibody is a "class switched" antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.

In certain embodiments, a chimeric antibody is a humanized antibody. Typically, a non-human antibody is humanized to reduce irmnunogenicity to humans, while retaining the specificity and affmity of the parental non-human antibody. Generally, a humanized antibody comprises one or more variable domains in which HV s, e.g. , CDR s, (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof} are derived from human antibody sequences. A humanized antibody optionally will also comprise at least a portion of a human constant region. In some embodiments, some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g. , the antibody from which the HVR residues are derived), e.g., to restore or improve antibody specificity or affinity. Humanized antibodies and methods of making them are reviewed, e.g. , in Almagro and Fransson, Froni. Biosci. 13: 1619-1633 (2008), and are further described, e.g., in Riechmann et al, Nature 332:323-329 (1988); Queen et a!., Proa Nat l. Acad. Sci. USA 86:10029-10033 (1989); US Patent Nos. 5, 821,337, 7,527,791 , 6,982,321 , and 7,087,409; Kaslnniri et al, Methods 36:25-34 (2005) (describing SDR (a-CDR) grafting); Padian, Mol Immunol 28:489-498 (1991) (describing

"resurfacing"); Dall'Acqua et l. Methods 36:43-60 (2005) (describing "FR shuffling"); and Osbourn et al, Methods 36:61-68 (2005) and Klimka et al, Br. J. Cancer, 83:252-260 (2000) (describing the "guided selection" approach to FR shuffling).

Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the "best-fit" method {see, e.g. , Suns et al J. Immunol 151 :2296 (1993)); framework regions derived from the consensus sequence of human antibodies of a particular subgroup of * light or heavy chain variable regions (see, e.g., Carter et al Proc. Natl Acad. Sci. USA, 89:4285 (1992); and Presta et al. J. Immunol, 151 :2623 (1 93)); human mature (somatically mutated) framework regions or human gerroiine framework regions [see, e.g., Aimagro and Fransson, Front. Biosci. 13:1619-1633 (2008)); and framework regions derived from screening FR libraries (see, e.g., Baca et al , J Biol. Ghent. 212 : 10678 » 10684 (1 97) and Rosok et al , J. Biol Chem. 2 1 :22611 -22618 (1996)).

4. Human Antibedies

In certain embodiments, an antibody provided herein is a human antibody. Human antibodies can be produced using various techniques known in fee art. Human antibodies are described generally in van Dijk and van de Winkel, Curr. Opin. Pharmacol. 5: 368-74 (2001) and Lonberg, Cisrr. Opin. Immunol 20:450-459 (2008).

Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge. Such animals typically contain all or a portion of the human immunoglobulin loci, which replace the endogenous immunoglobulin loci, or which are present extraehroniosomally or integrated, randomly into the animal's chromosomes, in such transgenic mice, the endogenous immunoglobulin loci have generally been inactivated. For review of methods for obtaining human antibodies from transgenic animals, see Lonberg, Nat. Biotech. 23:1 1 17-1 125 (2005). See also, e.g., U.S. Patent Nos. 6,075,181 and 6,150,584 describing XENOMOUSE™ technology; U.S. Patent No, 5,770,429 describing HUMAB® technology; U.S. Patent No. 7,041 ,870 describing K-M MOUSE® technology, and U.S. Patent Application Publication No. US 2007/0061900, describing VELOCiMouSE® technology). Human variable regions from intact antibodies generated by such animals may be further modified, e.g., by combining with a different human constant region.

Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeioma ceil lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor J Immunol, 133: 3001 (1984); Brodeur ei al., Monoclonal Antibody Production Techniques ami Applications, pp. 51-63 (Marcel Dekker, inc., New York, 19S7); and Boemer et al., J. Immunol., 147: 86 (1991).) Human antibodies generated via human B-cel! hybridorna technology are also described in Li et al, Proc. Natl Acad. Sci. USA, 103:3557-3562 (2006).

Additional methods include those described, for example, in U.S. Patent No. 7,189,826 (describing production of monoclonal human IgM antibodies from hybridorna cell lines) and Ni, Xiandai

Mianyixue, 26(4):265-268 (2006) (describing human-human hybridomas), Human hybridorna technology (Trioma technology) is also described in Vollmers and Brandiein, Histology and

Histopathology, 20(3):927-937 (2005) and Voiimera and Brandiein, Methods and Findings in

Experimental and Clinical Pharmacology, 27(3):185~91 (2005).

Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below,

5. Library-Derived Antibodies

Antibodies provided herein may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. For example, a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, e.g., hi Hoogenboom et al in Methods in

Molecular Biology 178:1 -37 (O'Brien et al, ed., Human Press, Totowa, NI. 2001) and further described, e.g. , in the McCafferty et al. Nature 348:552-554; Clackson et al, Nature 352: 624-628 (1991): Marks et al, J. Mol Biol. 222: 581-597 (1992); Marks and Bradbury, in Methods in Molecular Biology 248: 161-175 (Lo, ed., Human Press, Totowa, NJ, 2003); Sidhu et al,, J, Mol. Biol. 338(2): 299- 310 (2004); Lee et al, J. Mol. Biol 340(5): 1073-1093 (2004); FeJlouse, Proc. Natl. Acad. Sci. USA 101(34): 12467-12472 (2004); and Lee et al, J. Immunol. Methods 284(1 -2): 119-132(2004).

in certain phage dispiay methods, repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PGR) and recombined randomly in phage libraries, which can then be screened for antigen-binding phage as described h Whiter et αΙ. , Απη, Rev. Immunol, 12: 433-455 (1994), Phage iypicaliy display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments, Libraries from immunized sources provide high-affinity antibodies to the i niunogen without the requirement of constructing hybridomas. Alternatively, the naive repertoire can be cloned (e.g., from human) to provide a single source of antibodies to a wide range of non-self and also self antigens without any immunization as described by Griffiths et al. EMBO J. 12: 725-734 (1993). Finally, naive libraries can also be made synthetically by cloning unrearranged V~gene segments from stem cells, and using PGR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom and Winter, J. Mol. Biol, 227: 3S1-3S8 (1992). Patent publications describing human antibody phage libraries include, for example: US Patent No, 5,750,373, and US Patent Publication Nos. 2005/0079574, 2005/0 Π 455, 2005/0266000, 2007/01 17126, 2007/0160598, 2007/0237764, 2007/0292936, and 2009/0002360,

Antibodies or antibody fragments isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.

6. Muitispeeific Aittih&dies

In certain embodiments, an antibody provided herein is a muitispeeific antibody, e.g. a bispecific antibody, Muitispeeific antibodies are monoclonal antibodies that have binding specificities for at least two different sites. In certain embodiments, one of the binding specificities is for FcRHS and the other is for any other antigen. In certain embodiments, one of the binding specificities is for FcRH5 and the other is for CD3. See, e.g., U.S. Patent No. 5,821,337. In certain embodiments, bispecific antibodies may bind to two different epitopes of FcRHS. Bispecific antibodies may also be used to localize cytotoxic agents to ceils which express FcRHS. Bispecific antibodies can be prepared as full length antibodies or antibody fragments.

In some embodiments, the FcRHS antibodies are FcRHS bispecific antibodies. Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Exemplars' bispecific antibodies may bind to two different epitopes of an FcRHS protein as described herein. Other such antibodies may combine an FcRH5 binding site with a binding site for another protein.

Alternatively, an anti-FeRHS ami may be combined with an ann which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD3), or Fc receptors for IgG (FcyR), such as FeyRl (CD64), FcyRII (CD32) and FcyRIH (CD16), so as to focus and localize cellular defense mechanisms to the FcRHS -expressing cell, Bispecific antibodies may also be used to localize cytotoxic agents to cells which express FcRHS. These antibodies possess an FeRH5-binding ann and an arm which binds die cytotoxic agent {e.g., saporin, anti-interferon a vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten). Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g., Ffab') 2 bispecific antibodies). In some embodiments, die anti- FcRH.5 antibody binds an isoform c-specific region of the extracellular domain of FcRHSc. In some embodiments, the anti-FeRHS antibodies binds Ig-like domain 9 of FcRIISc,

in some embodiments, the FcRHS bispecific antibody comprises a first arm, wherein the first arm binds FcRHS and a second arm, wherei the second ann binds a Fc, The second arm of the FcRHS bispecific antibody may be any anti-Fc antibody known in the art. For example, WO 96/16673 describes a bispecific anti-ErbB2/anti-FeyRin antibody and U.S. Pat. No. 5,837,234 discloses a bispecific anti~ErbB2/anti~Fe¾Rl antibody. A bispecific ant ErbB2/Fc antibody is shown in

WO98/02463. In some embodiments, the anti-FeRHS antibody binds an isoform e-speeific region of the extracellular domain of FcRHSc, In some embodiments, the anti-FeRHS antibodies binds Ig-like domain 9 of FcRHSc.

In some embodiments, the FcRHS bispecific antibody comprises a first arm, wherein the first arm binds FcRH5 and a second arm, wherein the second arm binds CD3. The second ann of the FcRH5 bispecific antibody may be any anti-CD3 antibody known in the art. U.S, Pai, Nos, 5,821,337 and 6,407,21 .3 teach bispecific an†i~ErbB2/anii-CD3 antibodies. Additional bispecific antibodies that bind an epitope on the CDS antigen and a second epitope have heen described. See, for example, U.S. Fat. No. 5,078,998 (anti-CD3/tumor cell antigen); U.S. Fat. No. 5,601 ,819 (antt-CD3/IL-2R; anti- CD3/CD28; anti-CD3/CD45); U.S. Pai, No, 6,129,914 (anti-CD3/maiignant B-cell antigen); U.S. Pat. No. 7,1 12,324 (anti-CD3/CD19); U.S. Pat. No. 6,723,538 (anti-CD3/CCR5); U.S. Pat. No. 7,235,641 (anti-CD3 EpCAM); U.S. Pat. No. 7,262,276 (anti-CD3/ovariaa tumor antigen); and U.S. Pat. No. 5,731 ,1.68 (anii-CD3/CD4igG), which are incorporated by reference in their entirety, In some embodiments, the anti-CD3 antibody of the second arm is an antibody described in any one of WO 2005/i 18635, WO2007/042261, WO2008/1 19567, US5929212, US6750325, US6491916, US79942S9, US7993641, US6706265, US55S5097, US5968509, US5932448, US6129914, US7381803,

US5S34597, andUS7862813, which are incorporated by reference in their entirety, in some embodiments, the anii-FcRH5 antibody binds an isoforrn c-speciftc region of the extracellular domain of FcRHSc. In some embodiments, the anti-.FcRH5 antibodies binds Ig-!ike domain 9 of FcRH5e.

Techniques for making rnuitispecifk antibodies include, but are not limited to, recombinant co- expression of two imn¾mogk>bulin heavy chain- light chain pairs having different specificities {see Milstein and Cuello, Nature 305: 537 (1983)), WO 93/08829, and Traunecker ef a/., EMBO J. 10: 3655 (1991 )), and "knob-in-hole" engineering {see, e.g., U.S. Patent No. 5,731 ,168). Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc- heterodimeric molecules (WO 2009/089004A1): cross-linking two or more antibodies or fragments

{see, e.g. , US Patent No. 4.676.980. and Brennan el L, Science, 229: 81 (1985)); using leucine zippers to produce bi-specific antibodies {see, e.g., Kostelny et ai, J. Immunol, 148(5): 1547-1553 (1992)); using "diabody" technology for making bispecific antibody fragments {see, e.g., Hoilinger et !., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993)); and usmg single-chain Fv (sFv) dimers (see, e.g. Gather et aL, J, Immunol., 152:5368 (1994)): and preparing trispecific antibodies as described, e.g., in Tutt et «/, J. Immunol. 147: 60 (1991).

Engineered antibodies with three or more functional antigen binding sites, including "Octopus antibodies," are also included herein (see, e.g. US 2006/0025576A1 ).

The antibody or fragment herein also includes a "Dual Acting FAb" or "DAF" comprising an antigen binding site that binds to FcRH5 as well as another, different antigen (see, US 2008/0069820, for example).

According to a different approach, antibody variable domains with the desired binding specificities (antibody-antigen combining sites) are fused to immunoglobulin constant domain sequences. Preferably, the fusion is with an Ig heavy chain constant domain, comprising at least part of the hinge, C ii2 , and C f o regions. It is preferred to have the first heavy-chain constant region ( m) containing the site necessary for light chain bonding, present in at least one of the fusions, DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host cell. This provides for greater flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments when unequal ratios of the three polypeptide chains used in the construction provide the optimum yield of the desired bispecifie antibody. It is, however, possible to insert the coding sequences for two or all three polypeptide chains into a single expression vector when the expression of at least two polypeptide chains in equal ratios results in high yields or when the ratios have no significant affect on the yield of the desired chain combination.

In some embodiments, the bispecifie antibodies are composed of a hybrid nrtmunogiobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second bmding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecifie compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecifie molecule provides for a facile way of separation. This approach is disclosed in WO 94/04690. For further details of generating bispecifie antibodies see, for example, Suresh et a!., Methods in

Enzymology 121 :210 (1986).

According to another approach described in U.S. Pat. No. 5,731.168. the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture. The preferred interface comprises at least a part of the C H j domain, hi this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g., tyrosine or tryptophan). Compensatory

"cavities'- of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g., alanine or tlireonine). This provides a mechanism for increasing the yield of the heterodimer over other uawanted end-products such as homodimers, Bispecifie antibodies produced in accordance with this approach are referred to herein as "protuberanee-into-eavity" antibodies.

Bispecifie antibodies include cross-linked or "heteroconjugate" antibodies. For example, one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Pat. No. 4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Pat. No. 4,676,980, along with number of cross- linking techniques.

Techniques for generating bispecifie antibodies from antibody fragments have also been described in the literature. For example, bispecifie antibodies can be prepared using chemical linkage, Brerman et a!. , Science 229:81 ( 1985) describe a procedure wherein intact antibodies are proteolytiealiy cleaved to generate F(ab')2 fragments. These fragments are reduced in the presence of the dithiol compiexing agent, sodium arsenite, to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab' fragments generated are then converted to tbionitrobenzoate (TNB) derivatives. One of the Fab'-TNB derivatives is then reconverted to the Fab'-thioi by reduction with

mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispeciiic antibody. The bispeciiic antibodies produced can be used as agents for the selective immobilization of enzymes.

Fab'-SH fragments from E. coli can be directly recovered and chemically coupled to form bispeciiic antibodies. Shalaby et al, J. Exp. Med. 175: 21 7-225 (1992) describe the production of a fully humanized bispeciiic antibody F(ab¾ molecule. Each Fab' fragment was separately secreted from E. coii and subjected to directed chemical coupling in vitro to form the bispeeific antibody. The bispeciiic antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T ceils, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets.

Various techniques for making and isolating bispeeific antibody fragments directly from recombinant cell culture have also been described. For example, bispeeific antibodies have been produced using leucine zippers. Kostelny et al,, J, Immunol. 148(5): 1547-1553 (1992). The leucine zipper peptides from the Fos and .Tun proteins were linked to the Fab ! portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The "diabody" technology described by Hollinger et al., Proc. Natl. Acad. Set. USA 90:6444-6448 (1993) has provided an alternative mechanism for making bispeeific antibody fragments, The fragments comprise a VH connected to a VL by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and VL domains of one fragment are forced to pair with the complementary VL and V domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispeeific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See Gruber et al. , J.

Immunol., 152:5368 (1994).

Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et a!., J. Immunol. 147:60 (1991),

& Antibody Variants

In certain embodiments, amino acid sequence variants of the antibodies provided herein are contemplated, For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody, Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody, Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding. a) Substitution Insertion;. ¾s¾d Deletion Variants

In certain embodiments, antibody variants having one or more amino acid substitutions are provided. Sites of interest for substitutional mutagenesis include the HVRs and FRs. Conservative substitutions are shown in Table I under the heading of "preferred substitutions." More substantial changes are provided in Table 1 under the heading of "exemplary substitutions," and as further described below in reference to amino acid side chain classes. Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g. , retamed'improved antigen binding, decreased immunogenicity, or improved ADCC or CDC.

TABLE 1

Amino acids may be grouped according to common side-chain properties;

(1 ) hydrophobic: Norleucine, Met, Ala, Val, Leu, lie;

(2) neutral hydrophilic: Cys, Ser, Thr, Asn, Gin; (3) acidic: Asp, Glu;

(4) basic: His, Lys, Arg;

(5) residues that influence chain orientation: Gly, Pro;

(6) aromatic: Trp, Tyr, Phe.

Non-conservative substitutions will entail exchanging a member of one of these classes for another class.

One type of substitutional variant involves substituting one or more hypervariabie region residues of a parent antibody (e.g. a humanized or human antibody). Generally, the resulting variant(s) selected for further study will have modifications (e.g. , Improvements) in certain biological properties (e.g., increased affinity, reduced immunogenicity) relative to the parent antibody and/or will have substantially retained certain biological properties of the parent antibody. An exemplary substitutional variant is an affinity matured antibody, which may be conveniently generated, e.g. , using phage display-based affinity maturation techniques such as those described herein. Briefly, one or more HVR residues are mutated and the variant antibodies displayed on phage and screened for a particular biological activity (e.g. binding affinity).

Alterations (e.g., substitutions) may he made in HVRs, e.g., to improve antibody affinity. Such alterations may be made in HVR "hotspots," i.e., residues encoded by codcns that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol.

207:179-196 (2008)), and/or SDRs (a-CDRs), with the resulting variant VH or VL being tested for binding affinity. Affinity maturation by constructing and reselecting from secondary libraries has been described, e.g., in Hoogenboom et ai in Methods in Molecular Biology 178:1-37 (O'Brien et al, ed,, Human Press, Totowa, NJ, (2001).) in some embodiments of affinity maturation, diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oiigonucieotide-directed mutagenesis), A secondary library is then created. The librar is then screened to identify any antibody variants with the desired affinity. Another method to introduce diversity involves HVR-directed approaches, in which several HVR residues (e.g. , 4-6 residues at a time) are randomized, HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted.

In certain embodiments, substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen. For example, conservative alterations (e.g., conservative substitutions as provided herein) that do not substantially reduce binding affinity may be made hi HVRs. Such alterations may be outside of HVR ''hotspots" or SDRs. In certain embodiments of the variant VH and VL sequences provided above, each HVR either is unaltered, or contains no more than one, two or three amino acid substitutions.

A useful method for identification of residues or regions of an antibody that may be targeted for mutagenesis is called "alanine scanning mutagenesis" as described by Cunningham and Weils (1 89) Science, 244:1081-1085. In this method, a residue or group of target residues (e.g., charged residues such as arg, asp, his, lys, and glu) are identified and replaced by a neutral or negatively charged amino acid (e.g., alanine or poiyalanine) to determine whether the interaction of the antibody with antigen, is affected. Further substitutions may be introduced at the amino acid locations demonstrating functional sensitivity to the initial substitutions. Alternatively, or additionally, a crystal structure of an antigen- antibody complex is used to identify contact points between the antibody and antigen. Such contact residues and neighboring residues may be targeted or eliminated as candidates for substitution. Variants may be screened to determine whether they contain the desired properties.

Amino acid sequence insertions include amino- and/or carboxyl-terrninal fusions ranging in length from one residue to polypeptides containing a himdred or more residues, as well as inirasequence insertions of single or multiple amino acid residues. Examples of terrninai insertions include an antibody with an N-tenninai methionyi residue. Other insertionai variants of the antibody molecule include the fusion to the N- or€ -terminus of the antibody to an enzyme (e.g. for ADEPT) or a polypeptide which incre -life of the antibody.

hi certain embodiments, an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated. Addition or deletion of glvcosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glvcosylation sites is created or removed.

Where the antibody comprises an Fc region, tire carbohydrate attached thereto may be altered. Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N~hnkage to Asn297 of the CK2 domain of the Fc region. See, e.g., Wright et al. TIBTECH 15:26-32 (1997). The oligosaccharide may include various carbohydrates, e.g. , mannose, N-acetyf glucosamine (GicNAc), galactose, and sialic acid, as well as a fucose attached to a GicNAc in the "stem" of the biantennary oligosaccharide structure. In some embodiments, modifications of the oligosaccharide in an antibody provided herein may be made in order to create antibody variants with certain improved properties.

In one embodiment, antibody variants are provided having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region. For example, the amount of fucose in such antibody may be from 1% to 80%, from 1 % to 65%, from 5% to 65% or from. 20% to 40%. The amount of fucose is determined by calculating the average amount of fucose widiin the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TQF mass spectrometry, as described in WO 2008/077546, for example. Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ± 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies. Such fucosylation variants may have improved ADCC function. See, e.g. , US Patent Publication Nos. US 2003/0157108 (Presta, I,); US 2004/0093621 (Kyowa Hakko Kogyo Co., Ltd). Examples of publications related to "defucosylated" or "fucose-deficieni" antibody variants include: US 2003/01571.08; WO 2000/61739; WO 2001/29246; US 2003/0115614; US 2002/0164328; US 2004/0093621 ; US 2004/0132140; US 2004/01 10704; US 2004/01 10282; LJS 2004/0109865; WO 2003/08511 ; WO 2003/0S4570; WO 2005/035586; WO 2005/035778; WO2005/053742;

WO2002 031140; Okazaki et l J. Mol Biol 336: 12394249 (2004); Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004). Examples of cell lines capable of producing defucosylated a tibodies include Led 3 CHO ceils deficient in protein fucosyiation (Ripka et l. Arch, Biochem. Biophys. 249:533-545 (1986); US Pat Appl No US 2003/0157108 Al , Presta, L; and WO 2004/056312 Al , Adams et al., especially at Example 1 1), and knockout cell lines, such as alpha- 1 ,6-fucosyltransferase gene, FUT8, knockout CHO ceils (see, e.g., Yamane-Ohnuki et al Biotech. Bioeng. 87: 614 (2004); Kanda, Y. et al, Bwtechnol Bioeng., 94(4):680-688 (2006); and WO2003/08 107).

Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/01 1878 (Jean-Mairet et al); US Patent No. 6,602,684 (Umana et l.); and US 2005/0123546 (Umana et al). Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function. Such antibody variants are described, e.g. , in WO 1 97/30087 (Pat el et al); WO 1998/58964 (Raju, S.); and WO 1999/22764 ( aju, S.),

e) Fc . regies _ variants

In certain embodiments, one or more amino acid modifications may be introduced i to the Pc region of an antibody provided herein, thereby generating an Fc region variant. The Fc region variant may comprise a human Fc region sequence (e.g., a human IgGl , IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification (e.g. a substitution) at one or more amino acid positions.

In certain embodiments, the invention contemplates an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half-life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious. In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depiction of CDC and/or ADCC activities. For example, Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FeyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability. The primary cells for mediating ADCC. NK ceils, express Fc(RHI only, whereas monocytes express Fe(RI, Fc(PJI and Fe(RUI. FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Ki et, Amu. Rev. Immunol. 9:457-492 (1991 ). Non-limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in U.S. Patent No, 5,500,362 (see, e.g. Heilstrom, I. et al. Proc. Nat l Acad, Set. USA 83:7059-7063 (1986)) and Hellstrom, I et al, Proc. Nat l Acad. ScL USA 82: 1499-1502 (1985); 5,821 ,337 (see Bruggemann, M. et a!., J. Exp, Med. 166:1351 -1361 (1987)). Alternatively, non-radioaciive assays methods may be employed (see, for example, ACTF non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, CA; and CytoTox 96* non-radioactive cytotoxicity assay (Promega, Madison, WT), Useful effector ceils for such assays include peripheral blood mononuclear ceils (FBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g. , in an animal model such as that disclosed in Clynes et al Proc. Nat'! Acad, Sci. USA. 95:652-656 (1998). Cl q binding assays may also be carried out to confirm that the antibody is unable to bind Clq and hence lacks CDC activity. See, e.g., Clq and C3c binding ELISA in WO 2006/029879 and WO 2005/100402. To assess complement activation, a CDC assay may be performed {see, for example, Gazzano-Santcro e.t al. , J. Immunol. Methods 202 : 163 (1996); Cragg, M.S. et al, Blood 101 : 1045-1052 (2003); and Cragg, M.S. and MJ. Giennie, Blood 103:2738-2743 (2004)). FcRn binding and in vivo clearance/half-life determinations can also be performed using methods known in the art (see, e.g., Petkova, S.B. et αί, ίηί'ί. Immunol. \ 8(12); 1759- 1769 (2006)).

Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Patent No. 6,737,056). 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 to alanine (US Patent No. 7,332,581).

Certain antibody variants with improved or diminished binding to FcRs are described. (See, e.g. , U.S. Patent No. 6,737,056; WO 2004/056312, and Shields et al., J. Biol. Chem. 9(2): 6591 -6604 (2001).)

in certain embodiments, an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC. e.g., substitutions at positions 298, 333, and/or 334 of the Fc region (EU numbering of residues).

In some embodiments, alterations are made in the Fc region that result in altered (i.e., either improved or diminished) Clq binding and/or Complement Dependent Cytotoxicity (CDC), e.g., as described in US Patent No. 6,194,551 , WO 99/51642, and idusogie et ai J. Immunol. 164: 4178-4184 (2000).

Antibodies with increased half-lives and improved binding to the neonatal Fc receptor (FcRn), which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al. , J. Immunol. 1 17:587 (1976) and Kim et al, J. Immunol 24:249 (1994)), are described in US2G05/G014934A1 (Hinton et a!.}. Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn. Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311 , 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US Patent No. 7,371 ,826). See a!so Duncan & Winter, Nature 322:738-40 ( 1988); U.S. Patent No. 5,648,260; U.S. Patent No. 5.624,821 ; and WO of Fc region variants.

in certain embodiments, it may be desirable to create cysteine engineered antibodies, e.g., "thioMAhs," in which one or more residues of an antibody are substituted with cysteine residues. In particular embodiments, the substituted residues occur at accessible sites of the antibody. By substituting those residues with cysteine, reactive thiol groups are thereby positioned at accessible sites of the antibody nd may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an innnunoconjugate, as described further herein, fc certain

embodiments, any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; .41 18 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region. Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No. 7,521 ,541.

e) A hodyJ>griy¾t|ygs

in certain embodiments, an antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available. The moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers. Non- limiting examples of water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethykeiiulose, dexiran, polyvinyl alcohol, polyvinyl pyrroiidone, poiy~l , 3-dioxolane, poly- 1 ,3,6-irioxane, ethyiene/maleic anhydride copolymer, po!yaminoaeids (either homopoiymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)polyethylene glycol, propropylene glycol homopoiymers, proiypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyois (e.g., glycerol), polyvinyl alcohol, and mixtures thereof Polyethylene glycol pro ionaldehyde may have advantages in manufacturing due to its stability in water. The polymer may be of any molecular weight, and may be branched or unbranched. The number of polymers attached to the antibody may vary, and if more than one polymer are attached, they can be the same or different molecules, in general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.

In another embodiment, conjugates of an antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided. In one embodiment, the nonproteinaceous moiety is a carbon nanotube (Kam ei αί, Proc. Natl Acad. Sci. USA 102: 1 ] 600-11605 (2005)). The radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody-nonproteinaceous moiety are killed. B. Recombisast Methods asid Compositions

Antibodies may be produced using recombinant methods and compositions, e.g., as described in U.S. Patent No. 4,816,567, In one embodiment, isolated nucleic acid encoding an anti-FcRH5 antibody described herein is provided. Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody). In a further embodiment, one or more vectors (e.g., expression vectors) comprising such nucleic acid are provided, ϊη a further embodiment, a host ceil comprising such nucleic acid is provided, in one such embodiment, a host cell comprises (e.g. , has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the Vli of the antibody. In one embodiment, the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NSO, Sp20 cell). In one embodiment, a method of making an anti-FcRHS antibody is provided, wherein the method comprises cuituring a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).

For recombinant production of an anti-FcRH5 antibody, nucleic acid encoding an antibody, e,g., as described above, is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell. Such nucleic acid 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 antibody),

Suitable host ceils for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein. For example, antibodies may he produced in bacteria, in particular when glycosylation and Fc effector function are not needed. For expression of antibody fragments and polypeptides in bacteria, see, e.g., U.S. Patent Nos. 5,648,237, 5,789,199, and 5,840,523. (See also Chariton, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NI, 2003), pp. 245-254, describing expression of antibody fragments in E. coli.) After expression, the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.

n addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose

glycosylation pathways have been "humanized," resulting in the production of an antibody with a partially or folly human glycosylation pattern. See Gemgross, Nat. Biotech. 22: 1409-1414 (2004), and Li et αί, Nat. Biotech. 24:210-215 (2006).

Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate ceils iiiciude plant and

36 insect cells. Numerous bacuioviral strains have been identified which may be used in conjunction with insect ceils, particularly for transfection of Spodoptera fruglperda ceils.

Plant cell cultures can also be utilized as hosts. See, e.g. , US Patent Nos, 5,959, 177, 6,040,498.

6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIES™ technology for producing antibodies in transgenic plants).

Vertebrate cells may also be used as hosts. For example, mammalian cell lines that are adapted to grow in suspension may be useful. Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham et at, J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK); mouse Sertoli cells (TM4 cells as described, e.g. , in Mather, Biol. Reprod. 23:243-251 (1980)): monkey kidney cells (CV1); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3A); human lung cells (Wl 38); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); T ' Rl cells, as described, e.g. , in Mather et a!,, Annals N. Y. Acad. Sci. 383 :44-68 (1 82); MRC 5 cells; and FS4 cells. Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR ' CHO cells (IJriaub et ai, Proc. Natl. Acad. Sci. USA 77:4216 (1980)); and myeloma cell lines such as Y0, NSO and Sp2/0. For a review of certain mammalian host cell lines suitable for antibody production, see, e.g. , Yazaki and Wu,

Methods in Molecular Biology. Vol 248 (B.K.C, Lo. ed., Humana Press, Totowa, NJ)„ pp. 255-268 (2003).

C. Assays

Anti-FcRH5 antibodies provided herein may be identified, screened for, or characterized for their physical/chemical properties and/or biological activities by various assays known in the art.

In one aspect, a antibody provided herein may be tested for its antigen binding activity, e.g., by known methods such as ELiSA, BIACore* 1 . FACS, or Western blot.

In another aspect, competition assays may be used to identify an antibody that competes with any of the antibodies described herein for binding to FcRHS. In certain embodiments, such a competing antibody binds to the same epitope (e.g., a linear or a conformational epitope) that is bound by an antibody described herein. Detailed exemplary methods for mapping an epitope to which an antibody binds are provided in Morris (1996) "Epitope Mapping Protocols," in Methods in Molecular Biology vol. 66 (Humana Press, Totowa, NJ),

In an exemplary competition assay, immobilized FcRHS is incubated in a solution comprising a first labeled antibody that binds to FcRH5 {e.g. , any of the antibodies described herein) and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to FcRHS. The second antibody may be present in a hyhridoma supernatant. As a control, immobilized FcRHS is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After incubation under conditions permissive for binding of the first antibody to FcRHS, excess unbound antibody is removed, and the amount of label associated with immobilized FcRHS is measured. If the amount of label associated with immobiiized FcRH5 is substantially reduced in the test sample relative to the control sample, then that indicates that the second antibody is competing with the first antibody for binding to FcRH5. See Harlow and Lane (1988) Antibodies: A Laboratory Manual ch.14 (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY), In some embodiments, the FcRH5 is FcRHSc. In some embodiments, the anti-FcRH5 antibody binds an isoform c-specific region of the extracellular domain of FcRHSc. In some embodiments, the anti-FcRH5 antibodies binds Ig-like domain 9 of FcRHSc.

B. Inamunoeojijssgates

Also provided herein are immunoconjugates comprising an anti-FcRH5 antibody herein conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins {e.g., protein toxins, enzymaticaliy active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes {i.e., a radioconjugate). In some embodiments, the anti-FeRH5 antibody binds an isoform c-specific region of the extracellular domain of FeRH5c. In some embodiments, the anti-FcRH5 antibodies binds Ig-like domain 9 of FcRHSc.

Im i ocGnjugates allow for the targeted delivery of a drug moiety to a tumor, and, in some embodiments intracellular accumulation therein, where systemic administration of unconjugated drugs may result in unacceptable levels of toxicity to normal cells (Poiakis P. (2005) Current Opinion in Pharmacology 5:382-387),

Antibody-drug conjugates (ADC) are targeted chemotherapeutic molecules which combine properties of bom antibodies and cytotoxic drugs by targeting potent cytotoxic drugs to antigen- expressing tumor cells (Teicher, B.A, (2009) Current Cancer Drug Targets 9:982-1004), thereby enhancing the therapeutic index by maximizing efficacy and mmimiaing off-target toxicity (Carter, FJ. and Senter P.D. (2008) The Cancer Jour. 14(3): 154-169; Chart R.V. (2008) A . Chem. Res. 41 :98- 107.

The ADC compounds provided herein include those with anticancer activity. In some embodiments, the ADC compounds include an antibody conjugated, i.e. covalently attached, to the drug moiety. In some embodiments, the antibody is covalently attached to the drug moiety through a linker. The antibody-drug conjugates (ADC) provided herein selectively deliver an effective dose of a drug to tumor tissue whereby greater selectivity, i.e. a lower efficacious dose, may be achieved while increasing the therapeutic index ("therapeutic window").

The drug moiety (D) of the antibody-drug conjugates (ADC) may include any compound, moiety or group that has a cytotoxic or cytostatic effect. Drug moieties may impart their cytotoxic and cytostatic effects by mechanisms including but not limited to tubulin binding, DNA binding or intercalation, and inhibition of RNA polymerase, protein synthesis, and/or topoisomerase. Exemplary drug moieties include, but are not limited to, a maytansinoid, dolastatin, auristatin, caiieheamicin, pyrrolobenzodiazepine (FBD), nemorubicin and its derivatives, PNU-159682, anthracycline, duocarmycin, vinca alkaloid, taxane, trichothecene. CC 1065, camptothecin, elinafide, and antibody (Ab) which targets a tumor cell, a drag moiety (D), and a linker moiety (L) that attaches Ah to D, In some ernbodin ents, the antibody is attaciied to the linker moiety (L) through one or more amino acid residues, such as lysine arid/or cysteine.

An exemplary ADC has Formula I:

Ah"(i. i¾, I

where p is 1 to about 20. hi some embodiments, the number of drug moieties that can be conjugated to an antibody is limited by the number of free cysteine residues, hi some embodiments, free cysteine residues are introduced into the antibody amino acid sequence by the methods described herein.

Exemplary ADC of Formula I include, but are not limited to. antibodies that have 1 , 2, 3, or 4 engineered cysteine amino acids (Lyon, R. et al (2012) Methods in Enzym, 502:123-138). In some embodiments, one or more free cysteine residues are already present in an antibody, without the use of engineering, in which case the existing free cysteine residues may be used to conjugate the antibody to a dmg, In some embodiments, an antibody is exposed to reducing conditions prior to conjugation of the antibody in order to generate one or more free cysteine residues. In some embodiments, the anti-FcRH5 antibody binds an isoform c-specific region of the extracellular domain of FcRHSc, In some embodiments, the anti-Fc ig~iike domain 9 of FcRH5c,

A "Linker" (L) is a bifunctional or multifunctional moiety that can be used to link one or more drug moieties (D) to an antibody (Ah) to form an antihody-drag conjugate (ADC) of Formula I. In some embodiments, antihody-drug conjugates (ADC) can be prepared using a Linker having reactive functionalities for covalently attaching to the drug and to the antibody. For example, in some embodiments, a cysteine thiol of an antibody (Ab) can form a bond with a reactive functional group of a linker or a drug-linker intermediate to make an ADC.

In one aspect, a linker has a functionality that is capable of reacting with a free cysteine present, on an antibody to form a covalent bond. Nonprinting exemplary such reactive functionalities include maleimide, haioacetamides, σ.-haioaeetyl, activated esters such as suceinimide esters. 4-nitrophenyl esters, pentafluorophenyi esters, ietrafluorophenyl esters, anhydrides, acid chlorides, sulfon l chlorides, isocyanates, and isothiocyanates. See, e.g. , the conjugation method at page 766 of lussman, et al (2004), Bioconjiigale Chemistry i5(4):765-773, and the Examples herein.

In some embodiments, a linker has a functionality that is capable of reacting with an electrophilic group present on an antibody. Exemplary such eiectrophilic groups include, but are not limited to, aldehyde and ketone carbonyl groups. In some embodiments, a heteroatorn of the reactive functionality of the linker can react with an electrophilic group on an antibody and form a covalent bond to an antibody unit. Nonlimiting exemplary such reactive functionalities include, but are not limited to, hydrazide, oxime, amino, hydrazine, t iosemicarbazone, hydrazine carboxyiate, and arylhydrazide.

A linker may comprise one or more linker components. Exemplary linker components include 6-ma!eimidocaproyl ("MC"), maieiinidopropaiioyi ("MP"), vaiine-eitrulime ("vai-cit" or "vc"), alanine- phenylalanine ("ala-phe"), p-aminobenzyloxycarhonyl (a "PAB"), N-Succinimidyl 4-(2-pyridylthio) pentanoate ("SPP"), and 4~(N~roaie nidomefhyl) cyeiohexane-1 caiboxyiale ("MCC"). Various linker components are blown in the art, some of which are described below.

A linker may be a "cleavable linker, ' ' facilitating release of a drug. Nonlimiting exemplary cleavable linkers include acid-iabile linkers {e.g., comprising hydrazone), protease-sensitive {e.g., pepridase-sensitive) linkers, photoiahile linkers, or disulfide-conlaining linkers (Chari et al , Cancer Research 52:127431 (1992); US 5208020).

In certain embodi Formula ΪΪ:

II

wherein A is a "sireicher unit", and a is an integer from 0 to 1 ; W is an "amino acid unit", and w is an integer from 0 to 12; Y is a "spacer unit", and y is 0, 1, or 2, An ADC comprising the linker of Formula II has the Formula 1(A): Ab-(A a -W w -Y y -D)p, wherein Ah, D, and p are defined as above for Formula I. Exemplary embodiments of such linkers are described in U.S. Patent No. 7,498,298, which is expressly incorporated herein by reference.

In some embodiments, a linker component comprises a "stretcher unit" (A) that links an antibody to another linker component or to a drug moiety. Nonlimiting exemplary sireicher units are shown below (wherein the wavy line indicates sites of covaient attachment to an antibody, drug, or additional linker components):

n some embodiments, a linker component comprises an "amino acid unit" ( W), In some such embodiments , the amino acid unit allows for cleavage of the linker by a protease, thereby facilitating release of the drug from the immunoconjugate upon exposure to intracellular proteases, such as lysosomal enzymes (Doronina et al, (2003) Nat BiotechnoL 21 :778~784). Exemplary amino acid units include, but are not limited to, dipeptides, tripeptides, tetrapeptides, and pentapeptides. Exemplary dipeptides include, but are not limited to, vaime-citruiline (vc or vai-cit), alanine-phenylalanine (af or ala-phe); phenylalanine-lysine (fk or phe-iys); phenylalanme-homolysine (phe-homolys); and N- methyl-vaiine-citruiime (Me-val-cit). Exemplary tripeptides include, but are not limited to, glycine- valine-citrulline (giy-val-cit) and giyeine-giycme-glycine (gly-gly~giy). An amino acid unit may comprise amino acid residues that occur naturally and/or minor amino acids and/or non-naturaily occurring amino acid analogs, such as citrulline. Amino acid units can be designed and optimized for enzymatic cleavage by a particular enzyme, for example, a tumor-associated protease, eathepsin B, C and D, or a plasmin protease.

Typically, peptide-type linkers can be prepared by forming a peptide bond between two or more amino acids and/or peptide fragments, Such peptide bonds can be prepared, for example, according to a liquid phase synthesis method (e.g., E, Schroder and . Liibke (1965) "The Peptides", volume 1 , pp 76-136, Academic Press),

In some embodiments, a linker component comprises a "spacer unit" (Y) that links the antibody to a drug moiety, either directly or through a stretcher unit and/or an amino acid unit. A spacer unit may be "self-inunoiative" or a "non-self-immolative." A "non-seif-mimolative" spacer unit is one in which part or all of the spacer unit remains bound to the drug moiety upon cleavage of the ADC. Examples of non-seif-immolative spacer units include, but are not limited to, a glycine spacer unit and a glycine- glycine spacer unit. In some embodiments, enzymatic cleavage of an ADC contaixiing a glycine-glycine spacer unit by a tumor-ceil associated protease results in release of a glycine-glycine-drug moiety from the remainder of the ADC. In some such embodiments, the glycine-glycine-drug moiety is subjected to a hydrolysis step in the tumor cell, thus cleaving the glycine-glycine spacer unit from the drug moiety.

A "self-immolative" spacer unit allows for reiease of the drug moiety, in certain embodiments, a spacer unit of a linker comprises a p-aminobenzyl unit. In some such embodiments, a p-aminobenzyl alcohol is attached to an amino acid unit via an amide bond, and a carbamate, methylcarbamate, or carbonate is made between the benzyl alcohol and ihe drug (Hamann et al (2005) Expert Opin. Then Patents (2005) 15:1087-1103). in some embodiments, the spacer unit comprises p- aminobeiizyloxycarbonyl (PAB), in some embodiments, an ADC comprising a self-immolative linker has the structur

wherein Q is -CpCg alkyl. -0-(C C 8 alkyl), -halogen, -nitro, or -cyano; m is an integer ranging from 0 to 4: X may be one or more additional spacer units or may be absent; and p ranges from 1 to about 20. In some embodiments, p ranges from 1 to 10, 1 to 7, i to 5, or 1 to 4, Nonlirniting exemplary X spacer units include:

/ \ ^ \/ ^γ^ \^

\ / i II

1 7 and *¾ 0 ; wherein ¾ and R 2 are independently selected from H and Q-Cs alkyl. In some embodiments. Ri and R2 are each -Cl¾.

Other examples of self-immolative spacers include, but are not limited to, aromatic compounds that are eiectronically similar to the PAB group, such as 2-ainiiioimidazol-5-metlianol derivatives (U.S. Patent No. 7,375,078; Hay et al. (1999) Bioorg. Med. Chem. Lett. 9:2237) and oriho- or para- aminobenzylaeetals. In some embodiments, spacers can be used that undergo eyciization upon amide bond hydrolysis, such as substituted and unsubstiruted 4~aminohutyric acid amides (Rodrigues et al (1995) Chemistry Biology 2:223), appropriately substituted bicyclo[2.2.I] and bicycio[2.2.2] ring systems (Storm et al (1972) J. Amer. Chem., Soc. 94:5815) and 2-aminophenylpropionic acid amides (Amsberry, et al (3990) J. Org. Chem. 55:5867). Linkage of a drug to the a-carbon of a glycine residue is another example of a self-i nmolative spacer that may be useful in ADC (Kingsbury et al (1984) J, Med. Chem. 27:1447).

In some embodiments, linker L may be a dendritic type linker for covaient attachment of more tiian one drug moiety to an antibody through a branching, multifunctional linker moiety (Sun et al (2002) Bioorganic & Medicinal Chemistry Letters 12:2213-221 ; Sun et al (2003) Bioorg nic &

Medicinal Chemistry 1 1 : 1761 -1768), Dendritic linkers can increase the molar ratio of drug to antibody, i.e. loading, which is related to the potency of the ADC. Tims, where an antibody bears only one reactive cysteine thiol group, a multitude of drug moieties may be attached through a dendritic linker.

Nonlimiting exemplary linkers are shown below in the context of an ADC of Formula Ϊ:

; wherein R ; and

R 2 are independently selected from IT and C C 6 alkyi. In some embodiments, Rl and R2 are each - Phe-homoLys-PAB~Ab; wherein n is 0 to 12. in some embodiments, n is 2 to 10. In some embodiments, n is 4 to 8.

onlirnitmg exemplary ABCs include the siractures:

where X is:

each is independently H or Cj-Ce a!kyl; and n is 1 to 12,

In some embodiments, a linker is substituted with groups that modulate solubility and/or reactivity. As a noniirniting example, a charged substituent such as sulfonate (-SO;, ' ) or ammonium may increase water solubility of the linker reagent and facilitate the coupling reaction of the linker reagent with the antibody and/or the drug moiety, or facilitate the coupling reaction of Ata-L (antibody-linker intermediate) with D, or D~L (drug-linker intermediate) with Ab, depending on the synthetic route employed to prepare the ADC. In some embodiments, a portion of the linker is coupled to the antibody and a portion of the linker is coupled to the drug, and then the Ab-(lmker portion) 3 is coupled to drug- (imker portion)* 5 to form the ADC of Formula I.

The compounds provided herein expressly contemplate, but are not limited to, ADC prepared with the following linker reagents: bis-maleirnido-trioxyethylene glycol (BMPEO), Ν-(β- maieimidopropyloxy)~N-hydroxy succinimide ester (BMPS), N-(e-maieimidocaproyioxy) succinimide ester (EMCS), N-[y~maleitnidob tyryloxy]succmimide ester (GMBS), 1 ,6~hexane~bis-vinylsulfone

(HBVS), succinimidyl 4-( -maieimidomethyl)cyelohexane-l ~carhoxy~(6~amidoeaproaie) (LC-SMCC), m-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS), 4-(4-N-Maleiniidophenyl)buiyrie acid hydrazide (MPBH), succinimidyl 3-(bromoacetamido)propionate (SBAP), succinimidyl iodoacetate (SIA), succinimidyl (4~iodoacetyl)aminobenzoate (SLAB), N~succininiidyl~3~(2-pyridy ithio) propionate (SPDP), N-succmimidyl-4~(2-pyridylthio)pentanoate (SPP), succinimidyl 4-(N- maieimidometliyl)cyciohexane- 1 -carboxyiate (SMCC), succinimidyl 4-(p-maleiniidoph«aiyl)butyrate (SMPB), succinimidyl 6-[(beta-maleimidopropionaniido)hexanoate] (SMPH), iminothiolane (IT), sutfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfoMBS, sulfo-SIAB, sulfo-SMCC, and su!fo-SMPB, and suceimnrndyl-(4-vinylsulfone)benzoate (5VSB), and including bis-maleimide reagents:

dithiobismaieimidoethane (DTME), 1 ,4-Bismaleimidobutane (BMB), 1 ,4 BismaIeimidyl-2,3- dihydroxybutane (BMDB), bismaleimidohexane (BMH), bismale nidoethane (BMOE), BM(PEG)j (shown below), and ΒΜ(ΡΕΟ) 3 (shown below); bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuecmirnidyi suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenz»yi)-ethylenedia3Xiiiie), diisoeyanates (such as toluene 2,6- diisocyanate), and bis-active fluorine compounds (such as l ,5-diiluoro-2,4-dinitrobenzene). In some embodiments, bis-maleimide reagents allow the attachment of the thiol group of a cysteine in the antibody to a thioi-containing drug moiety, linker, or linker-drug intermediate. Other functional groups that are reactive with thiol groups include, but are not limited to, iodoaceiamide, bromoaceiamide, vinyl pyridine, disulfide, pyridyl disulfide, isocyanate, and isothiocyanate.

B (PEG) 2 B (PEG) 3

Certain useful linker reagents can be obtained from various commercial sources, such as Pierce Biotechnology, inc. (Rockford, IL), Molecular Biosciences Inc.fBoulder, CO), or synthesized in accordance with procedures described in the art; for example, in Tola et ai (2002) J. Org. Chem, 67:1866-1872; Dubowchik, et al (1997) Tetrahedron Letters, 38:5257-60; Walker, M.A. (1995) J, Org. Chem. 60:5352-5355; Frisch et al (1996) Bioconjugate Chem. 7:180-186; US 6214345; WO 02/088172; US 2003130189; US2003096743: WO 03/026577; WO 03/043583; and WO 04/032828.

Carbors-i 4-labeled l-isothiocyanaiobenzyl-3-meihyldieihylene triaminepentaaeetie acid (MX-

DTP A) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See, e.g., WO94/1 1026.

(1) Maytansine and maytansinoids

In some embodiments, an immunoeonjugate comprises an antibody conjugated to one or more maytansinoid molecules. Maytansinoids are derivatives of maytansine, and are mitototic inhibitors which act by inhibiting tubulin polymerization. Maytansine was first isolated from the east African shrub Maytenus serrata (U.S. Patent No. 389611.1 ). Subsequently, it was discovered that certain microbes also produce maytansinoids, such as maytansinol and C-3 maytansinol esters (U.S. Patent. No. 4,151 ,042). Synthetic maytansinoids are disclosed, for example, in U.S. Patent os. 4.137.230;

4,248,870; 4,256,746; 4,260,608; 4,265,814; 4,294,757; 4,307,016; 4,308,268; 4,308,269; 4,309,428; 4,313,946; 4,315,929; 4,317,821 ; 4,322,348; 4,331 ,598; 4,361 ,650; 4,364,866; 4,424,219; 4,450,254; 4,362,663; and 4,371 ,533.

Maytansinoid drug moieties are attractive drug moieties in antibody-drug conjugates because they are: (i) relatively accessible to prepare by fenneniation or chemical modification or derealization of fenneniation products, (ii) amenable to derealization with functional groups suitable for conjugation through non-disuifide linkers to antibodies, (hi) stable in plasma, and (iv) effective against a variety of tumor cell lines.

Certain maytansinoids suiiabie for use as maytansinoid drug moieties are known in the art and can be isolated from natural sources according to known methods or produced using genetic engineering techniques {see, e.g., Yu et ai (2002) PNAS 99:7968-7973). Maytansinoids may also be prepared synthetically according to known methods.

Maytansinoid drug moieties include, but are not limited to, those having a modified aromatic ring, such as: C-1 ~dechloro (US Pat. No. 4256746) (prepared, for example, by lithium aluminum hydride reduction of ansamytoein P2); C-20-hydroxy (or C-20-demethyl) +/-C-19~deehtoro (US Fat. Nos. 4361650 and 430701 ) (prepared, for example, by demethylation using Streptomyces or

Actinomyces or dechlorination using LAH); and C-20-dernethoxy, C-20-acyloxy (-OCOR), +/-dechloro (U.S. Fat, No. 4,294,757) (prepared, for example, by acylation using acyl chlorides), and those having modifications at other positions of the aromatic ring.

Maytansinoid drug moieties also include those having modifications such as: C-9-SH (US Pat.

No. 4424219) (prepared, for example, by the reaction of maytansinol with ¾5 or PiSj); C-14- alkoxymeihyl(demetlioxy CH : O )(US 4331598); C~14-hydroxymethyl or acyioxymethyl (CH 2 OH or C¾OAc) (US Pat. No. 4450254) (prepared, for example, from Noeardia); C-15-hydroxy/acyloxy (US 4364866) (prepared, for example, by the conversion of maytansinol by Strepiomyces): C-lS-rnethoxy (US Pat. Nos. 4313946 and 4315929) (for example, isolated ftom Trewia nudfflora): C-18-N-demethyl (US Pat. Nos. 4362663 and 4322348) (prepared, for exainpie, by the deraeihyiation of maytansinol by Strepiomyces); and 4,5-deoxy (US 4371533) (prepared, for example, by the titanium trichloride/LAH reduction of maytansinol).

Many positions on maytansinoid compounds are useful as the linkage position. For example, an ester linkage may be formed by reaction with a hydroxy! group using conventional coupling techniques. In some embodiments, the reaction may occur at the C-3 position having a hydroxyl group, the C-14 position modified with hydroxymethyl, the€-15 position modified with a hydroxyl group, and the C~20 position having a hydroxyl group. In some embodiments, the linkage is formed at the C~3 position of maytansinol or a maytansinol analogue.

Maytansino

where the wavy line indicates the covalent attachment of the sulfur atom of the maytansinoid drug moiety to a linker of an ADC. Each may independently be H or a C|-C 6 alkyi. The alkylene chain attaching the amide group to the sulfur atom may be mefhanyi, ethanyl, or propyl, i.e., m is 1 , 2, or 3 (US 633410; US .5208020; Chari et al (1992) Cancer Res. 52:127-1 31 ; Liu et al (1 96) Proc. Na . Acad. Sci USA 93:8618-3623).

All stereoisomers of the maytansinoid drug moiety are contemplated for the ADC provided herein, i.e. any combination of R and S configurations at the ehiral carbons (US 7276497; US 69 3748; US 6441 163; LIS 633410 (RE39151 ); US 5208020; Widdison et al (2006) J. Med, Chem. 49:4392- 4408, which are incorporated by reference in their entirety). In some embodiments, the maytansinoid drug moiety has the following stereochemistry:



wherein the wavy iine indicates the eovaient attachment of the sulfur atom of the drug to a linker (L) of an antibody-drag conjugate,

Other exemplaiy maytansinoid antibody-drug conjugates have the following structures and abbreviations (wherein Ab is antibody and p is 1 to about 20, In some embodiments, p is 1 to 10, p is 1 to 7, p is 1 to 5, or p is 1 to 4):

Ab-SMCC-DMl Exemplary antibody-drug conjugates where DM1 is linked through a BM.FEO linker to a thiol

where Ab is antibody: n is 0, 1 , or 2; and p is 1 to about 20, In some embodiments, p is i to 10, p is 1 to 7, p is 1 to 5, or p is 1 to 4,

Xrnmunoconjugates containing maytansinoids, methods of making the same, and their therapeutic use are disclosed, for example, in U.S. Patent Nos. 5.208,020 and 5,416,064; US

2005/0276812 AI ; and European Pateut EP 0 425 235 Bl , the disclosures of which are hereby expressly incorporated by reference. See also Liu et al Proc. Natl. Acad, Sci. USA 93:8618-8623 (1996); and Chari et l Cancer Research 52: 127431 (1992).

In some embodiments, atitibody-maytansinoid conjugates may be prepared by chemically linking ati antibody to a maytansinoid molecule without significantly diEnkiishing the biological activity of either the antibody or the maytansinoid molecule. See, e.g., U.S. Patent No. 5.208.020 (the disclosure of which is hereby expressly incorporated by reference). In some embodiments, ADC with an average of 3-4 maytansinoid molecules conjugated per antibody molecule has shown efficacy in enhancing cytotoxicity of target cells without negatively affecting the function or solubility of the antibody. In some instances, even one molecule of toxin antibody is expected to enhance cytotoxicity over the use of naked antibody.

Linking groups for making antibody-maytansinoid conjugates include, for example, those described herein and those disclosed in U.S. Patent No. 5208020; EP Patent 0 425 235 Bl ; Chari et al Cancer Research 52: 127-131 (1 92); US 2005/0276812 Al ; and US 2005/016993 Al , the disclosures of which are hereby expressly incorporated by reference.

(2) Auristatins and dolastatins

Drug moieties include dolastatins, auristatins, and analogs and derivatives thereof (US

5635483; US 5780588; US 5767237; US 6124431). Auristatins are derivatives of the marine mollusk compound dolastatin-10. While not intending to be bound by any particular theory, dolastatins and auristatins have been shown to interfere with microtubule dynamics, OTP hydrolysis, and nuclear and cellular division (Woyke et ai (2001) Antimicrob. Agents and Chemother. 45(1 2):3580-3584) and have anticancer (US 5663149) and antifungal activity (Pettit et al (1 98) Antimicrob. Agents Chemother. 42:2961 -2965). r .Fhe dolastalin/auristatin drug moiety may be attached to the antibody through the N (amino) terminus or the C (carboxyl) terminus of the peptidic drug moiety (WO 02/088172; Doronina et ai (2003) Nature Biotechnology 21 (7):778-784; Francisco et ai (2003) Shod 102(4):1458- 1465).

Exemplary auristatin embodiments include the N-terminus linked monomethylauristatin drug moieties D E and Dp, disclosed in US 7498298 and US 7659241 , the disclosures of which are expressly incorporated by reference in their entirety:

wherein the wavy line of DE and Dp indicates the covalent attachment site to an antibody or antibody- linker component, and independently at each location:

R 2 is selected from H and C r Cs alkyl;

R 3 is selected from H, Q-Cs aikyl, Cj-Cs carbocycle, aryl, Cj -Cg alkyl-aryl, CpCg alkyl~(Cj-Cg carbocycle), C\-C 3 heterocycle and C r Q alkyl~(Ci-C 3 heterocycle);

R 4 is selected from H, C Cg alkyl, Cj-Cs carbocycle, aryl. C r g alkyl-aryl, C Q a3kyi-(C ? ,-C 3 carbocycle), C 3 -C 8 heterocycle and Cj-Cs alkyl~(C Cs heterocycle);

R 5 is selected from H and methyl;

or R 4 and R 5 jointly form a carbocyelie ring and have the formula -(CR a R b )„- wherein R a and R u are independently selected from H, C r C 8 alkyl and C 3 - carbocycle and n is selected from 2. 3, 4, 5 and 6; R 6 is selected from H and Ci -Q alkyl;

R' is selected from H, O-Cg alkyl, C3-CS carbocycle, aryl, Cj - g alkyl-aryl, C:-C 8 alkyl-(Cj-C a carbocycle), C ¾ -Cg heterocycle and C Ca alkyi-(C 3 -C 3 heterocycle);

each R* is independently selected from H, OH, C r C g alkyl, Cj-Cg carbocycle and 0-(C Q alkyl); R' is selected from H and C Cg alkyl;

R i is selected from aryl or CVC S heterocycle;

Z is O, S, H, or NR 1" , wherein R !i is C r C 8 alkyl:

R" is selected from H, C C 2t j alkyl, aryl, C C g heterocycle, ~(R ; 'Q)„ r R !4 , or ^R 1 5 0) ffi -CH(R'¾; m is an integer ranging from 1 -1000; R is C 2 -C 8 alkyl:

R l4 is H or C r Q alkyl;

each occurrence of R i S is independently H s COOH, ~(CH ? .) n -N(R 16 ) 2 , -<CH 2 ) E -S0 3 H, or -(CH 2 ) [r SO . Cj-Cs alkyl;

each occurrence of R i6 is independently H, C r C s alkyl, or -(CIL COOH;

R , s is selected from ~C(R s ),~C( 8 ) -aryl, --C( 8 ) -C(R s ) 2 -(C 6 -C 8 heterocycle), and

-C(R 8 ) 2 -C(R s ) 2 -iC,-C H earbocycle); and

n is an integer ranging from 0 to 6.

In one embodiment, R 3 , R 4 and R 7 are independently isopropyl or see-butyl and R 5 is -H or methyl, i an exemplary embodiment, R 3 and R 4 are each isopropyl, R s is -H, and R 7 is see -butyl.

In yet anodier embodiment, R 2 and R° are each methyl, and R s is -H,

In still another embodiment, each occurrence of R 8 is -OCH,.

In some embodiments, R J and R ' ' are each isopropyl, R 2 and R ¾ are each methyl, R is -H, R'' is sec-butyl, each occurrence of R ¾ is -QCHj, and R 9 is -H,

In one embodiment, Z is ~Q- or ~NH~.

In one embodiment, R i0 is aryl.

In an exemplary embodiment, R 1'3 is -phenyl.

In an exemplary embodiment, when Z is ~G~. R i : is -H, methyl or t-butyl,

In one embodiment, when Z is -NIL R n is ~CH(R I5 ) 2 , wherein R 15 is -(CH 2 ) n -N(R ,6 ) 2 , and R !6 is -C: -Cg alkyl or -(CH 2 )a-COOH.

In another embodiment, when Z is -NH, R ! 1 is -CHf ^, wherein R 15 is ~(CH 2 ) ;! ~S0 3 H.

An exemplary auristatin embodiment of formula D t ; is MMAE, wherein the wavy line indicates the covalent attachment to a linker (L) of an antibody-drug conjugate:

An exemplary auristatin embodiment of formula D F is MMAF, wherein the wavy line indicates the covalent attachment to a linker (L) of an antibody-drug conjugate:

Other exemplary embodiments include monoraethylvaline compounds having phenylalanine carboxy modifications at the C-tenninus of the pentapeptide auristatin drag moiety (WO 2007/008848) and monomethyl valine compounds having phenylalanine sidechain modifications at the C-temiinus of the pentapeptide auristatin drug moiety (WO 2007/008603), Monlimiting exemplary embodiments of ADC of Formula Ϊ comprising MMAE or MMAF and various linker components have the following structures and abbreviations (wherein "Ab" is an

Ab-MC-vc-PAB-MMAF

Ab-MC-vc-PAB-MMAE

Ab-MC-MMA

Ab-MC-MMAF

oniimiting exemplary embodiments of ADCs of Fonnula I comprising MMAF and various linker components further include Ab-MC-PAB-MMAF and Ab-PAB-MMAF. Immunoconjugates comprising MM/VF attached to an antibody by a Sinker that is not proteo lyrically cieavabie have been shown to possess activity comparable to immunoconjugates comprising MMAF attached to an antibody by a proteolytieally cieavabie linker (Doroniiia et id. (2006) Bioconjugate Chem. 17: 1 14-124), In some such embodiments, drug release is believed to be effected by antibody degradation in the cell.

Typically, peptide-based drug moieties can be prepared by forming a peptide bond between two or more amino acids and/or peptide fragments, Such peptide bonds can be prepared, for example, according to a liquid phase synthesis method see, e.g., E. Schroder and . Liibke, "The Peptides", volume 1 , pp 76-136, 1 65, Academic Press), Auristafiri/dolastatin drug moieties may, in some embodiments, be prepared according to the methods of: US 7498298; US 5635483; US 57805SS; Pettit et al ( 1989) J. Am. Chem. Soc. 1 1 1 :5463-5465 ; Pettit et al ( 1 98) Anti-Cancer Drug Design 13 :243- 277; Pettit, G.R., et al. Synthesis, 1996, 719-725; Pettit et ai (1996) J, Chem. Soc. Perkin Trans. 1 5:859-863; and Doronina (2003) Nat. BiotechnoL 21 (7):778-784.

In some embodiments, auristatm/dolastatin drug moieties of formulas D E such as MMAE, and D F; such as M.MAF, and drug-linker intermediates and derivatives thereof, such as MC-MMAF, MO MMAE, MC-vc-PAB-MMAF, and MC-vc-PAB-MMAE, may be prepared using methods described in US 7498298; Doronina et al. (2006) Bioconjugate Chem. 17: 1 14-124; and Doronina et al (2003) Nat Biotech. 21 :778-784and then conjugated to an antibody of interest.

(3) Calieheamiein

In some embodiments, the immune-conjugate comprises an antibody conjugated to one or more calieheamiein molecules. The calieheamiein family of antibiotics, and analogues thereof, are capable of producing double-stranded DNA breaks at sub-pieomolar concentrations (Hinman et at, (1993) Cancer Research 53:3336-3342; Lode et al, (1998) Cancer Research 58:2925-2928). Calieheamiein has intracellular sites of action but, in certain instances, does not readily cross the plasma membrane.

Therefore, cellular uptake of these agents through antibody-mediated internalization may, in some embodiments, greatly enhances their cytotoxic effects. Nonii niiing exemplary methods of preparing antibody-drug conjugates with a calieheamiein drug moiety are described, for example, in US 571 2374: US 5714586; US 57391 16; and US 5767285.

(4) Pyrrolobenzodi zepines

In some embodiments, an ADC comprises a pyrroEobenzodiazepine (PBD). In some embodiments, PDB diniers recognize and bind to specific DNA sequences. The natural product anihramycin, a PBD, was first reported in 1965 (Leimgruber, et al, (1.965) J. Am. Chem. Soc, 87:5793- 5795; Leimgruber, et al, (1965) /. Am. Chem. Soc, 87:5791 -5793). Since then, a number of PBDs, both naturally-occurring and anaEognes, have been reported (Thurston, et al, (1994) Chem. Rev. 1994, 433-465 including diniers of the tricyclic PBD scaffold (US 6884799; US 704931 1 ; US 706751 1 ; US 7265105; US 7511032; US 7528126; US 7557099). Without intending to be bound by any particular theory, it is believed that the dimer structure imparts the appropriate three-dimensional shape for isohelicity with the minor groove of B~form D A, leading to a snug fit at the binding site ( ohn, in Antibiotics III, Springer- Verlag, New York, pp. 3-1 1 (1975); Hurley and Needham-VartDevanter, (1986) Acc, Chem. Res., 1 :230-237). Dirneric PBD compounds bearing C2 aryl substituents have been shown to be useful as cytotoxic agents (Hartley et al (2010) Cancer Res. 7G(17):6849-6858; Antonow (2010) J. Med. Chem. 53(7):292?-2941 ; Howard et ai (2009) Bioorganic and Med. Chem. Letters 19(22):6463-6466).

PBD dimers have been conjugated to antibodies and the resulting ADC shown to have anticancer properties. Nonlimiting exemplary linkage sites on the PBD dimer include the five-rnernbered pyrrolo ring, the tether between the PBD units, and the N10-C11 imine group (WO 2009/016516; US 2009/304710; US 2010/047257; US 2009/036431 ; US 201 1 /0256157; WO 201 1 /130598). onlimiting exemplary FBI dirtier components o: 2s are of Formula A:

and salts and solvates thereof, wherein:

the wavy line indicates the covalent attachment site to the linker;

the dotted lines indicate the optional presence of a double bond between CI and C2 or C2 and C3;

R 2 is independently selected from H, OH, =0, =€¾, CN, R, OR, ~CH-R D , -C(R D ) 2 , 0~S0 2 ~R, C(¾R and COR, and optionally farther selected from halo or dihalo, wherein R D is independently selected from R, C0 2 R, COR, CHO, CQ 2 H, and halo;

R 6 and R 9 are independently selected from H, R, OH, OR, SH, SR, N¾, NHR, NRR', NO,,

Me 3 Sn and halo;

R 7 is independently selected from II, R, OH, OR, SH, SR, N¾. NHR, NRR', NO,, M¾Sn and halo;

Q is independently selected from O, S and NR.;

R ! 1 is either H, or R or, where Q is O, SOj.M, where M is a metal cation;

R and R" are each independently selected from optionally substituted C; B alkyl, CE -E2 alkyl, heteroeyclyl,€ 3 . heterocycle, and C5.20 aryl groups, and optionally in relation to the group NRR', R and R' together with the nitrogen atom to which they are attached form an optionally substituted 4-, 5-, 6- or 7~membered heterocyclic ring;

R n , R i6 , R 19 and R 17 are as defined for R 2 . R 6 , R 8 and R v respectively;

R" is a C 5 .;2 alkylene group, which chain may be interrupted by one or more heteroatoms, e.g. O, S, (H), NMe and/or aromatic rings, e.g. benzene or pyridine, which rings are optionally substituted; and

X and X' are independently selected from O, S and N(H).

in some embodiments, R and R * are each independently selected from optionally substituted CV is alkyl, C3.20 heterocycle, and C5.20 aryl groups, and optionally in relation to the group NRR', R and ' together with the nitrogen atom to which they are attached form an optionally substituted 4-, 5-, 6- or 7-niembered heterocyclic ring.

In some embodiments, R 9 and R :3 are H.

in some embodiments, R 6 and R :6 are H.

In some embodiments, R 7 are R 1 7 are both GR /A , where R 7A is optionally substituted Cj. alkyl. in some embodiments, R' A is Me. In some embodiments, R' A is is ChiPh, where Ph is a phenyl group.

In some embodiments, X is O. In some embodiments, R 1 is H.

in some embodiments, there is a double bond between C2 and C3 in each monomer unit, in some embodiments, R 2 and R ! " are independently selected from H and R. In some embodiments, R" and R ' ~ are independently R. in some embodiments, R 2 and R i2 are independently optionally substituted C5.20 aryl or Cj. aryi or C g .. !0 a l- In some embodiments, R 2 and R !J are independently optionally substituted phenyl, thienyl, napthyl, pyridyi, quinolinyl, or isoquinoiinyl. In some embodiments, R 2 and R 13 are independently selected from ~-Q, ::: C¾, ~ CH-R U , and ~C(R D ) 2 . In some embodiments, R " and R each ~CH 2 . In some embodiments, R" and R.'~ are each H, In some embodiments, R ^ and R i2 are each =0. In some embodiments, R " and R ! ' are each ^CF-.. In some embodiments, R * and/or ' £ are independently in some embodiments, R 2 and/or R 12 are independently = : CH-R i> .

In some embodiments, when R 2 and/or R ! " is =CH-R D , each group may independently have either configuration shown below:

in)

In some embodiments, a ~ CK~R D is in configuration (I).

In some embodiments, R ff is a C 3 alkylene group or a C s alkyiene group.

In some embodiments, an exemplary PSD dimer component of an ABC has the structure of l

wherein n is 0 or 1.

In some embodiments, an exemplary FBD dimer component of an ADC has the structure of Formul

wherein n is 0 or 1 , In some embodiments, an exemplary PBD dimer component of an ADC has the structure of Formula Α(

wherein R £ and R £ are each independently selected from H or R D , wherein R D is defined as above; and wherein n is 0 or 1 ,

in some embodiments, n is 0, In some embodiments, n is i . In some embodiments, E and/or R £ is H. in some embodiments, R = and R £ are H, In some embodiments, R E and/or R E is R , wherein R D is optionally substituted€j.; 2 alkyi. in some embodiments, R E and/or R 11 is R D , wherein R D is methyl

In some embodiments, an exemplars' PBD dimer component of an ADC has the structure of Fo

A(IV);

wherein Ar' and Ar are each independently optionally substituted C 5 . J O aryl; wherein Ar' and Ar 2 may ¬ be the same or different; and

wherein n is 0 or 1.

In some embodiments, an exemplars' PBD dimer component of an ADC has the structure of Formula A(V):

wherein Ar ! and Ar 2 are each independently optionally substituted C 5-2 o ar l; wherein Ar 1 and Ar may be the same or different; and

wherein n is 0 or 1 ,

In some embodiments, Ar ! and Ar are each independently selected from optionally substituted phenyl, furanyl, thiophenyl and pyridyl. In some embodiments, Ar 1 and Ar are each independently optionally substituted phenyl. In some embodiments, Ar ! and Ar ' are each independently optionally substituted thien-2~yi or thien-3-yl. In some embodiments. Ar 1 and Ar are each independently optionally substituted quinolinyl or isoquinoiinyl. The quinolinyl or isoquinoiinyl group may be bound to the PBD core through any available ring position. For example, the quinoiinyi may be quinolin-2-yl, quroolm~3-yl, quinoiin-4yl, qiainoiin-5-yL quinoiin-6-yl, quinoiin~7-yl and quinolm~8~yL in some embodiments, the quinoiinyi is selecied from qumoIin-3-yl and quinoiin-6-yL The isoquinoiinyi may be isoquinolin-l -yl, isoquinobn~3-yl, isoquinohn-4yl s isoquinolin-5-yi, isoquinoln 6~yl, isoquinolin-7-yi and isoquinoiin-S-yi. in some embodiments, die isoquinoiinyi is selected from isoquinoIin-3-yl and isoq inolin-6-yl.

Further nonhmiting exemplary FBD dinier components of ADCs are of Formula B:

B and salts and solvates thereof, wherein:

the wavy line indicates the covalent attachment site to the linker;

the wavy line connected to the OH indicates the S or R configuration;

R Vi and R. v2 are independently selected from H, methyl, ethyl and phenyl (which phenyl may be optionally substituted with fltioro, particularly in the 4 position) and C<_ (i heterocyclyi; wherein R Vi and R V2 may be the same or different; and

n is 0 or 1.

In some embodiments, R l and R V2 are independently selected from H, phenyl, and 4~ fluorophenyl.

In some embodiments, a linker may be attached at one of various sites of the FBD dimer drug moiety, including the Nl 0 imine of the B ring, the C-2 endo/exo position of the C ring, or the tether unit linking the A rings (see structures C(I) and C(II) below).

Nonlimi (I) and C(II):

C(I)

cai) Formulas€(ϊ) and C(II) are shown in their N10-C1 1 imine form. Exemplary PBD drug moieties also include the carbinolamine and protected earbmoiamine forms as well, as shown in the table below:

wherein:

X is C¾ (n = 1 to 5), N, or O;

Z and Z' are independently selected from OR and NR 2 , where R is a primary, secondary or tertiary alkyl chain containing 1. to 5 carbon atoms;

Rt, R'i, R.2 and R'j are each independently selected from H, C r Cg alkyl, C 2 -C 8 alkenyl, C 2 ~Cg alkynyl,

C5.20 aryl (including substituted aryls), C s . 2 o heteroaryl groups, -NH 2 , -NHMe, -OH. and -SH, where, in some embodiments, alkyl, alkenyl and alkynyl chains comprise up to 5 carbon atoms;

R 3 and R\ are independently selected from H, OR, NHR, and R ?! where R is a primary, secondary or tertiaiy alkyl chain containing 1 to 5 carbon atoms;

R4 and R are independently selected from H, Me, and OMe;

Rs is selected from Cj-Cg alkyl, C 2 ~Cg alkenyl, C 2 ~C S alkynyl, C5.20 aryl (including aryls substituted by halo, nitro, cyano, alkoxy, alkyl, heterocyclyl) and Cs_?o heteroaryl groups, where, in some

embodiments, aikyi, alkenyl and alkynyl chains comprise up to 5 carbon atoms;

Rs I is H. Cj-Cg alkyl, or a protecting group (such as acetyl, trifluoroacetyl, t-butoxycarbonyl (HOC), benzyioxycarbonyl (CBZ), 9~fiuorenylmethylenoxycarbonyl (Fmoc), or a moiety comprising a self- immolating unit such as val e-citruliine-PAB);

R; 2 is Is H, C]-Cs aikyi, or a protecting group;

wherein a hydrogen of one of R 3 , R 5 U R ; . R%, or R, 2 or a hydrogen of the -CX¾CH 2 (X) n CH 2 CH 2 0- spacer between the A rings is replaced with a bond connected to the linker of the ADC.

Exemplary PDB dimer portions of ADC include, but are not limited to (tire wavy line indicates the site of covalent attachment to the linker):

PBD dimer;

Nonlimiting exemplary embodiments of ADCs comprising PBD dimers have the following structures:

PBD dimer-val-cii-PAB-Ab:

PBD dimer-Phe-homoLys-PAB-Ab, wherein: n is 0 to 12. In some embodiraenis, n is 2 to 10. In some embodiments, n is 4 to 8. In some

embodiments, n is selected from 4, 5, 6, 7, and 8. The linkers of PBD dimer-val-cit-PAB-Ab and the PBD dimer-Phe-homoLys~PAB~Ab are protease eleavabie, while the linker of PBD dimer-maieirnide-acetal is acid-labile,

PBD dimers and ADC comprising PBD dimers may be prepared according to methods known in the art. See, e.g., WO 2009/016516; US 2009/304710; US 2010/047257; US 2009/036431 ; US 201 1/0256157; WO 201 1/130598.

(5) Anthracyclines

in some embodiments, an ADC comprisi g anthracydine. Anthracyclines are antibiotic compounds diat exhibit cytotoxic activity; While not intending to be bound by any particular theory, studies have indicated that anthracyclines may operate to kill cells by a number of different mechanisms, including: 1) intercalation of the drug molecules into the DNA of the cell thereby inhibiting DNA-dependent nucleic acid synthesis; 2) production by the drug of free radicals which then react with cellular macroniolecules to cause damage to the cells, and/or 3) interactions of the drug molecules with the cell membrane (see, e.g., C. Peterson et ai, "Transport And Storage Of

Anthracydine In Experimental Systems And Human Leukemia" in Atithracyciine . Antibiotics In Cancer Therapy: N.R, Bachur, "Free Radical Damage" id, at pp,97~lQ2). Because of their cytotoxic potential anthracyclines have been used in the treatment of numerous cancers such as leukemia, breast carcinoma, lung carcinoma, ovarian adenocarcinoma and sarcomas {see e.g. , P.H- Wieraik, in

Anthracydine: Current Status And New Developments p 1 1).

Nonlimiting exemplar}' anthracyclines include doxorubicin, epirubicin, idarubicin, daunomyein, nemorubicin, and derivatives thereof, mmunoeonjugates and prodrugs of daunorubicin and doxorubicin have been prepared and studied (Kratz et al (2006) Current Med. Chem. 13:477-523; Jeffrey et al (2006) Bioorganic & Med. Chem. Letters 1.6:358-362; Torgov et al (2005) Bioconj. Chem. 16:717-721 ; Nagy et al (2000) Proc. Nad. Acad, Sci. USA 97:829-834; Dubowchik et al (2002) Bioorg. & Med. Chem. Letters 12: 1529-1532; King et al (2002) J. Med. Chem. 45:4336-4343; HP 0328147; US 6630579). The antibody-drug conjugate BR96-doxorubicin reacts specifically with the tumor- associated antigen Lewis- Y and has been evaluated in phase I. and II. studies (Saleh et ai (2000) J. CHn. Oncology 18:2282-2292; Ajani et al (2000) Cancer. Jour. 6:78-81 ; Tolcher et al (1999) J. Clin.

Oncology 17:478-484).

F IJ-159682 is a potent metabolite (or derivative) of nernoruhicin (Quintieri, et al. (2005) Clinical Cancer Research 11(4): 1608-1617). Nemorubicin is a semisynthetic analog of doxorubicin with a 2- 6ώο γηιο ^ιϋηο group on the glycoside amino of doxorubicin and has been under clinical evaluation (Grandi et ai (1 90) Cancer Treat. Rev, 17: 133; Ripamonti et al (1992) Brit J. Cancer 65:703; ), including phase Will trials for hepatocellular carcinoma (Sun et al (2003) Proceedings of the American Society for Clinical Oncology 22, Absl448; Quintieri (2003) Proceedings of die American Association of Cancer Research, 44: 1 st Ed, Abs 4649; Pacciarini et al (2006) Jour. CHn. Oncology 24:14116). A noniimiiiiig exemplary ADC comprising nemorabicin or nemorubicm derivatives is shown in Formula la:

wherein R| is hydrogen atom, hydroxy or methoxy group and Rj is a C C 5 alkoxy group, or a pharmaceutically acceptable salt thereof;

Lj and Z together are a linker (L) as described herein;

T is an antibody (Ab) as described herein: and

m is 1 to about 20. In some embodiments, m is 1 to 10, 1 to 7, 1 to 5, or 1 to 4.

in some embodiments, R) and R 2 are both methoxy (-QMe),

A further nonlimiting exemplary ADC comprising nen orubicin or nemorabicin derivatives is shown i Formula lb:

wherein Rj is hydrogen atom, hydroxy or methoxy group and R> is a C r C 5 alkoxy group, or a pharmaceutically acceptable salt thereof;

1.2 and Z together are a linker (L) as described herein;

T is an antibody (Ab) as described herein; and

m is 1 to about 20. In some embodiments, m is 1 to 10, 1 to 7, 1 to 5, or 1 to 4.

In some embodiments, Rj and R? are both methoxy (-QMe). In some embodiments, the nemorubicin component of a nemombicm-eontaining ADC is PNU- 159682. In some such embodiments, the drug portion of the ADC may have one of the following structures:

,0 wherein the wavy line indicates the attachment to the linker (L),

Anthracyc nes, including PNU- 159682, may be conjugated to antibodies through several linkage sites and a variety of linkers (US 201 1/0076287; WO2009/099741 ; US 2010/0034837; WO 2010/009124) , including the linkers described herein.

ut are not limited to:

PNU-l 59682-val-cit-PAB-Ab;

PNU- 159682-val-cit-PAB-spacer-Ab;

P U-159682-val-cii~FAB~spacer(R 1 R 2 )-Ab ! wherein:

i and R 2 are independently selected from H and C t -Q alkyl: and

PNU-159682-maieimide-Ab.

Tlie linker of PNU-159682 maleimide aceial-Ab is acid-labile, while the linkers of PNU- 159682-val-cit-PAB-Ab, PNU-159682-val-cit-PAB-spacer-Ab, and PNU-159682-val-cit-PAB- spacer{R ! R )-Ab are protease cleavable.

(6) Other Drug Moieties

Drug moieties also include geidanamyein (Mandler et al (2000) ./. Nat. Cancer Inst,

92(19):1573-1581 ; Mandler et al (2000) Bioorgamc & Med. Chem. Letters 10: 10254028; Mandler ei al (2002) Bioconjugate Chem, 13:786-791); and enzyniaticaily active toxins and fragments thereof, including, but not limited to, diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytoiaea arnerkiana proteins (PAPL ΡΑΡΠ, and PAP-S), momordica charantia inhibitor, eurein, cretin, sapaonaria officinalis inhibitor, gelonin, mitoge!lin, restrietoein, phenomy in, enomycin and the tricotheeenes, See, e.g., WO 93/21232.

Drug moieties also include compounds with nucieo lytic activity (e.g., a ribonuclease or a DNA endonuclease).

hi certain embodiments, an smmunoconjugate may comprise a highly radioactive atom. A variety of radioactive isotopes are available for the production of radio-conjugated antibodies, Examples include At 2U , I 13t , Γ 25 , Y 90 , Re 186 , Re m , Bi 2n , P 32 , Pb 2 ' 2 and radioactive isotopes of Lu. In some embodiments, when an inimunoconjugate is used for detection, it may comprise a radioactive atom for scintigraphic studies, for example Te" or I !"" \ or a spin label for nuclear magnetic resonance ( MR) imaging (also known as magnetic resonance imaging, MRI), such as zirconiurn-89, iodine-123, iodine- 131 , indium- 1 1 1, fluorine- 19, earbot I3, nitrogen-] 5, oxygen-17, gadolinium, manganese or iron. Zirconium~89 may be omplexed to various metai chelating agents and conjugated to antibodies, e.g., for PET imaging (WO 2011/056983).

The radio- or other labels may be incorporated in the inimunoconjugate in known ways. For example, a peptide may be biosynthesized or chemically synthesized using suitable amino acid precursors comprising, for example, one or more fluorine- 19 atoms in place of one or more hydrogens, hi some embodiments, labels such as Tc", 1 L'" \ Re' 86 , Re l s8 and In' 1 ! can be attached via a cysteine residue in the antibody. In some embodiments, yttrium-90 can be attached via a lysine residue of the antibody. In some embodiments, the IODQQEN method (Fraker et al (1 78) Biochem. Biophys. Res. Co mun. SO: 49-57 can be used to incorporate iodine- 123. "Monoclonal Antibodies in

Immunoscintigraphy" (Chatal, CRC Press 1989) describes certain other methods,

In certain embodiments, an immunoeonjugate may comprise an antibody conjugated to a prodrug-activating enzyme. In some such embodiments, a prodrug-aetivating enzyme converts a prodrug (e.g., a peptidyl chemotherapeutic agent, see WO 81/01 145) to an active drug, such as an anticancer drug. Such immunoeonjugates are useful, in some embodiments, in antibody-dependent enzyme- mediated prodrug therapy ("ADEPT"). Enzymes that may be conjugated to an antibody include, but are not limited to, alkaline phosphatases, which are useful for converting phosphate-containing prodrugs into free drugs; arylsuifatases, which are useful for converting sul fate-containing prodrugs into free drugs; cytosine deaminase, which is useful for converting non-toxic 5-fiuoroeytosine into the anticancer drug, 5-fluorouracil; proteases, such as serratia protease, ihermolysin, subtilisin,

carboxypeptidases and eathepsins (such as cathepsins B and L), which are useful for converting peptide-containing prodrugs into free drugs; Ό-alan.ylcarboxypeptidases, which are useful for converting prodrugs that contain D-amino acid substituents; carbohydrate-cleaving enzymes such as β- galactosidase and neuraminidase, which are useful for converting glycosylated prodrugs into free drugs; β-lactamase, which is useful for converting drugs derivatszed with β-lactams into free drugs; and penicillin amidases, such as penicillin V amidase and penicillin G amidase, which are useful for converting drugs derivatized at their amine nitrogens with phenoxyaeetyl or phenylaeetyl groups, respectively, into free drugs. In some embodiments, enzymes may be eovalently bound to antibodies by recombinant DNA techniques well known in the art. See, e.g. , Neuberger et al. , Nature 312:604-608 (1984).

Drug loading is represented by p, the a verage number of drug moieties per antibody in a molecule of Formula I. Drug loading may range from 1 to 20 drug moieties (D) per antibody. ADCs of Formula Ϊ include collections of antibodies conjugated with a range of dnig moieties, from 1 to 20, The average number of drug moieties per antibody in preparations of ADC from conjugation reactions may he characterized by conventional means such as mass spectroscopy, ELISA assay, and HPLC. '.The quantitative distribution of ADC in terms of p may also be determined, .m some instances, separation, purification, and characterization of homogeneous ADC where p is a certain value from ADC with other drug loadings may be achieved by means such as reverse phase HPLC or electrophoresis.

For some antibody-drug conjugates, p may be limited by the number of attachment si tes on the antibody. For example, where the attachment is a cysteine thiol, as in certain exemplary embodiments above, an antibody may have only one or several cysteine thiol groups, or may have only one or several sufficiently reactive thiol groups through which a linker may be attached. In certain embodiments, higher drug loading, e.g. p >5, may cause aggregation, insolubility, toxicity, or loss of cellular permeability of certain antibody-drug conjugates. In certain embodiments, the average dnig loading for an ADC ranges from 1 to about 8; from about 2 to about 6; or from about 3 to about 5. indeed, it has been shown that for certain ADCs, the optimal ratio of drug moieties per antibody may be less than 8, and may be about 2 to about 5 (US 7498298).

In certain embodiments, fewer than the theoretical maximum of drug moieties are conjugated to an antibody during a conjugation reaction. An antibody may contain, for example, lysine residues that do not react with the drug-linker intermediate or linker reagent, as discussed below. Generally, antibodies do not contain many free and reactive cysteine thiol groisps which may be linked to a drug moiety; indeed most cysteine thiol residues in antibodies exist as disulfide bridges. In certain embodiments, an antibody may be reduced with a reducing agent such as dithiotiireitoi (DTT) or triearbonylethylphosphme (TCEP), under partial or total reducing conditions, to generate reactive cysteine thiol groups. In certain embodiments, an antibody is subjected to denaturing conditions to reveal reactive nucleopbilic groups such as lysine or cysteine.

The loading (drug/antibody ratio) of an. ADC may be controlled in different ways, and for example, by: (i) limiting the molar excess of drug-linker intermediate or linker reagent relative to antibody, (ii) limiting the conjugation reaction time or temperature, and (iii) partial or limiting reductive conditions for cysteine thiol modification. It is to be understood that where more than one nucieophilic group reacts with a drug-linker intermediate or linker reagent, then the resulting product is a mixture of ADC compounds with a distribution of one or more drug moieties attached to an antibody. The average rrumber of drugs per antibody may be caicisiated from the mixture by a dual ELISA antibody assay, which is specific for antibody and specific for the drug. Individual ADC molecules may be identified in the mixture by mass spectroscopy and separated by HPLC, e.g. hydrophobic interaction chromatography (see, e.g.,

McDonagli et al (2006) Prot, Engr. Design & Selection 19(7):299~307; Hamblett et al (2004) Clin. Cancer Res, 10:7063-7070; Hamblett, K.J., et al. "Effect of drug loading on the pharmacology, pharmacokinetics, and toxicity of an anti-CD30 antibody-drug conjugate," Abstract No. 624, American Association for Cancer Research, 2004 Annual Meeting, March 27-31 , 2004, Proceedings of the AACR, Volume 45, March 2004; Alley, S.C., ei al. "Controlling the location of drug attachment in antibody-drug conjugates," Abstract No, 627, American Association for Cancer Research, 2004 Annual Meeting, March 27-31, 2004, Proceedings of the AACR, Volume 45, March 2004), In certain embodiments, a homogeneous ADC with a single loading value may be isolated from the conjugation mixture by electrophores

An ADC of Formula I may be prepared by several routes employing organic chemistry reactions, conditions, and reagents known to those skilled in the art, including: (1) reaction of a nucieophilic group of an antibody with a bivalent linker reagent to form Ab-L via a covalent bond, followed by reaction with a drug moiety D; and (2) reaction of a nucieophilic group of a drug moiety with a bivalent linker reagent, to form D-L, via a covalent bond, followed by reaction with a nucieophilic group of an antibody. Exemplary methods for preparing an ADC of Formula I via the latter route are described in US 7498298, which is expressly incorporated herein by reference.

Nucieophilic groups on antibodies include, but are not limited to: (i) N-terminal amine groups, (ii) side chain amine groups, e.g. lysine, (iii) side chain thiol groups, e.g. cysteine, and (iv) sugar hydroxy! or amino groups where the antibody is glycosylated. Amine, thiol, and hydroxy! groups are nucieophilic and capable of reacting to form covalent bonds with electrophilic groups on linker moieties and linker reagents including: (i) active esters such as NHS esters, HOBt esters, haloformates, and acid halides; (ii) alkyl and benzyl halides such as haioaeetamides; and (iii) aldehydes, ketones, carboxyi, and maieimide groups. Certain antibodies have reducible interchain disulfides, i.e. cysteine bridges. Antibodies may be made reactive for conjugation with linker reagents by treatment with a reducing agent such as DTT (dithiothreitoi) or tricarbonylethylphosphine (TCEP), such that the antibody is fully or partially reduced. Each cysteine bridge will thus form, theoretically, two reactive thiol nucleophiles. Additional nucieophilic groups can be introduced into antibodies tlirough modification of lysine residues, e.g., by reacting lysine residues with 2-iminothiolane (Traut's reagent), resulting in conversion of an amine into a thiol. Reactive thiol groups may also be introduced into an antibody by introducing one, two, three, four, or more cysteine residues (e.g., by preparing variant antibodies comprising one or more non-native cysteine amino acid residues).

Antibody-drug conjugates provided herein may also be produced by reaction between an eiectrophilic group on an antibody, such as an aldehyde or ketone carbonyl group, with a nucleophiiic group on a linker reagent or drug. Useful nucleophiiic groups on a linker reagent include, but are not limited to, hydrazide, oxime, amino, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide. In one embodiment, an antibody is modified to introduce eleeirophiiic moieties that are capable of reacting with nucleophiiic substituents on the linker reagent or drug. In another embodiment, the sugars of glycosylated antibodies may be oxidized, e.g. with periodate oxidizing reagents, to forra aldehyde or ketone groups which may react with the amine group of linker reagents or drug moieties. The resulting inline Sc.hiff base groups may form a stable linkage, or may be reduced, e.g. by borohydride reagents to form stable amine linkages. In one embodiment, reaction of the carbohydrate portion of a glycosylated antibody with either galactose oxidase or sodium meta-periodate may yield carbonyl (aldehyde and ketone) groups in die antibody that can react with appropriate groups on the drug (Hermanson, Bioeonjugate Techniques), In another embodiment, antibodies containing N-terrninal serine or threonine residues can react with sodium meta-periodate, resulting in production of an aldehyde in place of the first amino acid (Geoghegan & Stroh, (1992) Bioeonjugate Chem. 3: 138-146; US 5362852), Such an aldehyde can be reacted with a drug moiety or linker nueleophiie.

Exemplary nucleophiiic groups on a drug moiety include, but are not limited to: amine, thiol, hydroxyl, hydrazide, oxime, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide groups capable of reacting to form eovalent bonds with eiectrophilic groups on linker moieties and linker reagents including: (i) active esters such as NHS esters, HOBt esters, haloformates, and acid halides; (ii) alkyl and benzyl halides such as haloacetamides; (iii) aldehydes, ketones, carboxyl, and maleixni.de groups.

Nonlimiting exemplary cross-linker reagents that may be used to prepare ADC are described herein in the section titled "ExernpLary Linkers." Methods of using such cross-linker reagents to link two moieties, including a proteinaceous moiety and a chemical moiety are known in the art. In some embodiments, a fusion protein comprising an antibody and a cytotoxic agent may be made, e.g., by recombinant, techniques or peptide synthesis, A recombinant DNA molecule may comprise regions encoding the antibody and cytotoxic portions of the conjugate either adjacent to one another or separated by a region encoding a linker peptide which does not destroy the desired properties of the conjugate.

n yet another embodiment, an antibody may be conjugated to a "receptor'" (such as streptavidin) for utilization in tumor pre-targeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand" (e.g., avidin) which is conjugated to a cytotoxic agent (e.g., a drug or radionucieotide). E. Methods aad Cempositioss for Diagnostics ass t Detection

In certain embodiments, any of the anti-Fc H5 antibodies provided herein is useful for detecting the presence of FcRHS (e.g., FcRHS } in a biological sample. The terra "detecting" as used herein encompasses quantitative or qualitative detection, hi certain embodiments, a biological sample comprises a ceil or tissue. In certain embodiments, such tissues include normal and/or cancerous tissues that express FcRHS at higher levels relative to other tissues, for example, B-eells and/or B-celi associated tissues. In some embodiments, the anti-FcRHS antibody binds an isofonn e-speeific region of the extracellular domain of FcRHSc. In some embodiments, the anti-FcRHS antibodies binds Ig~like domain 9 of FcRHSc.

In one aspect, provided herein are methods of detecting the presence of FcRHS in a biological sample. In certain embodiments, the method comprises contacting the biological sample with an anti- FcRHS antibody under conditions permissive for binding of the anti-FcRHS antibody to FcRHS, and detecting whether a eompiex is formed between the anti~FeRH5 antibody and FcRHS. in one aspect, the invention provides a method of diagnosing a disorder associated with increased expression of FcRHS. In certain embodiments, the method comprises contacting a test cell with an anti-FcRHS antibody; determining the level of expression (either quantitatively or qualitatively) of FcRHS by the test cell by detecting binding of the anti-FcRHS antibody to FcRHS; and comparing the level of expression of FcRHS by the test cell with the level of expression of FcRHS by a control cell (e.g., a normai cell of the same tissue origin as the test cell or a cell that expresses FcRHS at levels comparable to such a normal cell), wherein a higher level of expression of FcRHS by the test cell as compared to the control cell indicates the presence of a disorder associated with increased expression of FcRHS, In certain embodiments, the test cell is obtained from an individual suspected of having a disorder associated with increased expression of FcRHS. In certain embodiments, the disorder is a ceii proliferative disorder, such as a cancer or a rumor. In some embodiments, the FcRHS is FcRHSc. In some embodiments, the anti-FcRHS antibody binds an isofo m c-specifie region of the extracellular domain of FcRHSc. In some embodiments, the anti-FcRHS antibodies binds Ig-iike domain 9 of FcRHSc.

Exemplary cell proliferative disorders that may be diagnosed using an antibody described herein include a B-cell disorder and/or a B-cell proliferative disorder including, but not limited to, lymphoma, multiple myeloma non-Hodgkin's lymphoma (NHL), aggressive NHL, relapsed aggressive NHL, relapsed indolent NHL, refractory NHL, refractory indolent NHL, chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma, leukemia, hairy cell leukemia (HCL), acute lymphocytic leukemia (ALL), and mantle cell lymphoma.

In one embodiment, an anti-FcRHS an tibody for use in a method of diagnosis or detection is provided. In a further aspect, a method of detecting the presence of FcRHS in a biological sample is provided, in certain embodiments, the method comprises contacting the biological sample with an anti- FcRHS antibody as described herein under conditions permissive for binding of the anti-FcRHS antibody to FcRHS, and detecting whether a eompiex is formed between the an†.i~FcRH5 antibody and FcRH5 in the biological sample. Such method may be an in vitro or in vivo method, In one embodiment, an anti-FcRJ-15 antibody is used to select subjects eligible for therapy with an anti-FcRHS antibody, e.g. where FcRHS is a biomarker for selection of patients. In a further embodiment, the biological sample is a cell or tissue (e.g., biopsy material). In some embodiments, the and -FcRHS antibody binds an isoform c-speciftc region of the extracellular domain of FeRHSc, In some

embodiments, the anti~FcRH5 antibodies binds Ig-iike domain 9 of FeRHSc.

In a further embodiment, an ami -FcRHS antibody is used in vivo to detect, e.g., by in vivo imaging, an FcRHS -positive cancer in a subject, e.g., for the purposes of diagnosing, prognosing, or staging cancer, determining the appropriate course of therapy, or monitoring response of a cancer to therapy. One method known in the art for in vivo detection is immuno-positron emission tomography (immuno-PET). as described, e.g., in van Dongen et al. The Oncologist 12:1379-1389 (2007) and Verel et a!., J. NucL Med. 44:1271 -1281 (2003). In such embodiments, a method is provided for detecting an FcRHS -positive cancer in a subject, the method comprising administering a labeled anti-FcRHS antibody to a subject having or suspected of having an FcRHS-positive cancer, and detecting the labeled anti-FeRH5 antibody in the subject, wherein detection of the labeled anti-FcRHS antibody indicates an FcRHS -positive cancer in the subject. In certain of such embodiments, the labeled anti- FcRHS antibody comprises an anti-FcRHS antibody conjugated to a positron emitter, such as 6 *Ga, ' 8 F, 6 Cu, 86 Y, 76 Br, 89 Zr, and i24 l. In a particular embodiment, the positron emitter is 89 Zr. In some embodiments, the anti-FcRHS antibody binds an isoform e-speeifk region of the extracellular domain of FcRHS c. In some embodiments, the anti-FcRHS antibodies binds ig-like domain 9 of FeRHSc.

In further embodiments, a method of diagnosis or detection comprises contacting a first anii- FcRHS antibody immobilized to a substrate with a biological sample to be tested for the presence of FcRHS, exposing the substrate to a second anti-FcRHS antibody, and detecting whether the second anti- FeRHS is bound to a complex between the first anti-FcRHS antibody and FcRHS in the biological sample. A substrate may be any supportive medium, e.g., glass, metal, ceramic, polymeric beads, slides, chips, and other substrates, in certain embodiments, a biological sample comprises a cell, blood, or tissue (e.g. , biopsy material)

Exemplary disorders that may be diagnosed or detected according to any of the above embodiments include FcRHS-positive cancers, such as FcRH -positive B-cell proliferative disease, FcRHS-positive plasma cell neoplasm, and FcRHS-positive multiple myeloma. In some embodiments, an FcRHS-positive cancer is detected by anti-FcRHS immunohistochemistry (IHC) or in situ hybridization (ISI-I). In some embodiments, an FcRHS-positive cancer is a cancer that expresses FcRHS according to a reverse- transcriptase PGR (RI ' -PCR) assay that detects FcRH5 mRNA. hi some embodiments, the RI ' -PCR is quantitative RT-PCR.

In certain embodiments, labeled anti-FcRFIS antibodies are provided. In some embodiments, the anti-FcRHS antibody binds an isoform c-speciftc region of the extracellular domain of FeRHSc. In some embodiments, the anti-FcRHS antibodies binds ig-iike domain 9 of FeRHSc. Labels include, but are not limited to, labels or moieties that are detected directly (such as fluorescent, chxornophoric, electron-dense, chemiluminescent, and radioactive labels), as well as moieties, such as enzymes or ligands, that are detected indirectly, e.g., through an enzymatic reaction or molecular interaction, Exemplary labels include, but are not limited to, the radioisotopes 5J P, l C, 2 % 3 H, and 131 1, fluorophores such as rare earth chelates or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbeihferone, lueiferases, e.g., firefly luciferase and bacterial luciferase (U.S. Patent No.

4,737,456), iuciferiti, 2,3-dihydrophthalazinediones, horseradish peroxidase (H P), alkaline phosphatase, p-galaetosidase, glucoamylase, lysozyme, saccharide oxidases, e.g., glucose oxidase, galactose oxidase, and glucose~6~phospha†e dehydrogenase, heterocyclic oxidases such as uncase and xanthine oxidase, coupled with an eiizyme that employs hydrogen peroxide to oxidize a dye precursor such as HRP, iaeioperoxidase, or rnieroperoxidase, hiotin/avidin, spin labels, bacteriophage labels, stable free radicals, and the like. In another embodiment, a label is a positron emitter. Positron emitters include but are not limited to 6" Ga. , 8 F, 64 Cu, 86 Y, 75 Br, ~3 Zr, and l2 "l. In a particular embodiment, a positron emitter is Zr.

F. Pharmaceutical Foriuulatiosis

Pharmaceutical formuiaiions of an anti~FcRH5 antibody or immunoeonjugate as described herein are prepared by mixing such antibody or immunoeonjugate having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Remington's Pharmaceutical Sciences 16th edition, Osoi, A, Ed, (3980)), in the form of lyophilized formulations or aqueous solutions.

Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and

concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as oetadeeyldimethyibenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride;

benzethonium chloride; phenol, butyl or benzyl alcohol: alkyl parabens sisch as methyl or propyl paraben; catechol; resorcinol; cyciohexanoi; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins;

hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutarnine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, rnannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn~ protein complexes): and/or non-ionic surfactants such as polyethylene glycol (PEG), Exemplary pharmaceutically acceptable carriers herein further include msterstifial drug dispersion agents such as soluble neutral-active hyaiuronidase glycoproteins (sHASEGP), for example, human soluble PH-20 hyaiuronidase glycoproteins, such as rIIuPH20 (HYLENEX"", Baxter International, Inc.). Certain exemplary sIlASEGPs and methods of use, including rHuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968, In one aspect, a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases. In some embodiments, the anti~FcRH5 antibody binds an isoform c-specific region of the extracellular domain of FcRH5e, In some embodiments, the anti-FcRHS antibodies binds Ig-like domain 9 of FcRHSc,

Exemplary iyophilized aniibody or immunoconjugate formulations are described in US Patent No, 6,267,958, Aqueous antibody or immunoconjugate formulations include those described in LIS Patent No. 6,171 ,586 and WO2006/044908, the latter formulations including a histidme-aeetate buffer.

The formulation herein may also contain more than one active ingredient as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.

Active ingredients may be entrapped in microcapsules prepared, for example, by eoaeervation techniques or by interfacial polymerization, for example, hydroxymethyiceliulose or gelatin- microcapsules and poiy-(methylmethaeylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemuisions, nano-particles and nanocapsulea) or in maeroemulsions. Such tec miques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).

Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the aniibody and/or immunoconjugate, which matrices are in the form of shaped articles, e.g. films, or

microcapsules.

The formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.

G, Therapesitic Methods ass Compositions

Any of the anti-FcRH5 antibodies (e.g., FcRIIS bispecific antibodies) and/or

im.munoconju gates provided herein may be used in methods, e.g., therapeutic methods. In some embodiments, the anti -FcRHS antibody binds an isoform c-specific region of the extracellular domain of FcRHSc. In some embodiments, the anti-FcRHS antibodies binds ig-like domain 9 of FcRHSc.

In one aspect, an anti-FcRHS antibody (e.g., FcRHS bispecific antibody) and/or

immunoconjugate provided herein is used in a method of inhibiting proliferation of an FcRH5~positive ceil, the mediod comprising exposing the ceil to the anti-FcRHS antibody (e.g., FcRHS bispecific antibody) and/or immunoconjugate under conditions permissive for binding of the anti-FcRHS antibody (e.g., FcRHS bispecific antibody) and/or hrmiunoconjugate to FcRHS (e.g., FcRHSc) on the surface of the ceil, thereby inhibiting the proliferation of the cell. In certain embodiments, the method is an in vitro or an in vivo method. In further embodiments, the ceil is a B-cel! proliferative disorder. In certain embodiments, the cell proliferative disorder is associated with increased expression and/or activity of FcRHS (e.g., FcRHSc). For example, in certain embodiments, lite B-celi proliferative disorder is associated with increased expression of FcRHS on the surface of a B-cell. In certain embodiments, the B-cell proliferative disorder is a tumor or a cancer. In some embodiments, the B-cell proliferative disorder is a plasma cell neoplasm, In some embodiments, the plasma cell neoplasm is multiple myeloma, plasmacytoma, and/or MGUS, Examples of B-cell proliferative disorders to be treated by the antibodies and/or imrnunoconiugates of the invention include, but are not limited to, lymphoma, multiple myelomanon-H.odgkms lymphoma (NHL), aggressive NHL, relapsed aggressive NHL, relapsed indolent NHL, refractory NHL, refractory indolent NHL, chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma, leukemia, hairy cell leukemia (HC.L), acute lymphocytic leukemia (ALL), and/or mantle cell lymphoma.

Presence of various biomarkers in a sample can be analyzed by a number of methodologies, many of which are known in the art and understood by the skilied artisan, including, but not limited to, i munohistochemistry ("IHC"), Western blot analysis, immunoprecipitation, molecular binding assays, ELISA, BLIP A, fluorescence activated cell sorting ("FACS"), MassARRAY, proteomies, quantitative blood based assays (as for example Serum ELISA), biochemical enzymatic activity assays, in situ hybridization, Southern analysis, Northern analysis, whole genome sequencing, polymerase chain reaction ("PCR") including quantitative real time PCR ("qRT-PCR") and other amplification type detectio methods, such as, for example, branched DNA, SISBA, TMA arid the like, RNA-Seq, FISH, microarray analysis, gene expression profiling, and/or serial analysis of gene expression ("SAGE"), as well as any one of the wide variety of assays that can be performed by protein, gene, and/or tissue array analysis. Typical protocols for evaluating the status of genes and gene products are found, for example i Ausubel et al, eds., 1995, Current Protocols In Molecular Biology, Units 2 (Northern Blotting), 4 (Southern Blotting), 15 (irnmunoblotting) and 18 (PCR Analysis). Multiplexed immunoassays such as those available from Rules Based Medicine or Meso Scale Discovery ("MSD") may also be used.

Inhibi tion of cell proliferation in vitro may be assayed using the CellTiter-Gio™ Luminescent Cell Viability Assay, which is commercially available from Promega (Madison, WI). That assay determines the number of viable ceils in culture based on quantitation of ATP present, which is an indication of metabolically active cells. See Crouch et al. (1993) J. Immunol, Meth. 1 0:81-88, US Pat, No. 6602677. The assay may be conducted in 96- or 384~well format, making it amenable to automated high-throughput screening (HTS). See Cree et al (1995) Anticancer Drugs 6:398-404. The assay procedure involves adding a single reagent (CellTiter-Gio 30 Reagent) directly to cultured cells. This results in cell lysis and generation of a luminescent signal produced by a iuciferase reaction. The luminescent signal is proportional to the amount of ATP present, which is directly proportional to the number of viable cells present in culture. Data can be recorded by luminometer or CCD camera imaging device, The luminescence output is expressed as relative light units (RLU).

in another aspect, an anti-FcRHS antibody (e.g., FcRH5 bispecific antibody) and/or immunoconjugate for use as a medicament is provided. In further aspects, an anti-FcRHS antibody (e.g., FcRHS bispecific antibody) and/or immunoconjugate for use in a method of treatment is provided, In certain embodiments, an anti-FcRI15 antibody (e.g., FcRHS bispecific antibody) and/or immunoconjugate for use in treating FcRHS (e.g,, FcRH5e)-positive cancer is provided, hi certain embodiments, provided herein the anti-FcRHS antibody (including FcRHS bispecific antibody) and/or irnmunoeonjugate for use in a method of treating an individual having an FcRHS (e.g., FcRHSc)- positive cancer, die method comprising administering to the individual an effective amount of the anti- FcRH5 antibody and/or immunoconjugaie. in some embodiments, the anti~FcRH5 antibody binds an isoform c-speeifie region of the extracellular domain of FcRHSc. In some embodiments, the anti- FcRHS antibodies binds Ig-iike domain 9 of FcRH5c. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below.

in a further aspect, provided herein are uses of an anti-FcRH5 antibody (e.g.. FcRMS bispeciiic antibody) and/or immunoconjugaie in the manufacture or preparation of a medicament. In one embodiment, the medicament is for treatment of FcRHS (e.g., FcRH5c)-positive cancer. In a further embodiment, the medicament is for use in a method of treating FcRHS (e.g., FcRHSeVpositive cancer, the method comprising administering to an individual having FcRHS (e.g., FcRH5 expositive cancer an effective amount of the medicament. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below. In some embodiments, the anti-FcRH5 antibody binds an isoform c-specifie region of the extracellular domain of FcRHSc. In some embodiments, the anti-FcRH5 antibodies binds Ig-like domain 9 of FcRHSc.

In a further aspect, provided herein are methods for treating FcRHS (e.g., FcRH5e)-positive cancer, fa. one embodiment, the method comprises administering to an mdividuai having such FcRHS (e.g., FcRH5c)-positive cancer an effecti ve amount of an anti-FcRHS antibody (e.g., FcRHS bispeciiic antibody) and/or immunoconjugaie. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, as described below. . In some embodiments, the anti-Fe.RH5 antibody binds an isoform c~specifi.e region of the extracellular domain of FcRHSc. In some embodiments, the anti-FeRH5 antibodies binds Ig-iike domain 9 of FcRHSc.

An FcRH5-positive cancer according to any of the above embodiments may be, e.g., FcRHS - positive B~ceil proliferative disorder, FcRH5-positive plasma ceil neoplasm, and/or FcRHS -positive multiple myeloma. In some embodiments, an FcRHS-positive cancer is detected by anti-FcRHS immunohistochemistry (EE-IC) or in situ hybridization (ISH). In some embodiments, an FcRHS-positive cancer is a cancer that expresses FcRHS according to a reverse-transeriptase PCR (RT-PCR) assay that detects FcRHS mRNA, In some embodiments, the RT-PCR is quantitative RT-PCR.

In some embodiments of any of the above embodiments, the individual may be a human.

In a further aspect, provided herein are pharmaceutical formulations comprising any of the anti- FcRHS antibodies and'or immunoconjugaie provided herein, e.g., for use in any of the above therapeutic methods. In one embodiment, a pharmaceutical formulation comprises any of the anti- FcRHS antibodies (e.g., bispeeifie antibodies) and/or immunoconjugates provided herein and a pharmaceutically acceptable carrier. In another embodiment, a pharmaceutical formulation comprises any of the anti-FeRHS antibodies (e.g., bispecific antibodies) and/or irnmunoconjugates provided herein and at least one additional therapeutic agent, e.g., as described below.

Antibodies (e.g., bispecific antibodies) and/or irnmunoconjugates provided herein can be used either alone or in combination with other agents in a therapy. Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the antibody or immunoeonjugate provided herein can occur prior to, simultaneously, and or following, administration of the additional therapeutic agent and/or adjuvant. Antibodies and/or irnmunoconjugates provided herein can also be used in combination with radiation therapy.

An antibody (including bispecific antibody) and/or un unoeonjugate provided herein (and any additional therapeutic agent) can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesiona! administration, Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic. Various dosing schedules including but not limited to single or multiple administrations over various time- points, bolus administration, and pulse infusion are contemplated herein.

Antibodies (e.g., bispecific antibodies) and/or irnmunoconjugates provided herein would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The antibody (e.g., bispecific antibodies) and/or immunoeonjugate need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody or

immunoeonjugate present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.

For the prevention or treatment of disease, the appropriate dosage of an antibody (e.g., bispecific antibody) and/or immunoeonjugate provided herein (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the type of antibody or immunoeonjugate, the severity and course of the disease, whether the antibody (e.g., bispecific antibody) and/or immunoeonjugate is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody or

immunoeonjugate, and the discretion of the attending physician. The antibody (e.g., bispecific antibody) and/or immunoeonjugate are suitably administered to the patient at one tune or over a series of treatments. Depending on the type and severity of the disease, about 1 &'¾ to 15 mg/kg (e.g.

O. lmg kg-lOmg kg) of antibody or immunoconjugate can be an initial candidate dosage for

administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 μ§ ¾¾ to 100 mg kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs. One exemplary dosage of the antibody (e.g., bispecific antibody) and/or immunoconjugate would be in the range from about 0.05 mg/kg to about 10 mg/kg, Thus, one or more doses of about 0,5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient. Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about sis doses of the antibody). An initial higher loading dose, followed by one or more lower doses may be administered. However, other dosage regimens may be useful The progress of this therapy is easily monitored by conventional techniques and assays.

it is understood thai any of the above formulations or therapeutic methods may be carried out using both art immunoconjugate provided herein and an anti~FcRH5 antibody, In some embodiments, the anti~FcRH5 antibody binds an isoform c-speeific region of the extracellular domain of FeRH5c. Irt some embodiments, the anti-FeRH5 antibodies binds Ig-like domain 9 of FcRH5e.

II. Articles of Mamrfactsjre

In another aspect provided herein, an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided, The article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the disorder and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is an antibody or immunoconjugate provided herein. The label or package insert indicates that the composition is used for treating the condition of choice, Moreover, the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an FcRHS antibody {e.g., bispecific antibody) and/or Fc.RH5 immunoconjugate provided herein: and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent. The article of manufacture in this embodiment provided herein may further comprise a package insert indicating that the compositions can be used to treat a particular condition. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI). phosphate-buffered saline, Ringer's solution or dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes. In some embodiments, the anti-FcRK5 antibody binds an isoform c-specific region of the extracellular domain of FcRHSe. in some embodiments, the anti-FcRH5 antibodies binds Ig-like domain 9 of FcRH5e,

III. EXAMPLES

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 above.

Materials end Methods

Immunogen (El 1 -flag)

Amino acids 745-850 of human FcRH5e (SEQ ID NOT) were cloned into mammalian expression vector pRK5.NT.Flag using standard protocols and expressed transiently in CHO cells. The recombinant protein with N-terminal Flag-expression tag was purified using anti-flag and size exclusion chromatography on an S200 Superdex column.

Development and characterization of mouse anti-FcRHS Ell antibodies

Balb/c mice (Charles River, Hollister, CA) were immunized with 2 μg human FcRlIS El 1 ECD protein (amino acid residues 743-850 of SEQ ID NO: l) (Genentech, South San Francisco. CA) mixed with MPL+TDM (Ribi) adjuvant via footpad injection. Mice received nine doses, followed by a prefusion boost in PBS alone via footpad and IV routes three days prior to fusion.

Popliteal lymph nodes were harvested and lymphocytes from these mice, ail of whose sera demonstrated strong binding titers to the immunisation protein by ELISA and showed strong FACS reactivity to SVT2 cells transfected with the human FcRH5 El 1 ECD, were fused with X63~Ag8.653 mouse myeloma ceils (American Type Culture Collection. Rockville, MD) via electrofusion (Harvard Apparatus, Holliston, MA). Fused cells were incubated at 37°C, 7% CO;, overnight in Medium C (StemCell Technologies, Vancouver, BC, Canada), before resuspension in semi-solid Medium D (StemCell Technologies) containing 0,01 mg/mi FfTC labeled anti-mouse IgG (Jackson

ImmunoReseareh, West Grove, PA) and plating into Omniwell trays (Thermo Fisher Scientific, Rochester, NY). Nine days after plating, fluorescent colonies were seiected and transferred into 96-w l plates containing Medium E (StemCell Technologies) using a Clonepix FL (Genetix, New Milton, Hampshire, UK). Supematants were screened by ELISA against anti-mouse IgG (MP Biomedicals, Santa Ana, CA) seven days after picking,

Hybridomas demonstrating mouse IgG expression by ELISA were expanded and screened by FACS against SVT2 cells overexpressing full-length human FcRHS, cyno FcRH5, and human FcRH5 El 1 ECD. Strong FACS positive clones were subcioned by single-ceil sorting using a FACSAria (BD S Franklin Fakes, NJ), Final clones demonstrating the highest ELISA and FACS binding of interest after one or two rounds of subcloning were expanded for large-scale production in bioreactors (Integra Biosciences, Chur, Switzerland). Supematants were then purified by Protein A affinity chromatography as previously described (Hongo et al, 2000). Production of bisFabs

BisFabs were generated by crosslinking a Fab' of the anti~FcRH5 Mab to a Fab" of the anti- CD3 (UCH.T1 ,v9) Mab at the hinge cysteine residues, To generate the Fab' 2 fragments from the hybridoma Abs different digestion conditions were used: Abs of the mlgGl isotype were digested with 1 :50 (w/w) pepsin at pH 3.5 for 1-2 H at 37°C; mouse IgG2a Abs were digested with Lysin C endopeptidase at 1 1 ;500 (w/w) ratio, pH 8, for 2-4 h at 37°C; and mouse IgG2b Abs were digested with Lysin C at a 1 :100 (w/w) ratio overnight at 37°C. in all eases the F( b 1 )2 fragment was isolated from the reaction mixture by capture with a SP column and elution with 10 column volumes of a lineal gradient (0-100%) of 1 M sodium chloride. Under the digestion conditions mentioned above mlgGl and rrjJgG2b produced a F^ab 1 )? fragment containing three Cysteine residues in the hinge, while the F(ab')2 from mIgG2a showed two cysteine residues in the hinge. To generate Fab' with a single reactive Cys two different methods were used. For fragments containing an odd (3) number of hinge cysteines (mlgG l and migG2h) the isolated F(ab')2s were reduced in 25 mM sodium acetate, pH 5. 150 mM sodium chloride, 2 mM EDTA, 2 mM TCEP for 2-6-H at T. After the reduction step was complete, the sample was diluted to 0,2 mg ml, the pH was raised to 7.5 by adding Tris pH 8 and 5 mM dehydroascorbie acid (DIIAA) was added to drive re-oxidation of the cysteines. After an overnight incubation at room temperature the presence of reduced Thiols was evaluated by probing with an excess of NEM and analyzing the MW shift by mass spectrometry. After confirming tiie presence of only one reactive Cysteine per molecule, the Fab' was purified by gel filtration to remove small amounts of homodimers.

For F(ab')2 fragments derived from mIgG2a and containing 2 Cysteine residues in the hinge, a single reactive Cysteine was produced by partial blocking with N-ethyl maieimide (NEM) as described in Seheer et ai (in press). Briefly, the antibody was digested with pepsin (1 % w/w) by treatment in sodium acetate buffer at pH 4.5. After digestion for 1 hour, the F(ab'¾ was isolated from the digestion mixture by capture on an SP-HP cation exchange resin and purified by a 10 CV salt gradient of 0-1 M NaCl. The F(ab' )2 was then reduced with 1 mM TCEP in a buffer containing 25 mm MES, pH 5.8, 2 mM EDTA, and 300 mM NaCl and the Fabs were oxidized by the addition of 5 mM dehydroaeorbic acid (DHAA) to reform tlie disulfide bond, between the heavy chain and light chain.

' Fl e effector arm of the bisfabs (UCHT1.v9) was generated by pepsin digestion, partial NEM blocking and conjugation to bismaieirnide as described before (Seheer et al; in press), Briefly, the two thiols (cys residues) at the hinge were then reacted with 1 equivalent of N-e&ylmaleirnide (NEM) (Sigma Aldrieh). Tlie different anti-FCRH5 Fab's containing a single reactive Cysteine were incubated with tlie anti-CD 3 Fab * conjugated to the bismaieirnide crosshnker overnight at room temperature, Tlie -lOOkDa crosslinked Fabs were separated from tlie unreacted species by gel filtration and then characterized by SDS-PAGE, mass spectrometry and analytical size exclusion chromatography. TDB expression and purification

TDBs were produced by two different approaches: co-culture of bacteria expressing each of the two antibody am s or by expressing each ami seoarately and then anrteling them in vitro. The strategies have been described in Christoph Spiess et al. 2012 and described in PCT/US10/58958 filed on 31 May 20 i 1 , which is incorporated by reference. Breifiy for the coeulture strategy E. coli expressing anti-CD3 (hole) and Ecoii expressing anti-tumor target (knob) were grown together in shaker flasks at a predetermined ratio such that it produced similar amounts o feach hemimer. ' H e coeultured bacterial broth was then harvested, the cells disrupted in a microtluidizer and the antibodies purified by Protein A affinity, it has been observed that during n icrofluidizing and protein A capture the two arms annealed and formed the hing inter-chain disulfide bridges (Christoph Spiess et al, 2012). Alternatively, the antibody hemimers were grown separately by high-ceil density fermentation and independently isolated by Protein A chromatography. I ' he purified hemimers were than combined at a 1 : 1 molar ration and inciubated in 50 niM Tris, pH 8.5 in the presence of 2 mM DTT for 4 hours to allow annealing and the reduction of disulfides in the hing region. Dialysis against the same buffer without DTT for 24-48 hours resulted in the formation of the inter-chain disulfide bonds, For both production strategies the bispecific antibody was purified from contaminants by hydrophobic interaction chromatography (HIC) as described in Christoph Spiess et ai. 2012. The resulting material was analyzed for endotoxin levels using an Endosafe protabie etest system and when needed, the endotoxin content was reduced by washing the protein with 0.1 % Triton X-l 14.

TDB characterization

The molecular weight of the bispecific antibody was analyzed by mass spectrometry (LC- ES17TCF) as described before (Jackrnan et al, 2010). The antibodies were also analyzed by analytical size exclusion chromatography in Zenix SEC-300 column (Sepax Technologies USA) using an Agilent 1 : 100 HFLC system. The presence of residual antibody fragments was quantified by electrophoresis using a 21.00 Bioanaiyzer and a Protein 230 Chip,

Blood ceil fractionation

PBMCs were separated from the blood of healthy volunteers using lymphocyte separation medium (MP biomedicals, Solon, OH), CD8+ cells were extracted from PBMC using human CD8+ Isolation Kit from Miltenyi (#130-094-156) by negative selection.

In vitro cytotoxicity assays (T ceil killing)

For in vitro cytotoxicity assays I lO 4 target ceils were plated on 96 well plates and incubated overnight. 3x10 4 CDS - T-ce!ls were added with or without TDB or BisFab and incubated 48 hours in +37 e C. T cells were removed by washing twice with growth media, Cell viability was measured using CellTiter-Glo® Luminescent Cell Viability Assay (Promega, Madison, WI).

Alternatively, in vitro cytotoxicity was monitored by flow cytometry. Target cells were labeled with carhoxyfluorescein suecinimidyl ester (CFSE) according to manufacturer's protocol (Invitrogen, #C34554), The CFSE -labeled target ceils and purified CD8+ T cells from human PBMC were mixed in 3:1 E:T ratio and incubated with TDB or BisFab for 48 hours. At the end of the incubation, the cells were lifted by trypsin and collected from the plate, The cells were resuspended in equal volume of PBS ÷ 2% FBS + 1 mM EOT A + propidfum iodine (PI). Flow cytometry analysis was done on a

FACSCalibur in automation format The number of live target cells was counted by gating on

CFSE+/PI negative cells. The percentage of cytotoxicity was calculated as follows: % cytotoxicity (live target cell number w/o TDB - live target cell number w/TDB) / (live target ceil number w/o TDB) x 100,

Analysis of T cell activation

Target cells and purified CD8+ T ceils were mixed in the presence or absence of TDB and T cell activation was analyzed by flow cytometry. At the end of the incubation, cells were stained with CD8-FITC (BD Biosciences, 555634) and CD69-PE (BD Biosciences, 555531),

Binding of subclone supernatants, monoclonal antibodies, bisFabs and TDBs

To test binding to endogenousiy FcRHS expressing cancer cells or FcRH5 transfected cancer cells, cells were lifted using EDTA/PBS. IxlO 5 cells were suspended in 100 ul and incubated h with primary antibodies (1 volume of non-IgG quantitated subclone supernatant, 4 ug ' mi IgG quantified subclone supernatant or 2ug/ul purified monoclonal antibodies), Cells were washed twice with FACS buffer (PBS 1 %BSA 2mM EDTA) and incubated with 1 : 1000 dilution of goat anti-mouse secondary labeled with PE or 1 : 100 of goat anti-mouse APC. Ceils were washed twice with FACS buffer and Flow cytometry analysis was done on a FACSCalibur. Direct Xenon-labeling of antibodies was done according to manufacturer's protocol (Invitrogen), when indicated, To analyze binding to NK or B cells, 1 million human PBM.C were incubated with 4 ug ml IgG quantified subclone supernatants for 60 min, washed and incubated with 1 : 100 dilution of goat anti-mouse secondary labeled with APC. Ceils were then washed again twice axid stained using artti~CD56 (FE; BD Biosciences #555516) and anfi- CD19 (PE; BD Biosciences #340364) prior flow cytometry and analysis of binding to human CD56+ and GDI 9÷ ceils.

Results

Initially to produce isoform specific antibodies for the membrane proximal ig-domain, mice were immunized with recombinant baeulovirus produced El. 1 protein (amino acids 745-848 of SEQ ID NO: l) of human FcRHSc and C -terminal His-expression tag). This immunization strategy did not result to significant immune response to FcRHS and failed to produce monoclonal anti-FcRH5 antibodies. The second immunization strategy was DNA-immunization with plasmid encoding amino acids 745- 977 of FcRHSc (SEQ ID NO:l) encoding membrane proximal ig-domain, transmembrane domain and intracellular domains of human FcRHS, This immunization strategy did not result to significant immune response to FcRHS and failed to produce monoclonal anti-FcRHS antibodies, The third immunization strategy utilized peptides corresponding to membrane proximal Ig-domain of FcRHS, that were homologous to cyno FcRHS and non-homologous to human FcRHl , FcRH2, FcRHS . and FcRH4, This- immunization strategy did not result to significant immune response to FcRHS and failed to produce monoclonal anti-FcRH5 antibodies.

For the fourth immunization strategy Ei 1 protein was produced in CHO-cells consisting of the membrane proximal ig-domaia of human FcRHS (amino acid residues 745-850 of SEQ ID NO:I) with N-tenninal Flag expression tag. The above recombinant protein was used to immunize mice, immunization, development and characterization of mouse anti-FcRRS El 1 antibodies was performed as described in detail above.

After 6 doses of the recombinant El 1 (ammo acid residues 745-850 of SEQ ID NO:l ), serum was analyzed for FcRHS binding antibodies using FACS. Significant reactivity was detected to SVT2 ceils that express human full length FcRHS, cyno full length FcRHS, or the human El 1 domain transmembrane domain and cytoplasmic domains but not vector transfected SVT2 cells indicating that FcRHS reactive antibodies were present in the sera of all 5 immunized mice,

After 9 doses, lymphocytes from the immunized mice were electro fused with X63~Ag8.653 mouse myeloma cells. 323 IgG positive hybridoma subclones were selected for further screening, Clones were tested for binding to recombinant El 1 protein (amino acid residues 745-850 of SEQ I ' D NO: l ) by ELISA (not shown) and binding to SYT2 cells that express human full length FcRHS, cyno foil length FcRHS or human El 1 domain transmembrane domain and cytoplasmic domains of FcRHS by FACS. A total of 26 clones were identified that bound to cells that express human FcRHS and cells that express cyno FcRHS, indicative of cross-species reactivity (Table 2). Subclone superaatants were further characterized for binding to A) multiple myeloma cells transfected with human FcRHS, B) cells that express human FcRHS endogenousiy (MOLP-2 myeloma ceils, peripheral human CD 19+ B-cells from healthy donors), C) SVT2 cells transfected to express human FcRHl, FcRH2, FcRIO or FcRH4, D) 293 ceils that express truncated version of human FcRHS (lacking 4 Ig-domains including H11 ; amino acids 464-850 of SEQ ID NO:l) and E) NK-cells. In addition, binding of supematants to soluble FcRHS a was analyzed by ELISA. Based on these analysis monoclonal antibodies were selected for purification.

Table 2.

Figure 2 shows the dose-range of binding of five purified E l 1 antibodies, non-isoform selective anti-FcRH5 antibody 10A8 (which binds Ig-ii.ke domains 4-5 of FcRHSc) and a control antibody specific to the N-terminal gD~tag to the SVT2 cells expressing either human FcR.H5 (Figure 2A) or eyno FcRH5 (Figure 2B). Antibodies in this assay were directly labeled with APC-fluorophore according to manufacturer's protocol (Invitrogen #z25051 , z25151 , z25251). Binding of representative El 1 aniibody 5A10 to human FcRHS transfected EJM (Figure 3A) and OPM2 (Figure 3B) multiple myeloma cell lines was found to be similar or better compared previously described non-isoform selective FcRHS antibodies 10A8 and 7D 1 1 (both bind Ig-like domains 4 5 of FcRHSc) (Elkins et a!., 2012; Poison et a I., 2006). MOLP-2 cells are one of the very few known multiple myeloma cell lines that express low levels of FcRHS endogenousiy. 5A10, 5F1, 3G7 and 6D2 subclone supematartts stained MOLP-2 cells with intensity similar to 7D1 1 (Figures 3C-F),

Two separate tests were designed to address dependency of binding on the presence of membrane proximal Ig-domain 9 (El 1). First a truncated human FeRHSe mutant was generated that lacks Ig~domains 6-9 (amino acids 464-850 of SEQ ID NO: 1) including the expected binding site for the antibodies derived from El 1 immunization, ' Ibis construct with N-terminal gD-tag was expressed in 293 cells and subjected to 2.5 ug/ml subclone supematants followed by PE labeled goat anti-mouse secondary antibody (1 : 1000 dilution). None of the tested subclones bound to 293 cells that express the truncated human FcRHSc (Figure 4A). In contrast binding was detected to 293 cells that express wild type human FcRHSc. Binding of gD or non-isoform selective antibody clone (10A8) was not altered by the mutation. This result demonstrates that binding site of the El 1 antibodies was included in Ig- doniains 6-9.

Isoform selectivity was further demonstrated by testing binding to the soluble FcRHSa isoform. For this, 293 cells were transfected to express the soluble isoform with C -terminal HiS-expression tag. Expression of FcRHSa protein was confirmed with Western blot analysis using anti-HIS antibody. A 65 kD band was detected in conditioned media from. FcRHSa but not vector transfected ceils (not shown). For the ELISA, plates were coated with anti-HTS capture antibody and incubated Ihour with 1 :10 diluted conditioned media including the HIS-tagged soluble FcRHSa isoform. The El 1 monoclonal antibodies were used for detection in 1 - 0.001 ug/ral concentration, incubated for 1 hour followed by incubation with goat anti-mouse HRP antibody and finally with TMB-substrate. While clones 2H7 and 5A10 demonstrate considerable reactivity to soluble FeRH5a 5 the other tested monoclonal antibodies do not show any detectable binding (Figure 5 A), This result confirms that the ig-domain 9 (El 1) is required for binding of the antibodies 1G7, 3A4, 3B12, 3G7 and 5F1, and therefore these antibodies are selective for full the length FcRH5 isoform (FcRHSc),

FcRH5 is expressed endogenously in B-ceils (Hatzivassiliou et at, 2001 ; Poison et al, 2006). To evaluate binding of subclone supematanis to 8-ceils, PBMCs were extracted from the blood of healthy donors. 1 million human PBMC were incubated with 4 ug l subclone supernatants for 60 min, washed and incubated with 1 TOO dilution of goat anti-mouse secondary labeled with APC, Cells were then washed again twice and stained PE-labeled anti-CD 19 (BD Biosciences #340364) prior flow cytometry and analysis of binding to CD 1 + cells. Most of the supematanis induced a significant shift in the APC signal in CD 19+ ceils (Figur 5B) over the controls (no primary antibody. anti-gD) indicative of binding to B cells.

Fc receptor homo log (FcRH) family molecules have a high degree of homology to one another (Miller et al, 2002). The homology is especially high between the membrane proximal domains, which the El 1 antibodies target (Miller et al, 2002). To investigate the cross reactivity to family members, FcRHl, FeRH2, FcRH3 and FeRH4 (ail including an N- terminal gD-expression tag) were expressed in SVT2 cells and cells were stained with subclone supernatants and goat anti~mouse~PE secondary antibody. Expression of the transfected FcRH was confirmed by a signal from anti-gD antibody in all cell lines. None of t ie supernatants bound significantly to FcRH2 expressing ceils as compared to staining with the gD antibody (Figure 6B). IBS, 1H11 , 3C10, 4G8 and 6D2 demonstrated a low level of binding to FcRHl (Figure 6A) and 1 F4 bound to FeRH4 (Figure 6D). Overall, the signals from FcRH3- expressing SVT2 cells were low, including the gD control antibody, indicative of low expression level. Low level of binding to FcRlD -expressing SVT2 cells was detected for 1 F4 and 4H8 supernatants (Figure 6C).

Since the overall signal in the FcRH3-expressing SVT2 ceils was low, further testing was done using PBMCs from healthy donors. PBMCs were stained as described above, but instead of CD 19, CD56 (BD Biosciences #555516) was used to gate the investigated ceil population to NK cells. NK- cells express endogenously FcRH3 (Poison et l , 2006), and as expected, were stained by a previously described monoclonal anti-FcRH3 antibody (Poison et al, 2006). FcRHl expression was also detected in CD56+ cells, but none of the El 1 subclone supematanis significantly stained the NK cells (Figure 7) demonstrating lack of cross reactivity to endogenously expressed FcRJO.

The cross reactivity of the family members were re-tested using the identical protocol dseribed above in SVT2 cells but using fresh reagents and re-transfecting SVT2 cells with FcRHl , FcRH2, FeRIB, and Fc H4, Re-testing the purified antibodies as described above resulted in significantly different results than the first series of experiments, These updated results are summarized in Table 4, Rather than showing little to no cross-reactivity with other Fc H family members, all but one ant body (1G7) showed significant binding to both FcRH ' 5 and at least one or more other family members.

Without being bound by theory, this amount of antibody cross-reactivity is what would be expected, given the sequence similarity of the last Xg-like domain in the various FcRH. family members,

CD8÷ T cells are among the most potent immune effector ceils, The activity of T cells can be recruited to kill tumor cells by using bispecific antibodies (or antibody fragments) that simultaneously bind both T ceil and a tumor antigen. The dual binding can iead to a polyclonal activation of T cells and specific killing of tumor antigen expressing ceils (Lm ei aL, 1985: Shaiaby et al , 1992). Several tumor targets and several bispecific antibody platforms have demonstrated general flexibility and preclinical feasibility for this approach, Importantly, promising clinical activity has been demonstrated with a CD19 targeting, T cell activating bispecific seFv antibody fragment blinatumomab (MT103;

MicroMet). Treatment with doses as low as 60 ug/mf/day results in prolonged responses in clinical trials for treatment of relapsed non-Hodgkin's lymphoma and acute lymphoblastic leukemia (Bargou et al , 2008; Dreier et al , 2002)

The ability of the FcRI-15 antibodies to activate T cell and mediate killing in bispecific antibody format was investigated by generating bispecific bisFab molecules. In short, these bispecific molecules are generated by proteolytical cleavage of the antibody, followed by reduction, re-oxidation reactions and conjugation of Fab-fragments using bis-maieamide (Scheer et al, 2012b and as described above), Anti-CD3 antibody clone UCHTl binds to human CD3 that incorporates to T ceil receptor, UCHTl .v9 has previously been shown to be efficient T ceil binding arm (Junttila et al., 2012 and as described above; Zhu et aL, 1995) and therefore was used to the FcRH ' 5 bisFabs. ine anti-FcRH5 antibody clones (1 G7, 2R7, 3G7, 5A10, 5F1 , 6D2, 3B12, 3C10, 3F10) from the El 1 immunization were chosen for the target arm and conjugated with UCHTl , 9 to result in CD3~FeRJI5 bispecific bisFab molecules.

In addition to bisFab molecules, also full length bispecific antibodies (T cell dependent bispecific antibodies; TDBs) were produced using knobs-into-hoies technology (Merchant et al, 1998), which relies on a pair of complementary engineered Fc regions that drive heterodimerization of antibody hemimers. As in the ease of bisFabs, the UCHTl .v9 (Zhu et al., 1995) was used as the anti- CD3 (hole). For the target arm (knob), antibody clones from the FcRH5 El 1 -immunization, a non- isoforra selective anti-FcRH5 clone (10A8) (Eikins et al, 2012) or aoti-HER2 clone 4D5 (trastuzumab) (Carter et aL, 1 92) were used. Generation and purification of the TDBs has been described in detail (junttila et aL, 2012; Scheer et al., 2012a and as described above).

The ability of the bispecific molecules to mediate killing of FcRH5 transfeeted 293 target cells was investigated by incubating the targets with CD8+ T cells (effector cells) for 48 hours and measuring the killing activity using Cell Titer Glo assay or FACS killing assay (assays described above). All nine bisFabs mat incorporated an anti~FeRH5 EI 1 target arm were efficient in mediating target cell killing (FIG. SA-B), Killing activity was detected as low as 1 -10 ng/ ' ml concentrations and saturated at 10-100 ng/ml concentration. Maximal killing activity exceeded 80% for most of the clones. The kiiling activity was similar compared to the HER2-TDB (FIG, 8A-B). Human HER2 is expressed in the 293 ceils on low level (data not shown), in contrast, killing activity far exceeded the non-isoform selective Fc H5-TDB (10A8), which was capable in killing only approximately 20% of the targets (FIG. 8A-B). Similar robust activity was detected using a foil length TDB format incorporating 2H7, 3G7 and 5A10 FcRH5-EI 1 clones as target arms (FIG. 8C-D). No significant difference was detected between TDB and bisFab versions of 2H7 and 3G7 indicating that Fc is neither necessary for the activity nor inhibitory for the killing activity. FcRHS bisFabs and full length TDBs incorporating 2H7 and 3G7 as target arm were also able to efficiently mediate killing of MOLP2 cells, which express endogenousiy low levels of FcRHS (FIG. 9A). T cell activation was followed in fee reactions measuring the proportion of CD8+ cells that express CD69 on the cell membrane. T cell activation corresponded the killing activity and was similar for botli bisFabs and TDBs (FIG. 9B). A summary of the results are shown in Table 3.

Table 3

'fable 4

References

Bargou, R. et al (2008). Science 32 L 974-977,

Carter, P. et ai (1 92). Proc Natl Acad Sci U S A 89, 4285-4289.

Dreier, T. et al. (2002), Ini J Cancer 100, 690-697.

Elkins, K. et al (2012). Mo! Cancer T er /, 2222-2232.

Hatzivassiliou, G, et al (2001). immunity 14, 277-289.

Liu, M. A. et at. (1985). Proc Natl Acad Sci U S A 82, 8648-8652.

Merchant, A, M. et al, (1 98). Nat Biotechnol 16, 677-681.

Miller, I et al. (2002). Blood 99, 2662-2669.

Poison, A. G. et l. (2006). Expression pattern of the human FeKH/IRTA receptors in normal tissue and in B-chronic lymphocytic leukemia. International immunology 18, 1363-1373.

Shalaby, M. R, et al (1992). J Exp Med 175, 217-225.

Zhu, Z. ef a/. (1 95). Int J Cancer 62, 319-324.

Alihougli the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention. The disclosures of all patent and scientific literature cited herein are expressly incorporated in their entirety by reference.

Variable Light Chain Domain

1CS.1

DiVMTQSQRFMSTSLGD VSVTC ASQINiVIT iVAWYQQ PGQSP ALIYSASY YSGVPDRFTGSGSGTDFTLTIS VQSEDLAEYFCQQYT YPMWTFGGGTRLEI RTVA (SEQ SD NO:110}

1G7.2

DSV TQSHKiSV!STSVGDRVSSTC ASQDVS SVVWFQQKPGQSPNLLIYSASYRYTGVPORFTGSGSGTOFTFTISSV QAEDLAVYYCQGHY8SPYTFG6GTKLES RTVAA (SEQ SD NO:112) 2H7.3

EIVLTQSPATLSVTPGDSVSLSCRASQM!RN JLHWYQQ SHESPRLL!KFTSQSSSG!PSRFTGSGSGTDFTLSINSVETE DFG YFCQQSNNWPQYTFGGGTKLEI RTVAA (SEQ. SD NO:114)

3A4.2

DSQMTQSPATLSVTPGDSVSLSCRASQSiS^ LHWYQQ SHESPRLLS FASQSSSGSPSRFSGSGSGTDFTLSiNSVETE DFGMYFCQQSNNWPQYTFGGGTKLELKRTVAA (SEQ SD N0:116)

3B12.1.1

DIQMTQSPASLSASVGEmiTC ASENIYSNlAWYQL QG SPQLLVYGAANLAEGVPSRISGSGSGTQYSLKINSLQ SEDFGTYYCQHFWGIPWTFGGGT LEIKRTVAA (SEQ SD SM0:118|

3G10

D!Q TQTPLSLPVTLGOQAS!SCRSSQSLVHRNGNTYLHWYLQ PGQSP LL!Y VSNRFSGVPDRFSGSGSGTDFTL SSRVEAEDL6VYFCSQSTHVPPTFGGGT LELKRTVAA (SEQ ID O:120)

3F10

DlVMTC^PASLSASVGEmiTCRASENIYSNLAWYQL QG SPQLLVYGAANLAEGVPSRISGSGSGTQYSL INSLQ

SED FGTYYCQH F WG ! P WTFG GT LE i RTVAA (SEQ SD O:122) 3G3

DSV TQSPASLSASVGEWTSTCRASE !YS LAWYQL QG SPQLLVYGAA LAEGVPSRISGSGSGTQYSL !fsiSLQ SED FGTYYCQH FWG I WTFGG GTKLE! KRTVAA (SEQ !D NO: 124)

3G7.1

DIVLIQSPATLSVTLGGSVSLSCRASClSISNNLHWYQQKSHESPRLLIKFASQSIS GiPSRFRGSGSGTDFTLTINSVETED

F S YFCQQSM WPQYTFGGGTKLE L RTVA A (SEQ ID O:126)

5A10.1.3

DiVLTQSPANLSViPGDSVSLSCRASQNiRNNLHWYQQ SQESPRLLIKFASQSMSGTPSRFTGSGSGTDFTLT!NTVE TEDFGMYFCQQSNNWPQYTFGGGTKLEIKRTVAA (SEQ ID NO:128)

5F1.1.5

QAVVTQESALTTSPGETWLTCRSSTG TTS FA WVQE PDHLFTGLIGGTS RAPGVPARFSGSLIGD AALTST GAQTEDEAiYFCVLWCSNLWVFGGGTKLTVLGQP AA {SEQ ID NQ;130)

6D2

DIV TQSH FMSTSVGDRVSITC ASQDVGTAVAWYQQKPGQSP LLIFWPSTRHTGVPDRFTGSGSGTDFTLT!G

NVQSEDLADYFCQQFSSLPHTFGGGT LESKRTVAA (SEQ \Ό NQ:132) 1G7'

DIVMT(¾HKIMS 5VGDRVSITCKASQDVSNIWWFQQKPGQSPNLUYSASYRYTGVPDRFTGSGSGTDFTFTI SSV QAEDLAVYYCQQHYSSPYTFGGGTKLEIK (SEQ ID 1ΜΟ:134) Variabie Heavy Chain Domain

1C8.1

EVQLQQSGPELV PGASMKISCEASGYSFTAYi WVKQSRG NLEW!GL!NPYNGETTYNQKF G ATLTVDQSSS TAYMELLSLTSEDSAVYFCARGLYWFPYWGQGTLVTVSAAST GPSVFPLAP (SEQ Ώ NO:lll)

1G7.2

EVQLQESGPGLVQPSQSLS!TCTVSGFSLTRFGVHWVRQSPG GLEWLGViW GGSTDYNAAFMS LTSTKD!SiS S QVFFKLNSL VDDTAIYYCSNHYYGSSDYALDNWGQGTSVTVSSASTKGPSVFPLAP (SEQ ΪΌ Η0Λ13) 2H7.3

EVQLQQSGPELW PGASVK SC ASGYT OYYM WV QTHGKSLEWiGDiNPNNGETFYSQKFKGKATLTVDKS STTAYMQLNSLTSEDSAVYYCARGLYRFDYWGQGTTLTVSSASTKGPSVFPLAP (SEQ ID NO:1155

3A4.2

EVQLQaSGPELVKSGASVK SCKASGYTFTDYY WV QSHG SLEWlGDi PYNGETFYINiQ L G ATLTV ' DKSS MTVF QLNSLTSEDSAVYYCARGLYFFAYWGQGTTLTVSSASTKGPSVFPLAP (SEQ !D M0:117)

3812.1.1

EVQLQQSGPELV PGASVKSSC TSG\TFTEYTiHWV QSHG SLERSGG^P MDAVSY QRFRG ATLTVDKSSST AYMELRSLT5EDSAVYYCA LGRGYYFDYWGQGTTLTVSSAST GPSVFPLAP (SEQ ID N0:119)

3C10

QVQLQQPGAELVRPGASVKISC TSGYTFISYWINWV QRPGQGLEW!GNIYPSDSYTNYNQ F DKATLTVDTSSS

TAY QLTSPTSEDSAVYYCTRSLYGYDASYFDYWGQGTTLTVSSAST GPSVFPLAP {SEQ ID N0:121)

3F10

QV LQQSGPELVKPGASV ISCiaSGnFTEYTIHWVKC^HGKSLERlGGINPNNDAISYNQKFRG ATLTVD SSSTA

Y ELRSLTSEDSAVYYCAKLGRGYYFDYWGRGTTLTVSSAST GPSVFPLAP (SEQ ID NO:123) 3G3

EVQLQQSGPELVKPGASV !SCKTSGYTFTEYTIHWVKQSHGKSLERIGG!NPNNDA!SYNQ FRG ATLTVDKSSSTA YMELRSLTSEDSAVYYCA LGRGYYFDYWGRGTTLTVSSAST GPSVFPLAP (SEQ \Ώ NO:125)

3G7.1.5

EVQLQQSGPELV PGASVK SCKASGYTF DYYMKWVRQNHG RLEW!GDiNPYNGDTFYNQKFKDKATLTVDKS SSTAY QFNSLTSEDSAVYYCARGLYFFHYWGQGTTLTVSSAST GPSVFPLAP (SEQ ID NO:127)

5A10.1.3

EVQLQQSGPELW PGASV MSC ASGYTFTDYY WV QSHG SLEWiGD!NPNNGETFYNQ F G ATLTVD S TSTAYMELNSLTTEDSAVYYCARGLYRFDYWGQGTTLTVSSAAST GPSVFPLAP {SEQ ID NQ:129)

5F1.1.5

QVQLQ ^GADLVRPGTSVKVSCKASGYAFTNYLIEWV QRPGQGLEWIGVINPGSGGTNYNEKF GKATLTAD SS

5TAY QLSSLTSDDSAVYFCARTRNY6YVIDYWGQGTTLTVSSAST GPSVFPLAP (SEQ !D 0:131)

602

QVQLQQSGPELVKPGASVKiSCKASGFSFTAYFMNWVKC^HGKSPEWIGRINPYNGETFF NQNFKDKATLTVDKSS

NTAH M E LLSLTSDDSAVYYCG RG LYYLNYWGQGTTLTVSSAST G PSVFP LAP {SEQ SD NG:133)

1G7'

QVQL QSGPGLVQPSQSLSITCTVSGFSLTRFGVHWVRQSPG GLEWLGViWRGGSTDYNAAFMSRLTiTKDNS S QVFFKLNSL VDDTAfYYCSNHYYGSSDYALDNWGQGISVTVSS (SEQ ID NO:135) KabatCDRLl (HVR-Ll)

Kabai CDR L2 (HVR-L2)

Kabai CDR L3 (HVR-L3)

KabatCDRHl (HVR-H1)

Kabat CDR H2 (HVR-H2)

CDR H2 (HVR-B.2)

Kabat CDR H3 (HVR-H3) FcRHSc

MIi.,WVII.;LVI.APVSGQFARTPRPIIFLQPP^ TVFQGERVTLTCKGFRFYSPOKT WIIRYLG

KEILRETPDNII FA / QESGFA ' RCQAQGSPLSSPVHLDFSSASL1L0 ?LSVFEGDSVVLRCRAK/\E

VTLNNTIYKlSiD IAFLNKRTOFHIPHACLro

PVIIIASSFQPISGNPVTLTCETQLSLE^

GFYWCKA, l¾4PYSVISDSPRS¾lQVQIPASHPVi;rLSPF ALNFECrrKVTI^CETQEDSLRTL

YRFYHEGVPLRHKSVRCERGASiSFSLTTENSGNYTCTADNGLGAKPSKAVSLSVlV PVSHPV

L ESSPEDLlFEG KVTLIiCEAQRGSLPILYQFFn-IEGAALERRSANSAGGVAISFSLTAEHSGN

YYCrAD GFGPQRS AVSLSVTVPVSHPVLTLSSAEALTFEGATVTLFICEVORGSPQILYOFY

HEDMPLWSSS1PSVGRVSFSFSETEGHSGNYY^CT/ NGFGPQRSEVVSL.FVT TVSRPILTLRV

PRAQAVVCmiJ LHCEAPRGSPPILYWFYHEDVTLGSSS^SGGEASFNLSLTAEIlSGNYSCE

A NGLVAQHSDTISLSViVPVSRPlLTFRAPRAQAVVGDLLEEHCEAERGSSPILYWFYHED VT

LGKISAPSGGGASFNLSLTTEHSGIYSCEADNGIEAQRSEMVTI VAVPVSRPVLTLRAPGra

AAVGDLLELHCEALRGSPLILYREFHEDVTiXi RSSPSGGASLNI.SI.. ' rAEHSGNYSCEADNGL

GAQRSET\ ? TLYITGLT NRSGPFATGVAGGLLSIAGLAAGAI J LI,YCWLSRKAGRKPASDPARS

PSDSDSQEPTYHNVPAWEELQPVYTNA>nPRGENVVYSEVR^

PIIYSEVkVASTPVSGSLFLASSAPHR (SEQ ID NO: I)