Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
IRTA-2 ANTIBODIES AND THEIR USES
Document Type and Number:
WIPO Patent Application WO/2006/076691
Kind Code:
A2
Abstract:
The present invention provides isolated monoclonal antibodies, particularly human monoclonal antibodies, that specifically bind to IRTA-2 with high affinity. Nucleic acid molecules encoding the antibodies of the invention, expression vectors, host cells and methods for expressing the antibodies of the invention are also provided. Immunoconjugates, bispecific molecules and pharmaceutical compositions comprising the antibodies of the invention are also provided. The invention also provides methods for detecting IRTA-2, as well as methods for treating various B cell malignancies, including non-Hodgkin's lymphoma.

Inventors:
GRAZINO ROBERT (US)
KING DAVID J (US)
SRINIVASAN MOHAN (US)
CARDARELLI JOSEPHINE (US)
HUANG HAICHUN (US)
Application Number:
PCT/US2006/001444
Publication Date:
July 20, 2006
Filing Date:
January 12, 2006
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MEDAREX INC (US)
GRAZINO ROBERT (US)
KING DAVID J (US)
SRINIVASAN MOHAN (US)
CARDARELLI JOSEPHINE (US)
HUANG HAICHUN (US)
International Classes:
C07K16/00; A61K39/395; C12Q1/68; C07H21/04; C07K17/00
Domestic Patent References:
WO2001038490A2
Other References:
See references of EP 1846449A4
Attorney, Agent or Firm:
FEHLNER, Paul F. et al. (30 Rockefeller PlazaNew York, NY, US)
Download PDF:
Claims:
We claim:

1. An isolated monoclonal antibody, or an antigen-binding portion thereof, wherein the antibody:

(a) binds to human IRTA-2 with a K D of 1x10 "7 M or less;

(b) does not substantially bind to human IRTA-3 or IRTA-4; and

(c) binds to Granta 519 tumor cells and does not substantially bind to Raji or Ramos tumor cells.

2. The antibody of claim 1, which is a human antibody.

3. The antibody of claim 1, which is a chimeric or humanized antibody.

4. The antibody of claim 2, which is full-length antibody of an IgGl or IgG4 isotype.

5. The antibody of claim 2, which is an antibody fragment or a single chain antibody.

6. The antibody of claim 2, wherein said antibody binds to human IRTA-2 with a KD of 5 xlO 's M or less.

7. The antibody of claim 2, wherein said antibody binds to human IRTA-2 with a KD of 5 xlO "9 M or less.

8. The antibody of claim 2, wherein human IRTA-2 comprises a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 25 [Genbank Ace. No. NP_112571].

9. The antibody of claim 2, wherein human IRTA-3 comprises a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 27 [Genbank Ace. No. AAL59390].

10. The antibody of claim 2, wherein human IRTA-4 comprises a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 28 [Genbank Ace. No. AAL60249].

11. The antibody of claim 2, wherein the antibody substantially binds to SU-DHL-6 or JEKO-I tumor cells.

12. The antibody of claim 2, wherein the antibody does not substantially bind to Daudi, IM-9, Karpas 1106P or SU-DHL-4 tumor cells.

13. An isolated monoclonal antibody, or antigen binding portion thereof, wherein the antibody cross-competes for binding to IRTA-2 with a reference antibody, wherein the antibody:

(a) binds to human IRTA-2 with a K 0 of IxIO "7 M or less;

(b) does not substantially bind to human IRTA-3 or IRTA-4; and

(c) binds to Granta 519 tumor cells and does not substantially bind to Raji or Ramos tumor cells.

14. The antibody of claim 13, wherein the reference antibody comprises:

(a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 13; and

(b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15.

15. The antibody of claim 13, wherein the reference antibody comprises:

(a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 14; and

(b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 16.

16. The antibody of claim 13, wherein human IRTA-2 comprises a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 25 [Genbank Ace. No. NPJ 12571].

17. The antibody of claim 13, wherein human IRTA-3 comprises a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 27 [Genbank Ace. No. AAL59390].

18. The antibody of claim 13, wherein human IRTA-4 comprises a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 28 [Genbank Ace. No. AAL60249].

19. An isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a heavy chain variable region that is the product of or derived from a human VH 3-23 gene or a human VH 1-8 gene, wherein the antibody specifically binds IRT A-2.

20. An isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a light chain variable region that is the product of or derived from a human VK L6 gene or a human VK Ll 8 gene, wherein the antibody specifically binds IRTA-2.

21. The isolated monoclonal antibody or an antigen-binding portion thereof of claim 20 further comprising a heavy chain variable region that is the product of or derived from a human VH 3-23 gene or a human VH 1-8 gene.

22. The antibody of claim 1, which comprises:

(a) a heavy chain variable region CDRl comprising SEQ ID NO: 1;

(b) a heavy chain variable region CDR2 comprising SEQ ID NO: 3;

(c) a heavy chain variable region CDR3 comprising SEQ ID NO: 5;

(d) a light chain variable region CDRl comprising SEQ ID NO: 7;

(e) a light chain variable region CDR2 comprising SEQ ID NO: 9; and

(f) a light chain variable region CDR3 comprising SEQ ID NO: 11.

23. The antibody of claim 1, which comprises:

(a) a heavy chain variable region CDRl comprising SEQ ID NO: 2;

(b) a heavy chain variable region CDR2 comprising SEQ ID NO: 4;

(c) a heavy chain variable region CDR3 comprising SEQ ID NO: 6;

(d) a light chain variable region CDRl comprising SEQ ID NO: 8;

(e) a light chain variable region CDR2 comprising SEQ ID NO: 10; and

(f) a light chain variable region CDR3 comprising SEQ ID NO: 12.

24. An isolated monoclonal antibody, or antigen binding portion thereof comprising:

(a) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 13 and 14; and

(b) a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 15 and 16; wherein the antibody specifically binds IRTA-2.

25. An isolated monoclonal antibody or antigen binding portion thereof comprising:

(a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 13; and

(b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15.

26. An isolated monoclonal antibody or antigen binding portion thereof comprising:

(a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 14; and

(b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 16.

27. A composition comprising the antibody of claim 1, or antigen-binding portion thereof, and a pharmaceutically acceptable carrier.

28. An immunocoηjugate comprising the antibody of claim 1, or antigen-binding portion thereof, linked to a therapeutic agent.

29. A composition comprising the immunoconjugate of claim 28 and a pharmaceutically acceptable carrier.

30. The immunocoηjugate of claim 28, wherein the therapeutic agent is a cytotoxin.

31. A composition comprising the immunoconjugate of claim 30 and a pharmaceutically acceptable carrier.

32. The immunoconjugate of claim 28, wherein the therapeutic agent is a radioactive isotope.

33. A composition comprising the immunoconjugate of claim 32 and a pharmaceutically acceptable carrier.

34. An isolated nucleic acid molecule encoding the antibody of claim 1, or antigen- binding portion thereof.

35. An expression vector comprising the nucleic acid molecule of claim 34.

36. A host cell comprising the expression vector of claim 35.

37. A method for preparing an anti-IRTA-2 antibody which comprises expressing the antibody in the host cell of claim 36 and isolating the antibody from the host cell.

38. A method of inhibiting growth of tumor cells expressing IRTA-2, comprising contacting the cells with the antibody of claim 1, or antigen-binding portion thereof, in an amount effective to inhibit growth of the tumor cells.

Description:

IRTA-2 ANTIBODIES AND THEIR USES

Background of the Invention

The Immune Receptor Translocation Associated (IRTA) genes/proteins, also known as Fc Receptor Homolog (FcRH) genes, consist of a five-member family of immunoglobulin- like cell surface receptors (Miller et al, (2002) Blood. 99:2662; Davis et al, (2002) Immunological Reviews. 190:123). The IRTAs were initially discovered by analysis of the breakpoints of a multiple myeloma cell line which contained a Iq21 chromosomal rearrangement (Hatzivassiliou et al, (2001) Immunity. 14:277). Each of the IRTA glycoproteins contains between 3 to 9 extracellular Ig-like domains (Miller, 2002, supra). IRTAs are also characterized by having a cytoplasmic domain containing 3 to 5 tyrosine residues contained within particular motifs, suggesting the presence of immunotyrosine inhibitory motifs (ITIM) and immunotyrosine activation-like (ITAM-like) motifs (Miller, 2002, supra; Hatzivassiliou, 2001, supra).

IRTAs typically are expressed in peripheral lymphoid tissues, including lymph nodes, tonsils, resting peripheral B cells and normal germinal center B cells (Davis et al, (2001) PNAS. 98:9772). IRTA-2, -3, -4, and -5 are all expressed at high levels in spleen, whereas, by comparison, IRTAl has been detected in lower levels in the spleen. IRTA expression has been analyzed within the B cell compartment of human tonsil tissue. IRTA-I is expressed outside of lymphoid follicles in the marginal zone pattern and in intraepithelial lymphocytes. IRTA-2 and -3 are expressed within the germinal center, with highest expression in the centocyte-rich light zone. IRT A-4 and -5 are expressed highest within mantle zones, indicating expression in naϊve B cells. (Miller, 2002, supra)

The IRTA genes have been shown to be highly expressed in B cell non-Hodgkin's lymphoma, chronic lymphocytic leukemias, follicular lymphomas, diffuse large cell lymphomas of B lineage, and multiple myelomas (Davis, 2001, supra).

Summary of the Invention

The present invention provides isolated monoclonal antibodies, in particular human monoclonal antibodies, that bind to IRTA-2 and that exhibit numerous desirable properties. These properties include high affinity binding to human IRTA-2, but lacking substantial cross-reactivity with either human IRT A-3 or IRTA-4. Still further, antibodies of the invention have been shown to bind to a B-cell tumor cell line.

In preferred embodiments of the invention, the human IRTA-2 comprises a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 25 [Genbank Ace. No. NP_112571]; the human IRTA-I comprises a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 26 [Genbank Ace. No. NP_112572]; the human IRTA-3 comprises a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 27 [Genbank Ace. No. AAL59390]; the human IRTA-4 comprises a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 28 [Genbank Ace. No. AAL60249]; and/or the human IRTA-5 comprises a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 29 [Genbank Ace. No. AAL60250].

In one aspect, the invention pertains to an isolated monoclonal antibody, or an antigen-binding portion thereof, wherein the antibody:

(a) binds to human IRTA-2 with a K 0 of IxIO "7 M or less;

(b) binds to human CHO cells transfected with IRTA-2;

(c) does not substantially bind to human IRTA-3 or IRTA-4; and

(d) binds to Granta 519 tumor cells and does not substantially bind to Raji or Ramos tumor cells.

Preferably the antibody is a human antibody, although in alternative embodiments the antibody can be a murine antibody, a chimeric antibody or humanized antibody.

In a preferred embodiment, the antibody does not substantially bind to Daudi, IM-9, Karpas 1106P, or SU-DHL-4 tumor cells.

In more preferred embodiments, the antibody binds to human IRTA-2 with a K D of 5 x 10 ~8 M or less, binds to human IRTA-2 with a KD of 2 X lO "8 M or less, binds to human IRTA-2 with a K 0 of 1 x 10 "8 M or less, binds to human IRTA-2 with a K 0 of 5x10 "9 M or less, binds to human IRTA-2 with a K D of 4x10 "9 M or less, binds to human IRTA-2 with a KD of 3x10 "9 M or less, or binds to human IRTA-2 with a K 0 of 2.1 x 10 ~9 M or less.

In another embodiment, the invention provides an isolated monoclonal antibody, or antigen binding protion thereof, wherein the antibody cross-competes for binding to IRTA-2 with a reference antibody comprising:

(a) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 13 and 14; and

(b) a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 15 and 16.

In various embodiments, the reference antibody comprises:

(a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 13; and

(b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15; or the reference antibody comprises:

(a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 14; and

(b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 16.

In another aspect, the invention pertains to an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a heavy chain variable region that is the product of or derived from a human VH 3-23 gene, wherein the antibody specifically binds IRT A-2. The invention also provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a heavy chain variable region that is the product of or derived from a human VH 1-8 gene, wherein the antibody specifically binds IRT A-2. The invention still further provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a light chain variable region that is the product of or derived from a human VK L6 gene, wherein the antibody specifically binds IRTA-2. The invention even further provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a light chain variable region that is the product of or derived from a human VK Ll 8 gene, wherein the antibody specifically binds IRTA-2.

In a preferred embodiment, the invention provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising:

(a) a heavy chain variable region of a human VH 3-23 or 1-8 gene; and

(b) a light chain variable region of a human V K L6 or VK Ll 8; wherein the antibody specifically binds to IRTA-2.

In a preferred embodiment, the antibody comprises a heavy chain variable region of a human VH 3-23 gene and a light chain variable region of a human VK L6 gene. In another preferred embodiment, the antibody comprises a heavy chain variable region of a human V H 1-8 gene and a light chain variable region of a human VK Ll 8 gene.

In another aspect, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof, comprising:

a heavy chain variable region that comprises CDRl, CDR2, and CDR3 sequences; and a light chain variable region that comprises CDRl, CDR2, and CDR3 sequences, wherein:

(a) the heavy chain variable region CDR3 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 5 and 6, and conservative modifications thereof;

(b) the light chain variable region CDR3 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequence of SEQ ID NOs: 11 and 12, and conservative modifications thereof;

(c) the antibody binds to human IRTA-2 with a K D of 1x10 "7 M or less;

(d) binds to human CHO cells transfected with IRTA-2;

(e) the antibody does not substantially bind to human IRTA-3 or IRTA-4; and

(f) the antibody binds to Granta 519 tumor cells and does not substantially bind to

Raji or Ramos tumor cells.

Preferably, the heavy chain variable region CDR2 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 3 and 4, and conservative modifications thereof; and the light chain variable region CDR2 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 9 and 10, and conservative modifications thereof. Preferably, the heavy chain variable region CDRl sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 1 and 2, and conservative modifications thereof; and the light chain variable region CDRl sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 7 and 8, and conservative modifications thereof.

In a preferred embodiment, the antibody does not substantially bind to Daudi, IM-9, Karpas 1106P, or SU-DHL-4 tumor cells.

In yet another aspect, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof, comprising a heavy chain variable region and a light chain variable region, wherein:

(a) the heavy chain variable region comprises an amino acid sequence that is at least

80% homologous to an amino acid sequence selected from the group consisting of

SEQ ID NOs: 13 and 14;

(b) the light chain variable region comprises an amino acid sequence that is at least 80% homologous to an amino acid sequence selected from the group consisting of SEQ ID NOs: 15 and 16;

(c) the antibody binds to human IRTA-2 with a K D of 1x10 ~7 M or less;

(d) binds to human CHO cells transfected with IRTA-2;

(e) the antibody does not substantially bind to human IRT A-3 or IRTA-4; and

(f) the antibody binds to Granta 519 tumor cells and does not substantially bind to

Raji or Ramos tumor cells.

In a preferred embodiment, the antibody does not substantially bind to Daudi, IM-9, Karpas 1106P, or SU-DHL-4 tumor cells.

In preferred embodiments, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof, comprising:

(a) a heavy chain variable region CDRl comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 and 2;

(b) a heavy chain variable region CDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 3 and 4;

(c) a heavy chain variable region CDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5 and 6;

(d) a light chain variable region CDRl comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 7 and 8;

(e) a light chain variable region CDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 9 and 10; and

(f) a light chain variable region CDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 11 and 12; wherein the antibody specifically binds IRTA-2. A preferred combination comprises:

(a) a heavy chain variable region CDRl comprising SEQ ID NO: 1;

(b) a heavy chain variable region CDR2 comprising SEQ ID NO: 3;

(c) a heavy chain variable region CDR3 comprising SEQ ID NO: 5;

(d) a light chain variable region CDRl comprising SEQ ID NO: 7;

(e) a light chain variable region CDR2 comprising SEQ ID NO: 9; and

(f) a light chain variable region CDR3 comprising SEQ ID NO: 11. Another preferred combination comprises:

(a) a heavy chain variable region CDRl comprising SEQ ID NO: 2;

(b) a heavy chain variable region CDR2 comprising SEQ ID NO: 4;

(c) a heavy chain variable region CDR3 comprising SEQ ID NO: 6;

(d) a light chain variable region CDRl comprising SEQ ID NO: 8;

(e) a light chain variable region CDR2 comprising SEQ ID NO: 10; and

(f) a light chain variable region CDR3 comprising SEQ ID NO: 12.

Other preferred antibodies of the invention, or antigen binding portions thereof comprise:

(a) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 13 and 14; and

(b) a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 15 and 16; wherein the antibody specifically binds IRTA-2. A preferred combination comprises:

(a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 13; and

(b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15.

Another preferred combination comprises:

(a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 14; and

(b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 16.

In another aspect of the invention, antibodies, or antigen-binding portions thereof, are provided that compete for binding to IRTA-2 with any of the aforementioned antibodies.

The antibodies of the invention can be, for example, full-length antibodies, for example of an IgGl or IgG4 isotype. Alternatively, the antibodies can be antibody fragments, such as Fab or Fab'2 fragments, or single chain antibodies.

The invention also provides an immunoconjugate comprising an antibody of the invention, or antigen-binding portion thereof, linked to a therapeutic agent, such as a cytotoxin or a radioactive isotope. The invention also provides a bispecific molecule comprising an antibody, or antigen-binding portion thereof, of the invention, linked to a second functional moiety having a different binding specificity than said antibody, or antigen binding portion thereof.

Compositions comprising an antibody, or antigen-binding portion thereof, or immunoconjugate or bispecific molecule of the invention and a pharmaceutically acceptable carrier are also provided.

Nucleic acid molecules encoding the antibodies, or antigen-binding portions thereof, of the invention are also encompassed by the invention, as well as expression vectors comprising such nucleic acids and host cells comprising such expression vectors. Moreover, the invention provides a transgenic mouse comprising human immunoglobulin heavy and light chain transgenes, wherein the mouse expresses an antibody of the invention, as well as hybridomas prepared from such a mouse, wherein the hybridoma produces the antibody of the invention.

In yet another aspect, the invention provides a method of treating a B cell malignancy in a subject in need of treatment comprising administering to the subject the antibody, or antigen-binding portion thereof, of the invention, such that the B cell malignancy in the subject is treated. The disease can be, for example, non-Hodgkin's lymphoma, chronic lymphocytic leukemias, follicular lymphomas, diffuse large cell lymphomas of B lineage, and multiple myelomas.

The invention also provides methods for making "second generation" anti-IRTA-2 antibodies based on the sequences of the anti-IRTA-2 antibodies provided herein. For example, the invention provides a method for preparing an anti-IRTA-2 antibody comprising:

(a) providing: (i) a heavy chain variable region antibody sequence comprising a CDRl sequence selected from the group consisting of SEQ ID NOs: 1 and 2, a CDR2 sequence selected from the group consisting of SEQ ID NOs: 3 and 4 and/or a CDR3 sequence selected from the group consisting of SEQ ID NOs: 5 and 6; and/or (ii) a light chain variable region antibody sequence comprising a CDRl sequence selected from the group consisting of SEQ ID NOs: 7 and 8, a CDR2 sequence selected from the group consisting of SEQ ID NOs: 9 and 10 and/or a CDR3 sequence selected from the group consisting of SEQ ID NOs: 11 and 12;

(b) altering at least one amino acid residue within the heavy chain variable region antibody sequence and/or the light chain variable region antibody sequence to create at least one altered antibody sequence; and

(c) expressing the altered antibody sequence as a protein.

Other features and advantages of the instant invention will be apparent from the following detailed description and examples which should not be construed as limiting. The

contents of all references, Genbank entries, patents and published patent applications cited throughout this application are expressly incorporated herein by reference.

Brief Description of the Drawings

Figure IA shows the nucleotide sequence (SEQ ID NO: 17) and amino acid sequence (SEQ ID NO: 13) of the heavy chain variable region of the 9D12 human monoclonal antibody. The CDRl (SEQ ID NO: 1), CDR2 (SEQ ID NO: 3) and CDR3 (SEQ ID NO: 5) regions are delineated and the V, D and J germline derivations are indicated.

Figure IB shows the nucleotide sequence (SEQ ID NO: 19) and amino acid sequence (SEQ ID NO: 15) of the light chain variable region of the 9D12 human monoclonal antibody. The CDRl (SEQ ID NO: 7), CDR2 (SEQ ID NO: 9) and CDR3 (SEQ ID NO: 11) regions are delineated and the V and J germline derivations are indicated.

Figure 2A shows the nucleotide sequence (SEQ ID NO: 18) and amino acid sequence (SEQ ID NO: 14) of the heavy chain variable region of the 8Al human monoclonal antibody. The CDRl (SEQ ID NO: 2), CDR2 (SEQ ID NO: 4) and CDR3 (SEQ ID NO: 6) regions are delineated and the V and J germline derivations are indicated.

Figure 2B shows the nucleotide sequence (SEQ ID NO: 20) and amino acid sequence (SEQ ID NO: 16) of the light chain variable region of the 8Al human monoclonal antibody. The CDRl (SEQ ID NO: 8), CDR2 (SEQ ID NO: 10) and CDR3 (SEQ ID NO: 12) regions are delineated and the V and J germline derivations are indicated.

Figure 3 shows the alignment of the amino acid sequence of the heavy chain variable region of 9D12 with the human germline VH 3-23 amino acid sequence (SEQ ID NO: 21).

Figure 4 shows the alignment of the amino acid sequence of the heavy chain variable region of 8Al with the human germline VH 1-8 amino acid sequences (SEQ ID NO: 22).

Figure 5 shows the alignment of the amino acid sequence of the light chain variable region of 9Dl 2 with the human germline V k L6 amino acid sequence (SEQ IDNO:23).

Figure 6 shows the alignment of the amino acid sequence of the light chain variable region of 8Al with the human germline V k Ll 8 amino acid sequence (SEQ ID NO:24).

Figure 7A-C shows the results of experiments demonstrating that the human monoclonal antibodies, 9Dl 2 and 8Al, directed against human IRTA-2, specifically bind to human IRTA-2. Figure 7A is a bar graph showing binding to CHO cells transfected with IRTA-2. Figure 7B is a bar graph showing lack of binding to CHO cells transfected with IRTA-3. Figure 7C is a bar graph showing lack of binding to CHO cells transfected with IRTA-4.

Figure 8 shows the results of flow cytometry experiments demonstrating that the human monoclonal antibodies 9Dl 2 and 8Al, directed against human IRTA-2, bind to CD19+ B cells.

Figure 9 shows the results of flow cytometry experiments demonstrating that the human monoclonal antibodies, 9Dl 2 and 8Al, directed against human IRTA-2, do not bind to the cell surface of the B-cell tumor cell lines Ramos, Raji, Daudi, or IM-9.

Figure 10 shows the results of flow cytometry experiments demonstrating binding of the human monoclonal antibodies 9Dl 2 and 8Al, directed against human IRTA-2, to the B- cell tumor lines Karpas 1106P, SU-DHL-4, and Granta 519.

Figure 11 shows the results of flow cytometry experiments demonstrating binding of the human monoclonal antibodies 9D12 and 8Al, directed against human IRTA-2, to the B- cell tumor lines SU-DHL-6 and JEKO-I.

Detailed Description of the Invention

The present invention relates to isolated monoclonal antibodies, particularly human monoclonal antibodies, that bind specifically to IRTA-2 with high affinity. In certain embodiments, the antibodies of the invention are derived from particular heavy and light chain germline sequences and/or comprise particular structural features such as CDR regions comprising particular amino acid sequences. The invention provides isolated antibodies, methods of making such antibodies, immunoconjugates and bispecifϊc molecules comprising such antibodies and pharmaceutical compositions containing the antibodies, immuncoηjugates or bispecific molecules of the invention. The invention also relates to methods of using the antibodies, such as to detect IRTA-2, as well as to treat diseases associated with expression of IRTA-2, such as B cell malignancies that express IRTA-2. Accordingly, the invention also provides methods of using the anti -IRTA-2 antibodies of the invention to treat B cell malignancies, for example, in the treatment of non-Hodgkin's lymphoma, chronic lymphocytic leukemias, follicular lymphomas, diffuse large cell lymphomas of B lineage, and multiple myelomas.

In order that the present invention may be more readily understood, certain terms are first defined. Additional definitions are set forth throughout the detailed description.

The terms "immunoglobulin superfamily receptor translocation associated gene 2" and "IRTA-2" are used interchangeably, and include variants, isoforms and species homologs of human IRTA-2. Accordingly, human antibodies of the invention may, in certain cases, cross-react with IRTA-2 from species other than human. In other cases, the antibodies may

be completely specific for human IRTA-2 and may not exhibit species or other types of cross-reactivity. The complete amino acid sequence of human IRTA-2 has Genbank accession number NPJ 12571 (SEQ ID NO: 25).

The terms "IRTA-I", "IRTA-3", "IRTA-4", and "IRTA-5" include variants, isoforms and species homologs of human "IRTA-I", "IRTA-3", "IRTA-4", and "IRTA-5", respectively. The complete amino acid sequence of human IRTA-I has Genbank accession number NP_112572 (SEQ ID NO: 26). The complete amino acid sequence of human IRTA- 3 has Genbank accession number AAL59390 (SEQ ID NO: 27). The complete amino acid sequence of human IRTA-4 has Genbank accession number AAL60249 (SEQ ID NO: 28). The complete amino acid sequence of human IRTA-5 has Genbank accession number AAL60250 (SEQ ID NO: 29).

The term "immune response" refers to the action of, for example, lymphocytes, antigen presenting cells, phagocytic cells, granulocytes, and soluble macromolecules produced by the above cells or the liver (including antibodies, cytokines, and complement) that results in selective damage to, destruction of, or elimination from the human body of invading pathogens, cells or tissues infected with pathogens, cancerous cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues.

A "signal transduction pathway" refers to the biochemical relationship between a variety of signal transduction molecules that play a role in the transmission of a signal from one portion of a cell to another portion of a cell. As used herein, the phrase "cell surface receptor" includes, for example, molecules and complexes of molecules capable of receiving a signal and the transmission of such a signal across the plasma membrane of a cell. An example of a "cell surface receptor" of the present invention is the IRTA-2 receptor.

The term "antibody" as referred to herein includes whole antibodies and any antigen binding fragment (i.e., "antigen-binding portion") or single chains thereof. An "antibody" refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CHI, CH 2 and CH 3 . Each light chain is comprised of a light chain variable region (abbreviated herein as V L ) and a light chain constant region. The light chain constant region is comprised of one domain, CL- The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is

composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FRl, CDRl, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (CIq) of the classical complement system.

The term "antigen-binding portion" of an antibody (or simply "antibody portion"), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., IRTA-2). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term "antigen-binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al, (1989) Nature 341:544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sd. USA 85:5879-5883, ) . Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.

An "isolated antibody", as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds IRTA-2 is substantially free of antibodies that specifically bind antigens other than IRTA-2). An isolated antibody that specifically binds IRTA-2 may, however, have cross-reactivity to other antigens, such as IRTA-2 molecules from other species. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.

The terms "monoclonal antibody" or "monoclonal antibody composition" as used herein refer to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.

The term "human antibody", as used herein, is intended to include antibodies having variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences. Furthermore, if the antibody contains a constant region, the constant region also is derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site- specific mutagenesis in vitro or by somatic mutation in vivo). However, the term "human antibody", as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.

The term "human monoclonal antibody" refers to antibodies displaying a single binding specificity which have variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences. In one embodiment, the human monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell.

The term "recombinant human antibody", as used herein, includes all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom (described further below), (b) antibodies isolated from a host cell transformed to express the human antibody, e.g., from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial human antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable regions in which the framework and CDR regions are derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies can be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant

antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.

As used herein, "isotype" refers to the antibody class (e.g., IgM or IgGl) that is encoded by the heavy chain constant region genes.

The phrases "an antibody recognizing an antigen" and "an antibody specific for an antigen" are used interchangeably herein with the term "an antibody which binds specifically to an antigen."

The term "human antibody derivatives" refers to any modified form of the human antibody, e.g., a conjugate of the antibody and another agent or antibody.

The term "humanized antibody" is intended to refer to antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. Additional framework region modifications may be made within the human framework sequences.

The term "chimeric antibody" is intended to refer to antibodies in which the variable region sequences are derived from one species and the constant region sequences are derived from another species, such as an antibody in which the variable region sequences are derived from a mouse antibody and the constant region sequences are derived from a human antibody.

As used herein, an antibody that "specifically binds to human IRTA-2" is intended to refer to an antibody that binds to human IRTA-2 with a KD of 1 x 10 "7 M or less, more preferably 5 x 10 "8 M or less, more preferably 3 x 10 "8 M or less, more preferably 1 x 10 "8 M or less, even more preferably 5 x 10 "9 M or less.

The term "K assoc " or "K 8 ", as used herein, is intended to refer to the association rate of a particular antibody-antigen interaction, whereas the term "K^ 3 " or "K d ," as used herein, is intended to refer to the dissociation rate of a particular antibody-antigen interaction. The term "KD", as used herein, is intended to refer to the dissociation constant, which is obtained from the ratio of Kd to K a (Ie,. K d /K a ) and is expressed as a molar concentration (M). K D values for antibodies can be determined using methods well established in the art. A preferred method for determining the KD of an antibody is by using surface plasmon resonance, preferably using a biosensor system such as a Biacore® system.

As used herein, the term "high affinity" for an IgG antibody refers to an antibody having a KD of 1 x 10 "7 M or less, more preferably 5 x 10 "8 M or less and even more preferably 5 x 10 "9 M or less for a target antigen. However, "high affinity" binding can vary

for other antibody isotypes. For example, "high affinity" binding for an IgM isotype refers to an antibody having a KD of 10 '6 M or less, more preferably 10 "7 M or less, even more preferably 10 "8 M or less.

As used herein, the term "subject" includes any human or nonhuman animal. The term "nonhuman animal" includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows chickens, amphibians, reptiles, etc.

Various aspects of the invention are described in further detail in the following subsections.

Anti-IRTA-2 Antibodies

The antibodies of the invention are characterized by particular functional features or properties of the antibodies. For example, the antibodies bind specifically to human IRTA-2. Preferably, an antibody of the invention binds to IRTA-2 with high affinity, for example with a KD of 1 x 10 "7 M or less. The anti-IRTA-2 antibodies of the invention preferably exhibit one or more of the following characteristics:

(a) binds to human IRTA-2 with a KD of IxIO "7 M or less;

(b) binds to human CHO cells transfected with IRTA-2;

(c) does not substantially bind to human IRTA-3 or IRTA-4; and/or

(d) binds to Granta 519 tumor cells and does not substantially bind to Raji or Ramos tumor cells.

Preferrably, the antibody binds to human IRTA-2 with a K D of 5 x 10 "8 M or less, binds to human IRTA-2 with a K D of 2 x 10 "8 M or less, binds to human IRTA-2 with a K D of 5x10 "9 M or less, binds to human IRTA-2 with a KD of 4x10 "9 M or less, binds to human IRTA-2 with a K D of 3xlO "9 M or less, or binds to human IRTA-2 with a K D of 2.1 x 10 "9 M or less.

In a preferred embodiment, the antibody does not substantially bind to Daudi, IM-9, Karpas 1106P, or SU-DHL-4 tumor cells.

Standard assays to evaluate the binding ability of the antibodies toward IRTA-2 are known in the art, including for example, ELISAs, Western blots, RIAs, and flow cytometry analysis. Suitable assays are described in detail in the Examples. The binding kinetics (e.g., binding affinity) of the antibodies also can be assessed by standard assays known in the art, such as by Biacore analysis.

Monoclonal Antibodies 9Dl 2 and 8Al

Preferred antibodies of the invention are the human monoclonal antibodies 9Dl 2 and 8Al, isolated and structurally characterized as described in Examples 1 and 2. The VH amino acid sequences of 9D12 and 8Al are shown in SEQ ID NOs: 13 and 14, respectively. The VL amino acid sequences of 9D12 and 8Al are shown in SEQ ID NOs: 15 and 16, respectively.

Given that each of these antibodies can bind to IRT A-2, the VH and VL sequences can be "mixed and matched" to create other anti-IRTA-2 binding molecules of the invention. IRTA-2 binding of such "mixed and matched" antibodies can be tested using the binding assays described above and in the Examples (e.g., ELISAs). Preferably, when VH and VL chains are mixed and matched, a VH sequence from a particular VH/VL pairing is replaced with a structurally similar VH sequence. Likewise, preferably a VL sequence from a particular VH/VL pairing is replaced with a structurally similar VL sequence.

Accordingly, in one aspect, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof comprising:

(a) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 13 and 14; and

(b) a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 15 and 16; wherein the antibody specifically binds IRTA-2, preferably human IRTA-2. Preferred heavy and light chain combinations include:

(a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 13; and (b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15; or

(a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 14; and (b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 16.

In another aspect, the invention provides antibodies that comprise the heavy chain and light chain CDRIs, CDR2s and CDR3s of 9Dl 2 and 8Al, or combinations thereof. The amino acid sequences of the VH CDRIS of 9D12 and 8Al are shown in SEQ ID NOs: 1 and 2. The amino acid sequences of the VH CDR2S of 9Dl 2 and 8Al are shown in SEQ ID NOs: 3 and 4. The amino acid sequences of the V H CDR3s of 9D12 and 8Al are shown in SEQ ID NOs: 5 and 6. The amino acid sequences of the V k CDRIs of 9D12 and 8Al are shown in SEQ ID NOs: 7 and 8. The amino acid sequences of the V k CDR2s of 9D12 and 8Al are shown in SEQ ID NOs: 9 and 10. The amino acid sequences of the V k CDR3s of 9D12 and

8Al are shown in SEQ ID NOs: 1 1 and 12. The CDR regions are delineated using the Kabat system (Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242).

Given that each of these antibodies can bind to IRT A-2 and that antigen-binding specificity is provided primarily by the CDRl, CDR2, and CDR3 regions, the V H CDRl, CDR2, and CDR3 sequences and V k CDRl, CDR2, and CDR3 sequences can be "mixed and matched" (i.e., CDRs from different antibodies can be mixed and match, although each antibody must contain a V H CDRl, CDR2, and CDR3 and a V k CDRl, CDR2, and CDR3) to create other anti-IRTA-2 binding molecules of the invention. IRTA-2 binding of such "mixed and matched" antibodies can be tested using the binding assays described above and in the Examples {e.g., ELISAs, Biacore analysis). Preferably, when VH CDR sequences are mixed and matched, the CDRl, CDR2 and/or CDR3 sequence from a particular VH sequence is replaced with a structurally similar CDR sequence(s). Likewise, when V k CDR sequences are mixed and matched, the CDRl, CDR2 and/or CDR3 sequence from a particular V k sequence preferably is replaced with a structurally similar CDR sequence(s). It will be readily apparent to the ordinarily skilled artisan that novel VH and VL sequences can be created by substituting one or more VH and/or VL CDR region sequences with structurally similar sequences from the CDR sequences disclosed herein for monoclonal antibodies antibodies 9Dl 2 and 8Al.

Accordingly, in another aspect, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof comprising:

(a) a heavy chain variable region CDRl comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 and 2;

(b) a heavy chain variable region CDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 3 and 4;

(c) a heavy chain variable region CDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5 and 6;

(d) a light chain variable region CDRl comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 7 and 8;

(e) a light chain variable region CDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 9 and 10; and

(f) a light chain variable region CDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 11 and 12; wherein the antibody specifically binds IRTA-2, preferably human IRTA-2.

In a preferred embodiment, the antibody comprises:

(a) a heavy chain variable region CDRl comprising SEQ ID NO: 1;

(b) a heavy chain variable region CDR2 comprising SEQ ID NO: 3;

(c) a heavy chain variable region CDR3 comprising SEQ ID NO: 5;

(d) a light chain variable region CDRl comprising SEQ ID NO: 7;

(e) a light chain variable region CDR2 comprising SEQ ID NO: 9; and

(f) a light chain variable region CDR3 comprising SEQ ID NO: 11. In another preferred embodiment, the antibody comprises:

(a) a heavy chain variable region CDRl comprising SEQ ID NO: 2;

(b) a heavy chain variable region CDR2 comprising SEQ ID NO: 4;

(c) a heavy chain variable region CDR3 comprising SEQ ID NO: 6;

(d) a light chain variable region CDRl comprising SEQ ID NO: 8;

(e) a light chain variable region CDR2 comprising SEQ ID NO: 10; and

(f) a light chain variable region CDR3 comprising SEQ ID NO: 12.

Antibodies Having Particular Germline Sequences

In certain embodiments, an antibody of the invention comprises a heavy chain variable region from a particular germline heavy chain immunoglobulin gene and/or a light chain variable region from a particular germline light chain immunoglobulin gene.

For example, in a preferred embodiment, the invention provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a heavy chain variable region that is the product of or derived from a human VH 3-23 gene, wherein the antibody specifically binds IRT A-2. In another preferred embodiment, the invention provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a heavy chain variable region that is the product of or derived from a human VH 1-8 gene, wherein the antibody specifically binds IRT A-2. In yet another preferred embodiment, the invention provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a light chain variable region that is the product of or derived from a human VK L6 gene, wherein the antibody specifically binds IRT A-2. In yet another preferred embodiment, the invention provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a light chain variable region that is the product of or derived from a human VK Ll 8 gene, wherein the antibody specifically binds IRTA-2. In yet another preferred embodiment, the invention provides an isolated monoclonal antibody, or antigen- binding portion thereof, wherein the antibody:

(a) comprises a heavy chain variable region that is the product of or derived from a human VH 3-23 or 1-8 gene (which genes encode the amino acid sequences set forth in SEQ ID NOs: 21 and 22, respectively);

(b) comprises a light chain variable region that is the product of or derived from a human VK L6 or VK Ll 8 gene (which genes encode the amino acid sequences set forth in SEQ ID NOs: 23 and 24, respectively); and

(c) specifically binds to IRTA-2, preferably human IRTA-2.

An example of an antibody having VH and VK of VH 3-23 and VK L6, respectively, is 9D12. An example of an antibody having VH and VK of VH 1-8 and VK L18, respectively, is 8Al.

As used herein, a human antibody comprises heavy or light chain variable regions that is "the product of or "derived from" a particular germline sequence if the variable regions of the antibody are obtained from a system that uses human germline immunoglobulin genes. Such systems include immunizing a transgenic mouse carrying human immunoglobulin genes with the antigen of interest or screening a human immunoglobulin gene library displayed on phage with the antigen of interest. A human antibody that is "the product of or "derived from" a human germline immunoglobulin sequence can be identified as such by comparing the amino acid sequence of the human antibody to the amino acid sequences of human germline immunoglobulins and selecting the human germline immunoglobulin sequence that is closest in sequence (i.e., greatest % identity) to the sequence of the human antibody. A human antibody that is "the product of or "derived from" a particular human germline immunoglobulin sequence may contain amino acid differences as compared to the germline sequence, due to, for example, naturally-occurring somatic mutations or intentional introduction of site-directed mutation. However, a selected human antibody typically is at least 90% identical in amino acids sequence to an amino acid sequence encoded by a human germline immunoglobulin gene and contains amino acid residues that identify the human antibody as being human when compared to the germline immunoglobulin amino acid sequences of other species (e.g., murine germline sequences). In certain cases, a human antibody may be at least 95%, or even at least 96%, 97%, 98%, or 99% identical in amino acid sequence to the amino acid sequence encoded by the germline immunoglobulin gene. Typically, a human antibody derived from a particular human germline sequence will display no more than 10 amino acid differences from the amino acid sequence encoded by the human germline immunoglobulin gene. In certain cases, the human antibody may display no more

than 5, or even no more than 4, 3, 2, or 1 amino acid difference from the amino acid sequence encoded by the germline immunoglobulin gene.

Homologous Antibodies

In yet another embodiment, an antibody of the invention comprises heavy and light chain variable regions comprising amino acid sequences that are homologous to the amino acid sequences of the preferred antibodies described herein, and wherein the antibodies retain the desired functional properties of the anti-IRTA-2 antibodies of the invention.

For example, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof, comprising a heavy chain variable region and a light chain variable region, wherein:

(a) the heavy chain variable region comprises an amino acid sequence that is at least 80% homologous to an amino acid sequence selected from the group consisting of SEQ ID NOs: 13 and 14;

(b) the light chain variable region comprises an amino acid sequence that is at least 80% homologous to an amino acid sequence selected from the group consisting of SEQ ID NOs: 15 and 16;

(c) the antibody binds to human IRTA-2 with a K D of 1x10 "7 M or less;

(d) the antibody binds to human CHO cells transfected with IRTA-2;

(e) the antibody does not substantially bind to human IRTA-3 or IRTA-4; and

(f) the antibody binds to Granta 519 tumor cells and does not substantially bind to Raji or Ramos tumor cells.

In various embodiments, the antibody can be, for example, a human antibody, a humanized antibody or a chimeric antibody.

In a preferred embodiment, the antibody does not substantially bind to Daudi, IM-9, Karpas 1106P, or SU-DHL-4 tumor cells.

In other embodiments, the VH and/or V L amino acid sequences may be 85%, 90%, 95%, 96%, 97%, 98% or 99% homologous to the sequences set forth above. An antibody having VH and VL regions having high (i.e., 80% or greater) homology to the VH and V L regions of the sequences set forth above, can be obtained by mutagenesis (e.g., site-directed or PCR-mediated mutagenesis) of nucleic acid molecules encoding SEQ ID NOs: 17, 18, 19, and 20, followed by testing of the encoded altered antibody for retained function (i.e., the functions set forth in (c) through (f) above) using the functional assays described herein.

As used herein, the percent homology between two amino acid sequences is equivalent to the percent identity between the two sequences. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences {i.e., % homology = # of identical positions/total # of positions x 100), taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described in the non-limiting examples below.

The percent identity between two amino acid sequences can be determined using the algorithm of E. Meyers and W. Miller {Comput. Appl. Biosci., 4:11-17 (1988)) which has been incorporated into the ALIGN program (version 2.0), using a PAM 120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. In addition, the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (J. MoI. Biol. 48:444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.

Additionally or alternatively, the protein sequences of the present invention can further be used as a "query sequence" to perform a search against public databases to, for example, identify related sequences. Such searches can be performed using the XBLAST program (version 2.0) of Altschul, et al. (1990) J. MoI. Biol. 211:403-10. BLAST protein searches can be performed with the XBLAST program, score = 50, wordlength = 3 to obtain amino acid sequences homologous to the antibody molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al, (1997) Nucleic Acids Res. 25(17):3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. See http://www.ncbi.nlm.nih.gov.

Antibodies with Conservative Modifications

In certain embodiments, an antibody of the invention comprises a heavy chain variable region comprising CDRl, CDR2 and CDR3 sequences and a light chain variable region comprising CDRl, CDR2 and CDR3 sequences, wherein one or more of these CDR sequences comprise specified amino acid sequences based on the preferred antibodies described herein (e.g., 9D12 or 8Al), or conservative modifications thereof, and wherein the

antibodies retain the desired functional properties of the anti-IRTA-2 antibodies of the invention. Accordingly, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof, comprising a heavy chain variable region comprising CDRl 5 CDR2, and CDR3 sequences and a light chain variable region comprising CDRl, CDR2, and CDR3 sequences, wherein:

(a) the heavy chain variable region CDR3 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 5 and 6, and conservative modifications thereof;

(b) the light chain variable region CDR3 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequence of SEQ ID NOs: 11 and 12, and conservative modifications thereof;

(c) the antibody binds to human IRTA-2 with a KD of 1x10 "7 M or less;

(d) the antibody binds to human CHO cells transfected with IRTA-2;

(e) the antibody does not substantially bind to human IRTA-3 or IRTA-4; and

(f) the antibody binds to Granta 519 tumor cells and does not substantially bind to Raji or Ramos tumor cells.

In a preferred embodiment, the heavy chain variable region CDR2 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 3 and 4, and conservative modifications thereof; and the light chain variable region CDR2 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 9 and 10, and conservative modifications thereof. In another preferred embodiment, the heavy chain variable region CDRl sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 1 and 2, and conservative modifications thereof; and the light chain variable region CDRl sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 7 and 8, and conservative modifications thereof.

In various embodiments, the antibody can be, for example, human antibodies, humanized antibodies or chimeric antibodies.

In a preferred embodiment, the antibody does not substantially bind to Daudi, IM-9, Karpas 1106P, or SU-DHL-4 tumor cells.

As used herein, the term "conservative sequence modifications" is intended to refer to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequence. Such conservative modifications include

amino acid substitutions, additions and deletions. Modifications can be introduced into an antibody of the invention by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, one or more amino acid residues within the CDR regions of an antibody of the invention can be replaced with other amino acid residues from the same side chain family and the altered antibody can be tested for retained function (i.e., the functions set forth in (c) through (f) above) using the functional assays described herein.

Antibodies that Bind to the Same Epitope as Anti-IRTA-2 Antibodies of the Invention In another embodiment, the invention provides antibodies that bind to the same epitope on human IRTA-2 as any of the IRT A-2 monoclonal antibodies of the invention (i.e., antibodies that have the ability to cross-compete for binding to IRTA-2 with any of the monoclonal antibodies of the invention). In preferred embodiments, the reference antibody for cross-competition studies can be the monoclonal antibody 9Dl 2 (having VH and VL sequences as shown in SEQ ID NOs: 13 and 15, respectively), or the monoclonal antibody 8Al (having VH and VL sequences as shown in SEQ ID NOs: 14 and 16, respectively). Such cross-competing antibodies can be identified based on their ability to cross-compete with 9D12 or 8Al in standard IRTA-2 binding assays. For example, BIAcore analysis, ELISA assays or flow cytometry may be used to demonstrate cross-competition with the antibodies of the current invention. The ability of a test antibody to inhibit the binding of, for example, 9D12 or 8Al, to human IRTA-2 demonstrates that the test antibody can compete with 9D12 or 8Al for binding to human IRTA-2 and thus binds to the same epitope on human IRTA-2 as 9Dl 2 or 8Al . In a preferred embodiment, the antibody that binds to the same epitope on human IRTA-2 as 9Dl 2 or 8Al is a human monoclonal antibody. Such human monoclonal antibodies can be prepared and isolated as described in the Examples.

Engineered and Modified Antibodies

An antibody of the invention further can be prepared using an antibody having one or more of the VH and/or VL sequences disclosed herein as starting material to engineer a modified antibody, which modified antibody may have altered properties from the starting antibody. An antibody can be engineered by modifying one or more residues within one or both variable regions (i.e., Y 11 and/or VL), for example within one or more CDR regions and/or within one or more framework regions. Additionally or alternatively, an antibody can be engineered by modifying residues within the constant region(s), for example to alter the effector function(s) of the antibody.

One type of variable region engineering that can be performed is CDR grafting. Antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarity determining regions (CDRs). For this reason, the amino acid sequences within CDRs are more diverse between individual antibodies than sequences outside of CDRs. Because CDR sequences are responsible for most antibody-antigen interactions, it is possible to express recombinant antibodies that mimic the properties of specific naturally occurring antibodies by constructing expression vectors that include CDR sequences from the specific naturally occurring antibody grafted onto framework sequences from a different antibody with different properties (see, e.g., Riechmann, L. et al. (1998) Nature 332:323-327; Jones, P. et al. (1986) Nature 321:522-525; Queen, C. et al. (1989) Proc. Natl. Acad. See. U.S.A. 86:10029-10033; U.S. Patent No. 5,225,539 to Winter, and U.S. Patent Nos. 5,530,101; 5,585,089; 5,693,762 and 6,180,370 to Queen et al.)

Accordingly, another embodiment of the invention pertains to an isolated monoclonal antibody, or antigen binding portion thereof, comprising a heavy chain variable region comprising CDRl, CDR2, and CDR3 sequences comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 and 2, SEQ ID NOs: 3 and 4, and SEQ ID NOs: 5 and 6, respectively, and a light chain variable region comprising CDRl, CDR2, and CDR3 sequences comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 7 and 8, SEQ ID NOs: 9 and 10, and SEQ ID NOs: 11 and 12, respectively. Thus, such antibodies contain the VH and V L CDR sequences of monoclonal antibodies 9Dl 2 or 8Al yet may contain different framework sequences from these antibodies.

Such framework sequences can be obtained from public DNA databases or published references that include germline antibody gene sequences. For example, germline DNA sequences for human heavy and light chain variable region genes can be found in the

"VBase" human germline sequence database (available on the Internet at www.mrc- cpe.cam.ac.uk/vbase). as well as in Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242; Tomlinson, I. M., et al (1992) "The Repertoire of Human Germline VH Sequences Reveals about Fifty Groups of VH Segments with Different Hypervariable Loops" J MoI. Biol. 227:776-79$; and Cox, J. P. L. et al. (1994) "A Directory of Human Germ-line VH Segments Reveals a Strong Bias in their Usage" Eur. J. Immunol. 24:827-836; the contents of each of which are expressly incorporated herein by reference. As another example, the germline DNA sequences for human heavy and light chain variable region genes can be found in the Genbank database. For example, the following heavy chain germline sequences found in the HCo7 HuMAb mouse are available in the accompanying Genbank accession numbers: 1-69 (NG_0010109, NT_024637 and BC070333), 3-33 (NG_0010109 and NT_024637) and 3-7 (NG_0010109 and NT_024637). As another example, the following heavy chain germline sequences found in the HCo 12 HuMAb mouse are available in the accompanying Genbank accession numbers: 1 -69 (NG_0010109, NT_024637 and BC070333), 5-51 (NG_0010109 and NT_024637), 4-34 (NG_0010109 and NT_024637), 3-30.3 (?) and 3-23 (AJ406678).

Preferred framework sequences for use in the antibodies of the invention are those that are structurally similar to the framework sequences used by selected antibodies of the invention, e.g., similar to the VH 3-23 framework sequences (SEQ ID NO: 21) and/or the VH 1-8 framework sequences (SEQ ID NO: 22) and/or the VK L6 framework sequences (SEQ ID NO: 23) and/or the V k Ll 8 framework sequence (SEQ ID NO: 24) used by preferred monoclonal antibodies of the invention. The VH CDRl, CDR2, and CDR3 sequences, and the VK CDRl, CDR2, and CDR3 sequences, can be grafted onto framework regions that have the identical sequence as that found in the germline immunoglobulin gene from which the framework sequence derive, or the CDR sequences can be grafted onto framework regions that contain one or more mutations as compared to the germline sequences. For example, it has been found that in certain instances it is beneficial to mutate residues within the framework regions to maintain or enhance the antigen binding ability of the antibody (see e.g., U.S. Patent Nos. 5,530,101; 5,585,089; 5,693,762 and 6,180,370 to Queen et al).

Another type of variable region modification is to mutate amino acid residues within the VH and/or VK CDRl, CDR2 and/or CDR3 regions to thereby improve one or more binding properties (e.g., affinity) of the antibody of interest. Site-directed mutagenesis or PCR-mediated mutagenesis can be performed to introduce the mutation(s) and the effect on

antibody binding, or other functional property of interest, can be evaluated in in vitro or in vivo assays as described herein and provided in the Examples. Preferably conservative modifications (as discussed above) are introduced. The mutations may be amino acid substitutions, additions or deletions, but are preferably substitutions. Moreover, typically no more than one, two, three, four or five residues within a CDR region are altered.

Accordingly, in another embodiment, the invention provides isolated anti-IRTA-2 monoclonal antibodies, or antigen binding portions thereof, comprising a heavy chain variable region comprising: (a) a VH CDRl region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 and 2, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 1 and 2; (b) a VH CDR2 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 3 and 4, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 3 and 4; (c) a VH CDR3 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5 and 6, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 5 and 6; (d) a VK CDRl region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 7 and 8, or an amino acid sequence having one, two, three, four or five amino ac\d substitutions, deletions or additions as compared to SEQ ID NOs: 7 and 8; (e) a VK CDR2 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 9 and 10, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 9 and 10; and (f) a VK CDR3 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 11 and 12, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 11 and 12.

Engineered antibodies of the invention include those in which modifications have been made to framework residues within VH and/or VR, e.g. to improve the properties of the antibody. Typically such framework modifications are made to decrease the immunogenicity of the antibody. For example, one approach is to "backmutate" one or more framework residues to the corresponding germline sequence. More specifically, an antibody that has undergone somatic mutation may contain framework residues that differ from the germline sequence from which the antibody is derived. Such residues can be identified by comparing the antibody framework sequences to the germline sequences from which the antibody is

derived. For example, for 9D12, amino acid residue #88 (within FR3) of VH is a valine whereas this residue in the corresponding VH 3-23 germline sequence is an alanine. To return the framework region sequences to their germline configuration, the somatic mutations can be "backmutated" to the germline sequence by, for example, site-directed mutagenesis or PCR-mediated mutagenesis (e.g., residue #88 (residue #22 of FR3) of the V H of 9Dl 2 can be "backmutated" from valine to alanine).

As another example, for 9D12, amino acid residue U9\ (within FR3) of VH is an alanine whereas this residue in the corresponding VH 3-23 germline sequence is a threonine. To return the framework region sequences to their germline configuration, for example, residue #91 (reside #25 of FR3) of the V H of 9D12 can be "backmutated" from alanine to threonine. Such "backmutated" antibodies are also intended to be encompassed by the invention.

As another example, for 9Dl 2, amino acid residue #93 (within FR3) of VH is a leucine whereas this residue in the corresponding VH 3-23 germline sequence is a valine. To return the framework region sequences to their germline configuration, for example, residue #93 (residue #27 of FR3) of the V H of 9D12 can be "backmutated" from leucine to valine. Such "backmutated" antibodies are also intended to be encompassed by the invention.

As yet another example, for 8Al, amino acid residue #2 (within FRl) of VH is a methionine whereas this residue in the corresponding VH 1-8 germline sequence is a valine. To return the framework region sequences to their germline configuration, for example, residue #2 within FRl of the VH of 8Al can be "backmutated" from methionine to valine. Such "backmutated" antibodies are also intended to be encompassed by the invention.

As yet another example, for 8Al, amino acid residue #30 (within FRl) of VH is a isoleucine whereas this residue in the corresponding VH 1-8 germline sequence is a threonine. To return the framework region sequences to their germline configuration, for example, residue #30 within FRl of the VH of 8Al can be "backmutated" from isoleucine to threonine. Such "backmutated" antibodies are also intended to be encompassed by the invention.

Another type of framework modification involves mutating one or more residues within the framework region, or even within one or more CDR regions, to remove T cell epitopes to thereby reduce the potential immunogenicity of the antibody. This approach is also referred to as "deimmunization" and is described in futher detail in U.S. Patent Publication No. 20030153043 by Carr et al.

In addition or alternative to modifications made within the framework or CDR regions, antibodies of the invention may be engineered to include modifications within the Fc

region, typically to alter one or more functional properties of the antibody, such as serum half-life, complement Fixation, Fc receptor binding, and/or antigen-dependent cellular cytotoxicity. Furthermore, an antibody of the invention may be chemically modified {e.g., one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation, again to alter one or more functional properties of the antibody. Each of these embodiments is described in further detail below. The numbering of residues in the Fc region is that of the EU index of Kabat.

In one embodiment, the hinge region of CHl is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased. This approach is described further in U.S. Patent No. 5,677,425 by Bodmer et al. The number of cysteine residues in the hinge region of CHl is altered to, for example, facilitate assembly of the light and heavy chains or to increase or decrease the stability of the antibody.

In another embodiment, the Fc hinge region of an antibody is mutated to decrease the biological half life of the antibody. More specifically, one or more amino acid mutations are introduced into the CH2-CH3 domain interface region of the Fc-hinge fragment such that the antibody has impaired Staphylococcyl protein A (SpA) binding relative to native Fc-hinge domain SpA binding. This approach is described in further detail in U.S. Patent No. 6,165,745 by Ward et al.

In another embodiment, the antibody is modified to increase its biological half life. Various approaches are possible. For example, one or more of the following mutations can be introduced: T252L, T254S, T256F, as described in U.S. Patent No. 6,277,375 to Ward. Alternatively, to increase the biological half life, the antibody can be altered within the CHl or CL region to contain a salvage receptor binding epitope taken from two loops of a CH2 domain of an Fc region of an IgG, as described in U.S. Patent Nos. 5,869,046 and 6,121,022 by Presta et al.

In yet other embodiments, the Fc region is altered by replacing at least one amino acid residue with a different amino acid residue to alter the effector function(s) of the antibody. For example, one or more amino acids selected from amino acid residues 234, 235, 236, 237, 297, 318, 320 and 322 can be replaced with a different amino acid residue such that the antibody has an altered affinity for an effector ligand but retains the antigen-binding ability of the parent antibody. The effector ligand to which affinity is altered can be, for example, an Fc receptor or the Cl component of complement. This approach is described in further detail in U.S. Patent Nos. 5,624,821 and 5,648,260, both by Winter et al.

In another example, one or more amino acids selected from amino acid residues 329, 331 and 322 can be replaced with a different amino acid residue such that the antibody has altered CIq binding and/or reduced or abolished complement dependent cytotoxicity (CDC). This approach is described in further detail in U.S. Patent Nos. 6,194,551 by Idusogie et al.

In another example, one or more amino acid residues within amino acid positions 231 and 239 are altered to thereby alter the ability of the antibody to fix complement. This approach is described further in PCT Publication WO 94/29351 by Bodmer et al.

In yet another example, the Fc region is modified to increase the ability of the antibody to mediate antibody dependent cellular cytotoxicity (ADCC) and/or to increase the affinity of the antibody for an Fcγ receptor by modifying one or more amino acids at the following positions: 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 301, 303, 305, 307, 309, 312, 315, 320, 322, 324, 326, 327, 329, 330, 331, 333, 334, 335, 337, 338, 340, 360, 373, 376, 378, 382, 388, 389, 398, 414, 416, 419, 430, 434, 435, 437, 438 or 439. This approach is described further in PCT Publication WO 00/42072 by Presta. Moreover, the binding sites on human IgGl for FcγRl, FcγRJI, FcγRIII and FcRn have been mapped and variants with improved binding have been described (see Shields, R.L. et al. (2001) J. Biol. Chem. 276:6591-6604). Specific mutations at positions 256, 290, 298, 333, 334 and 339 were shown to improve binding to FcγRIII. Additionally, the following combination mutants were shown to improve FcγRIII binding: T256A/S298A, S298A/E333A, S298A/K224A and S298A/E333A/K334A.

In still another embodiment, the glycosylation of an antibody is modified. For example, an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation). Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen. Such carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence. For example, one or more amino acid substitutions can be made that result in elimination of one or more variable region framework glycosylation sites to thereby eliminate glycosylation at that site. Such aglycosylation may increase the affinity of the antibody for antigen. Such an approach is described in further detail in U.S. Patent Nos. 5,714,350 and 6,350,861 by Co et al.

Additionally or alternatively, an antibody can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GlcNac structures. Such altered

glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies. Such carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies of the invention to thereby produce an antibody with altered glycosylation. For example, the cell lines Ms 704, Ms705, and Ms709 lack the fucosyltransferase gene, FUT8 (alpha (1,6) fucosyltransferase), such that antibodies expressed in the Ms704, Ms705, and Ms709 cell lines lack fucose on their carbohydrates. The Ms704, Ms705, and Ms709 FUT8 "7" cell lines were created by the targeted disruption of the FUT8 gene in CHO/DG44 cells using two replacement vectors (see U.S. Patent Publication No. 20040110704 by Yamane et al and Yamane-Ohnuki et al. (2004) Biotechnol Bioeng 87:614-22). As another example, EP 1,176,195 by Hanai et al. describes a cell line with a functionally disrupted FUT8 gene, which encodes a fucosyl transferase, such that antibodies expressed in such a cell line exhibit hypofucosylation by reducing or eliminating the alpha 1,6 bond-related enzyme. Hanai et al. also describe cell lines which have a low enzyme activity for adding fucose to the N-acetylglucosamine that binds to the Fc region of the antibody or does not have the enzyme activity, for example the rat myeloma cell line YB2/0 (ATCC CRL 1662). PCT Publication WO 03/035835 by Presta describes a variant CHO cell line, Led 3 cells, with reduced ability to attach fucose to Asn(297)-linked carbohydrates, also resulting in hypofucosylation of antibodies expressed in that host cell (see also Shields, R.L. et al. (2002) J. Biol. Chem. TTJ:2βm,-261AQι). PCT Publication WO 99/54342 by Umana et al. describes cell lines engineered to express glycoprotein-modifying glycosyl transferases {e.g., beta(l,4)-N-acetylglucosaminyltransferase III (GnTIII)) such that antibodies expressed in the engineered cell lines exhibit increased bisecting GlcNac structures which results in increased ADCC activity of the antibodies (see also Umana et al (1999) Nat. Biotech. YV.176-180). Alternatively, the fucose residues of the antibody may be cleaved off using a fucosidase enzyme. For example, the fucosidase alpha-L-fucosidase removes fucosyl residues from antibodies (Tarentino, A.L. et al. (1975) Biochem. 14:5516- 23).

Another modification of the antibodies herein that is contemplated by the invention is pegylation. An antibody can be pegylated to, for example, increase the biological {e.g., serum) half life of the antibody. To pegylate an antibody, the antibody, or fragment thereof, typically is reacted with polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to

the antibody or antibody fragment. Preferably, the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer). As used herein, the term "polyethylene glycol" is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (Cl-ClO) alkoxy- or aryloxy-polyethylene glycol or polyethylene glycol-maleimide. In certain embodiments, the antibody to be pegylated is an aglycosylated antibody. Methods for pegylating proteins are known in the art and can be applied to the antibodies of the invention. See for example, EP 0 154 316 by Nishimura et al. and EP 0 401 384 by Ishikawa et al.

Methods of Engineering Antibodies

As discussed above, the anti-IRTA-2 antibodies having V H and V K sequences disclosed herein can be used to create new anti-IRTA-2 antibodies by modifying the VH and/or V K sequences, or the constant region(s) attached thereto. Thus, in another aspect of the invention, the structural features of an anti-IRTA-2 antibody of the invention, e.g. 9Dl 2 or 8Al, are used to create structurally related anti-IRTA-2 antibodies that retain at least one functional property of the antibodies of the invention, such as binding to human IRTA-2. For example, one or more CDR regions of 9Dl 2 or 8Al, or mutations thereof, can be combined recombinantly with known framework regions and/or other CDRs to create additional, recombinantly-engineered, anti-IRTA-2 antibodies of the invention, as discussed above. Other types of modifications include those described in the previous section. The starting material for the engineering method is one or more of the V H and/or V K sequences provided herein, or one or more CDR regions thereof. To create the engineered antibody, it is not necessary to actually prepare {i.e., express as a protein) an antibody having one or more of the V H and/or V R sequences provided herein, or one or more CDR regions thereof. Rather, the information contained in the sequence(s) is used as the starting material to create a "second generation" sequence(s) derived from the original sequence(s) and then the "second generation" sequence(s) is prepared and expressed as a protein.

Accordingly, in another embodiment, the invention provides a method for preparing an anti-IRTA-2 antibody comprising:

(a) providing: (i) a heavy chain variable region antibody sequence comprising a CDRl sequence selected from the group consisting of SEQ ID NOs: 1 and 2, a CDR2 sequence selected from the group consisting of SEQ ID NOs: 3 and 4, and/or a CDR3 sequence selected from the group consisting of SEQ ID NOs: 5 and 6; and/or (ii) a light chain

variable region antibody sequence comprising a CDRl sequence selected from the group consisting of SEQ ID NOs: 7 and 8, a CDR2 sequence selected from the group consisting of SEQ ID NOs: 9 and 10, and/or a CDR3 sequence selected from the group consisting of SEQ ID NOs: 11 and 12;

(b) altering at least one amino acid residue within the heavy chain variable region antibody sequence and/or the light chain variable region antibody sequence to create at least one altered antibody sequence; and

(c) expressing the altered antibody sequence as a protein.

Standard molecular biology techniques can be used to prepare and express the altered antibody sequence.

Preferably, the antibody encoded by the altered antibody sequence(s) is one that retains one, some or all of the functional properties of the anti-IRTA-2 antibodies described herein, which functional properties include, but are not limited to:

(i) binds to human IRTA-2 with a K D of IxIO "7 M or less; (ii) binds to human CHO cells transfected with IRTA-2; (iii) does not substantially bind to human IRTA-3 or IRTA-4; and/or (iv) binds to Granta 519 tumor cells and does not substantially bind to Raji or Ramos tumor cells.

In a preferred embodiment, the antibody does not substantially bind to Daudi, IM-9, Karpas 1106P, or SU-DHL-4 tumor cells.

The functional properties of the altered antibodies can be assessed using standard assays available in the art and/or described herein, such as those set forth in the Examples (e.g., flow cytometry, binding assays).

In certain embodiments of the methods of engineering antibodies of the invention, mutations can be introduced randomly or selectively along all or part of an anti-IRTA-2 antibody coding sequence and the resulting modified anti-IRTA-2 antibodies can be screened for binding activity and/or other functional properties as described herein. Mutational methods have been described in the art. For example, PCT Publication WO 02/092780 by Short describes methods for creating and screening antibody mutations using saturation mutagenesis, synthetic ligation assembly, or a combination thereof. Alternatively, PCT Publication WO 03/074679 by Lazar et al. describes methods of using computational screening methods to optimize physiochemical properties of antibodies.

Nucleic Acid Molecules Encoding Antibodies of the Invention

Another aspect of the invention pertains to nucleic acid molecules that encode the antibodies of the invention. The nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form. A nucleic acid is "isolated" or "rendered substantially pure" when purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis and others well known in the art. See, F. Ausubel, et ah, ed. (1987) Current Protocols in Molecular Biology, Greene Publishing and Wiley Interscience, New York. A nucleic acid of the invention can be, for example, DNA or RNA and may or may not contain intronic sequences. In a preferred embodiment, the nucleic acid is a cDNA molecule.

Nucleic acids of the invention can be obtained using standard molecular biology techniques. For antibodies expressed by hybridomas (e.g., hybridomas prepared from transgenic mice carrying human immunoglobulin genes as described further below), cDNAs encoding the light and heavy chains of the antibody made by the hybridoma can be obtained by standard PCR amplification or cDNA cloning techniques. For antibodies obtained from an immunoglobulin gene library (e.g., using phage display techniques), nucleic acid encoding the antibody can be recovered from the library.

Preferred nucleic acids molecules of the invention are those encoding the VH and VL sequences of the 9D12 or 8Al monoclonal antibodies. DNA sequences encoding the VH sequences of 9D12 and 8Al are shown in SEQ ID NOs: 17 and 18, respectively. DNA sequences encoding the VL sequences of 9Dl 2 and 8Al are shown in SEQ ID NOs: 19 and 20, respectively.

Once DNA fragments encoding VH and VL segments are obtained, these DNA fragments can be further manipulated by standard recombinant DNA techniques, for example to convert the variable region genes to full-length antibody chain genes, to Fab fragment genes or to a scFv gene. In these manipulations, a VL- or VH-encoding DNA fragment is operatively linked to another DNA fragment encoding another protein, such as an antibody constant region or a flexible linker. The term "operatively linked", as used in this context, is intended to mean that the two DNA fragments are joined such that the amino acid sequences encoded by the two DNA fragments remain in-frame.

The isolated DNA encoding the VH region can be converted to a full-length heavy chain gene by operatively linking the VH-encoding DNA to another DNA molecule encoding heavy chain constant regions (CHl, CH2 and CH3). The sequences of human heavy chain

constant region genes are known in the art (see e.g., Kabat, E. A., el al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification. The heavy chain constant region can be an IgGl, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region, but most preferably is an IgGl or IgG4 constant region. For a Fab fragment heavy chain gene, the VH-encoding DNA can be operative Iy linked to another DNA molecule encoding only the heavy chain CHl constant region.

The isolated DNA encoding the VL region can be converted to a full-length light chain gene (as well as a Fab light chain gene) by operatively linking the VL-encoding DNA to another DNA molecule encoding the light chain constant region, CL. The sequences of human light chain constant region genes are known in the art (see e.g., Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification. The light chain constant region can be a kappa or lambda constant region, but most preferably is a kappa constant region.

To create a scFv gene, the VH- and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (GIy 4 -Ser) 3 , such that the VH and VL sequences can be expressed as a contiguous single- chain protein, with the VL and VH regions joined by the flexible linker (see e.g., Bird et al. (1988) Science TM-AlZAlβ; Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883; McCafferty et al, (1990) Nature 348:552-554).

Production of Monoclonal Antibodies of the Invention

Monoclonal antibodies (mAbs) of the present invention can be produced by a variety of techniques, including conventional monoclonal antibody methodology e.g., the standard somatic cell hybridization technique of Kohler and Milstein (1975) Nature 256: 495. Although somatic cell hybridization procedures are preferred, in principle, other techniques for producing monoclonal antibody can be employed e.g., viral or oncogenic transformation of B lymphocytes.

The preferred animal system for preparing hybridomas is the murine system. Hybridoma production in the mouse is a very well-established procedure. Immunization

protocols and techniques for isolation of immunized splenocytes for fusion are known in the art. Fusion partners (e.g., murine myeloma cells) and fusion procedures are also known.

Chimeric or humanized antibodies of the present invention can be prepared based on the sequence of a murine monoclonal antibody prepared as described above. DNA encoding the heavy and light chain immunoglobulins can be obtained from the murine hybridoma of interest and engineered to contain non-murine (e.g., human) immunoglobulin sequences using standard molecular biology techniques. For example, to create a chimeric antibody, the murine variable regions can be linked to human constant regions using methods known in the art (see e.g., U.S. Patent No. 4,816,567 to Cabilly et al.). To create a humanized antibody, the murine CDR regions can be inserted into a human framework using methods known in the art (see e.g., U.S. Patent No. 5,225,539 to Winter, and U.S. Patent Nos. 5,530,101; 5,585,089; 5,693,762 and 6,180,370 to Queen et al).

In a preferred embodiment, the antibodies of the invention are human monoclonal antibodies. Such human monoclonal antibodies directed against IRTA-2 can be generated using transgenic or transchromosomic mice carrying parts of the human immune system rather than the mouse system. These transgenic and transchromosomic mice include mice referred to herein as the HuMAb Mouse ® and KM Mouse ® , respectively, and are collectively referred to herein as "human Ig mice."

The HuMAb Mouse ® (Medarex ® , Inc.) contains human immunoglobulin gene miniloci that encode unrearranged human heavy (μ and γ) and K light chain immunoglobulin sequences, together with targeted mutations that inactivate the endogenous μ and K chain loci (see e.g., Lonberg, et al. (1994) Nature 368(6474): 856-859). Accordingly, the mice exhibit reduced expression of mouse IgM or K, and in response to immunization, the introduced human heavy and light chain transgenes undergo class switching and somatic mutation to generate high affinity human IgGκ monoclonal (Lonberg, N. et al. (1994), supra; reviewed in Lonberg, N. (1994) Handbook of Experimental Pharmacology 113:49-101; Lonberg, N. and Huszar, D. (1995) Intern. Rev. Immunol. 13: 65-93, and Harding, F. and Lonberg, N. (1995) Ann. NY. Acad. Sci. 764:536-546). preparation and use of the HuMAb Mouse ® , and the genomic modifications carried by such mice, is further described in Taylor, L. et al. (1992) Nucleic Acids Research 20:6287-6295; Chen, J. et al. (1993) International Immunology 5: 647-656; Tuaillon et al. (1993) Proc. Natl. Acad. Sci. USA 90:3720-3724; Choi et al. (1993) Nature Genetics 4:117-123; Chen, J. et al. (1993) EMBO J. U: 821-830; Tuaillon et al. (1994) J. Immunol. 152:2912-2920; Taylor, L. et al. (1994) International Immunology 6:

579-591; and Fishwild, D. et al (1996) Nature Biotechnology 14: 845-851, the contents of all of which are hereby specifically incorporated by reference in their entirety. See further, U.S. Patent Nos. 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,789,650; 5,877,397; 5,661,016; 5,814,318; 5,874,299; and 5,770,429; all to Lonberg and Kay; U.S. Patent No. 5,545,807 to Surani et al.; PCT Publication Nos. WO 92/03918, WO 93/12227, WO 94/25585, WO 97/13852, WO 98/24884 and WO 99/45962, all to Lonberg and Kay; and PCT Publication No. WO 01/14424 to Korman et al.

In another embodiment, human antibodies of the invention can be raised using a mouse that carries human immunoglobulin sequences on transgenes and transchomosomes, such as a mouse that carries a human heavy chain transgene and a human light chain transchromosome. Such mice, referred to herein as "KM mice™", are described in detail in PCT Publication WO 02/43478 to Ishida et al.

Still further, alternative transgenic animal systems expressing human immunoglobulin genes are available in the art and can be used to raise anti-IRTA-2 antibodies of the invention. For example, an alternative transgenic system referred to as the Xenomouse (Abgenix, Inc.) can be used; such mice are described in, for example, U.S. Patent Nos. 5,939,598; 6,075,181; 6,114,598; 6,150,584 and 6,162,963 to Kucherlapati et al.

Moreover, alternative transchromosomic animal systems expressing human immunoglobulin genes are available in the art and can be used to raise anti-IRTA-2 antibodies of the invention. For example, mice carrying both a human heavy chain transchromosome and a human light chain tranchromosome, referred to as "TC mice" can be used; such mice are described in Tomizuka et al. (2000) Proc. Natl. Acad. ScI USA 97:722- 727. Furthermore, cows carrying human heavy and light chain transchromosomes have been described in the art (Kuroiwa et al. (2002) Nature Biotechnology 20:889-894) and can be used to raise anti-IRTA-2 antibodies of the invention.

Human monoclonal antibodies of the invention can also be prepared using phage display methods for screening libraries of human immunoglobulin genes. Such phage display methods for isolating human antibodies are established in the art. See for example: U.S. Patent Nos. 5,223,409; 5,403,484; and 5,571,698 to Ladner et al.; U.S. Patent Nos. 5,427,908 and 5,580,717 to Dower et al; U.S. Patent Nos. 5,969,108 and 6,172,197 to McCafferty et al; and U.S. Patent Nos. 5,885,793; 6,521,404; 6,544,731; 6,555,313; 6,582,915 and 6,593,081 to Griffiths et al.

Human monoclonal antibodies of the invention can also be prepared using SCID mice into which human immune cells have been reconstituted such that a human antibody response

can be generated upon immunization. Such mice are described in, for example, U.S. Patent Nos. 5,476,996 and 5,698,767 to Wilson et al.

Immunization of Human Ig Mice

When human Ig mice are used to raise human antibodies of the invention, such mice can be immunized with a purified or enriched preparation of IRTA-2 antigen and/or recombinant IRTA-2, or an IRTA-2 fusion protein, as described by Lonberg, N. et al. (1994) Nature 368(6474): 856-859; Fishwild, D. et al. (1996) Nature Biotechnology 14: 845-851 ; and PCT Publication WO 98/24884 and WO 01/14424. Preferably, the mice will be 6-16 weeks of age upon the first infusion. For example, a purified or recombinant preparation (5- 50 μg) of IRTA-2 antigen can be used to immunize the human Ig mice intraperitoneally.

Detailed procedures to generate fully human monoclonal antibodies to IRTA-2 are described in Example 1 below. Cumulative experience with various antigens has shown that the transgenic mice respond when initially immunized intraperitoneally (IP) with antigen in complete Freund's adjuvant, followed by every other week IP immunizations (up to a total of 6) with antigen in incomplete Freund's adjuvant. However, adjuvants other than Freund's are also found to be effective. In addition, whole cells in the absence of adjuvant are found to be highly immunogenic. The immune response can be monitored over the course of the immunization protocol with plasma samples being obtained by retroorbital bleeds. The plasma can be screened by ELISA (as described below), and mice with sufficient titers of anti-IRTA-2 human immunoglobulin can be used for fusions. Mice can be boosted intravenously with antigen 3 days before sacrifice and removal of the spleen. It is expected that 2-3 fusions for each immunization may need to be performed. Between 6 and 24 mice are typically immunized for each antigen. Usually both HCo7 and HCol2 strains are used. In addition, both HCo7 and HCol2 transgene can be bred together into a single mouse having two different human heavy chain transgenes (HCo7/HCol2). Alternatively or additionally, the KM Mouse ® strain can be used, as described in Example 1.

Generation of Hybridomas Producing Human Monoclonal Antibodies of the Invention

To generate hybridomas producing human monoclonal antibodies of the invention, splenocytes and/or lymph node cells from immunized mice can be isolated and fused to an appropriate immortalized cell line, such as a mouse myeloma cell line. The resulting hybridomas can be screened for the production of antigen-specific antibodies. For example, single cell suspensions of splenic lymphocytes from immunized mice can be fused to one-

sixth the number of P3X63-Ag8.653 nonsecreting mouse myeloma cells (ATCC, CRL 1580) with 50% PEG. Cells are plated at approximately 2 x 10 5 in flat bottom microtiter plate, followed by a two week incubation in selective medium containing 20% fetal Clone Serum, 18% "653" conditioned media, 5% origen (IGEN), 4 mM L-glutamine, 1 mM sodium pyruvate, 5mM HEPES, 0.055 mM 2-mercaptoethanol, 50 units/ml penicillin, 50 mg/ml streptomycin, 50 mg/ml gentamycin and IX HAT (Sigma; the HAT is added 24 hours after the fusion). After approximately two weeks, cells can be cultured in medium in which the HAT is replaced with HT. Individual wells can then be screened by ELISA for human monoclonal IgM and IgG antibodies. Once extensive hybridoma growth occurs, medium can be observed usually after 10-14 days. The antibody secreting hybridomas can be replated, screened again, and if still positive for human IgG, the monoclonal antibodies can be subcloned at least twice by limiting dilution. The stable subclones can then be cultured in vitro to generate small amounts of antibody in tissue culture medium for characterization.

To purify human monoclonal antibodies, selected hybridomas can be grown in two- liter spinner-flasks for monoclonal antibody purification. Supernatants can be filtered and concentrated before affinity chromatography with protein A-sepharose (Pharmacia, Piscataway, NJ.) . Eluted IgG can be checked by gel electrophoresis and high performance liquid chromatography to ensure purity. The buffer solution can be exchanged into PBS, and the concentration can be determined by OD280 using 1.43 extinction coefficient. The monoclonal antibodies can be aliquoted and stored at -80° C.

Generation of Transfectomas Producing Monoclonal Antibodies of the Invention

Antibodies of the invention also can be produced in a host cell transfectoma using, for example, a combination of recombinant DNA techniques and gene transfection methods as is well known in the art (e.g., Morrison, S. (1985) Science 229:1202).

For example, to express the antibodies, or antibody fragments thereof, DNAs encoding partial or full-length light and heavy chains, can be obtained by standard molecular biology techniques (e.g., PCR amplification or cDNA cloning using a hybridoma that expresses the antibody of interest) and the DNAs can be inserted into expression vectors such that the genes are operatively linked to transcriptional and translational control sequences. In this context, the term "operatively linked" is intended to mean that an antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the antibody gene. The expression vector and expression control sequences are chosen to be compatible

with the expression host cell used. The antibody light chain gene and the antibody heavy chain gene can be inserted into separate vector or, more typically, both genes are inserted into the same expression vector. The antibody genes are inserted into the expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present). The light and heavy chain variable regions of the antibodies described herein can be used to create full- length antibody genes of any antibody isotype by inserting them into expression vectors already encoding heavy chain constant and light chain constant regions of the desired isotype such that the VH segment is operatively linked to the CH segment(s) within the vector and the VK segment is operatively linked to the CL segment within the vector. Additionally or alternatively, the recombinant expression vector can encode a signal peptide that facilitates secretion of the antibody chain from a host cell. The antibody chain gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the antibody chain gene. The signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide {i.e., a signal peptide from a non-immunoglobulin protein).

In addition to the antibody chain genes, the recombinant expression vectors of the invention carry regulatory sequences that control the expression of the antibody chain genes in a host cell. The term "regulatory sequence" is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the antibody chain genes. Such regulatory sequences are described, for example, in Goeddel (Gene Expression Technology. Methods in Enzymology 185, Academic Press, San Diego, CA (1990)). It will be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences, may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. Preferred regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV), Simian Virus 40 (SV40), adenovirus, (e.g., the adenovirus major late promoter (AdMLP) and polyoma. Alternatively, nonviral regulatory sequences may be used, such as the ubiquitin promoter or β-globin promoter. Still further, regulatory elements composed of sequences from different sources, such as the SRa promoter system, which contains sequences from the SV40 early promoter and the long terminal repeat of human T cell leukemia virus type 1 (Takebe, Y. et al. (1988) MoI. Cell. Biol. 8:466-472).

In addition to the antibody chain genes and regulatory sequences, the recombinant expression vectors of the invention may carry additional sequences, such as sequences that regulate replication of the vector in host cells {e.g., origins of replication) and selectable marker genes. The selectable marker gene facilitates selection of host cells into which the vector has been introduced (see, e.g., U.S. Pat. Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et al). For example, typically the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced. Preferred selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr- host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).

For expression of the light and heavy chains, the expression vector(s) encoding the heavy and light chains is transfected into a host cell by standard techniques. The various forms of the term "transfection" are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like. Although it is theoretically possible to express the antibodies of the invention in either prokaryotic or eukaryotic host cells, expression of antibodies in eukaryotic cells, and most preferably mammalian host cells, is the most preferred because such eukaryotic cells, and in particular mammalian cells, are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody. Prokaryotic expression of antibody genes has been reported to be ineffective for production of high yields of active antibody (Boss, M. A. and Wood, C. R. (1985) Immunology Today 6:12-13).

Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr- CHO cells, described in Urlaub and Chasin, (1980) Proc. Natl. Acad. ScL USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in R. J. Kaufman and P.A. Sharp (1982) MoI. Biol. /59:601-621), NSO myeloma cells, COS cells and SP2 cells. In particular, for use with NSO myeloma cells, another preferred expression system is the GS gene expression system disclosed in WO 87/04462, WO 89/01036 and EP 338,841. When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods.

Characterization of Antibody Binding to Antigen

Antibodies of the invention can be tested for binding to IRTA-2 by, for example, standard ELISA. Briefly, microtiter plates are coated with purified IRTA-2 at 0.25 μg/ml in PBS, and then blocked with 5% bovine serum albumin in PBS. Dilutions of antibody (e.g., dilutions of plasma from IRTA-2-immunized mice) are added to each well and incubated for 1-2 hours at 37 0 C. The plates are washed with PBS/Tween and then incubated with secondary reagent (e.g., for human antibodies, a goat-anti-human IgG Fc-specific polyclonal reagent) conjugated to alkaline phosphatase for 1 hour at 37 0 C. After washing, the plates are developed with pNPP substrate (1 mg/ml), and analyzed at OD of 405-650. Preferably, mice which develop the highest titers will be used for fusions.

An ELISA assay as described above can also be used to screen for hybridomas that show positive reactivity with IRTA-2 immunogen. Hybridomas that bind with high avidity to IRTA-2 are subcloned and further characterized. One clone from each hybridoma, which retains the reactivity of the parent cells (by ELISA), can be chosen for making a 5-10 vial cell bank stored at -140 0 C, and for antibody purification.

To purify anti-IRTA-2 antibodies, selected hybridomas can be grown in two-liter spinner-flasks for monoclonal antibody purification. Supernatants can be filtered and concentrated before affinity chromatography with protein A-sepharose (Pharmacia, Piscataway, NJ). Eluted IgG can be checked by gel electrophoresis and high performance liquid chromatography to ensure purity. The buffer solution can be exchanged into PBS, and the concentration can be determined by OD 28 O using 1.43 extinction coefficient. The monoclonal antibodies can be aliquoted and stored at -80 0 C.

To determine if the selected anti-IRTA-2 monoclonal antibodies bind to unique epitopes, each antibody can be biotinylated using commercially available reagents (Pierce, Rockford, IL). Competition studies using unlabeled monoclonal antibodies and biotinylated monoclonal antibodies can be performed using IRTA-2 coated-ELISA plates as described above. Biotinylated mAb binding can be detected with a strep-avidin-alkaline phosphatase probe.

To determine the isotype of purified antibodies, isotype ELISAs can be performed using reagents specific for antibodies of a particular isotype. For example, to determine the isotype of a human monoclonal antibody, wells of microtiter plates can be coated with 1 μg/ml of anti-human immunoglobulin overnight at 4°C. After blocking with 1% BSA, the plates are reacted with 1 μg /ml or less of test monoclonal antibodies or purified isotype

controls, at ambient temperature for one to two hours. The wells can then be reacted with either human IgGl or human IgM-specific alkaline phosphatase-conjugated probes. Plates are developed and analyzed as described above.

Anti-IRTA-2 human IgGs can be further tested for reactivity with IRT A-2 antigen by Western blotting. Briefly, IRTA-2 can be prepared and subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis. After electrophoresis, the separated antigens are transferred to nitrocellulose membranes, blocked with 10% fetal calf serum, and probed with the monoclonal antibodies to be tested. Human IgG binding can be detected using anti- human IgG alkaline phosphatase and developed with BCIP/NBT substrate tablets (Sigma Chem. Co., St. Louis, Mo.).

Immunoconiugates

In another aspect, the present invention features an anti-IRTA-2 antibody, or a fragment thereof, conjugated to a therapeutic moiety, such as a cytotoxin, a drug (e.g., an immunosuppressant) or a radiotoxin. Such conjugates are referred to herein as "immunoconjugates". Immunoconjugates that include one or more cytotoxins are referred to as "immunotoxins." A cytotoxin or cytotoxic agent includes any agent that is detrimental to (e.g., kills) cells. Examples include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1- dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents also include, for example, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5- fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine).

Other preferred examples of therapeutic cytotoxins that can be conjugated to an antibody of the invention include duocarmycins, calicheamicins, maytansines and auristatins, and derivatives thereof. An example of a calicheamicin antibody conjugate is commercially available (Mylotarg™; Wyeth-Ayerst).

Cytoxins can be conjugated to antibodies of the invention using linker technology available in the art. Examples of linker types that have been used to conjugate a cytotoxin to an antibody include, but are not limited to, hydrazones, thioethers, esters, disulfides and peptide-containing linkers. A linker can be chosen that is, for example, susceptible to cleavage by low pH within the lysosomal compartment or susceptible to cleavage by proteases, such as proteases preferentially expressed in tumor tissue such as cathepsins (e.g., cathepsins B, C, D).

For further discussion of types of cytotoxins, linkers and methods for conjugating therapeutic agents to antibodies, see also Saito, G. et al. (2003) Adv. Drug Deliv. Rev. 55:199-215; Trail, P.A. et al. (2003) Cancer Immunol. Immunother. 52:328-337; Payne, G. (2003) Cancer Cell 3:207-212; Allen, T.M. (2002) Nat. Rev. Cancer 2:750-763; Pastan, I. and Kreitman, R. J. (2002) Curr. Opin. Investig. Drugs 3:1089-1091; Senter, P.D. and Springer, CJ. (2001) Adv. Drug Deliv. Rev. 53:247-264.

Antibodies of the present invention also can be conjugated to a radioactive isotope to generate cytotoxic radiopharmaceuticals, also referred to as radioimmunoconjugates. Examples of radioactive isotopes that can be conjugated to antibodies for use diagnostically or therapeutically include, but are not limited to, iodine 131 , indium 111 , yttrium 90 and lutetium 177 . Method for preparing radioimmunconjugates are established in the art. Examples of radioimmunoconjugates are commercially available, including Zevalin™ (IDEC Pharmaceuticals) and Bexxar™ (Corixa Pharmaceuticals), and similar methods can be used to prepare radioimmunoconjugates using the antibodies of the invention.

The antibody conjugates of the invention can be used to modify a given biological response, and the drug moiety is not to be construed as limited to classical chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, an enzymatically active toxin, or active fragment thereof, such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor or interferon-γ; or, biological response modifiers such as, for example, lymphokines, interleukin-1 ("IL-I"), interleukin-2 ("IL-2"), interleukin-6 ("IL-6"), granulocyte macrophage colony stimulating factor ("GM- CSF"), granulocyte colony stimulating factor ("G-CSF"), or other growth factors.

Techniques for conjugating such therapeutic moiety to antibodies are well known, see, e.g., Arnon et al, "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56

(Alan R. Liss, Inc. 1985); Hellstrom et al, "Antibodies For Drug Delivery", in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker, Inc. 1987); Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review", in Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et al. (eds.), pp. 475-506 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy", in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al, "The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates", Immunol. Rev., 62:119-58 (1982).

Bispecific Molecules

In another aspect, the present invention features bispecific molecules comprising an anti-IRTA-2 antibody, or a fragment thereof, of the invention. An antibody of the invention, or antigen-binding portions thereof, can be derivatized or linked to another functional molecule, e.g., another peptide or protein (e.g., another antibody or ligand for a receptor) to generate a bispecific molecule that binds to at least two different binding sites or target molecules. The antibody of the invention may in fact be derivatized or linkd to more than one other functional molecule to generate multispecific molecules that bind to more than two different binding sites and/or target molecules; such multispecific molecules are also intended to be encompassed by the term "bispecific molecule" as used herein. To create a bispecific molecule of the invention, an antibody of the invention can be functionally linked {e.g., by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other binding molecules, such as another antibody, antibody fragment, peptide or binding mimetic, such that a bispecific molecule results.

Accordingly, the present invention includes bispecific molecules comprising at least one first binding specificity for IRT A-2 and a second binding specificity for a second target epitope. In a particular embodiment of the invention, the second target epitope is an Fc receptor, e.g., human FcγRI (CD64) or a human Fcα receptor (CD89). Therefore, the invention includes bispecific molecules capable of binding both to FcγR or FcαR expressing effector cells (e.g., monocytes, macrophages or polymorphonuclear cells (PMNs)), and to target cells expressing IRTA-2. These bispecific molecules target IRTA-2 expressing cells to effector cell and trigger Fc receptor-mediated effector cell activities, such as phagocytosis of

an IRTA-2 expressing cells, antibody dependent cell-mediated cytotoxicity (ADCC), cytokine release, or generation of superoxide anion.

In an embodiment of the invention in which the bispecific molecule is multispecifϊc, the molecule can further include a third binding specificity, in addition to an anti-Fc binding specificity and an anti-IRTA-2 binding specificity. In one embodiment, the third binding specificity is an anti-enhancement factor (EF) portion, e.g., a molecule which binds to a surface protein involved in cytotoxic activity and thereby increases the immune response against the target cell. The "anti-enhancement factor portion" can be an antibody, functional antibody fragment or a ligand that binds to a given molecule, e.g., an antigen or a receptor, and thereby results in an enhancement of the effect of the binding determinants for the F 0 receptor or target cell antigen. The "anti-enhancement factor portion" can bind an F c receptor or a target cell antigen. Alternatively, the anti-enhancement factor portion can bind to an entity that is different from the entity to which the first and second binding specificities bind. For example, the anti-enhancement factor portion can bind a cytotoxic T-cell (e.g. via CD2, CD3, CD8, CD28, CD4, CD40, ICAM-I or other immune cell that results in an increased immune response against the target cell).

In one embodiment, the bispecific molecules of the invention comprise as a binding specificity at least one antibody, or an antibody fragment thereof, including, e.g., an Fab, Fab', F(ab')2, Fv, or a single chain Fv. The antibody may also be a light chain or heavy chain dimer, or any minimal fragment thereof such as a Fv or a single chain construct as described in Ladner et αl. U.S. Patent No. 4,946,778, the contents of which is expressly incorporated by reference.

In one embodiment, the binding specificity for an Fcγ receptor is provided by a monoclonal antibody, the binding of which is not blocked by human immunoglobulin G (IgG). As used herein, the term "IgG receptor" refers to any of the eight γ-chain genes located on chromosome 1. These genes encode a total of twelve transmembrane or soluble receptor isoforms which are grouped into three Fcγ receptor classes: FcγRI (CD64), Fcγ RII(CD32), and FcγRIII (CD 16). In one preferred embodiment, the Fcγ receptor a human high affinity FcγRI. The human FcγRI is a 72 kDa molecule, which shows high affinity for monomeric IgG (10 8 - 10 9 M-').

The production and characterization of certain preferred anti-Fcγ monoclonal antibodies are described by Fanger et αl. in PCT Publication WO 88/00052 and in U.S. Patent No. 4,954,617, the teachings of which are fully incorporated by reference herein. These

antibodies bind to an epitope of FcγRI, FcγRII or FcγRIII at a site which is distinct from the Fcγ binding site of the receptor and, thus, their binding is not blocked substantially by physiological levels of IgG. Specific anti-FcγRI antibodies useful in this invention are mAb 22, mAb 32, mAb 44, mAb 62 and mAb 197. The hybridoma producing mAb 32 is available from the American Type Culture Collection, ATCC Accession No. HB9469. In other embodiments, the anti-Fcγ receptor antibody is a humanized form of monoclonal antibody 22 (H22). The production and characterization of the H22 antibody is described in Graziano, R.F. et al. (1995J J. Immunol 155 (10): 4996-5002 and PCT Publication WO 94/10332. The H22 antibody producing cell line was deposited at the American Type Culture Collection under the designation HA022CL1 and has the accession no. CRL 11177.

In still other preferred embodiments, the binding specificity for an Fc receptor is provided by an antibody that binds to a human IgA receptor, e.g., an Fc-alpha receptor (Fcα RI (CD89)), the binding of which is preferably not blocked by human immunoglobulin A (IgA). The term "IgA receptor" is intended to include the gene product of one α-gene (Fcα RI) located on chromosome 19. This gene is known to encode several alternatively spliced transmembrane isoforms of 55 to 110 kDa. FcαRI (CD89) is constitutive Iy expressed on monocytes/macrophages, eosinophilic and neutrophilic granulocytes, but not on non-effector cell populations. FcαRI has medium affinity (« 5 x 10 7 M" 1 ) for both IgAl and IgA2, which is increased upon exposure to cytokines such as G-CSF or GM-CSF (Morton, H.C. et al. (1996) Critical Reviews in Immunology ,16:423-440). Four FcαRI-specific monoclonal antibodies, identified as A3, A59, A62 and A77, which bind FcαRI outside the IgA ligand binding domain, have been described (Monteiro, R.C. et al. (1992) J. Immunol. 148:1764).

FcαRI and FcγRI are preferred trigger receptors for use in the bispecific molecules of the invention because they are (1) expressed primarily on immune effector cells, e.g., monocytes, PMNs, macrophages and dendritic cells; (2) expressed at high levels {e.g., 5,000- 100,000 per cell); (3) mediators of cytotoxic activities (e.g., ADCC, phagocytosis); (4) mediate enhanced antigen presentation of antigens, including self-antigens, targeted to them.

While human monoclonal antibodies are preferred, other antibodies which can be employed in the bispecific molecules of the invention are murine, chimeric and humanized monoclonal antibodies.

The bispecific molecules of the present invention can be prepared by conjugating the constituent binding specificities, e.g., the anti-FcR and anti-IRTA-2 binding specificities, using methods known in the art. For example, each binding specificity of the bispecific

molecule can be generated separately and then conjugated to one another. When the binding specificities are proteins or peptides, a variety of coupling or cross-linking agents can be used for covalent conjugation. Examples of cross-linking agents include protein A, carbodiimide, N-succinimidyl-S-acetyl-thioacetate (SATA), 5,5'-dithiobis(2-nitrobenzoic acid) (DTNB), o- phenylenedimaleimide (oPDM), N-succinimidyl-3-(2-pyridyldithio)propionate (SPDP), and sulfosuccinimidyl 4-(N-maleimidomethyl) cyclohaxane-1-carboxylate (sulfo-SMCC) (see e.g., Karpovsky et al. (1984) J. Exp. Med. 160:1686; Liu, MA et al (1985) Proc. Natl. Acad. Sci. USA 82:8648). Other methods include those described in Paulus (1985) Behring Ins. Mitt. No. 78, 118-132; Brennan et al (1985) Science 229:81-83), and Glennie et al (1987) J. Immunol. _I39: 2367-2375). Preferred conjugating agents are SATA and sulfo-SMCC, both available from Pierce Chemical Co. (Rockford, IL).

When the binding specificities are antibodies, they can be conjugated via sulfhydryl bonding of the C-terminus hinge regions of the two heavy chains. In a particularly preferred embodiment, the hinge region is modified to contain an odd number of sulfhydryl residues, preferably one, prior to conjugation.

Alternatively, both binding specificities can be encoded in the same vector and expressed and assembled in the same host cell. This method is particularly useful where the bispecific molecule is a mAb x mAb, mAb x Fab, Fab x F(ab')2 or ligand x Fab fusion protein. A bispecific molecule of the invention can be a single chain molecule comprising one single chain antibody and a binding determinant, or a single chain bispecific molecule comprising two binding determinants. Bispecific molecules may comprise at least two single chain molecules. Methods for preparing bispecific molecules are described for example in U.S. Patent Number 5,260,203; U.S. Patent Number 5,455,030; U.S. Patent Number 4,881,175; U.S. Patent Number 5,132,405; U.S. Patent Number 5,091,513; U.S. Patent Number 5,476,786; U.S. Patent Number 5,013,653; U.S. Patent Number 5,258,498; and U.S. Patent Number 5,482,858.

Binding of the bispecific molecules to their specific targets can be confirmed by, for example, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), FACS analysis, bioassay (e.g., growth inhibition), or Western Blot assay. Each of these assays generally detects the presence of protein-antibody complexes of particular interest by employing a labeled reagent (e.g., an antibody) specific for the complex of interest. For example, the FcR-antibody complexes can be detected using e.g., an enzyme-linked antibody or antibody fragment which recognizes and specifically binds to the antibody-FcR

complexes. Alternatively, the complexes can be detected using any of a variety of other immunoassays. For example, the antibody can be radioactively labeled and used in a radioimmunoassay (RIA) (see, for example, Weintraub, B., Principles of Radioimmunoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March, 1986, which is incorporated by reference herein). The radioactive isotope can be detected by such means as the use of a γ counter or a scintillation counter or by autoradiography.

Pharmaceutical Compositions

In another aspect, the present invention provides a composition, e.g., a pharmaceutical composition, containing one or a combination of monoclonal antibodies, or antigen-binding portion(s) thereof, of the present invention, formulated together with a pharmaceutically acceptable carrier. Such compositions may include one or a combination of (e.g., two or more different) antibodies, or immunoconjugates or bispecific molecules of the invention. For example, a pharmaceutical composition of the invention can comprise a combination of antibodies (or immunoconjugates or bispecifics) that bind to different epitopes on the target antigen or that have complementary activities.

Pharmaceutical compositions of the invention also can be administered in combination therapy, i.e., combined with other agents. For example, the combination therapy can include an anti-IRTA-2 antibody of the present invention combined with at least one other anti-inflammatory or immunosuppressant agent. Examples of therapeutic agents that can be used in combination therapy are described in greater detail below in the section on uses of the antibodies of the invention.

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

The pharmaceutical compounds of the invention may include one or more pharmaceutically acceptable salts. A "pharmaceutically acceptable salt" refers to a salt that

retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects (see e.g., Berge, S.M., et al. (1977) J. Pharm. Sci. 66:1-19). Examples of such salts include acid addition salts and base addition salts. Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorous and the like, as well as from nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like. Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and the like, as well as from nontoxic organic amines, such as N,N'-dibenzylethylenediamine, N-methylglucamine, chloroprocaine, choline, diethanolamine, ethylenediamine, procaine and the like.

A pharmaceutical composition of the invention also may include a pharmaceutically acceptable antioxidant. Examples of pharmaceutically acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.

Examples of suitable aqueous and nonaqueous carriers that may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.

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

Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the invention is contemplated. Supplementary active compounds can also be incorporated into the compositions.

Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.

Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by sterilization microfiltration. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying (lyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.

The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, and the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the composition which produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 0.01 per cent to about ninety-nine percent of active ingredient,

preferably from about 0.1 per cent to about 70 per cent, most preferably from about 1 per cent to about 30 per cent of active ingredient in combination with a pharmaceutically acceptable carrier.

Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.

For administration of the antibody, the dosage ranges from about 0.0001 to 100 mg/kg, and more usually 0.01 to 5 mg/kg, of the host body weight. For example, dosages can be 0.3 mg/kg body weight, 1 mg/kg body weight, 3 mg/kg body weight, 5 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg. An exemplary treatment regime entails administration once per week, once every two weeks, once every three weeks, once every four weeks, once a month, once every 3 months or once every three to 6 months. Preferred dosage regimens for an anti-IRTA-2 antibody of the invention include 1 mg/kg body weight or 3 mg/kg body weight via intravenous administration, with the antibody being given using one of the following dosing schedules: (i) every four weeks for six dosages, then every three months; (ii) every three weeks; (iii) 3 mg/kg body weight once followed by 1 mg/kg body weight every three weeks.

In some methods, two or more monoclonal antibodies with different binding specificities are administered simultaneously, in which case the dosage of each antibody administered falls within the ranges indicated. Antibody is usually administered on multiple occasions. Intervals between single dosages can be, for example, weekly, monthly, every three monthgs or yearly. Intervals can also be irregular as indicated by measuring blood levels of antibody to the target antigen in the patient. In some methods, dosage is adjusted to

achieve a plasma antibody concentration of about 1-1000 μg /ml and in some methods about 25-300 μg /ml.

Alternatively, antibody can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the antibody in the patient. In general, human antibodies show the longest half life, followed by humanized antibodies, chimeric antibodies, and nonhuman antibodies. The dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.

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

A "therapeutically effective dosage" of an anti-IRTA-2 antibody of the invention preferably results in a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction. For example, for the treatment of IRTA-2+ tumors, a "therapeutically effective dosage" preferably inhibits cell growth or tumor growth by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still more preferably by at least about 80% relative to untreated subjects. The ability of a compound to inhibit tumor growth can be evaluated in an animal model

system predictive of efficacy in human tumors. Alternatively, this property of a composition can be evaluated by examining the ability of the compound to inhibit, such inhibition in vitro by assays known to the skilled practitioner. A therapeutically effective amount of a therapeutic compound can decrease tumor size, or otherwise ameliorate symptoms in a subject. One of ordinary skill in the art would be able to determine such amounts based on such factors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of administration selected.

A composition of the present invention can be administered via one or more routes of administration using one or more of a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. Preferred routes of administration for antibodies of the invention include intravenous, intramuscular, intradermal, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, for example by injection or infusion. The phrase "parenteral administration" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.

Alternatively, an antibody of the invention can be administered via a non-parenteral route, such as a topical, epidermal or mucosal route of administration, for example, intranasally, orally, vaginally, rectally, sublingually or topically.

The active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.

Therapeutic compositions can be administered with medical devices known in the art. For example, in a preferred embodiment, a therapeutic composition of the invention can be administered with a needleless hypodermic injection device, such as the devices disclosed in U.S. Patent Nos. 5,399,163; 5,383,851; 5,312,335; 5,064,413; 4,941,880; 4,790,824; or 4,596,556. Examples of well-known implants and modules useful in the present invention

include: U.S. Patent No. 4,487,603, which discloses an implantable micro-infusion pump for dispensing medication at a controlled rate; U.S. Patent No. 4,486,194, which discloses a therapeutic device for administering medicants through the skin; U.S. Patent No. 4,447,233, which discloses a medication infusion pump for delivering medication at a precise infusion rate; U.S. Patent No. 4,447,224, which discloses a variable flow implantable infusion apparatus for continuous drug delivery; U.S. Patent No. 4,439,196, which discloses an osmotic drug delivery system having multi-chamber compartments; and U.S. Patent No. 4,475,196, which discloses an osmotic drug delivery system. These patents are incorporated herein by reference. Many other such implants, delivery systems, and modules are known to those skilled in the art.

In certain embodiments, the human monoclonal antibodies of the invention can be formulated to ensure proper distribution in vivo. For example, the blood-brain barrier (BBB) excludes many highly hydrophilic compounds. To ensure that the therapeutic compounds of the invention cross the BBB (if desired), they can be formulated, for example, in liposomes. For methods of manufacturing liposomes, see, e.g., U.S. Patents 4,522,811; 5,374,548; and 5,399,331. The liposomes may comprise one or more moieties which are selectively transported into specific cells or organs, thus enhance targeted drug delivery (see, e.g., V. V. Ranade (1989) J. Clin. Pharmacol. 29:685). Exemplary targeting moieties include folate or biotin (see, e.g., U.S. Patent 5,416,016 to Low et al); mannosides (Umezawa et at, (1988) Biochem. Biophys. Res. Commun. 153:1038); antibodies (P.G. Bloeman et al. (1995) FEBS Lett. 357:140; M. Owais et al. (1995) Antimicrob. Agents Chemother. 39:180); surfactant protein A receptor (Briscoe et al. ( 1995) Am. J. Physiol. 1233: 134); p 120 (Schreier et al. (1994) J. Biol. Chem. 269:9090); see also K. Keinanen; M.L. Laukkanen (1994) FEBS Lett. 346:123; JJ. Killion; IJ. Fidler (1994) Immunomethods 4:273.

Uses and Methods of the Invention

The antibodies, particulary the human antibodies, antibody compositions and methods of the present invention have numerous in vitro and in vivo diagnostic and therapeutic utilities involving the diagnosis and treatment of IRTA-2 mediated disorders. For example, these molecules can be administered to cells in culture, in vitro or ex vivo, or to human subjects, e.g., in vivo, to treat, prevent and to diagnose a variety of disorders. As used herein, the term "subject" is intended to include human and non-human animals. Non-human animals includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, sheep, dogs, cats, cows, horses, chickens, amphibians, and reptiles. Preferred subjects include

human patients having disorders mediated by IRTA-2 activity. The methods are particularly suitable for treating human patients having a disorder associated with aberrant IRTA-2 expression. When antibodies to IRTA-2 are administered together with another agent, the two can be administered in either order or simultaneously.

Given the specific binding of the antibodies of the invention for IRTA-2, compared to IRTA-3 or IRTA-4, the antibodies of the invention can be used to specifically detect IRTA-2 expression on the surface of cells and, moreover, can be used to purify IRTA-2 via immunoaffinity purification.

Furthermore, given the expression of IRTA-2 on various tumor cells (Davis et ah, (2002) Immunological Reviews. 190:123), the human antibodies, antibody compositions and methods of the present invention can be used to treat a subject with a tumorigenic disorder, e.g., a disorder characterized by the presence of tumor cells expressing IRTA-2 including, for example, Burkitt's lymphoma, anaplastic large-cell lymphomas (ALCL), cutaneous T-cell lymphomas, nodular small cleaved-cell lymphomas, lymphocytic lymphomas, peripheral T- cell lymphomas, Lennert's lymphomas, immunoblastic lymphomas, T-cell leukemia/lymphomas (ATLL), adult T-cell leukemia (T-ALL), entroblastic/centrocytic (cb/cc) follicular lymphomas cancers, diffuse large cell lymphomas of B lineage, angioimmunoblastic lymphadenopathy (AILD)-like T cell lymphoma, HIV associated body cavity based lymphomas, Embryonal Carcinomas, undifferentiated carcinomas of the rhino- pharynx (e.g., Schmincke's tumor), Castleman's disease, Kaposi's Sarcoma and other B-cell lymphomas.

In one embodiment, the antibodies (e.g., human monoclonal antibodies, multispecific and bispecific molecules and compositions) of the invention can be used to detect levels of IRTA-2, or levels of cells which contain IRTA-2 on their membrane surface, which levels can then be linked to certain disease symptoms. Alternatively, the antibodies can be used to inhibit or block IRTA-2 function which, in turn, can be linked to the prevention or amelioration of certain disease symptoms, thereby implicating IRTA-2 as a mediator of the disease. This can be achieved by contacting a sample and a control sample with the anti- IRTA-2 antibody under conditions that allow for the formation of a complex between the antibody and IRTA-2. Any complexes formed between the antibody and IRTA-2 are detected and compared in the sample and the control.

In another embodiment, the antibodies (e.g., human antibodies, multispecific and bispecific molecules and compositions) of the invention can be initially tested for binding activity associated with therapeutic or diagnostic use in vitro. For example, compositions of

the invention can be tested using the flow cytometric assays described in the Examples below.

The antibodies (e.g., human antibodies, multispecific and bispecific molecules, immunoconjugates and compositions) of the invention have additional utility in therapy and diagnosis of IRTA-2-related diseases. For example, the human monoclonal antibodies, the multispecific or bispecific molecules and the immunoconjugates can be used to elicit in vivo or in vitro one or more of the following biological activities: to inhibit the growth of and/or kill a cell expressing IRTA-2; to mediate phagocytosis or ADCC of a cell expressing IRTA-2 in the presence of human effector cells, or to block IRTA-2 ligand binding to IRTA-2.

In a particular embodiment, the antibodies (e.g., human antibodies, multispecific and bispecific molecules and compositions) are used in vivo to treat, prevent or diagnose a variety of IRTA-2-related diseases. Examples of IRTA-2-related diseases include, among others, cancer, non-Hodgkin's lymphoma, Burkitt's lymphoma, anaplastic large-cell lymphomas (ALCL), cutaneous T-cell lymphomas, nodular small cleaved-cell lymphomas, lymphocytic lymphomas, peripheral T-cell lymphomas, Lennert's lymphomas, immunoblastic lymphomas, T-cell leukemia/lymphomas (ATLL), adult T-cell leukemia (T-ALL), entroblastic/centrocytic (cb/cc) follicular lymphomas cancers, diffuse large cell lymphomas of B lineage, angioimmunoblastic lymphadenopathy (AILD)-like T cell lymphoma, HIV associated body cavity based lymphomas, Embryonal Carcinomas, undifferentiated carcinomas of the rhino- pharynx (e.g., Schmincke's tumor), Castleman's disease, Kaposi's Sarcoma and other B-cell lymphomas.

Suitable routes of administering the antibody compositions (e.g., human monoclonal antibodies, multispecific and bispecific molecules and immunoconjugates ) of the invention in vivo and in vitro are well known in the art and can be selected by those of ordinary skill. For example, the antibody compositions can be administered by injection (e.g., intravenous or subcutaneous). Suitable dosages of the molecules used will depend on the age and weight of the subject and the concentration and/or formulation of the antibody composition.

As previously described, human anti-IRTA-2 antibodies of the invention can be coadministered with one or other more therapeutic agents, e.g., a cytotoxic agent, a radiotoxic agent or an immunosuppressive agent. The antibody can be linked to the agent (as an immunocomplex) or can be administered separate from the agent. In the latter case (separate administration), the antibody can be administered before, after or concurrently with the agent or can be co-administered with other known therapies, e.g., an anti-cancer therapy, such as radiation. Such therapeutic agents include, among others, anti-neoplastic agents such as

doxorubicin (adriamycin), cisplatin bleomycin sulfate, carmustine, chlorambucil, and cyclophosphamide hydroxyurea which, by themselves, are only effective at levels which are toxic or subtoxic to a patient. Cisplatin is intravenously administered as a 100 mg/dose once every four weeks and adriamycin is intravenously administered as a 60-75 mg/ml dose once every 21 days. Co-administration of the human anti-IRTA-2 antibodies, or antigen binding fragments thereof, of the present invention with chemotherapeutic agents provides two anticancer agents which operate via different mechanisms which yield a cytotoxic effect to human tumor cells. Such co-administration can solve problems due to development of resistance to drugs or a change in the antigenicity of the tumor cells which would render them unreactive with the antibody.

Target-specific effector cells, e.g., effector cells linked to compositions (e.g., human antibodies, multispecific and bispecific molecules) of the invention can also be used as therapeutic agents. Effector cells for targeting can be human leukocytes such as macrophages, neutrophils or monocytes. Other cells include eosinophils, natural killer cells and other IgG- or IgA-receptor bearing cells. If desired, effector cells can be obtained from the subject to be treated. The target-specific effector cells can be administered as a suspension of cells in a physiologically acceptable solution. The number of cells administered can be in the order of 10 8 -10 9 but will vary depending on the therapeutic purpose. In general, the amount will be sufficient to obtain localization at the target cell, e.g., a tumor cell expressing IRTA-2, and to effect cell killing by, e.g., phagocytosis. Routes of administration can also vary.

Therapy with target-specific effector cells can be performed in conjunction with other techniques for removal of targeted cells. For example, anti-tumor therapy using the compositions (e.g., human antibodies, multispecific and bispecific molecules) of the invention and/or effector cells armed with these compositions can be used in conjunction with chemotherapy. Additionally, combination immunotherapy may be used to direct two distinct cytotoxic effector populations toward tumor cell rejection. For example, anti-IRTA-2 antibodies linked to anti-Fc-gamma RI or anti-CD3 may be used in conjunction with IgG- or IgA-receptor specific binding agents.

Bispecific and multispecific molecules of the invention can also be used to modulate FcγR or FcγR levels on effector cells, such as by capping and elimination of receptors on the cell surface. Mixtures of anti-Fc receptors can also be used for this purpose.

The compositions (e.g., human, humanized, or chimeric antibodies, multispecific and bispecific molecules and immunoconjugates) of the invention which have complement binding sites, such as portions from IgGl, -2, or -3 or IgM which bind complement, can also be used in the presence of complement. In one embodiment, ex vivo treatment of a population of cells comprising target cells with a binding agent of the invention and appropriate effector cells can be supplemented by the addition of complement or serum containing complement. Phagocytosis of target cells coated with a binding agent of the invention can be improved by binding of complement proteins. In another embodiment target cells coated with the compositions (e.g., human antibodies, multispecific and bispecific molecules) of the invention can also be lysed by complement. In yet another embodiment, the compositions of the invention do not activate complement.

The compositions (e.g., human, humanized, or chimeric antibodies, multispecific and bispecific molecules and immunoconjugates) of the invention can also be administered together with complement. Accordingly, within the scope of the invention are compositions comprising human antibodies, multispecific or bispecific molecules and serum or complement. These compositions are advantageous in that the complement is located in close proximity to the human antibodies, multispecific or bispecific molecules. Alternatively, the human antibodies, multispecific or bispecific molecules of the invention and the complement or serum can be administered separately.

Also within the scope of the present invention are kits comprising the antibody compositions of the invention (e.g., human antibodies, bispecific or multispecific molecules, or immunoconjugates) and instructions for use. The kit can further contain one ore more additional reagents, such as an immunosuppressive reagent, a cytotoxic agent or a radiotoxic agent, or one or more additional human antibodies of the invention (e.g., a human antibody having a complementary activity which binds to an epitope in the IRT A-2 antigen distinct from the first human antibody).

Accordingly, patients treated with antibody compositions of the invention can be additionally administered (prior to, simultaneously with, or following administration of a human antibody of the invention) with another therapeutic agent, such as a cytotoxic or radiotoxic agent, which enhances or augments the therapeutic effect of the human antibodies.

In other embodiments, the subject can be additionally treated with an agent that modulates, e.g., enhances or inhibits, the expression or activity of Fcγ or Fcγ receptors by, for example, treating the subject with a cytokine. Preferred cytokines for administration during

treatment with the multispecific molecule include of granulocyte colony-stimulating factor (G-CSF), granulocyte- macrophage colony-stimulating factor (GM-CSF), interferon-γ (IFN- γ), and tumor necrosis factor (TNF).

The compositions (e.g., human antibodies, multispecific and bispecific molecules) of the invention can also be used to target cells expressing FcγR or IRTA-2, for example for labeling such cells. For such use, the binding agent can be linked to a molecule that can be detected. Thus, the invention provides methods for localizing ex vivo or in vitro cells expressing Fc receptors, such as FcγR, or IRTA-2. The detectable label can be, e.g., a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.

In a particular embodiment, the invention provides methods for detecting the presence of IRTA-2 antigen in a sample, or measuring the amount of IRTA-2 antigen, comprising contacting the sample, and a control sample, with a human monoclonal antibody, or an antigen binding portion thereof, which specifically binds to IRTA-2, under conditions that allow for formation of a complex between the antibody or portion thereof and IRTA-2. The formation of a complex is then detected, wherein a difference complex formation between the sample compared to the control sample is indicative the presence of IRTA-2 antigen in the sample.

In other embodiments, the invention provides methods for treating an IRTA-2 mediated disorder in a subject, e.g., cancer, non-Hodgkin's lymphoma, Burkitt's lymphoma, anaplastic large-cell lymphomas (ALCL), cutaneous T-cell lymphomas, nodular small cleaved-cell lymphomas, lymphocytic lymphomas, peripheral T-cell lymphomas, Lennert's lymphomas, immunoblastic lymphomas, T-cell leukemia/lymphomas (ATLL), adult T-cell leukemia (T-ALL), entroblastic/centrocytic (cb/cc) follicular lymphomas cancers, diffuse large cell lymphomas of B lineage, angioimmunoblastic lymphadenopathy (AILD)-like T cell lymphoma, HIV associated body cavity based lymphomas, Embryonal Carcinomas, undifferentiated carcinomas of the rhino-pharynx (e.g., Schmincke's tumor), Castleman's disease, Kaposi's Sarcoma and other B-cell lymphomas, by administering to the subject the human antibodies described above. Such antibodies and derivatives thereof are used to inhibit IRTA-2 induced activities associated with certain disorders, e.g., proliferation and differentiation. By contacting the antibody with IRTA-2 (e.g., by administering the antibody to a subject), the ability of IRTA-2 to induce such activities is inhibited and, thus, the associated disorder is treated. The antibody composition can be administered alone or along with another therapeutic agent, such as a cytotoxic or a radiotoxic agent which acts in

conjunction with or synergistically with the antibody composition to treat or prevent the IRTA-2 mediated disease.

In yet another embodiment, immunoconjugates of the invention can be used to target compounds {e.g., therapeutic agents, labels, cytotoxins, radiotoxoins immunosuppressants, etc.) to cells which have IRTA-2 cell surface receptors by linking such compounds to the antibody. Thus, the invention also provides methods for localizing ex vivo or in vivo cells expressing IRTA-2 (e.g., with a detectable label, such as a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor). Alternatively, the immunoconjugates can be used to kill cells which have IRTA-2 cell surface receptors by targeting cytotoxins or radiotoxins to IRTA-2.

The present invention is further illustrated by the following examples which should not be construed as further limiting. The contents of all figures and all references, patents and published patent applications cited throughout this application are expressly incorporated herein by reference.

Examples

Example 1: Generation of Human Monoclonal Antibodies Against IRTA-2

Antigen

A recombinant fusion protein composed of the extracellular domain of the IRTA-2 linked to a non-IRTA-2 polypeptide was generated by standard recombinant methods and used as antigen for immunization.

Transgenic HuMAb Mouse ® and KM Mouse ®

Fully human monoclonal antibodies to IRTA-2 were prepared using HCo7/HCol2 strains of the transgenic HuMAb Mouse ® and the KM strain of transgenic transchromosomic mice, each of which express human antibody genes. In each of these mouse strains, the endogenous mouse kappa light chain gene has been homozygously disrupted as described in Chen et al. (1993) EMBO J. 12:811-820 and the endogenous mouse heavy chain gene has been homozygously disrupted as described in Example 1 of PCT Publication WO 01/09187. Each of these mouse strains carries a human kappa light chain transgene, KCo5, as described in Fishwild et al. (1996) Nature Biotechnology 14:845-851. The HCo7 strain carries the HCo7 human heavy chain transgene as described in U.S. Patent Nos. 5,545,806; 5,625,825;

and 5,545,807. The HCol2 strain carries the HCol2 human heavy chain transgene as described in Example 2 of PCT Publication WO 01/09187. The KM Mouse ® strain contains the SC20 transchromosome as described in PCT Publication WO 02/43478.

HuMab and KM Immunizations:

To generate fully human monoclonal antibodies to IRTA-2, mice of the HuMAb Mouse ® and KM Mouse ® were immunized with purified recombinant IRTA-2 fusion protein derived from NSO cells that had been transfected with an expression vector containing the gene encoding the fusion protein. General immunization schemes for the HuMAb Mouse ® are described in Lonberg, N. et al (1994; Nature 368(6474): 856-859; Fishwild, D. et al. (1996) Nature Biotechnology 14:845-851 and PCT Publication WO 98/24884. The mice were 6-16 weeks of age upon the first infusion of antigen. A purified recombinant preparation (5-50 μg) of IRTA-2 fusion protein was used to immunize the HuMab mice and KM mice ® intraperitonealy, subcutaneously (Sc) or via footpad injection.

Transgenic mice were immunized twice with antigen in complete Freund's adjuvant or Ribi adjuvant either intraperitonealy (IP), subcutaneously (Sc) or via footpad (FP), followed by 3-21 days IP, Sc or FP immunization (up to a total of 11 immunizations) with the antigen in incomplete Freund's or Ribi adjuvant. The immune response was monitored by retroorbital bleeds. The plasma was screened by ELISA (as described below), and mice with sufficient titers of anti-IRTA-2 human immunogolobulin were used for fusions. Mice were boosted intravenously with antigen 3 and 2 days before sacrifice and removal of the spleen. Typically, 10-35 fusions for each antigen were performed. Several dozen mice were immunized for each antigen.

Selection of HuMab or KM Mice ® Producing Anti-IRTA-2 Antibodies:

To select HuMab or KM mice ® producing antibodies that bound IRTA-2, sera from immunized mice was tested by ELISA as described by Fishwild, D. et al. (1996). Briefly, microtiter plates were coated with purified recombinant IRTA-2 at 1-2 μg /ml in PBS, 50 μl/wells incubated 4 0 C overnight then blocked with 200 μl/well of 5% chicken serum in PBS/Tween (0.05%). Dilutions of plasma from IRTA-2-immunized mice were added to each well and incubated for 1-2 hours at ambient temperature. The plates were washed with PBS/Tween and then incubated with a goat-anti-human IgG Fc polyclonal antibody conjugated with horseradish peroxidase (HRP) for 1 hour at room temperature. After

washing, the plates were developed with ABTS substrate (Sigma, A-1888, 0.22 mg/ml) and analyzed by spectrophotometer at OD 415-495. Mice that developed the highest titers of anti-IRTA-2 antibodies were used for fusions. Fusions were performed as described below and hybridoma supernatants were tested for anti-IRTA-2 activity by ELISA.

Generation of Hybridomas Producing Human Monoclonal Antibodies to IRTA-2

The mouse splenocytes, isolated from the HuMab mice and KM mice ® , were fused with PEG to a mouse myeloma cell line based upon standard protocols. The resulting hybridomas were then screened for the production of antigen-specific antibodies. Single cell suspensions of splenic lymphocytes from immunized mice were fused to one-fourth the number of SP2/0 nonsecreting mouse myeloma cells (ATCC, CRL 1581) with 50% PEG (Sigma). Cells were plated at approximately 1x10 5 /well in flat bottom microtiter plate, followed by about two week incubation in selective medium containing 10% fetal bovine serum, 10% P388D1 (ATCC, CRL TIB-63) conditioned medium, 3-5% origen (IGEN) in DMEM (Mediatech, CRL 10013, with high glucose, L-glutamine and sodium pyruvate) plus 5 mM HEPES, 0.055 mM 2-mercaptoethanol, 50 mg/ml gentamycin and Ix HAT (Sigma, CRL P-7185). After 1-2 weeks, cells were cultured in medium in which the HAT was replaced with HT. Individual wells were then screened by ELISA (described above) for human anti-IRTA-2 monoclonal IgG antibodies. Once extensive hybridoma growth occurred, medium was monitored usually after 10-14 days. The antibody secreting hybridomas were replated, screened again and, if still positive for human IgG, anti-IRTA-2 monoclonal antibodies were subcloned at least twice by limiting dilution. The stable subclones were then cultured in vitro to generate small amounts of antibody in tissue culture medium for further characterization.

Hybridoma clones 9Dl 2, generated from a HuMAb Mouse ® , and 8Al, generated from a KM Mouse ® , were selected for further analysis.

Example 2; Structural Characterization of Human Monoclonal Antibodies 9D12 and 8Al

The cDNA sequences encoding the heavy and light chain variable regions of the 9Dl 2 and 8Al monoclonal antibodies were obtained from the 9Dl 2 and 8Al hybridomas, respectively, using standard PCR techniques and were sequenced using standard DNA sequencing techniques.

The nucleotide and amino acid sequences of the heavy chain variable region of 9Dl 2 are shown in Figure IA and in SEQ ID NOs: 13 and 17, respectively.

The nucleotide and amino acid sequences of the light chain variable region of 9Dl 2 are shown in Figure IB and in SEQ ID NOs: 15 and 19, respectively.

Comparison of the 9D12 heavy chain immunoglobulin sequence to the known human germ line immunoglobulin heavy chain sequences demonstrated that the 9Dl 2 heavy chain utilizes a VH segment from human germline VH 3-23, a D segment from the human germline 7-27, and a JH segment from human germline JH 3b. The alignment of the 9D12 VH sequence to the germline VH 3-23 sequence is shown in Figure 3. Further analysis of the 9Dl 2 VH sequence using the Kabat system of CDR region determination led to the delineation of the heavy chain CDRl, CDR2 and CD3 regions as shown in Figures IA and 3, and in SEQ ID NOs: 1, 3 and 5, respectively.

Comparison of the 9Dl 2 light chain immunoglobulin sequence to the known human germline immunoglobulin light chain sequences demonstrated that the 9Dl 2 light chain utilizes a VL segment from human germline VK L6 and a JK segment from human germline JK 1. The alignment of the 9Dl 2 VL sequence to the germline VK L6 sequence is shown in Figure 5. Further analysis of the 9Dl 2 VL sequence using the Kabat system of CDR region determination led to the delineation of the light chain CDRl, CDR2 and CD3 regions as shown in Figures IB and 5, and in SEQ ID NOs: 7, 9 and 11, respectively.

The nucleotide and amino acid sequences of the heavy chain variable region of 8Al are shown in Figure 2A and in SEQ ID NOs: 14 and 18, respectively.

The nucleotide and amino acid sequences of the light chain variable region of 8Al are shown in Figure 2B and in SEQ ID NOs: 16 and 20, respectively.

Comparison of the 8Al heavy chain immunoglobulin sequence to the known human germline immunoglobulin heavy chain sequences demonstrated that the 8Al heavy chain utilizes a VH segment from human germline VH 1-8, a D segment from the human germline 3-10, and a JH segment from human germline JH 6b. The alignment of the 8Al VH sequence to the germline VH 1-8 sequence is shown in Figure 4. Further analysis of the 8Al VH sequence using the Kabat system of CDR region determination led to the delineation of the heavy chain CDRl, CDR2 and CD3 regions as shown in Figures 2A and 4, and in SEQ ID NOs: 2, 4 and 6, respectively.

Comparison of the 8Al light chain immunoglobulin sequence to the known human germline immunoglobulin light chain sequences demonstrated that the 8Al light chain utilizes a VL segment from human germline VK Ll 8 and a JK segment from human germline

JK 2. The alignment of the 8Al VL sequence to the germline VK Ll 8 sequence is shown in Figure 6. Further analysis of the 8Al VL sequence using the Kabat system of CDR region determination led to the delineation of the light chain CDRl, CDR2 and CD3 regions as shown in Figures 2B and 6, and in SEQ ID NOs: 8, 10 and 12, respectively.

Example 3: Characterization of Binding Specificity and Binding Kinetics of Anti-IRTA-2 Human Monoclonal Antibodies

In this example, binding affinity and binding kinetics of anti-IRTA-2 antibodies were examined by Biacore analysis. Also, binding specificity was examined by flow cytometry.

Binding affinity and kinetics

Anti-IRTA-2 antibodies were characterized for affinities and binding kinetics by Biacore analysis (Biacore AB, Uppsala, Sweden). Purified anti-IRTA-2 monoclonal antibodies were captured by anti-human IgG antibody covalently linked to a CM5 chip (carboxy methyl dextran coated chip) via primary amines, using standard amine coupling chemistry and kit provided by Biacore. Binding was measured by flowing the IRTA-2-Fc antigen (a fusion protein composed of the extracellular domain of the IRTA-2 linked to the Fc portion of human IgGl) in HBS EP buffer (provided by Biacore AB) at concentrations of 400, 300, 200, 100, and 50 nM at a flow rate of 12 μl/min. The antigen-antibody association kinetics was followed for either 20 minutes (for 8Al) or 5 minutes (for 9D12) and the dissociation kinetics was followed for ether 10 minutes (for 8Al) or 5 minutes (for 9D12). The association and dissociation curves were fit to a 1:1 Langmuir binding model using BI Ae valuation software (Biacore AB). The KD, k on and k off values that were determined are shown in Table 1.

Table 1. Biacore binding data for IRTA-2 HuMAbs.

Binding specificity by flow cytometry

Chinese hamster ovary (CHO) cell lines that express either the IRTA-2, IRTA-3 or IRTA-4 protein at the cell surface were developed and used to determine the specificity of the

IRTA-2 monoclonal antibodies by flow cytometry. CHO cells were transfected with expression plasmids containing full length cDNA encoding transmembrane forms of IRTA-2, IRTA-3, or IRTA-4. In addition, the transfected proteins contained a myc tag at the N- terminus for detection by anti-myc antibody. Binding of the two anti-IRTA-2 monoclonal antibodies was assessed by incubating the transfected cells with each of the IRTA-2 Abs at a concentration of 10 μg/ml. The cells were washed and binding was detected with a phycoerythrin-labeled anti-human IgG Ab. A murine anti-myc Ab followed by labeled anti- murine IgG was used as the positive control. Secondary antibody alone was used as a negative control. The results are depicted in Figures 7A-C. The IRTA-2 monoclonal antibodies 9Dl 2 and 8Al bound to the CHO line transfected with IRTA-2 but not to CHO lines expressing IRTA-3 or -4, as measured by the mean fluorescent intensity (MFI) of staining. These data demonstrate the specificity of the monoclonal antibodies for IRTA-2.

Example 4: Binding of the IRTA-2 antibodies to normal B cells and to B cell-derived tumor lines

Two colored immunofluorescence and flow cytometry was employed to demonstrate the binding of the IRTA-2 HuMAbs to peripheral blood B cells. CD19 is a cell surface marker that can be used to distinguish peripheral blood B lymphocytes. Human peripheral blood mononuclear cells were incubated with biotinylated 9Dl 2, biotinylated 8Al, or an isotype control biotinylated human Ab. Cells were washed and were incubated with FITC- labeled streptavidin together with a phycoerythrin-labeled anti-CD19 antibody. Cells were washed and anylyzed by flow cytometry. The results are depicted in Figure 8. The IRTA-2 monoclonal antibodies 9Dl 2 and 8Al show increased binding to CD 19+ cells compared to a control isotype antibody, as measured by the mean fluorescent intensity (MFI) of staining. These data demonstrate that the IRTA-2 protein, as assessed by the monoclonal antibodies 9D12 and 8Al, is expressed by normal peripheral blood B lymphocytes.

Binding of the IRTA-2 HuMAbs by flow cytometry to the B cell tumor lines Ramos (ATCC CRL- 1596), Raji (ATCC CCL-86), Daudi (ATCC CCL-213), IM-9 (ATCC CCL- 159), Karpas 1106P (DSMZ ACC 545), SU-DHL-4 (DSMZ ACC 495), Granta 519 (DSMZ ACC 342), SU-DHL-6 (DSMZ ACC 572) and JEKO-I (DSMZ ACC 553) was assessed. The Ramos, Raji, Daudi, and IM-9 B cell tumor lines were incubated with each of the IRTA-2 HuMAbs, an anti-B-cell positive control antibody, or an isotype matched negative control human antibody, washed and detected by a phycoerythrin-labeled anti-human secondary

antibody. Cells were washed and analyzed by flow cytometry. The results for binding of the 9D12 and 8Al antibodies are shown in Figure 9. The IRTA-2 HuMAbs did not substantially bind to any of the B cell tumor lines Ramos, Raji, Daudi, or IM-9. The low level of binding observed for 9Dl 2 and 8Al to the IM-9 cell line was comparable to the level of binding seen for the isotype control antibody, thus demonstrating the binding was not specific. The Karpas 1106P, SU-DHL-4, Granta 519, SU-DHL-6 and JEKO-I cell lines were incubated with each of the IRTA-2 HuMAbs or a control human antibody, washed and detected by a phycoerythrin-labeled anti-human secondary antibody. The results for binding of the 9Dl 2 and 8Al antibodies are shown in Figures 10 and 11. These data show that anti-IRTA-2 antibodies did bind to Granta 519, SU-DHL-6 and JEKO-I tumor cells as compared to the control human antibody, but did not substantially bind to Karpas 1106P or SU-DHL-4 tumor cells. Together, these data demonstrate that certain B-cell tumor lines express the IRTA-2 protein on the cell surface.

SEQUENCE LISTING

Amino acid sequence of VH CDRl of 9D12 (SEQ ID NO: 1)

1 ssams

Amino acid sequence of VH CDRl of 8Al (SEQ ID NO: 2) 1 sydin

Amino acid sequence of VH CDR2 of 9D12 (SEQ ID NO: 3) 1 sisgsgdtty yadsvkg

Amino acid sequence of VH CDR2 of 8Al (SEQ ID NO: 4) 1 wmhpnsgntg yaqkfqg

Amino acid sequence of VH CDR3 of 9Dl 2 (SEQ ID NO: 5) 1 nwgaafdi

Amino acid sequence of VH CDR3 of 8Al (SEQ ID NO: 6) 1 grgitmvrgg iiyygmdv

Amino acid sequence of VK CDRl of 9D12 (SEQ ID NO: 7)

1 rasqsvssyl a

Amino acid sequence of VK CDRl of 8Al (SEQ ID NO: 8) 1 rasqgissal a

Amino acid sequence of VK CDR2 of 9D12 (SEQ ID NO: 9) ldasnrat

Amino acid sequence of VK CDR2 of 8Al (SEQ ID NO: 10) 1 dassles

Amino acid sequence of VK CDR 3 of 9D12 (SEQ ID NO: 11) 1 qqrsnwprwt

Amino acid sequence of VK CDR3 of 8Al (SEQ ID NO: 12) 1 qqfnsypht

Amino acid sequence of VH of 9Dl 2 (SEQ ID NO: 13)

1 evqllesggg lvqpggslrl scaasgftfs ssamswvrqa pgkglewvss isgsgdttyy

61 adsvkgrfti srdnskntly lqmnslrved aalyycaanw gaafdiwgqg tmvtvss

Amino acid sequence of VH of 8Al (SEQ ID NO: 14)

1 qmqlvqsgae vkkpgasvkv sckasgytfi sydinwvrqa tgqglewmgw mhpnsgntgy

61 aqkfqgrvtm trntsistay melsslrsed tavyycargr gitmvrggii yygmdvwgqg ttvtvss

Amino acid sequence of VK of 9D12 (SEQ ID NO: 15)

1 eivltqspat lslspgerat lscrasqsvs sylawyqqkp gqaprlliyd asnratgipa

61 rfsgsgsgtd ftltisslep edfavyycqq rsnwprwtfg qgtkveik

Amino acid sequence of VK of 8Al (SEQ ID NO: 16)

1 aiqltqspss lsasvgdrvt itcrasqgis salawyqqkp gkapklliyd asslesgvps

61 rfsgsgsgtd ftltisslqp edfatyycqq fnsyphtfgq gtkleik

Nucleotide Sequence of VH of 9Dl 2 (SEQ ID NO: 17)

1 GAG GTG CAA CTG TTG GAG TCT GGG GGA GGC TTG GTA CAG CCTGGG GGG TCC CTGAGA CTC TCC TGT GCA GCC TCT GGATTC ACC TTTAGC AGC TCTGCC ATGAGC TGG GTC CGC CAG GCT CCA GGG AAG GGG CTG GAG TGG GTC TCA TCTATTAGT GGTAGT GGTGATACC ACA TAC TAC GCA GAC TCC GTG AAG GGC CGG TTC ACC ATC TCC AGA GAC AATTCC AAG AATACG CTG TAT CTG CAA ATGAAC AGC CTG AGA GTC GAG GAC GCG GCC TTATATTAC TGT GCG GCTAAC TGG GGA GCT GCTTTTGATATC TGG GGC CAA GGG ACA ATG GTC ACC GTC TCTTCA

Nucleotide Sequence of VH of 8Al (SEQ ID NO: 18)

CAAATG CAG CTG GTG CAG TCT GGG GCT GAG GTG AAG AAG CCT GGG GCC TCA GTG AAG GTC TCC TGC AAG GCTTCTGGATAC ACC TTC ATTAGT TAT GATATC AAC TGG GTG CGA CAG GCC ACT GGA CAA GGG CTT GAGTGGATG GGA TGG ATG CAC CCTAAC AGT GGTAAC ACA GGC TAT GCA CAG AAG TTC CAG GGC AGA GTC ACC ATG ACC AGGAAC ACC TCC ATA AGC ACA GCC TAC ATG GAA CTGAGC AGC CTG AGA TCT GAG GAC ACG GCC GTG TATTAC TGT GCG AGA GGC CGA GGAATTACTATG GTT CGG GGA GGTATTATC TAC TAC GGTATG GAC GTC TGG GGC CAA GGG ACC ACG GTC ACC GTC TCC TCA

Nucleotide Sequence of VK of 9D12 (SEQ ID NO: 19)

GAA ATT GTG TTG ACA CAG TCT CCA GCC ACC CTG TCT TTG TCT CCA GGG GAA AGA GCC ACCCCTC TCC TGC AGG GCC AGT CAG AGT GTT AGC AGC TAC TTA GCC TGG TAC CAA CAG AAA CCT GCC CAG GCT CCC AGG CTC CTC ATC TAT GAT GCA TCC AAC AGG GCC ACT GGC ATC ACA GCC AGG TTC AGT GGC AGT GGG TCT GGG ACA GAC TTC ACT CTC ACC ATC AGC AGC CTA GAG CCT GAA GAT TTT GCA GTT TAT TAC TGT CAG CAG CGT AGC AAC TGG CCT CGG TGG ACG TTC GGC CAA GGG ACC AAG G TG GAA ATC AAA

Nucleotide Sequence of VK of 8Dl (SEQ ID NO: 20)

GCC ATC CAG TTG ACC CAG TCT CCA TCC TCC CTG TCT GCA TCT GTA GGA GAC AGA GTC ACC ATC ACT TGC CGG GCA AGT CAG GGC ATT AGC AGT GCT TTA GCC TGG TAT CAG CAG AAA CCA GGG AAA GCT CCT AAG CTC CTG ATC TAT GAT GCC TCC AGT TTG GAA AGT GGG GTC CCA TCA AGG TTC AGC GGC

AGT GGA TCT GGG ACA GATTTC ACT CTC ACC ATC AGC AGC CTG CAG CCT GAA GATTTT GCAACTTATTAC TGT CAA CAGTTT AATAGTTAC CCTCAC ACTTTTGGC CAG GGGACC AAG CTG GAGATC AAA

Amino acid sequence of VH germline 3-23 (SEQ ID NO: 21)

1 evqllesggg lvqpggslrl scaasgftfs sylamswvrq apgkglewvs aisgsggsty

61 yadsvkgrft isrdnskntl ylqmnslrae dtavyyca

Amino acid sequence of VH germline 1-8 (SEQ ID NO: 22)

1 qvqlvqsgae vkkpgasvkv kvsckasgy tftsydinwv rqatgqglew mgwmnpnsgn

61 tgyaqkfqgr vtmtrntsis taymelsslr sedtavyycar g

Amino acid sequence of VK L6 germline (SEQ ID NO: 23)

1 eivltqspat lslspgerat lscrasqsvs sylawyqqkp gqaprlliyd asnratgipa

61 rfsgsgsgtd ftltisslep edfavyycqq rsnwp

Amino acid sequence of VK L18 germline (SEQ ID NO: 24)

1 aiqltqspss lsasvgdrvt itcrasqgis salawyqqkp gkapklliyd asslesgvps

61 rfsgsgsgtd ftltisslqp edfatyycqq fhsyp

Amino acid sequence of IRTA-2 (SEQ ID NO: 25) 1 mllwvillvl apvsgqfart prpiiflqpp wttvfqgerv tltckgfrfy spqktkwyhr 61 ylgkeilret pdnilevqes geyrcqaqgs plsspvhldf ssaslilqap Isvfegdsvv 121 lrcrakaevt lnntiykndn vlaflnkrtd fhiphaclkd ngayrctgyk esccpvssnt 181 vkiqvqepft rpvlrassfq pisgnpvtlt cetqlslers dvplrfrffr ddqtlglgws 241 lspnfqitam wskdsgfywc kaatmphsvi sdsprswiqv qipashpvlt lspekalnfe 301 gtkvtlhcet qedslrtlyr fyhegvplrh ksvrcergas isfslttens gnyyctadng 361 lgakpskavs lsvtvpvshp vlnlsspedl ifegakvtlh ceaqrgslpi lyqfhhedaa 421 lerrsansag gvaisfslta ehsgnyycta dngfgpqrsk avslsitvpv shpvltlssa 481 ealtfegatv tlhcevqrgs pqilyqfyhe dmplwssstp svgrvsfsfs lteghsgnyy 541 ctadngfgpq rsevvslfvt vpvsrpiltl rvpraqavvg dllelhceap rgsppilywf 601 yhedvtlgss sapsggeasf nlsltaehsg nysceanngl vaqhsdtisl svivpvsrpi 661 ltfrapraqa vvgdllelhc ealrgsspil ywfyhedvtl gkisapsggg asfnlsltte 721 hsgiyscead ngpeaqrsem vtlkvavpvs rpvltlrapg thaavgdlle lhcealrgsp

781 lilyrffhed vtlgnrssps ggaslnlslt aehsgnysce adnglgaqrs etvtlyitgl 841 tanrsgpfat gvaggllsia glaagallly cwlsrkagrk pasdparspp dsdsqeptyh 901 nvpaweelqp vytnanprge nvvysevrii qekkkhavas dprhlrnkgs piiysevkva 961 stpvsgslfl assaphr

Amino acid sequence of IRTA-I (SEQ ID NO: 26) 1 mllwasllaf apvcgqsaaa hkpvisvhpp wttffkgerv tltcngfqfy atekttwyhr 61 hywgekltlt pgntlevres glyrcqargs prsnpvrllf ssdslilqap ysvfegdtlv 121 lrchrrrkek ltavkytwng nilsisnksw dllipqassn nngnyrcigy gdendvfrsn 181 fkiikiqelf phpelkatds qptegnsvnl scetqlpper sdtplhfnff rdgevilsdw 241 stypelqlpt vwrensgsyw cgaetvrgni hkhspslqih vqripvsgvl letqpsggqa 301 vegemlvlvc svaegtgdtt fswhredmqe slgrktqrsl raelelpair qshaggyyct 361 adnsygpvqs mvlnvtvret pgnrdglvaa gatggllsal llavallfhc wrrrksgvgf 421 lgdetrlppa pgpgesshsi cpaqvelqsl yvdvhpkkgd lvyseiqttq lgeeeeants 481 rtlledkdvs vvysevktqh pdnsagkiss kdees

Amino acid sequence of IRTA-3 (SEQ ID NO: 27) 1 mllwllllil tpgreqsgva pkavlllnpp wstafkgekv alicssishs laqgdtywyh 61 dekllkikhd kiqitepgny qcktrgssls davhvefspd wlilqalhpv fegdnvilrc 121 qgkdnknthq kvyykdgkql pnsynlekit vnsvsrdnsk yhctayrkfy ildievtskp 181 lniqvqelfl hpvlrassst piegspmtlt cetqlspqrp dvqlqfslfr dsqtlglgws 241 rsprlqipam wtedsgsywc evetvthsik krslrsqirv qrvpvsnvnl eirptggqli 301 egenmvlics vaqgsgtvtf swhkegrvrs lgrktqrsll aelhvltvke sdagryycaa 361 dnvhspilst wirvtvripv shpvltfrap rahtvvgdll elhceslrgs ppilyrfyhe 421 dvtlgnssap sgggasfhls ltaehsgnys cdadnglgaq hshgvslrvt vpvsrpvltl 481 rapgaqavvg dllelhcesl rgsfpilywfyheddtlgni sahsgggasf nlslttehsg 541 nysceadngl gaqhskvvtl nvtgtsrnrt gltaagitgl vlsilvlaaa aallhyarar 601 rkpgglsatg tsshspsecq epsssrpsri dpqepthskp lapmelepmy snvnpgdsnp 661 iysqiwsiqh tkensancpm mhqeheeltv lyselkkthp ddsageassr graheeddee 721 nyenvprvll asdh

Amino acid sequence of IRTA-4 (SEQ ID NO: 28)

1 mllwsllvif davteqadsl tlvapssvfe gdsivlkcqg eqnwkiqkma yhkdnkelsv

61 fkkfsdfliq savlsdsgny fcstkgqlfl wdktsnivki kvqelfqrpv ltassfqpie

121 ggpvslkcet rlspqrldvq lqfcffrenq vlgsgwsssp elqisavwse dtgsywckae 181 tvthrirkqs lqsqihvqri pisnvsleir apggqvtegq klillcsvag gtgnvtfswy 241 reatgtsmgk ktqrslsael eipavkesda gkyycradng hvpiqskvvn ipvripvsrp 301 vltlrspgaq aavgdllelh cealrgsppi lyqfyhedvt lgnssapsgg gasfnlslta 361 ehsgnyscea nnglgaqcse avpvsisgpd gyrrdlmtag vlwglfgvlg ftgvalllya 421 lfhkisgess atneprgasr pnpqeftyss ptpdmeelqp vyvnvgsvdv dvvysqvwsm 481 qqpessanir tllenkdsqv iyssvkks

Amino acid sequence of IRTA-5 (SEQ ID NO: 29)

1 mlprllllic aplcepaelf liaspshpte gspvtltckm pflqssdaqf qfcffrdtra

61 Igpgwssspk lqiaamwked tgsywceaqt maskvlrsrr sqinvhrvpv advsletqpp

121 ggqvmegdrl vlicsvamgt gditflwykg avglnlqskt qrsltaeyei psvresdaeq

181 yycvaengyg pspsglvsit vripvsrpil mlrapraqaa vedvlelhce alrgsppily

241 wfyheditlg srsapsggga sftilslteeh sgnysceann glgaqrseav tlnftvptga

301 rsnhltsgvi egllstlgpa tvallfcygl krkigrrsar dplrslpspl pqeftylnsp

361 tpgqlqpiye nvnvvsgdev yslayynqpe qesvaaetlg thmedkvsld iysrlrkani

421 tdvdyedam

SUMMARY OF SEQUENCE LISTING