Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTIBODY THERAPEUTICS THAT BIND PSMA
Document Type and Number:
WIPO Patent Application WO/2016/145139
Kind Code:
A1
Abstract:
There is disclosed compositions and methods relating to or derived from anti-PSMA antibodies. More specifically, there is disclosed fully human antibodies that bind PSMA, PSMA-antibody binding fragments and derivatives of such antibodies, and PSMA-binding polypeptides comprising such fragments. Further still, there is disclosed nucleic acids encoding such antibodies, antibody fragments and derivatives and polypeptides, cells comprising such polynucleotides, methods of making such antibodies, antibody fragments and derivatives and polypeptides, and methods of using such antibodies, antibody fragments and derivatives and polypeptides, including methods of treating a disease. There is further disclosed targeted therapies for treatment of prostate cancer having the disclosed anti-PSMA antibodies and fragments thereof targeting the therapeutic agent or cell.

Inventors:
GROS EDWIGE (US)
ZHOU HEYUE (US)
YALAMANCHILI SILPA
Application Number:
PCT/US2016/021672
Publication Date:
September 15, 2016
Filing Date:
March 10, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SORRENTO THERAPEUTICS INC (US)
International Classes:
A61K39/395; C07K16/28
Domestic Patent References:
WO1999001475A21999-01-14
WO2009127046A12009-10-22
Foreign References:
US20110189093A12011-08-04
US20120316071A12012-12-13
US20090123476A12009-05-14
US8026344B22011-09-27
US8030457B22011-10-04
US6846634B12005-01-25
US6696245B22004-02-24
US20020002512A12002-01-03
US20040202995A12004-10-14
US20040038291A22004-02-26
US20040009507A12004-01-15
US20030039958A12003-02-27
US6010902A2000-01-04
US5582996A1996-12-10
US5959083A1999-09-28
US5807706A1998-09-15
Other References:
LANDIS, S. H. ET AL., CA CANCER J. CLIN., vol. 48, 1998, pages 6 - 29
PARKER ET AL., CA CANCER J. CLIN., vol. 47, 1997, pages 5 - 280
WINGO ET AL., CA CANCER J. CLIN., vol. 46, 1996, pages 113 - 25
CHANG, REV. UROLOGY, vol. 6, no. S10, 2004, pages S13 - S18
CHANG ET AL., CANCER RES., vol. 59, 1999, pages 3192 - 3198
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 26
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 83
BRUIIIMELL, BIOCHERN, vol. 32, 1993, pages 1180 - 1187
KOBAYASHI ET AL., PROTEIN ENG., vol. 12, no. 10, 1999, pages 879 - 884
BURKS ET AL., PROC. NATL. ACAD. SET. USA, vol. 94, 1997, pages 412 - 417
NEAR ET AL., MOL. IMMUNOL., vol. 30, 1993, pages 369 - 377
MILSTEIN ET AL., NATURE, vol. 305, 1983, pages 537
BRENNAN ET AL., SCIENCE, vol. 229, 1985, pages 81
GLENNIE ET AL., J. IMMUNOL., vol. 139, 1987, pages 2367
KOSTELNY ET AL., J. IMMUNOL., vol. 148, 1992, pages 1547
"Cloning Vectors: A Laboratory Manual", 1985, ELSEVIER
"Solid Phase Peptide Synthesis", 1984, THE PIERCE CHEMICAL CO.
BOWIE ET AL., SCIENCE, vol. 253, 1991, pages 164
"Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS & WILKINS
See also references of EP 3268039A4
Attorney, Agent or Firm:
COWLES, Cristin H. et al. (LLP265 Franklin Stree, Boston MA, US)
Download PDF:
Claims:
We claim:

1. An isolated fully human anti-PSMA antibody of an IgG class that binds to a PSMA epitope, said antibody comprising:

a heavy chain variable domain sequence that is at least 95% identical to the amino acid sequence of SEQ ID NO. 1; and

a light chain variable domain sequence that is at least 95% identical to an amino acid sequence selected from the group consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, and SEQ ID NO. 17.

2. The fully human antibody of claim 1, wherein the antibody comprises a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2 (PS Al l), SEQ ID NO. 1/SEQ ID NO. 3 (PSGB 11), SEQ ID NO.

1/SEQ ID NO. 4 (PSGB 12), SEQ ID NO. 1/SEQ ID NO. 5 ( PSGC8), SEQ ID NO. 1/SEQ ID NO. 6 (PSGC9), SEQ ID NO. 1/SEQ ID NO. 7 (PSGC12), SEQ ID NO. 1/SEQ ID NO. 8 (PSGD3), SEQ ID NO. 1/SEQ ID NO. 9 (PSGD4), SEQ ID NO. 1/SEQ ID NO. 10 (PSGD6 ), SEQ ID NO. 1/SEQ ID NO. 11 (PSGEIO), SEQ ID NO. 1/SEQ ID NO. 12 (PSGEl l), SEQ ID NO. 1/SEQ ID NO. 13 (PSGF9), SEQ ID NO. 1/SEQ ID NO. 14 (PSGF11), SEQ ID NO. 1/SEQ ID NO. 15 (PSGG6 ), SEQ ID NO. 1/SEQ ID NO. 16 (PSGH3), and SEQ ID NO. 1/SEQ ID NO. 17 (PSGH8).

3. The fully human antibody of claim 1 or 2, wherein the antibody has a KD of at least 1 x 10"6 M.

4. An anti-PSMA fully human antibody Fab fragment, comprising

a heavy chain variable domain comprising an amino acid sequence that is at least 95% identical to the amino acid sequence SEQ ID NO. 1; and

a light chain variable domain comprising an amino acid sequence that is at least 95% identical to an amino acid sequence selected from the group consisting of: SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, and SEQ ID NO. 17.

5. The fully human antibody Fab fragment of claim 4, wherein the antibody comprises a heavy chain/light chain variable domain sequence selected from the group consisting of: SEQ ID NO. 1/SEQ ID NO. 2, SEQ ID NO. 1/SEQ ID NO. 3, SEQ ID NO. 1/SEQ ID NO. 4, SEQ ID NO. 1/SEQ ID NO. 5, SEQ ID NO. 1/SEQ ID NO. 6, SEQ ID NO. 1/SEQ ID NO. 7, SEQ ID NO. 1/SEQ ID NO. 8, SEQ ID NO. 1/SEQ ID NO. 9, SEQ ID NO. 1/SEQ ID NO. 10, SEQ ID NO. 1/SEQ ID NO. 11, SEQ ID NO. 1/SEQ ID NO. 12, SEQ ID NO. 1/SEQ ID NO. 13, SEQ ID NO. 1/SEQ ID NO. 14, SEQ ID NO. 1/SEQ ID NO. 15, SEQ ID NO. 1/SEQ ID NO. 16, and SEQ ID NO. 1/SEQ ID NO. 17.

6. The fully human antibody Fab fragment of claim 4 or 5, wherein the antibody has a KD of at least 1 x 10"6 M.

7. An anti-PSMA single chain human antibody, comprising a heavy chain variable domain and a light chain variable domain connected by a peptide linker,

wherein the heavy chain variable domain comprises an amino acid sequence that is at least 95% identical to the amino acid sequence SEQ ID NO. 1; and

wherein the light chain variable domain comprises an amino acid sequence that is at least 95% identical to an amino acid sequence selected from the group consisting of: SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, and SEQ ID NO. 17.

8. The fully human single chain antibody of claim 7, wherein the single chain fully human antibody comprises a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2, SEQ ID NO. 1/SEQ ID NO. 3, SEQ ID NO. 1/SEQ ID NO. 4, SEQ ID NO. 1/SEQ ID NO. 5, SEQ ID NO. 1/SEQ ID NO. 6, SEQ ID NO. 1/SEQ ID NO. 7, SEQ ID NO. 1/SEQ ID NO. 8, SEQ ID NO. 1/SEQ ID NO. 9, SEQ ID NO. 1/SEQ ID NO. 10, SEQ ID NO. 1/SEQ ID NO. 11, SEQ ID NO. 1/SEQ ID NO. 12, SEQ ID NO. 1/SEQ ID NO. 13, SEQ ID NO. 1/SEQ ID NO. 14, SEQ ID NO. 1/SEQ ID NO. 15, SEQ ID NO. 1/SEQ ID NO. 16, and SEQ ID NO. 1/SEQ ID NO. 17.

9. The fully human single chain antibody of claim 7 or 8, wherein the antibody has a KD of at least 1 x 10"6 M.

10. A method for treating prostate cancer, said method comprising administering an anti-PSMA antibody or antibody fragment of any one of claims 1-9, to a subject in need thereof.

11. A method for treating cancer, said method comprising administering an anti- PSMA antibody, or antibody fragment of any one of claims 1-9, to a subject in need thereof.

12. The method of claim 11, wherein the cancer is selected from the group consisting of ovarian cancer, colon cancer, breast cancer, lung cancer, myeloma,

neuroblastic-derived CNS tumor, monocytic leukemia, B-cell derived leukemia, T-cell derived leukemia, B-cell derived lymphoma, T-cell derived lymphoma, and a mast cell derived tumor.

13. An isolated anti-PSMA antibody, or an antigen-binding fragment thereof, comprising

a heavy chain variable domain comprising complementarity determining regions

(CDRs) as set forth in SEQ ID NO. 1; and

a light chain variable domain comprising CDRs as set forth in a light chain variable region amino acid sequence selected from the group consisting of: SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, and SEQ ID NO. 17.

14. A pharmaceutical composition comprising the anti-PSMA antibody, or antibody fragment of any one of claims 1-9 or 13, and a pharmaceutically acceptable carrier.

15. A method of treating cancer associated with PSMA expression in a human subject in need thereof, comprising administering an effective amount of the anti-PSMA antibody, or antigen-binding fragment thereof, of any one of claims 1-9 or 13 to the subject, such that the cancer is treated.

16. The method of claim 15, wherein the cancer is prostate cancer.

17. The method of claim 15, wherein the cancer is selected from the group consisting of ovarian cancer, colon cancer, breast cancer, lung cancer, myeloma,

neuroblastic-derived CNS tumor, monocytic leukemia, B-cell derived leukemia, T-cell derived leukemia, B-cell derived lymphoma, T-cell derived lymphoma, and mast cell derived tumor.

Description:
Antibody Therapeutics That Bind PSMA

Related Applications

This application claims priority to United States Provisional Application No.

62/131,227, filed on March 10, 2015, the entire contents of which are incorporated by reference in its entirety herein.

Technical Field

The present disclosure provides compositions and methods relating to or derived from anti-PSMA (prostate-specific membrane antigen) antibodies. More specifically, the present disclosure provides fully human antibodies that bind PSMA, PSMA-antibody binding fragments and derivatives of such antibodies, and PSMA-binding polypeptides comprising such fragments. Further still, the present disclosure provides nucleic acids encoding such antibodies, antibody fragments and derivatives and polypeptides, cells comprising such polynucleotides, methods of making such antibodies, antibody fragments and derivatives and polypeptides, and methods of using such antibodies, antibody fragments and derivatives and polypeptides, including methods of treating a disease. The present disclosure further provides targeted therapies for treatment of prostate cancer having the disclosed anti-PSMA antibodies and fragments thereof targeting the therapeutic agent or cell.

Background

Prostate cancer is the most prevalent type of cancer and the second leading cause of death from cancer in American men (Landis, S. H. et al. CA Cancer J. Clin. 48:6-29 (1998)). The number of men diagnosed with prostate cancer is steadily increasing as a result of the increasing population of older men as well as a greater awareness of the disease leading to its earlier diagnosis (Parker et al., 1997, CA Cancer J. Clin. 47:5-280). The life time risk for men developing prostate cancer is about 1 in 5 for Caucasians, 1 in 6 for African Americans. High risk groups are represented by those with a positive family history of prostate cancer or African Americans.

Over a lifetime, more than ¾ of the men diagnosed with prostate cancer die of the disease (Wingo et al., 1996, CA Cancer J. Clin. 46: 113-25). Moreover, many patients who do not succumb to prostate cancer require continuous treatment to ameliorate symptoms such as pain, bleeding and urinary obstruction. Thus, prostate cancer also represents a major cause of suffering and increased health care expenditures.

Radiation therapy has also been used as an alternative to radical prostatectomy.

Patients generally treated by radiation therapy are those who are older and less healthy and those with higher- grade, more clinically advanced tumors. Particularly preferred procedures are external-beam therapy which in vol ves three dimensional, confocal radiation therapy where the field of radiation is designed to conform to the volume of tissue treated; interstitial- radiation therapy where seeds of radioactive compounds are implanted using ultrasound guidance: and a combination of external-beam therapy and interstitial-radiation therapy.

For treatment of patients with locally advanced disease, hormonal therapy before or fol lowing radical prostatectomy or radiation therapy has been utilized. Hormonal therapy is the main form of treating men with disseminated prostate cancer. Orchiectomy reduces serum testosterone concentrations, while estrogen treatment is similarly beneficial . Diethylstilbestrol from estrogen is another useful hormonal therapy which has a disadvantage of causing cardiovascular toxicity. When gonadotropin-releasing hormone agonists are administered testosterone concentrations are ultimately reduced. Flutamide and other nonsteroidal, anti- androgen agents block binding of testosterone to its intracellular receptors. As a result, it blocks the effect of testosterone, increasing serum testosterone concentrations and allows patients to remain potent, a significant problem after radical prostatectomy and radiation treatments.

Cytotoxic chemotherapy is largely ineffective in treating prostate cancer. Its toxicity makes such therapy unsuitable for elderly patients, in addition, prostate cancer is relatively resistant to cytotoxic agents.

Relapsed or more advanced disease is also treated with a ti-androgen therapy.

Unfortunately, almost all tumors become hormone-resistant and progress rapidly in the absence of any effective therapy.

Accordingly, there is a need for effective therapeutics for prostate cancer which are not overwhelmingly toxic to normal tissues of a patient, and which are effective in selectively eliminating prostate cancer cells.

Summary of the Invention

This invention pertains to binding proteins capable of binding to pro state- specific membrane antigen (PSMA), (e.g., human PSMA), including anti-PSMA antibodies, and antigen-binding fragments thereof.

In one aspect, the present disclosure provides an isolated fully human anti-PSMA antibody of an IgG class that binds to a PSMA epitope, said antibody comprising a heavy chain variable domain sequence that is at least 95% identical to the amino acid sequence SEQ ID NO. 1 ; and a light chain variable domain sequence that is at least 95% identical to an amino acid sequence selected from the group consisting of: SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, and SEQ ID NO. 17.

In another aspect, the present disclosure provides a fully human antibody of an IgG class that binds to a PSMA epitope with a binding affinity of at least 10 ~6 M, and comprises a heavy chain variable domain sequence that is at least 95% identical to the amino acid sequence SEQ ID NO. 1, and comprises a light chain variable domain sequence that is at least 95% identical to the amino acid sequence consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, SEQ ID NO. 17. In one embodiment, the fully human antibody comprises both a heavy chain and a light chain wherein the antibody has a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2 (called PSA11 herein), SEQ ID NO. 1/SEQ ID NO. 3 (called PSGB 11 herein), SEQ ID NO. 1/SEQ ID NO. 4 (called PSGB 12 herein), SEQ ID NO. 1/SEQ ID NO. 5 (called PSGC8 herein), SEQ ID NO. 1/SEQ ID NO. 6 (called PSGC9 herein), SEQ ID NO. 1/SEQ ID NO. 7 (called PSGC12 herein), SEQ ID NO. 1/SEQ ID NO. 8 (called PSGD3 herein), SEQ ID NO. 1/SEQ ID NO. 9 (called PSGD4 herein), SEQ ID NO. 1/SEQ ID NO. 10 (called PSGD6 herein), SEQ ID NO. 1/SEQ ID NO. 11 (called PSGEIO herein), SEQ ID NO. 1/SEQ ID NO. 12 (called PSGE11 herein), SEQ ID NO. 1/SEQ ID NO. 13 (called PSGF9 herein), SEQ ID NO. 1/SEQ ID NO. 14 (called PSGF11 herein), SEQ ID NO. 1/SEQ ID NO. 15 (called PSGG6 herein), SEQ ID NO. 1/SEQ ID NO. 16 (called PSGH3 herein), SEQ ID NO. 1/SEQ ID NO. 17 (called PSGH8 herein).

In another aspect, the present disclosure provides an anti-PSMA Fab fully human antibody fragment, having a variable domain region from a heavy chain and a variable domain region from a light chain, wherein the heavy chain variable domain comprises an amino acid sequence that is at least 95% identical to the amino acid sequence of SEQ ID NO. 1, and comprises a light chain variable domain sequence that is at least 95% identical to an amino acid sequence selected from the group consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, SEQ ID NO. 17. In one embodiment, the fully human antibody Fab fragment comprises both a heavy chain variable domain region and a light chain variable domain region wherein the antibody has a heavy chain/light chain variable domain sequence selected from the group consisting SEQ ID NO. 1/SEQ ID NO. 2, SEQ ID NO. 1/SEQ ID NO. 3, SEQ ID NO. 1/SEQ ID NO. 4, SEQ ID NO. 1/SEQ ID NO. 5, SEQ ID NO. 1/SEQ ID NO. 6, SEQ ID NO. 1/SEQ ID NO. 7, SEQ ID NO. 1/SEQ ID NO. 8, SEQ ID NO. 1/SEQ ID NO. 9, SEQ ID NO. 1/SEQ ID NO. 10, SEQ ID NO. 1/SEQ ID NO. 11, SEQ ID NO. 1/SEQ ID NO. 12, SEQ ID NO. 1/SEQ ID NO. 13, SEQ ID NO. 1/SEQ ID NO. 14, SEQ ID NO. 1/SEQ ID NO. 15, SEQ ID NO. 1/SEQ ID NO. 16, SEQ ID NO. 1/SEQ ID NO. 17.

In another aspect, the present disclosure provides an anti-PSMA single chain human antibody, comprising a variable domain region from a heavy chain and a variable domain region from a light chain and a peptide linker connection the heavy chain and light chain variable domain regions, wherein the heavy chain variable domain comprises an amino acid sequence that is at least 95% identical to the amino acid sequence of SEQ ID NO. 1, and comprises a light chain variable domain sequence that is at least 95% identical to an amino acid sequence selected from the group consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, SEQ ID NO. 17. In one embodiment, the fully human single chain antibody comprises both a heavy chain variable domain region and a light chain variable domain region, wherein the single chain fully human antibody comprises a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2, SEQ ID NO. 1/SEQ ID NO. 3, SEQ ID NO. 1/SEQ ID NO. 4, SEQ ID NO. 1/SEQ ID NO. 5, SEQ ID NO. 1/SEQ ID NO. 6, SEQ ID NO. 1/SEQ ID NO. 7, SEQ ID NO. 1/SEQ ID NO. 8, SEQ ID NO. 1/SEQ ID NO. 9, SEQ ID NO. 1/SEQ ID NO. 10, SEQ ID NO. 1/SEQ ID NO. 11, SEQ ID NO. 1/SEQ ID NO. 12, SEQ ID NO. 1/SEQ ID NO. 13, SEQ ID NO. 1/SEQ ID NO. 14, SEQ ID NO. 1/SEQ ID NO. 15, SEQ ID NO. 1/SEQ ID NO. 16, and SEQ ID NO. 1/SEQ ID NO. 17.

In one embodiment, the invention features an isolated anti-PSMA antibody, or an antigen-binding fragment thereof, comprising a heavy chain variable domain comprising complementarity determining regions (CDRs) as set forth in SEQ ID NO. 1 ; and comprising a light chain variable domain comprising CDRs as set forth in a light chain variable region amino acid sequence selected from the group consisting of: SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, and SEQ ID NO. 17. In one embodiment, the isolated anti- PSMA antibody, or antigen-binding fragment thereof, is human. The present disclosure further provides a method for treating a prostate cancer, comprising administering an anti-PSMA antibody used to target a chemical or cellular therapeutic payload, wherein the anti-PSMA antibody has a heavy chain variable domain sequence that is at least 95% identical to the amino acid sequence SEQ ID NO. 1, and that has a light chain variable domain sequence that is at least 95% identical to the amino acid consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, SEQ ID NO. 17, combinations thereof.

The present disclosure further provides a method for treating a prostate cancer, comprising administering an anti-PSMA antibody Fab fully human fragment used to target a chemical or cellular therapeutic payload, wherein the Fab fully human antibody fragment comprises a heavy chain variable domain sequence that is at least 95% identical to the amino acid sequence SEQ ID NO. 1, and comprises a light chain variable domain sequence that is at least 95% identical to an amino acid sequence selected from the group consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, SEQ ID NO. 17.

The present disclosure also provides a method for treating a prostate cancer, comprising administering an anti-PSMA single chain human antibody used to target a chemical or cellular therapeutic payload, wherein the anti-PSMA antibody single chain human antibody comprises a heavy chain variable domain sequence that is at least 95% identical to the amino acid sequence SEQ ID NO. 1, and comprises a light chain variable domain sequence that is at least 95% identical to the amino acid sequence consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, SEQ ID NO. 17.

In one embodiment, the fully human antibody comprises both a heavy chain and a light chain, wherein the antibody has a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2 (called PSA11 herein), SEQ ID NO. 1/SEQ ID NO. 3 (called PSGB l l herein), SEQ ID NO. 1/SEQ ID NO. 4 (called PSGB 12 herein), SEQ ID NO. 1/SEQ ID NO. 5 (called PSGC8 herein), SEQ ID NO. 1/SEQ ID NO. 6 (called PSGC9 herein), SEQ ID NO. 1/SEQ ID NO. 7 (called PSGC12 herein), SEQ ID NO. 1/SEQ ID NO. 8 (called PSGD3 herein), SEQ ID NO. 1/SEQ ID NO. 9 (called PSGD4 herein), SEQ ID NO. 1/SEQ ID NO. 10 (called PSGD6 herein), SEQ ID NO. 1/SEQ ID NO. 11 (called PSGE10 herein), SEQ ID NO. 1/SEQ ID NO. 12 (called PSGE11 herein), SEQ ID NO. 1/SEQ ID NO. 13 (called PSGF9 herein), SEQ ID NO. 1/SEQ ID NO. 14 (called PSGF11 herein), SEQ ID NO. 1/SEQ ID NO. 15 (called PSGG6 herein), SEQ ID NO. 1/SEQ ID NO. 16 (called PSGH3 herein), and SEQ ID NO. 1/SEQ ID NO. 17 (called PSGH8 herein). In one embodiment, the fully human single chain antibody has both a heavy chain variable domain region and a light chain variable domain region, wherein the single chain fully human antibody has a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2, SEQ ID NO. 1/SEQ ID NO. 3, SEQ ID NO. 1/SEQ ID NO. 4, SEQ ID NO. 1/SEQ ID NO. 5, SEQ ID NO. 1/SEQ ID NO. 6, SEQ ID NO. 1/SEQ ID NO. 7, SEQ ID NO. 1/SEQ ID NO. 8, SEQ ID NO. 1/SEQ ID NO. 9, SEQ ID NO. 1/SEQ ID NO. 10, SEQ ID NO. 1/SEQ ID NO. 11, SEQ ID NO. 1/SEQ ID NO. 12, SEQ ID NO. 1/SEQ ID NO. 13, SEQ ID NO. 1/SEQ ID NO. 14, SEQ ID NO. 1/SEQ ID NO. 15, SEQ ID NO. 1/SEQ ID NO. 16, and SEQ ID NO. 1/SEQ ID NO. 17.

The present invention further provides in another aspect, a method for treating cancer, said method comprising administering an anti-PSMA antibody, or antibody fragment of any one of the aspects or embodiments described herein, to a subject in need thereof. In one embodiment, the cancer is selected from the group consisting of ovarian cancer, colon cancer, breast cancer, lung cancer, myeloma, neuroblastic-derived CNS tumor, monocytic leukemia, B-cell derived leukemia, T-cell derived leukemia, B-cell derived lymphoma, T-cell derived lymphoma, and mast cell derived tumor.

In certain embodiments, the anti-PSMA antibody is used to treat a broad spectrum of mammalian cancers, including, for example, ovarian, colon, breast, lung cancers, myelomas, neuroblastic-derived CNS tumors, monocytic leukemias, B-cell derived leukemias, T-cell derived leukemias, B-cell derived lymphomas, T-cell derived lymphomas, mast cell derived tumors, and combinations thereof.

The present invention also provides in another aspect, a method for treating prostate cancer, said method comprising administering an anti-PSMA antibody, or antibody fragment of any one of the aspects or embodiments described herein, to a subject in need thereof.

In certain embodiments, the antibody, or antigen-binding fragment thereof, of the invention has a binding affinity (KD) of at least 1 x 10 "6 M. In other embodiments, the antibody, or antigen-binding fragment thereof, of the invention has a KD of at least 1 x 10 " M. In other embodiments, the antibody, or antigen-binding fragment thereof, of the invention has a K D of at least 1 x 10 "8 M. In certain embodiments, the antibody is an IgGl isotype. In other embodiments, the antibody is an IgG4 isotype.

In certain embodiments, the antibody, or antigen-binding fragment, described herein is recombinant.

In certain embodiments, the antibody, or antigen-binding fragment, described herein is a human antibody, or antigen binding fragment of an antibody.

The invention also provides pharmaceutical compositions comprising an effective amount of an anti-PSMA antibody or fragment disclosed herein, and a pharmaceutically acceptable carrier.

Description of the Drawings

Figures 1A to ID graphically depict binding of anti-PSMA antibodies to PSMA- positive cells (LNCaP) (as described in Figures 1A and IB), and PSMA-negative normal human fibroblast cells (PC3), (as described in Figures 1C and ID).

Figures 2A to 2C illustrate cytotoxic potential of anti-PSMA antibodies PSGF9, PSGC9, PSGD6, (shown in Figure 2A); PSGD3, PSGE10, PSGH3, PSGB 11, (shown in

Figure 2B); and PSGD4 and PSGB 12 (shown in Figure 2C) complexed with Protein G (PG)- MMAF molecules on PSMA- overexpressing LNCaP cancer cells.

Figure 2D illustrates the non-specific cell killing effect observed on normal human fibroblasts (PC3) with various anti-PSMA antibodies complexed with Protein G -MMAF molecules.

Figure 3 graphically depicts affinity binding characteristics of anti-PSMA antibody

PSA11.

Figure 4 graphically depicts affinity binding characteristics of anti-PSMA antibody PSGB 11.

Detailed Description

Definitions

The terms "peptide," "polypeptide" and "protein" each refers to a molecule comprising two or more amino acid residues joined to each other by peptide bonds. These terms encompass, e.g., native and artificial proteins, protein fragments and polypeptide analogs (such as muteins, variants, and fusion proteins) of a protein sequence as well as post- translationally, or otherwise covalently or non-covalently, modified proteins. A peptide, polypeptide, or protein may be monomeric or polymeric. A "variant" of a polypeptide (for example, a variant of an antibody) comprises an amino acid sequence wherein one or more amino acid residues are inserted into, deleted from and/or substituted into the amino acid sequence relative to another polypeptide sequence. Disclosed variants include, for example, fusion proteins.

A "derivative" of a polypeptide is a polypeptide (e.g., an antibody) that has been chemically modified, e.g., via conjugation to another chemical moiety (such as, for example, polyethylene glycol or albumin, e.g., human serum albumin), phosphorylation, and glycosylation. Unless otherwise indicated, the term "antibody" includes, in addition to antibodies comprising two full-length heavy chains and two full-length light chains, derivatives, variants, fragments, and muteins thereof, examples of which are described below.

An "antigen binding protein" is a protein comprising a portion that binds to an antigen and, optionally, a scaffold or framework portion that allows the antigen binding portion to adopt a conformation that promotes binding of the antigen binding protein to the antigen. Examples of antigen binding proteins include antibodies, antibody fragments (e.g., an antigen binding portion of an antibody), antibody derivatives, and antibody analogs. The antigen binding protein can comprise, for example, an alternative protein scaffold or artificial scaffold with grafted CDRs or CDR derivatives. Such scaffolds include, but are not limited to, antibody-derived scaffolds comprising mutations introduced to, for example, stabilize the three-dimensional structure of the antigen binding protein as well as wholly synthetic scaffolds comprising, for example, a biocompatible polymer. See, for example, Korndorfer et al., 2003, Proteins: Structure, Function, and Bioinformatics, Volume 53, Issue 1: 121-129; Roque et al., 2004, Biotechnol. Prog. 20:639-654. In addition, peptide antibody mimetics ("PAMs") can be used, as well as scaffolds based on antibody mimetics utilizing fibronection components as a scaffold.

An antigen binding protein can have, for example, the structure of an

immunoglobulin. An "immunoglobulin" is a tetrameric molecule composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kDa) and one "heavy" chain (about 50-70 kDa). The amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function. Human light chains are classified as kappa or lambda light chains. Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively. Preferably, the anti-PSMA antibodies disclosed herein are characterized by their variable domain region sequences in the heavy V H and light V L amino acid sequences. Within light and heavy chains, the variable and constant regions are joined by a "J" region of about 12 or more amino acids, with the heavy chain also including a "D" region of about 10 more amino acids. See generally, Fundamental

Immunology Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989)). The variable regions of each light/heavy chain pair form the antibody binding site such that an intact immunoglobulin has two binding sites.

The variable regions of immunoglobulin chains exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs. From N-terminus to C-terminus, both light and heavy chains comprise the domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each domain is in accordance with the definitions of Kabat et al. in Sequences of Proteins of Immunological Interest, 5 th Ed., US Dept. of Health and Human Services, PHS, NIH, NIH Publication no. 91-3242, 1991. Other numbering systems for the amino acids in immunoglobulin chains include IMGT.RTM. (international

ImMunoGeneTics information system; Lefranc et al, Dev. Comp. Immunol. 29: 185-203; 2005) and AHo (Honegger and Pluckthun, J. Mol. Biol. 309(3):657-670; 2001).

An "antibody" refers to an intact immunoglobulin or to an antigen binding portion thereof that competes with the intact antibody for specific binding, unless otherwise specified. In one embodiment, an antibody comprises two identical heavy chains each comprising a heavy chain variable domain and heavy chain constant regions C H I, Cm and C H3 ; and comprises two identical light chains each comprising a light chain variable domain and a light chain constant region (C L ). In one embodiment, an antibody of the invention comprises a heavy and light chain variable domain sequence selected from those described herein in Table 3.

In certain embodiments, antibodies can be obtained from sources such as serum or plasma that contain immunoglobulins having varied antigenic specificity. If such antibodies are subjected to affinity purification, they can be enriched for a particular antigenic specificity. Such enriched preparations of antibodies usually are made of less than about 10% antibody having specific binding activity for the particular antigen. Subjecting these preparations to several rounds of affinity purification can increase the proportion of antibody having specific binding activity for the antigen. Antibodies prepared in this manner are often referred to as "monospecific."

The term "monospecific", as used herein, refers to an antibody that displays an affinity for one particular epitope. Monospecific antibody preparations can be made up of about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 99%, or 99.9% antibody having specific binding activity for the particular antigen.

In certain embodiments, an antigen binding protein, such as an antibody, may have one or more binding sites. If there is more than one binding site, the binding sites may be identical to one another or may be different. For example, a naturally occurring human immunoglobulin typically has two identical binding sites, while a "bispecific" or

"bifunctional" antibody has two different binding sites.

An "antibody fragment" or "antigen binding fragment of an antibody" comprises a portion of an intact antibody, and preferably comprises the antibody antigen binding or variable domains. Examples of an antibody fragment include a Fab, an Fab', an F(ab')2, an Fv fragment, and a linear antibody.

Antigen binding portions (or fragments) of an antibody may be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies. Antigen binding portions include, inter alia, Fab, Fab', F(ab') 2 , Fv, domain antibodies (dAbs), and complementarity determining region (CDR) fragments, single-chain antibodies (scFv), chimeric antibodies, diabodies, triabodies, tetrabodies, and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide.

A Fab fragment is a monovalent fragment having the V L , V R , C L and C H I domains; a F(ab') 2 fragment is a bivalent fragment having two Fab fragments linked by a disulfide bridge at the hinge region; a Fd fragment has the V H and C H I domains; an Fv fragment has the V L and V H domains of a single arm of an antibody; and a dAb fragment has a V H domain, a V L domain, or an antigen-binding fragment of a V H or V L domain (U.S . Patents 6,846,634;

6,696,245, US App. Pub.20/0202512; 2004/0202995; 2004/0038291; 2004/0009507 ;20 03/0039958, and Ward et al., Nature 341:544-546, 1989).

A single-chain antibody (scFv) is an antibody in which a V L and a V H region are joined via a linker {e.g., a synthetic sequence of amino acid residues) to form a continuous protein chain wherein the linker is long enough to allow the protein chain to fold back on itself and form a monovalent antigen binding site (see, e.g., Bird et al., 1988, Science

242:423-26 and Huston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-83).

Diabodies are bivalent antibodies comprising two polypeptide chains, wherein each polypeptide chain comprises V H and V L domains joined by a linker that is too short to allow for pairing between two domains on the same chain, thus allowing each domain to pair with a complementary domain on another polypeptide chain (see, e.g., Holliger et al., 1993, Proc. Natl. Acad. Sci. USA 90:6444-48, and Poljak et al., 1994, Structure 2: 1121-23). If the two polypeptide chains of a diabody are identical, then a diabody resulting from their pairing will have two identical antigen binding sites. Polypeptide chains having different sequences can be used to make a diabody with two different antigen binding sites. Similarly, triabodies and tetrabodies are antibodies comprising three and four polypeptide chains, respectively, and forming three and four antigen binding sites, respectively, which can be the same or different.

The term "human antibody" includes antibodies that have one or more variable and constant regions derived from human immunoglobulin sequences. In one embodiment, all of the variable and constant domains of the antibody are derived from human immunoglobulin sequences (referred to as "a fully human antibody"). These antibodies may be prepared in a variety of ways, examples of which are described below, including through the immunization with an antigen of interest of a mouse that is genetically modified to express antibodies derived from human heavy and/or light chain-encoding genes. In a preferred embodiment, a fully human antibody is made using recombinant methods such that the glycosylation pattern of the antibody is different than an antibody having the same sequence if it were to exist in nature.

A "humanized antibody" has a sequence that differs from the sequence of an antibody derived from a non-human species by one or more amino acid substitutions, deletions, and/or additions, such that the humanized antibody is less likely to induce an immune response, and/or induces a less severe immune response, as compared to the non-human species antibody, when it is administered to a human subject. In one embodiment, certain amino acids in the framework and constant domains of the heavy and/or light chains of the non- human species antibody are mutated to produce the humanized antibody. In another embodiment, the constant domain(s) from a human antibody are fused to the variable domain(s) of a non-human species. In another embodiment, one or more amino acid residues in one or more CDR sequences of a non-human antibody are changed to reduce the likely immunogenicity of the non-human antibody when it is administered to a human subject, wherein the changed amino acid residues either are not critical for immuno specific binding of the antibody to its antigen, or the changes to the amino acid sequence that are made are conservative changes, such that the binding of the humanized antibody to the antigen is not significantly worse than the binding of the non-human antibody to the antigen. Examples of how to make humanized antibodies may be found in U.S. Patents 6,054,297, 5,886,152 and 5,877,293. The term "chimeric antibody" refers to an antibody that contains one or more regions from one antibody and one or more regions from one or more other antibodies. In one embodiment, one or more of the CDRs are derived from a human anti-PSMA antibody. In another embodiment, all of the CDRs are derived from a human anti-PSMA antibody. In another embodiment, the CDRs from more than one human anti-PSMA antibodies are mixed and matched in a chimeric antibody. For instance, a chimeric antibody may comprise a CDRl from the light chain of a first human anti-PAR-2 antibody, a CDR2 and a CDR3 from the light chain of a second human anti-PSMA antibody, and the CDRs from the heavy chain from a third anti-PSMA antibody. Other combinations are possible.

Further, the framework regions may be derived from one of the same anti-PSMA antibodies, from one or more different antibodies, such as a human antibody, or from a humanized antibody. In one example of a chimeric antibody, a portion of the heavy and/or light chain is identical with, homologous to, or derived from an antibody from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is/are identical with, homologous to, or derived from an antibody (-ies) from another species or belonging to another antibody class or subclass. Also included are fragments of such antibodies that exhibit the desired biological activity (i.e., the ability to specifically bind PSMA).

A "neutralizing antibody" or an "inhibitory antibody" is an antibody that inhibits the proteolytic activation of PSMA when an excess of the anti-PSMA antibody reduces the amount of activation by at least about 20% using an assay such as those described herein in the Examples. In various embodiments, the antigen binding protein reduces the amount of amount of proteolytic activation of PSMA by at least 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 99%, and 99.9%.

A "CDR grafted antibody" is an antibody comprising one or more CDRs derived from an antibody of a particular species or isotype and the framework of another antibody of the same or different species or isotype.

A "multi- specific antibody" is an antibody that recognizes more than one epitope on one or more antigens. A subclass of this type of antibody is a "bi-specific antibody" which recognizes two distinct epitopes on the same or different antigens.

An antigen binding protein "specifically binds" to an antigen (e.g., PSMA) if it binds to the antigen with a dissociation constant of 1 nanomolar or less.

An "antigen binding domain, "antigen binding region," or "antigen binding site" is a portion of an antigen binding protein that contains amino acid residues (or other moieties) that interact with an antigen and contribute to the antigen binding protein's specificity and affinity for the antigen. For an antibody that specifically binds to its antigen, this will include at least part of at least one of its CDR domains.

The term "Fc polypeptide" includes native and mutein forms of polypeptides derived from the Fc region of an antibody. Truncated forms of such polypeptides containing the hinge region that promotes dimerization also are included. Fusion proteins comprising Fc moieties (and oligomers formed therefrom) offer the advantage of facile purification by affinity chromatography over Protein A or Protein G columns.

An "epitope" is the portion of a molecule that is bound by an antigen binding protein (e.g., by an antibody). An epitope can comprise non-contiguous portions of the molecule (e.g., in a polypeptide, amino acid residues that are not contiguous in the polypeptide's primary sequence but that, in the context of the polypeptide's tertiary and quaternary structure, are near enough to each other to be bound by an antigen binding protein).

The "percent identity" or "percent homology" of two polynucleotide or two polypeptide sequences is determined by comparing the sequences using the GAP computer program (a part of the GCG Wisconsin Package, version 10.3 (Accelrys, San Diego, Calif.)) using its default parameters.

The terms "polynucleotide," "oligonucleotide" and "nucleic acid" are used

interchangeably throughout and include DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), analogs of the DNA or RNA generated using nucleotide analogs (e.g., peptide nucleic acids and non-naturally occurring nucleotide analogs), and hybrids thereof. The nucleic acid molecule can be single- stranded or double- stranded. In one embodiment, the nucleic acid molecules of the invention comprise a contiguous open reading frame encoding an antibody, or a fragment, derivative, mutein, or variant thereof.

Two single- stranded polynucleotides are "the complement" of each other if their sequences can be aligned in an anti-parallel orientation such that every nucleotide in one polynucleotide is opposite its complementary nucleotide in the other polynucleotide, without the introduction of gaps, and without unpaired nucleotides at the 5' or the 3' end of either sequence. A polynucleotide is "complementary" to another polynucleotide if the two polynucleotides can hybridize to one another under moderately stringent conditions. Thus, a polynucleotide can be complementary to another polynucleotide without being its

complement.

A "vector" is a nucleic acid that can be used to introduce another nucleic acid linked to it into a cell. One type of vector is a "plasmid," which refers to a linear or circular double stranded DNA molecule into which additional nucleic acid segments can be ligated. Another type of vector is a viral vector (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), wherein additional DNA segments can be introduced into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors comprising a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. An "expression vector" is a type of vector that can direct the expression of a chosen polynucleotide.

A nucleotide sequence is "operably linked" to a regulatory sequence if the regulatory sequence affects the expression (e.g., the level, timing, or location of expression) of the nucleotide sequence. A "regulatory sequence" is a nucleic acid that affects the expression (e.g., the level, timing, or location of expression) of a nucleic acid to which it is operably linked. The regulatory sequence can, for example, exert its effects directly on the regulated nucleic acid, or through the action of one or more other molecules (e.g., polypeptides that bind to the regulatory sequence and/or the nucleic acid). Examples of regulatory sequences include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Further examples of regulatory sequences are described in, for example, Goeddel, 1990, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif, and Baron et al., 1995, Nucleic Acids Res. 23:3605-06.

A "host cell" is a cell that can be used to express a nucleic acid, e.g., a nucleic acid of the invention. A host cell can be a prokaryote, for example, E. coli, or it can be a eukaryote, for example, a single-celled eukaryote (e.g., a yeast or other fungus), a plant cell (e.g., a tobacco or tomato plant cell), an animal cell (e.g., a human cell, a monkey cell, a hamster cell, a rat cell, a mouse cell, or an insect cell) or a hybridoma. Examples of host cells include the COS-7 line of monkey kidney cells (ATCC CRL 1651) (see Gluzman et al., 1981, Cell 23: 175), L cells, C127 cells, 3T3 cells (ATCC CCL 163), Chinese hamster ovary (CHO) cells or their derivatives such as Veggie CHO and related cell lines which grow in serum-free media (see Rasmussen et al., 1998, Cytotechnology 28:31) or CHO strain DX-B 11, which is deficient in DHFR (see Urlaub et al., 1980, Proc. Natl. Acad. Sci. USA 77:4216-20), HeLa cells, BHK (ATCC CRL 10) cell lines, the CV1/EBNA cell line derived from the African green monkey kidney cell line CV1 (ATCC CCL 70) (see McMahan et al., 1991, EMBO J. 10:2821), human embryonic kidney cells such as 293,293 EBNA or MSR 293, human epidermal A431 cells, human Colo205 cells, other transformed primate cell lines, normal diploid cells, cell strains derived from in vitro culture of primary tissue, primary explants, HL-60, U937, HaK or Jurkat cells. In one embodiment, a host cell is a mammalian host cell. In one embodiment, a host cell is a mammalian host celL_but is not a human host cell.

Typically, a host cell is a cultured cell that can be transformed or transfected with a polypeptide-encoding nucleic acid, which can then be expressed in the host cell. The phrase "recombinant host cell" can be used to denote a host cell that has been transformed or transfected with a nucleic acid to be expressed. A host cell also can be a cell that comprises the nucleic acid but does not express it at a desired level unless a regulatory sequence is introduced into the host cell such that it becomes operably linked with the nucleic acid. It is understood that the term host cell refers not only to the particular subject cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to, e.g., mutation or environmental influence, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.

The term "recombinant antibody" refers to an antibody that is prepared according to standard recombinant expression methods. A recombinant antibody can, for example, be expressed from a cell or cell line transfected with an expression vector (or possibly more than one expression vector) comprising the coding sequence of the antibody, where said coding sequence is not naturally associated with the cell. In one embodiment, a recombinant antibody has a glycosylation pattern that is different than the glycosylation pattern of an antibody having the same sequence if it were to exist in nature. In one embodiment, a recombinant antibody is expressed in a mammalian host cell which is not a human host cell. Notably, individual mammalian host cells have unique glycosylation patterns.

The term "effective amount" as used herein, refers to that amount of an antibody, or an antigen binding portion thereof that binds PSMA, which is sufficient to effect treatment of a disease when administered to a subject. A therapeutically effective amount of an antibody, or fragment, provided herein will vary depending upon the relative activity of the antibodies and depending upon the subject and disease condition being treated, the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.

The term "isolated" refers to a protein (e.g., an antibody) that is substantially free of other cellular material. In one embodiment, an isolated antibody is substantially free of other proteins from the same species. In one embodiment, an isolated antibody is expressed by a cell from a different species and is substantially free of other proteins from the different species. A protein may be rendered substantially free of naturally associated components (or components associated with the cellular expression system used to produce the antibody) by isolation, using protein purification techniques well known in the art. In one embodiment, the anti-PSMA antibodies, or antigen binding fragments, of the invention are isolated.

PSMA Antigen Binding Proteins

The present invention pertains to PSMA binding proteins, particularly anti-PSMA antibodies, or antigen-binding portions thereof, that bind PSMA, e.g., human PSMA, and uses thereof. Various aspects of the invention relate to antibodies and antibody fragments, pharmaceutical compositions, nucleic acids, recombinant expression vectors, and host cells for making such antibodies and fragments. Methods of using the antibodies of the invention to detect human PSMA, to inhibit PSMA activity, either in vitro or in vivo, and to prevent or treat disorders such as cancer are also encompassed by the invention. In one embodiment, the antibody of the invention is a human antibody.

As described in Table 3 below, included in the invention are novel human antibody heavy and light chain variable regions that are specific to PSMA. In one embodiment, the invention provides an anti-PSMA antibody, or an antigen-binding fragment thereof, that comprises a heavy chain having a variable domain comprising an amino acid sequence as set forth in SEQ ID NO: l. In one embodiment, the invention provides an anti-PSMA antibody, or an antigen-binding fragment thereof, that comprises a light chain having a variable domain comprising an amino acid sequence as set forth in any one of SEQ ID Nos: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 and 17. In one embodiment, the invention provides an anti- PSMA antibody, or an antigen-binding fragment thereof, that comprises a light chain having a variable domain comprising an amino acid sequence as set forth in any one of SEQ ID Nos: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 and 17; and a heavy chain having a variable domain comprising an amino acid sequence as set forth in SEQ ID NO: 1.

Complementarity determining regions (CDRs) are known as hypervariable regions both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework (FR). Complementarity determining regions (CDRs) and framework regions (FR) of a given antibody may be identified using the system described by Kabat et al. supra; Lefranc et al., supra and/or Honegger and Pluckthun, supra. Also familiar to those in the art is the numbering system described in Kabat et al. (1991, NIH Publication 91-3242, National Technical Information Service, Springfield, Va.). In this regard Kabat et al. defined a numbering system for variable domain sequences that is applicable to any antibody. One of ordinary skill in the art can unambiguously assign this system of "Kabat numbering" to any variable domain amino acid sequence, without reliance on any experimental data beyond the sequence itself.

In certain embodiments, the present invention provides an anti-PSMA antibody comprising the CDRs of the heavy and light chain variable domains described in Table 3 (SEQ ID Nos: 1 to 17). For example, the invention provides an anti-PSMA antibody, or antigen-binding fragment thereof, comprising a heavy chain variable region having the CDRs described in an amino acid sequence as set forth in SEQ ID NO: l. In one embodiment, the invention provides an anti-PSMA antibody, or antigen-binding fragment thereof, comprising a light chain variable region having the CDRs described in an amino acid sequence as set forth in any one of SEQ ID Nos: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 and 17. In one embodiment, the invention provides an anti-PSMA antibody, or antigen-binding fragment thereof, comprising a light chain variable region having the CDRs described in an amino acid sequence as set forth in any one of SEQ ID Nos: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 and 17; and a heavy chain variable region having the CDRs described in an amino acid sequence as set forth in SEQ ID NO: l.

In one embodiment, the present disclosure provides an isolated anti-PSMA antibody, or antigen binding fragment thereof, that comprises a heavy chain comprising a variable domain comprising the amino acid sequence of SEQ ID NO: 1, and a light chain comprising a variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, SEQ ID NO. 17.

In one embodiment, the present disclosure provides a fully human antibody of an IgG class that binds to a PSMA epitope with a binding affinity of 10 "6 M or less, that has a heavy chain variable domain sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to the amino acid sequence SEQ ID NO. 1, and that has a light chain variable domain sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to the amino acid sequence consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, SEQ ID NO. 17, and combinations thereof.

In one embodiment, the fully human antibody has both a heavy chain and a light chain wherein the antibody has a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2 (called PSA11 herein), SEQ ID NO. 1/SEQ ID NO. 3 (called PSGB 11 herein), SEQ ID NO. 1/SEQ ID NO. 4 (called PSGB 12 herein), SEQ ID NO. 1/SEQ ID NO. 5 (called PSGC8 herein), SEQ ID NO. 1/SEQ ID NO. 6 (called PSGC9 herein), SEQ ID NO. 1/SEQ ID NO. 7 (called PSGC12 herein), SEQ ID NO. 1/SEQ ID NO. 8 (called PSGD3 herein), SEQ ID NO. 1/SEQ ID NO. 9 (called PSGD4 herein), SEQ ID NO. 1/SEQ ID NO. 10 (called PSGD6 herein), SEQ ID NO. 1/SEQ ID NO. 11 (called PSGE10 herein), SEQ ID NO. 1/SEQ ID NO. 12 (called PSGE11 herein), SEQ ID NO. 1/SEQ ID NO. 13 (called PSGF9 herein), SEQ ID NO. 1/SEQ ID NO. 14 (called PSGFl l herein), SEQ ID NO. 1/SEQ ID NO. 15 (called PSGG6 herein), SEQ ID NO. 1/SEQ ID NO. 16 (called PSGH3 herein), SEQ ID NO. 1/SEQ ID NO. 17 (called PSGH8 herein), and combinations thereof.

In one embodiment, the invention provides an anti-PSMA antibody, or an antigen- binding fragment thereof, comprising a heavy chain comprising a CDR3 domain as set forth in SEQ ID NO: 1, and comprising a variable domain comprising an amino acid sequence that is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence as set forth in SEQ ID NO: 1. In one embodiment, the invention provides an anti- PSMA antibody, or an antigen-binding fragment thereof, comprising a light chain comprising a CDR3 domain as set forth in any one of SEQ ID Nos: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 and 17 and comprising a light chain variable domain comprising an amino acid sequence that has at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence as set forth in any one of SEQ ID Nos: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 and 17. Thus, in certain embodiments, the CDR3 domain is held constant, while variability may be introduced into the remaining CDRs and/or framework regions of the heavy and/or light chains, while the antibody, or antigen binding fragment thereof, retains the ability to bind to PSMA and retains the functional characteristics, e.g., binding affinity, of the parent.

In one embodiment, the substitutions made within a heavy or light chain that is at least 95% identical (or at least 96% identical, or at least 97% identical, or at least 98% identical, or at least 99% identical) are conservative amino acid substitutions. A

"conservative amino acid substitution" is one in which an amino acid residue is substituted by another amino acid residue having a side chain (R group) with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of a protein. In cases where two or more amino acid sequences differ from each other by conservative substitutions, the percent sequence identity or degree of similarity may be adjusted upwards to correct for the conservative nature of the substitution. Means for making this adjustment are well-known to those of skill in the art. See, e.g. , Pearson (1994) Methods Mol. Biol. 24: 307-331, herein incorporated by reference. Examples of groups of amino acids that have side chains with similar chemical properties include (1) aliphatic side chains: glycine, alanine, valine, leucine and isoleucine; (2) aliphatic-hydroxyl side chains: serine and threonine; (3) amide-containing side chains: asparagine and glutamine; (4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; (5) basic side chains: lysine, arginine, and histidine; (6) acidic side chains: aspartate and glutamate, and (7) sulfur-containing side chains are cysteine and methionine.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having the antigen binding regions of any of the antibodies described in Table 3.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSA11. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 2. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 2. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 2. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGB 11. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 3. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 3. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 3. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGB 12. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 4. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO:4. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 4. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGC8. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 5. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 5. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 5. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGC9. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 6. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 6. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 6. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGC12. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 7. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 7. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 7. The antibody may further be an IgGl or an IgG4 isotype. In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGD3. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 8. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 8. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 8. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGD4. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 9. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 9. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 9. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGD6. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 10. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 10. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 10. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGE10. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 11. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 11. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 11. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGE11. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 12. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 12. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 12. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGF9. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 13. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 13. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 13. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGF11. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 14. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 14. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 14. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGG6. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 15. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 15. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 15. The antibody may further be an IgGl or an IgG4 isotype.

In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGH3. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 16. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 16. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 16. The antibody may further be an IgGl or an IgG4 isotype. In one embodiment, the present invention is directed to an antibody, or an antigen binding fragment thereof, having antigen binding regions of antibody PSGH8. In one embodiment, the invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable domain sequence as set forth in SEQ ID NO: 1, and a light chain variable domain sequence as set forth in SEQ ID NO: 17. In one embodiment, the invention is directed to an antibody having a heavy chain variable domain comprising the CDRs of SEQ ID NO: 1, and a light chain variable domain comprising the CDRs of SEQ ID NO: 17. In one embodiment, the invention features an isolated human antibody, or antigen- binding fragment thereof, that comprises a heavy chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 1 and comprises a light chain variable region having an amino acid sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the sequence set forth in SEQ ID NO: 17. The antibody may further be an IgGl or an IgG4 isotype.

Antigen binding proteins (e.g., antibodies, antibody fragments, antibody derivatives, antibody muteins, and antibody variants) are polypeptides that bind to PSMA.

Antigen-binding fragments of antigen binding proteins of the invention may be produced by conventional techniques. Examples of such fragments include, but are not limited to, Fab and F(ab')2 fragments.

Single chain antibodies may be formed by linking heavy and light chain variable domain (Fv region) fragments via an amino acid bridge (short peptide linker), resulting in a single polypeptide chain. Such single-chain Fvs (scFvs) have been prepared by fusing DNA encoding a peptide linker between DNAs encoding the two variable domain polypeptides (V L and V H ). The resulting polypeptides can fold back on themselves to form antigen -binding monomers, or they can form multimers (e.g., dimers, trimers, or tetramers), depending on the length of a flexible linker between the two variable domains (Kortt et al., 1997, Prot. Eng. 10:423; Kortt et al., 2001, Biomol. Eng. 18:95-108). By combining different V L and V H - comprising polypeptides, one can form multimeric scFvs that bind to different epitopes (Kriangkum et al., 2001, Biomol. Eng. 18:31-40). Techniques developed for the production of single chain antibodies include those described in U.S. Patent 4,946,778; Bird, 1988, Science 242:423; Huston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879; Ward et al., 1989, Nature 334:544, de Graaf et al., 2002, Methods Mol. Biol. 178:379-87. In certain embodiments, the present disclosure provides a Fab fully human antibody fragment, having a variable domain region from a heavy chain and a variable domain region from a light chain, wherein the heavy chain variable domain sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to the amino acid sequence SEQ ID NO. 1, and that has a light chain variable domain sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to the amino acid sequence consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, SEQ ID NO. 17, and

combinations thereof. In one embodiment, the fully human antibody Fab fragment has both a heavy chain variable domain region and a light chain variable domain region wherein the antibody has a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2, SEQ ID NO. 1/SEQ ID NO. 3, SEQ ID NO. 1/SEQ ID NO. 4, SEQ ID NO. 1/SEQ ID NO. 5, SEQ ID NO. 1/SEQ ID NO. 6, SEQ ID NO. 1/SEQ ID NO. 7, SEQ ID NO. 1/SEQ ID NO. 8, SEQ ID NO. 1/SEQ ID NO. 9, SEQ ID NO. 1/SEQ ID NO. 10, SEQ ID NO. 1/SEQ ID NO. 11, SEQ ID NO. 1/SEQ ID NO. 12, SEQ ID NO. 1/SEQ ID NO. 13, SEQ ID NO. 1/SEQ ID NO. 14, SEQ ID NO. 1/SEQ ID NO. 15, SEQ ID NO. 1/SEQ ID NO. 16, SEQ ID NO. 1/SEQ ID NO. 17, and combinations thereof.

In one embodiment, the present disclosure provides a single chain human antibody, having a variable domain region from a heavy chain and a variable domain region from a light chain and a peptide linker connection the heavy chain and light chain variable domain regions, wherein the heavy chain variable domain sequence is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to the amino acid sequence of SEQ ID NO. 1, and wherein the light chain variable domain sequence is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to an amino acid sequence selected from the group consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, SEQ ID NO. 17, and combinations thereof. In one embodiment, the fully human single chain antibody has both a heavy chain variable domain region and a light chain variable domain region, wherein the single chain fully human antibody has a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2, SEQ ID NO. 1/SEQ ID NO. 3, SEQ ID NO. 1/SEQ ID NO. 4, SEQ ID NO. 1/SEQ ID NO. 5, SEQ ID NO. 1/SEQ ID NO. 6, SEQ ID NO. 1/SEQ ID NO. 7, SEQ ID NO. 1/SEQ ID NO. 8, SEQ ID NO. 1/SEQ ID NO. 9, SEQ ID NO. 1/SEQ ID NO. 10, SEQ ID NO. 1/SEQ ID NO. 11, SEQ ID NO. 1/SEQ ID NO. 12, SEQ ID NO. 1/SEQ ID NO. 13, SEQ ID NO. 1/SEQ ID NO. 14, SEQ ID NO. 1/SEQ ID NO. 15, SEQ ID NO. 1/SEQ ID NO. 16, SEQ ID NO. 1/SEQ ID NO. 17, and combinations thereof.

Techniques are known for deriving an antibody of a different subclass or isotype from an antibody of interest, i.e., subclass switching. Thus, IgG antibodies may be derived from an IgM antibody, for example, and vice versa. Such techniques allow the preparation of new antibodies that possess the antigen-binding properties of a given antibody (the parent antibody), but also exhibit biological properties associated with an antibody isotype or subclass different from that of the parent antibody. Recombinant DNA techniques may be employed. Cloned DNA encoding particular antibody polypeptides may be employed in such procedures, e.g., DNA encoding the constant domain of an antibody of the desired isotype (Lantto et al., 2002, Methods Mol. Biol. 178:303-16). Moreover, if an IgG4 is desired, it may also be desired to introduce a point mutation (CPSC->CPPC) in the hinge region (Bloom et al., 1997, Protein Science 6:407) to alleviate a tendency to form intra-H chain disulfide bonds that can lead to heterogeneity in the IgG4 antibodies. Thus, in one embodiment, the antibody of the invention is a human IgGl antibody. Thus, in one embodiment, the antibody of the invention is a human IgG4 antibody.

The present disclosure provides a number of antibodies structurally characterized by the amino acid sequences of their variable domain regions. However, the amino acid sequences can undergo some changes while retaining their high degree of binding to their specific targets. More specifically, many amino acids in the variable domain region can be changed with conservative substitutions and it is predictable that the binding characteristics of the resulting antibody will not differ from the binding characteristics of the wild type antibody sequence. There are many amino acids in an antibody variable domain that do not directly interact with the antigen or impact antigen binding and are not critical for

determining antibody structure. For example, a predicted nonessential amino acid residue in any of the disclosed antibodies is preferably replaced with another amino acid residue from the same class. Methods of identifying amino acid conservative substitutions which do not eliminate antigen binding are well- known in the art (see, e.g., Brummell et a!., Biochem. 32: 1180-1187 (1993); Kobayashi et al. Protein Eng. 12(10):879-884 (1999); and Burks et al Proc. Nail. Acad. Set USA 94:412-417 (1997)). Near et al. Mol. Immunol. 30:369-377, 1993 explains how to impact or not impact binding through site-directed mutagenesis. Near et al. only mutated residues that they thought had a high probability of changing antigen binding. Most had a modest or negative effect on binding affinity (Near et al. Table 3) and binding to different forms of digoxin (Near et al. Table 2). Thus, the invention also includes, in certain embodiments, variable sequences having at least 95% identity, at least 96% identity, at least 97% identity, at least 98% identity, or at least 99% identity to those sequences disclosed herein.

In certain embodiments, an antibody, or antigen-binding fragment thereof, provided herein has a binding affinity (K D ) of 1 x 10 ~6 M or less; 5 x 10 ~7 M or less' 1 x 10 ~7 M or less; 5 x 10 ~8 M or less; 1 x 10 ~8 M or less; 5 x 10 "9 M or less; or 1 x 10 "9 M or less. In one embodiment, the antibody, or antigen-binding fragment thereof, of the invention as a K D from

1 x 10 " 7' M to 1 x 10 " 1 1 0 U M. In one embodiment, the antibody, or antigen-binding fragment thereof, of the invention as a K D from 1 x 10 ° M to 1 x 10 "1U M.

Those of ordinary skill in the art will appreciate standard methods known for determining the K D of an antibody, or fragment thereof. For example, in one embodiment, K D is measured by a radiolabeled antigen binding assay (RIA). In one embodiment, an RIA is performed with the Fab version of an antibody of interest and its antigen. For example, solution binding affinity of Fabs for antigen is measured by equilibrating Fab with a minimal concentration of ( 125 I)-labeled antigen in the presence of a titration series of unlabeled antigen, then capturing bound antigen with an anti-Fab antibody-coated plate (see, e.g., Chen et al, J. Mol. Biol. 293:865-881(1999)).

According to another embodiment, K D is measured using a BIACORE surface plasmon resonance assay. The term "surface plasmon resonance", as used herein, refers to an optical phenomenon that allows for the analysis of real-time interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the

BIACORE system (Biacore Life Sciences division of GE Healthcare, Piscataway, NJ).

In particular embodiments, antigen binding proteins of the present invention have a K A for PSMA of at least 10 6 . In other embodiments, the antigen binding proteins exhibit a K A of at least 10 7 , at least 108 , at least 109 , or at least 1010. In another embodiment, the antigen binding protein exhibits a K A substantially the same as that of an antibody described herein in the Examples.

In another embodiment, the present disclosure provides an antigen binding protein that has a low dissociation rate from PSMA. In one embodiment, the antigen binding protein has a K 0 ff of 1 X 10 "4 to 1 or lower. In another embodiment, the K 0 ff is 5 X 10 "5 to 1 or lower. In another embodiment, the K 0 ff is substantially the same as an antibody described herein. In another embodiment, the antigen binding protein binds to PSMA with substantially the same Koff as an antibody described herein.

In another aspect, the present disclosure provides an antigen binding protein that inhibits an activity of PSMA. In one embodiment, the antigen binding protein has an IC 50 of 1000 nM or lower. In another embodiment, the IC 50 is 100 nM or lower; in another embodiment, the IC 50 is 10 nM or lower. In another embodiment, the IC 50 is substantially the same as that of an antibody described herein in the Examples. In another embodiment, the antigen binding protein inhibits an activity of PSMA with substantially the same IC 50 as an antibody described herein.

In another aspect, the present disclosure provides an antigen binding protein that binds to PSMA expressed on the surface of a cell and, when so bound, inhibits PSMA signaling activity in the cell without causing a significant reduction in the amount of PSMA on the surface of the cell. Any method for determining or estimating the amount of PSMA on the surface and/or in the interior of the cell can be used. In other embodiments, binding of the antigen binding protein to the PSMA-expressing cell causes less than about 75%, 50%, 40%, 30%, 20%, 15%, 10%, 5%, 1%, or 0.1% of the cell-surface PSMA to be internalized.

In another aspect, the present disclosure provides an antigen binding protein having a half-life of at least one day in vitro or in vivo (e.g., when administered to a human subject). In one embodiment, the antigen binding protein has a half-life of at least three days. In another embodiment, the antigen binding protein has a half-life of four days or longer. In another embodiment, the antigen binding protein has a half-life of eight days or longer. In another embodiment, the antigen binding protein is derivatized or modified such that it has a longer half-life as compared to the underivatized or unmodified antigen binding protein. In another embodiment, the antigen binding protein contains one or more point mutations to increase serum half life, such as described in WO00/09560, incorporated by reference herein.

The present disclosure further provides multi-specific antigen binding proteins, for example, bispecific antigen binding protein, e.g., antigen binding protein that bind to two different epitopes of PSMA, or to an epitope of PSMA and an epitope of another molecule, via two different antigen binding sites or regions. Moreover, bispecific antigen binding protein as disclosed herein can comprise a PSMA binding site from one of the herein- described antibodies and a second PSMA binding region from another of the herein-described antibodies, including those described herein by reference to other publications. Alternatively, a bispecific antigen binding protein may comprise an antigen binding site from one of the herein described antibodies and a second antigen binding site from another PSMA antibody that is known in the art, or from an antibody that is prepared by known methods or the methods described herein.

Numerous methods of preparing bispecific antibodies are known in the art. Such methods include the use of hybrid-hybridomas as described by Milstein et al., 1983, Nature 305:537, and chemical coupling of antibody fragments (Brennan et al., 1985, Science 229:81; Glennie et al., 1987, J. Immunol. 139:2367; U.S. Patent 6,010,902). Moreover, bispecific antibodies can be produced via recombinant means, for example by using leucine zipper moieties {i.e., from the Fos and Jun proteins, which preferentially form heterodimers;

Kostelny et al., 1992, J. Immunol. 148: 1547) or other lock and key interactive domain structures as described in U.S. Patent 5,582,996. Additional useful techniques include those described in U.S. Patents 5,959,083; and 5,807,706.

In another aspect, the antigen binding protein comprises a derivative of an antibody. The derivatized antibody can comprise any molecule or substance that imparts a desired property to the antibody, such as increased half-life in a particular use. The derivatized antibody can comprise, for example, a detectable (or labeling) moiety {e.g., a radioactive, colorimetric, antigenic or enzymatic molecule, a detectable bead (such as a magnetic or electrodense {e.g., gold) bead), or a molecule that binds to another molecule {e.g., biotin or streptavidin), a therapeutic or diagnostic moiety {e.g., a radioactive, cytotoxic, or

pharmaceutically active moiety), or a molecule that increases the suitability of the antibody for a particular use {e.g., administration to a subject, such as a human subject, or other in vivo or in vitro uses). Examples of molecules that can be used to derivatize an antibody include albumin {e.g., human serum albumin) and polyethylene glycol (PEG). Albumin-linked and PEGylated derivatives of antibodies can be prepared using techniques well known in the art. In one embodiment, the antibody is conjugated or otherwise linked to transthyretin (TTR) or a TTR variant. The TTR or TTR variant can be chemically modified with, for example, a chemical selected from the group consisting of dextran, poly(n-vinyl pyrrolidone), polyethylene glycols, propropylene glycol homopolymers, polypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols and polyvinyl alcohols.

An alternative approach to antibody-targeted therapy is to utilize anti-PSMA antibodies of the invention for delivery of cytotoxic drugs specifically to PSMA antigen- expressing cancer cells. In one embodiment, an anti-PSMA antibody, or fragment, of the invention is conjugated to a cytotoxic agent via a linker, to form an anti-PSMA Antibody Drug Conjugate (ADC). An alternative approach to antibody-targeted therapy is to utilize anti-PSMA antibodies for delivery of cytotoxic drugs specifically to PSMA antigen- expressing cancer cells. Various cytotoxic drugs are known in the art which can be conjugated with any of the antibodies disclosed herein to form an ADC, including, but not limited to, an auristatin. Auristatins, such as Monomethyl auristatin E (MMAE) and

Monomethyl auristatin F (MMAF) are antimitotic agents that inhibit cell division by blocking the polymerization of tubulin (Francisco et al. Blood. 2003 Aug 15; 102(4): 1458-65; Smith et al. Mol Cancer Ther June 2006 5; 1474-82). Antibody-drug conjugates (ADCs) composed of the auristatin MMAF linked to an anti-PSMA antibody, as described in Example 2 below, showed potent anti-tumor activity in PSMA-expressing tumor cell lines. Thus, in one embodiment, an anti-PSMA antibody, or fragment thereof, of the invention is conjugated to an auristatin, e.g., MMAF.

Oligomers that contain one or more antigen binding proteins may be employed as PSMA antagonists. Oligomers may be in the form of covalently-linked or non-covalently- linked dimers, trimers, or higher oligomers. Oligomers comprising two or more antigen binding protein are contemplated for use, with one example being a homodimer. Other oligomers include heterodimers, homotrimers, hetero trimers, homotetramers, heterotetramers, etc.

One embodiment is directed to oligomers comprising multiple antigen binding proteins joined via covalent or non-covalent interactions between peptide moieties fused to the antigen binding proteins. Such peptides may be peptide linkers (spacers), or peptides that have the property of promoting oligomerization. Leucine zippers and certain polypeptides derived from antibodies are among the peptides that can promote oligomerization of antigen binding proteins attached thereto, as described in more detail below.

In particular embodiments, the oligomers comprise from two to four antigen binding proteins. The antigen binding proteins of the oligomer may be in any form, such as any of the forms described above, e.g., variants or fragments. Preferably, the oligomers comprise antigen binding proteins that have PSMA binding activity.

In one embodiment, an oligomer is prepared using polypeptides derived from immunoglobulins. Preparation of Fusion Proteins Comprising Certain Heterologous

Polypeptides Fused to Various Portions of antibody-derived polypeptides (including the Fc domain) has been described, e.g., by Ashkenazi et al., 1991, Proc. Natl. Acad. Sci. USA 88: 10535; Byrn et al., 1990, Nature 344:677; and Hollenbaugh et al., 1992 "Construction of Immunoglobulin Fusion Proteins", in Current Protocols in Immunology, Suppl. 4, pages 10.19.1-10.19.11. One embodiment is directed to a dimer comprising two fusion proteins created by fusing a PSMA binding fragment of an anti-PSMA antibody to the Fc region of an antibody. The dimer can be made by, for example, inserting a gene fusion encoding the fusion protein into an appropriate expression vector, expressing the gene fusion in host cells transformed with the recombinant expression vector, and allowing the expressed fusion protein to assemble much like antibody molecules, whereupon interchain disulfide bonds form between the Fc moieties to yield the dimer.

Another method for preparing oligomeric antigen binding proteins involves use of a leucine zipper. Leucine zipper domains are peptides that promote oligomerization of the proteins in which they are found. Leucine zippers were originally identified in several DNA- binding proteins (Landschulz et al., 1988, Science 240: 1759), and have since been found in a variety of different proteins. Among the known leucine zippers are naturally occurring peptides and derivatives thereof that dimerize or trimerize. Examples of leucine zipper domains suitable for producing soluble oligomeric proteins are described in WO 94/10308, and the leucine zipper derived from lung surfactant protein D (SPD) described in Hoppe et al., 1994, FEBS Letters 344: 191. The use of a modified leucine zipper that allows for stable trimerization of a heterologous protein fused thereto is described in Fanslow et al., 1994, Semin. Immunol. 6:267-78. In one approach, recombinant fusion proteins comprising an anti- PSMA antibody fragment or derivative fused to a leucine zipper peptide are expressed in suitable host cells, and the soluble oligomeric anti-PSMA antibody fragments or derivatives that form are recovered from the culture supernatant.

Antigen binding proteins directed against PSMA can be used, for example, in assays to detect the presence of PSMA polypeptides, either in vitro or in vivo. The antigen binding proteins also may be employed in purifying PSMA proteins by immunoaffinity

chromatography. Blocking antigen binding proteins can be used in the methods disclosed herein. Such antigen binding proteins that function as PSMA antagonists may be employed in treating any PSMA-induced condition, including but not limited to various cancers.

Antigen binding proteins may be employed in an in vitro procedure, or administered in vivo to inhibit PSMA-induced biological activity. Disorders caused or exacerbated (directly or indirectly) by the proteolytic activation of PSMA, examples of which are provided herein, thus may be treated. In one embodiment, the present invention provides a therapeutic method comprising in vivo administration of a PSMA blocking antigen binding protein to a mammal in need thereof in an amount effective for reducing a PSMA-induced biological activity. In certain embodiments of the invention, antigen binding proteins include fully human monoclonal antibodies that inhibit a biological activity of PSMA.

Antigen binding proteins, including antibodies and antibody fragments described herein, may be prepared by any of a number of conventional techniques. For example, they may be purified from cells that naturally express them (e.g., an antibody can be purified from a hybridoma that produces it), or produced in recombinant expression systems, using any technique known in the art. See, for example, Monoclonal Antibodies, Hybridomas: A New Dimension in Biological Analyses, Kennet et al. (eds.), Plenum Press, New York (1980); and Antibodies: A Laboratory Manual, Harlow and Land (eds.), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., (1988).

Any expression system known in the art can be used to make the recombinant polypeptides, including antibodies and antibody fragments described herein, of the invention. In general, host cells are transformed with a recombinant expression vector that comprises DNA encoding a desired polypeptide. Among the host cells that may be employed are prokaryotes, yeast or higher eukaryotic cells. Prokaryotes include gram negative or gram positive organisms, for example E. coli or bacilli. Higher eukaryotic cells include insect cells and established cell lines of mammalian origin. Examples of suitable mammalian host cell lines include the COS-7 line of monkey kidney cells (ATCC CRL 1651) (Gluzman et al., 1981, Cell 23: 175), L cells, 293 cells, C127 cells, 3T3 cells (ATCC CCL 163), Chinese hamster ovary (CHO) cells, HeLa cells, BHK (ATCC CRL 10) cell lines, and the

CV1/EBNA cell line derived from the African green monkey kidney cell line CV1 (ATCC CCL 70) as described by McMahan et al., 1991, EMBO J. 10: 2821. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described by Pouwels et al. (Cloning Vectors: A Laboratory Manual, Elsevier, N.Y., 1985).

The transformed cells can be cultured under conditions that promote expression of the polypeptide, and the polypeptide recovered by conventional protein purification procedures. One such purification procedure includes the use of affinity chromatography, e.g., over a matrix having all or a portion (e.g., the extracellular domain) of PSMA bound thereto.

Polypeptides contemplated for use herein include substantially homogeneous recombinant mammalian anti-PSMA antibody polypeptides substantially free of contaminating

endogenous materials.

Antigen binding proteins may be prepared, and screened for desired properties, by any of a number of known techniques. Certain of the techniques involve isolating a nucleic acid encoding a polypeptide chain (or portion thereof) of an antigen binding protein of interest (e.g., an anti-PSMA antibody), and manipulating the nucleic acid through recombinant DNA technology. The nucleic acid may be fused to another nucleic acid of interest, or altered (e.g., by mutagenesis or other conventional techniques) to add, delete, or substitute one or more amino acid residues, for example.

Polypeptides of the present disclosure can be produced using any standard methods known in the art. In one example, the polypeptides are produced by recombinant DNA methods by inserting a nucleic acid sequence (e.g., a cDNA) encoding the polypeptide into a recombinant expression vector and expressing the DNA sequence under conditions promoting expression.

Nucleic acids encoding any of the various polypeptides disclosed herein may be synthesized chemically. Codon usage may be selected so as to improve expression in a cell. Such codon usage will depend on the cell type selected. Specialized codon usage patterns have been developed for E. coli and other bacteria, as well as mammalian cells, plant cells, yeast cells and insect cells. See for example: Mayfield et al., Proc. Natl. Acad. Sci. USA. 2003 100(2):438-42; Sinclair et al. Protein Expr. Purif. 2002 (1):96-105; Connell N D. Curr. Opin. Biotechnol. 2001 12(5):446-9; Makrides et al. Microbiol. Rev. 1996 60(3):512-38; and Sharp et al. Yeast. 1991 7(7):657-78.

In one embodiment, the invention features nucleic acids encoding the antibodies or antibody fragments described herein. For example, in one embodiment, the invention includes a nucleic acid encoding a heavy chain variable domain as set forth in SEQ ID NO: 1 and/or a light chain variable domain as set forth in any one of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, and SEQ ID NO. 17.

General techniques for nucleic acid manipulation are described for example in

Sambrook et al., Molecular Cloning: A Laboratory Manual, Vols. 1-3, Cold Spring Harbor Laboratory Press, 2 ed., 1989, or F. Ausubel et al., Current Protocols in Molecular Biology (Green Publishing and Wiley-Interscience: New York, 1987) and periodic updates, herein incorporated by reference. The DNA encoding the polypeptide is operably linked to suitable transcriptional or translational regulatory elements derived from mammalian, viral, or insect genes. Such regulatory elements include a transcriptional promoter, an optional operator sequence to control transcription, a sequence encoding suitable mRNA ribosomal binding sites, and sequences that control the termination of transcription and translation. The ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants is additionally incorporated.

The recombinant DNA can also include any type of protein tag sequence that may be useful for purifying the protein. Examples of protein tags include but are not limited to a histidine tag, a FLAG tag, a myc tag, an HA tag, or a GST tag. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts can be found in Cloning Vectors: A Laboratory Manual, (Elsevier, N.Y., 1985).

The expression construct is introduced into the host cell using a method appropriate to the host cell. A variety of methods for introducing nucleic acids into host cells are known in the art, including, but not limited to, electroporation; transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances;

microprojectile bombardment; lipofection; and infection (where the vector is an infectious agent). Suitable host cells include prokaryotes, yeast, mammalian cells, or bacterial cells.

Suitable bacteria include gram negative or gram positive organisms, for example, E. coli or Bacillus spp. Yeast, preferably from the Saccharomyces species, such as S. cerevisiae, may also be used for production of polypeptides. Various mammalian or insect cell culture systems can also be employed to express recombinant proteins. Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers, (Bio/Technology, 6:47, 1988). Examples of suitable mammalian host cell lines include endothelial cells, COS-7 monkey kidney cells, CV-1, L cells, C127, 3T3, Chinese hamster ovary (CHO), human embryonic kidney cells, HeLa, 293, 293T, and BHK cell lines. Purified polypeptides are prepared by culturing suitable host/vector systems to express the

recombinant proteins. In some embodiments, the small size of many of the polypeptides disclosed herein would make expression in E. coli as the preferred method for expression. The protein is then purified from culture media or cell extracts.

Proteins disclosed herein can also be produced using cell-translation systems. For such purposes the nucleic acids encoding the polypeptide must be modified to allow in vitro transcription to produce mRNA and to allow cell-free translation of the mRNA in the particular cell-free system being utilized (eukaryotic such as a mammalian or yeast cell-free translation system or prokaryotic such as a bacterial cell-free translation system.

PSMA-binding polypeptides can also be produced by chemical synthesis {e.g., by the methods described in Solid Phase Peptide Synthesis, 2nd ed., 1984, The Pierce Chemical Co., Rockford, 111.). Modifications to the protein can also be produced by chemical synthesis. The polypeptides of the present disclosure can be purified by isolation/purification methods for proteins generally known in the field of protein chemistry. Non-limiting examples include extraction, recrystallization, salting out (e.g., with ammonium sulfate or sodium sulfate), centrifugation, dialysis, ultrafiltration, adsorption chromatography, ion exchange chromatography, hydrophobic chromatography, normal phase chromatography, reversed-phase chromatography, gel filtration, gel permeation chromatography, affinity chromatography, electrophoresis, countercurrent distribution or any combinations of these. After purification, polypeptides may be exchanged into different buffers and/or concentrated by any of a variety of methods known to the art, including, but not limited to, filtration and dialysis. The purified polypeptide is preferably at least 85% pure, more preferably at least 95% pure, and most preferably at least 98% pure. Regardless of the exact numerical value of the purity, the polypeptide is sufficiently pure for use as a pharmaceutical product.

In certain embodiments, the present disclosure provides monoclonal antibodies that bind to PSMA. Monoclonal antibodies may be produced using any technique known in the art, e.g., by immortalizing spleen cells harvested from the transgenic animal after completion of the immunization schedule. The spleen cells can be immortalized using any technique known in the art, e.g., by fusing them with myeloma cells to produce hybridomas. Myeloma cells for use in hybridoma-producing fusion procedures preferably are non-antibody- producing, have high fusion efficiency, and enzyme deficiencies that render them incapable of growing in certain selective media which support the growth of only the desired fused cells (hybridomas). Examples of suitable cell lines for use in mouse fusions include Sp-20, P3- X63/Ag8, P3-X63-Ag8.653, NS l/l.Ag 4 1, Sp210-Agl4, FO, NSO/U, MPC-11, MPC11- X45-GTG 1.7 and S 194/5XX0 Bui; examples of cell lines used in rat fusions include

R210.RCY3, Y3-Ag 1.2.3, IR983F and 48210. Other cell lines useful for cell fusions are U- 266, GM1500-GRG2, LICR-LON-HMy2 and UC729-6.

Fragments or analogs of antibodies can be readily prepared by those of ordinary skill in the art following the teachings of this specification and using techniques known in the art. Preferred amino- and carboxy-termini of fragments or analogs occur near boundaries of functional domains. Structural and functional domains can be identified by comparison of the nucleotide and/or amino acid sequence data to public or proprietary sequence databases. Computerized comparison methods can be used to identify sequence motifs or predicted protein conformation domains that occur in other proteins of known structure and/or function. Methods to identify protein sequences that fold into a known three-dimensional structure are known. See, Bowie et al., 1991, Science 253: 164. Post-Translational Modifications of Polypeptides

In certain embodiments, the binding polypeptides of the invention may further comprise post-translational modifications. Exemplary post-translational protein modifications include phosphorylation, acetylation, methylation, ADP-ribosylation, ubiquitination, glycosylation, carbonylation, sumoylation, biotinylation or addition of a polypeptide side chain or of a hydrophobic group. As a result, the modified soluble polypeptides may contain non-amino acid elements, such as lipids, poly- or mono-saccharide, and phosphates. A preferred form of glycosylation is sialylation, which conjugates one or more sialic acid moieties to the polypeptide. Sialic acid moieties improve solubility and serum half-life while also reducing the possible immunogeneticity of the protein. See Raju et al. Biochemistry. 2001 31; 40(30):8868-76.

In one embodiment, modified forms of the subject soluble polypeptides comprise linking the subject soluble polypeptides to nonproteinaceous polymers. In one embodiment, the polymer is polyethylene glycol ("PEG"), polypropylene glycol, or polyoxyalkylenes, in the manner as set forth in U.S. Patents 4,640,835; 4,496,689; 4,301,144; 4,670,417;

4,791,192 or 4,179,337.

PEG is a water soluble polymer that is commercially available or can be prepared by ring-opening polymerization of ethylene glycol according to methods well known in the art (Sandler and Karo, Polymer Synthesis, Academic Press, New York, Vol. 3, pages 138-161). The term "PEG" is used broadly to encompass any polyethylene glycol molecule, without regard to size or to modification at an end of the PEG, and can be represented by the formula: X-0(CH 2 CH 2 0) n -CH 2 CH 2 OH (1), where n is 20 to 2300 and X is H or a terminal modification, e.g., a C 1-4 alkyl. In one embodiment, the PEG of the invention terminates on one end with hydroxy or methoxy, i.e., X is H or CH 3 ("methoxy PEG"). A PEG can contain further chemical groups which are necessary for binding reactions; which results from the chemical synthesis of the molecule; or which is a spacer for optimal distance of parts of the molecule. In addition, such a PEG can consist of one or more PEG side-chains which are linked together. PEGs with more than one PEG chain are called multi-armed or branched PEGs. Branched PEGs can be prepared, for example, by the addition of polyethylene oxide to various polyols, including glycerol, pentaerythriol, and sorbitol. For example, a four-armed branched PEG can be prepared from pentaerythriol and ethylene oxide. Branched PEG are described in, for example, EP-A 0 473 084 and U.S. Patent. 5,932,462. One form of PEGs includes two PEG side-chains (PEG2) linked via the primary amino groups of a lysine (Monfardini et al., Bioconjugate Chem. 6 (1995) 62-69). The serum clearance rate of PEG-modified polypeptide may be decreased by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or even 90%, relative to the clearance rate of the unmodified binding polypeptide. The PEG-modified polypeptide may have a half-life (ti /2 ) which is enhanced relative to the half-life of the unmodified protein. The half-life of PEG-binding polypeptide may be enhanced by at least 10%, 20%, 30%, 40%, 50%, 60%,

70%, 80%, 90%, 100%, 125%, 150%, 175%, 200%, 250%, 300%, 400% or 500%, or even by 1000% relative to the half-life of the unmodified binding polypeptide. In some embodiments, the protein half-life is determined in vitro, such as in a buffered saline solution or in serum. In other embodiments, the protein half-life is an in vivo half life, such as the half-life of the protein in the serum or other bodily fluid of an animal.

Therapeutic Methods, Formulations and Modes of Administration

The present disclosure provides a method for treating cancer, comprising

administering an anti-PSMA antibody, or antibody fragment, as described herein. The present disclosure further provides a method for treating a prostate cancer, comprising administering an anti-PSMA antibody, or an antibody fragment as described herein.

Studies have demonstrated PSMA expression in many types of prostate tissue and increased PSMA expression in cancer tissue (Chang, S. Rev Urol. 2004; 6(Suppl 10): S 13— S 18; Silver et al. Clin Cancer Res. 1997;3:81-85; Troyer et al. Int J Cancer. 1995;62:552- 558; Haffner et al. Hum Pathol. 2009 Dec;40(12): 1754-61). Accordingly, in one

embodiment, the anti-PSMA antibodies and antibody fragments of the invention are used to treat cancer associated with increased PSMA expression.

Any of the antibodies disclosed herein may be used in such methods. For example, the methods may be performed using a fully human antibody of an IgG class that binds to a PSMA epitope with a binding affinity of at least 10 "6 M, a Fab fully human antibody fragment, having a variable domain region from a heavy chain and a variable domain region from a light chain, a single chain human antibody, having a variable domain region from a heavy chain and a variable domain region from a light chain and a peptide linker connection the heavy chain and light chain variable domain regions, including the heavy and light chain variable regions (and CDRs within said sequences) described in SEQ ID Nos. 1-17 (Table 3).

In one embodiment, the antibodies disclosed herein are used to target a chemical or cellular therapeutic payload, wherein the anti-PSMA antibody comprises a heavy chain variable domain sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to the amino acid sequence SEQ ID NO. 1, and comprising a light chain variable domain sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to an amino acid sequences selected from the group consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, and SEQ ID NO. 17.

In one embodiment, the methods described herein include the use of a fully human Fab antibody fragment comprising a heavy chain variable domain sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to the amino acid sequence SEQ ID NO. 1, and comprising a light chain variable domain sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to an amino acid sequence selected from the group consisting SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO.

11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, and SEQ ID NO. 17.

In one embodiment, the methods described herein include the use of a single chain human antibody comprising a heavy chain variable domain sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to the amino acid sequence SEQ ID NO. 1, and comprising a light chain variable domain sequence that is at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical, to an amino acid sequence selected from the group consisting of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 8, SEQ ID NO. 8, SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, SEQ ID NO. 12, SEQ ID NO. 13, SEQ ID NO. 14, SEQ ID NO. 15, SEQ ID NO. 16, and SEQ ID NO. 17.

In one embodiment, the fully human antibody has both a heavy chain and a light chain wherein the antibody comprises a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2, SEQ ID NO. 1/SEQ ID NO. 3, SEQ ID NO. 1/SEQ ID NO. 4, SEQ ID NO. 1/SEQ ID NO. 5, SEQ ID NO. 1/SEQ ID NO. 6, SEQ ID NO. 1/SEQ ID NO. 7, SEQ ID NO. 1/SEQ ID NO. 8, SEQ ID NO. 1/SEQ ID NO. 9, SEQ ID NO. 1/SEQ ID NO. 10, SEQ ID NO. 1/SEQ ID NO. 11, SEQ ID NO. 1/SEQ ID NO.

12, SEQ ID NO. 1/SEQ ID NO. 13, SEQ ID NO. 1/SEQ ID NO. 14, SEQ ID NO. 1/SEQ ID NO. 15, SEQ ID NO. 1/SEQ ID NO. 16, and SEQ ID NO. 1/SEQ ID NO. 17. In one embodiment, the fully human antibody Fab fragment comprises both a heavy chain variable domain region and a light chain variable domain region wherein the antibody comprises a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2, SEQ ID NO. 1/SEQ ID NO. 3, SEQ ID NO. 1/SEQ ID NO. 4, SEQ ID NO. 1/SEQ ID NO. 5, SEQ ID NO. 1/SEQ ID NO. 6, SEQ ID NO. 1/SEQ ID NO. 7, SEQ ID NO. 1/SEQ ID NO. 8, SEQ ID NO. 1/SEQ ID NO. 9, SEQ ID NO. 1/SEQ ID NO. 10, SEQ ID NO. 1/SEQ ID NO. 11, SEQ ID NO. 1/SEQ ID NO. 12, SEQ ID NO. 1/SEQ ID NO. 13, SEQ ID NO. 1/SEQ ID NO. 14, SEQ ID NO. 1/SEQ ID NO. 15, SEQ ID NO. 1/SEQ ID NO. 16, and SEQ ID NO. 1/SEQ ID NO. 17.

In one embodiment, the fully human single chain antibody comprises both a heavy chain variable domain region and a light chain variable domain region, wherein the single chain fully human antibody comprises a heavy chain/light chain variable domain sequence selected from the group consisting of SEQ ID NO. 1/SEQ ID NO. 2, SEQ ID NO. 1/SEQ ID NO. 3, SEQ ID NO. 1/SEQ ID NO. 4, SEQ ID NO. 1/SEQ ID NO. 5, SEQ ID NO. 1/SEQ ID NO. 6, SEQ ID NO. 1/SEQ ID NO. 7, SEQ ID NO. 1/SEQ ID NO. 8, SEQ ID NO. 1/SEQ ID NO. 9, SEQ ID NO. 1/SEQ ID NO. 10, SEQ ID NO. 1/SEQ ID NO. 11, SEQ ID NO. 1/SEQ ID NO. 12, SEQ ID NO. 1/SEQ ID NO. 13, SEQ ID NO. 1/SEQ ID NO. 14, SEQ ID NO. 1/SEQ ID NO. 15, SEQ ID NO. 1/SEQ ID NO. 16, and SEQ ID NO. 1/SEQ ID NO. 17.

In one embodiment, cancer which can be treated using the antibodies and fragments disclosed herein include, but are not limited to, ovarian cancer, colon cancer, breast cancer, lung cancer, myelomas, neuroblastic-derived CNS tumors, monocytic leukemias, B-cell derived leukemias, T-cell derived leukemias, B-cell derived lymphomas, T-cell derived lymphomas, and mast cell derived tumors.

In one embodiment, the anti-PSMA antibodies and antibody fragments of the invention are used to treat prostate cancer.

The PSMA antibodies, and antibody fragments, described herein are useful in treating, delaying the progression of, preventing relapse of or alleviating a. symptom of a. cancer or other neoplastic condition, including, hematological malignancies and/or PSMA+ tumors. The PSMA antibodies described herein are useful in treating a cancer selected from the group consisting of non-Hodgkin's lymphoma (NHL), acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), multiple myeloma (MM), breast cancer, ovarian cancer, head and neck cancer, bladder cancer, melanoma, colorectal cancer, pancreatic cancer, lung cancer, leiomyoma, leiomyosarcoma, glioma, glioblastoma, and solid tumors, wherein solid tumors are selected from the group consisting of breast tumors, ovarian tumors, lung tumors, pancreatic tumors, prostate tumors, melanoma tumors, colorectal tumors, lung tumors, head and neck tumors, bladder tumors, esophageal tumors, liver tumors, and kidney tumors.

As used herein, "hematological cancer" refers to a cancer of the blood, and includes leukemia, lymphoma and myeloma among others. "Leukemia" refers to a cancer of the blood in which too many white blood cells that are ineffective in fighting infection are made, thus crowding out the other parts that make up the blood, such as platelets and red blood cells, it is understood that cases of leukemia are classified as acute or chronic. As such, antibodies and fragments of the invention may be used to treat a subject having a hematological cancer.

Certain forms of leukemia include, acute lymphocytic leukemia (ALL); acute myeloid leukemia (AMI,); chronic lymphocytic leukemia. (CLL); chronic myelogenous leukemia. (CML); Myeloproliferative disorder/neoplasm (MPDS); and myelodysplasia syndrome. 'Lymphoma." may refer to a. Hodgkin * s lymphoma, both indolent and aggressive non- Hodgkin's lymphoma, Burkitt's lymphoma, and follicular lymphoma (small cell and large cell), among others. Myeloma may refer to multiple myeloma (MM), giant cell myeloma, heavy-chain myeloma, and light chain or Bence- Jones myeloma.

The present disclosure features methods for treating or preventing the S. aureus infection comprising administering an anti-PSMA polypeptide. Techniques and dosages for administration vary depending on the type of specific polypeptide and the specific condition being treated but can be readily determined by the skilled artisan. In general, regulatory agencies require that a protein reagent to be used as a therapeutic is formulated so as to have acceptably low levels of pyrogens. Accordingly, therapeutic formulations will generally be distinguished from other formulations in that they are substantially pyrogen free, or at least contain no more than acceptable levels of pyrogen as determined by the appropriate regulatory agency (e.g., FDA).

Therapeutic compositions of the present disclosure may be administered with a pharmaceutically acceptable diluent, carrier, or excipient, in unit dosage form. Administration may be parenteral (e.g., intravenous, subcutaneous), oral, or topical, as non-limiting examples. In addition, any gene therapy technique, using nucleic acids encoding the polypeptides of the invention, may be employed, such as naked DNA delivery, recombinant genes and vectors, cell-based delivery, including ex vivo manipulation of patients' cells, and the like.

The composition can be in the form of a pill, tablet, capsule, liquid, or sustained release tablet for oral administration; or a liquid for intravenous, subcutaneous or parenteral administration; gel, lotion, ointment, cream, or a polymer or other sustained release vehicle for local administration.

In certain embodiments, the disclosed antibodies are administered by inhalation, but aerosolization of full IgG antibodies may prove limiting due to their molecular size

(~150kDa).

Methods well known in the art for making formulations are found, for example, in "Remington: The Science and Practice of Pharmacy" (20th ed., ed. A. R. Gennaro A R., 2000, Lippincott Williams & Wilkins, Philadelphia, Pa.). Formulations for parenteral administration may, for example, contain excipients, sterile water, saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated napthalenes. Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or

polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds. Nanoparticulate formulations (e.g., biodegradable nanoparticles, solid lipid nanoparticles, liposomes) may be used to control the biodistribution of the compounds. Other potentially useful parenteral delivery systems include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. The concentration of the compound in the formulation varies depending upon a number of factors, including the dosage of the drug to be administered, and the route of administration.

The polypeptide may be optionally administered as a pharmaceutically acceptable salt, such as non-toxic acid addition salts or metal complexes that are commonly used in the pharmaceutical industry. Examples of acid addition salts include organic acids such as acetic, lactic, pamoic, maleic, citric, malic, ascorbic, succinic, benzoic, palmitic, suberic, salicylic, tartaric, methanesulfonic, toluenesulfonic, or trifluoroacetic acids or the like; polymeric acids such as tannic acid, carboxymethyl cellulose, or the like; and inorganic acid such as hydrochloric acid, hydrobromic acid, sulfuric acid phosphoric acid, or the like. Metal complexes include zinc, iron, and the like. In one example, the polypeptide is formulated in the presence of sodium acetate to increase thermal stability.

Formulations for oral use include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, inert diluents or fillers (e.g., sucrose and sorbitol), lubricating agents, glidants, and anti-adhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc). Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium.

A therapeutically effective dose refers to a dose that produces the therapeutic effects for which it is administered. The exact dose will depend on the disorder to be treated, and may be ascertained by one skilled in the art using known techniques. In general, the polypeptide is administered at about 0.01 μg/kg to about 50 mg/kg per day, preferably 0.01 mg/kg to about 30 mg/kg per day, most preferably 0.1 mg/kg to about 20 mg/kg per day. The polypeptide may be given daily (e.g., once, twice, three times, or four times daily) or preferably less frequently (e.g., weekly, every two weeks, every three weeks, monthly, or quarterly). In addition, as is known in the art, adjustments for age as well as the body weight, general health, sex, diet, time of administration, drug interaction, and the severity of the disease may be necessary, and will be ascertainable with routine experimentation by those skilled in the art.

A PSMA binding polypeptide, as disclosed herein, can be administered alone or in combination with one or more additional therapies such as chemotherapy radiotherapy, immunotherapy, surgical intervention, or any combination of these. Long-term therapy is equally possible as is adjuvant therapy in the context of other treatment strategies, as described above.

In certain embodiments of such methods, one or more polypeptide therapeutic agents can be administered, together (simultaneously) or at different times (sequentially). In addition, polypeptide therapeutic agents can be administered with another type of compounds for treating cancer or for inhibiting angiogenesis.

In certain embodiments, the subject anti-PSMA antibodies agents of the invention can be used alone.

In certain embodiments, the binding polypeptides of fragments thereof can be labeled or unlabeled for diagnostic purposes. Typically, diagnostic assays entail detecting the formation of a complex resulting from the binding of a binding polypeptide to PSMA. The binding polypeptides or fragments can be directly labeled, similar to antibodies. A variety of labels can be employed, including, but not limited to, radionuclides, fluorescers, enzymes, enzyme substrates, enzyme cofactors, enzyme inhibitors and ligands (e.g., biotin, haptens). Numerous appropriate immunoassays are known to the skilled artisan (see, for example, U.S. Patents. 3,817,827; 3,850,752; 3,901,654; and 4,098,876). When unlabeled, the binding polypeptides can be used in assays, such as agglutination assays. Unlabeled binding polypeptides can also be used in combination with another (one or more) suitable reagent which can be used to detect the binding polypeptide, such as a labeled antibody reactive with the binding polypeptide or other suitable reagent (e.g., labeled protein A).

In one embodiment, the binding polypeptides of the present invention can be utilized in enzyme immunoassays, wherein the subject polypeptides are conjugated to an enzyme. When a biological sample comprising a PSMA protein is combined with the subject binding polypeptides, binding occurs between the binding polypeptides and the PSMA protein. In one embodiment, a sample containing cells expressing a PSMA protein (e.g., endothelial cells) is combined with the subject antibodies, and binding occurs between the binding polypeptides and cells bearing a PSMA protein recognized by the binding polypeptide. These bound cells can be separated from unbound reagents and the presence of the binding polypeptide-enzyme conjugate specifically bound to the cells can be determined, for example, by contacting the sample with a substrate of the enzyme which produces a color or other detectable change when acted on by the enzyme. In another embodiment, the subject binding polypeptides can be unlabeled, and a second, labeled polypeptide (e.g., an antibody) can be added which recognizes the subject binding polypeptide.

In certain aspects, kits for use in detecting the presence of a PSMA protein in a biological sample can also be prepared. Such kits will include a PSMA binding polypeptide which binds to a PSMA protein or portion of said receptor, as well as one or more ancillary reagents suitable for detecting the presence of a complex between the binding polypeptide and the receptor protein or portions thereof. The polypeptide compositions of the present invention can be provided in lyophilized form, either alone or in combination with additional antibodies specific for other epitopes. The binding polypeptides and/or antibodies, which can be labeled or unlabeled, can be included in the kits with adjunct ingredients (e.g., buffers, such as Tris, phosphate and carbonate, stabilizers, excipients, biocides and/or inert proteins, e.g., bovine serum albumin). For example, the binding polypeptides and/or antibodies can be provided as a lyophilized mixture with the adjunct ingredients, or the adjunct ingredients can be separately provided for combination by the user. Generally these adjunct materials will be present in less than about 5% weight based on the amount of active binding polypeptide or antibody, and usually will be present in a total amount of at least about 0.001% weight based on polypeptide or antibody concentration. Where a second antibody capable of binding to the binding polypeptide is employed, such antibody can be provided in the kit, for instance in a separate vial or container. The second antibody, if present, is typically labeled, and can be formulated in an analogous manner with the antibody formulations described above.

Polypeptide sequences are indicated using standard one- or three-letter abbreviations. Unless otherwise indicated, each polypeptide sequence has amino termini at the left and a carboxy termini at the right; each single- stranded nucleic acid sequence, and the top strand of each double-stranded nucleic acid sequence, has a 5' termini at the left and a 3' termini at the right. A particular polypeptide sequence also can be described by explaining how it differs from a reference sequence.

Having now described the present invention in detail, the same will be more clearly understood by reference to the following examples, which are included for purposes of illustration only and are not intended to be limiting of the invention.

Example 1

Human anti-PSMA antibodies were identified, and the amino acid sequences of the light chain and heavy chain variable regions are described in Table 3 below.

This example illustrates the binding of anti-PSMA antibodies to endogenous human

PSMA expressed on LNCaP human prostate cancer cells, as assayed by flow cytometry. EC 50 values for antibodies were determined as follows.

PSMA expressing LNCaP cells were harvested with enzyme-free Cell Dissociation Buffer (GIBCO) and transferred to V-Bottom 96 well-plates (50,000 cells/ well). Cells were incubated on ice for 45 min with serial dilutions of anti-PSMA antibodies PSGB 11, PSGB 12, PSGC12, PSGD4, PSGD6 and PSA11 in FACS buffer (PBS+ 2% FBS) + NaN 3 . The control antibody (cig) was a negative control, and was an isotype matched, nonspecific (i.e., does not bind to PSMA and also did not bind the cells) antibody. After 1 wash in FACS buffer, a 1: 1000 dilution of Phycoerythrin conjugated anti-Human IgG (γ-chain specific) was added and incubated for 30 min. Following a final wash, fluorescence intensity was measured on an Intellicyt High Throughput Flow Cytometer (HTFC).

Data were analyzed using Graphpad Prism software and non-linear regression fit. Data points are shown as the median fluorescence intensity (MFI) of positively labeled cells +/- Standard Error. EC 50 values are reported as the concentration of antibody to achieve 50% of maximal PSMA antibodies binding to PSMA expressing cells.

In parallel, cell binding was also assessed with the same method on PC3, a prostate cancer cell line that does not express human PSMA endogenously, and thus served as a negative control. The results from the binding assays for both cancer, PSMA-specific LNCaP cells and negative control PC3 cells are provided in Figure 1 (A-D). As shown in Figure 1A and B, anti-PSMA antibodies PSGB l l, PSGB 12, PSGC12, PSGD4, PSGD6 and PSA11 strongly bound PSMA-expressing LNCaP cells relative to the negative IgG control (see Figure 1A). Furthermore, as described in Figures (C) and (D), anti-PSMA antibodies PSGB 11, PSGB 12, PSGC12, PSGD4, PSGD6 and PSA11 showed little to no binding to cells not expressing human PSMA (PC3 cells), as the cells showed similar results to the negative control antibody (see Figure 1(C)). Thus, the results in Figure l(A-D) show that the antibodies were specific to PSMA expressing cells, including PSMA expressing cancer cells.

In addition, the anti-PSMA antibodies identified herein displayed EC 50 values in the nanomolar range for LNCaP PSMA expressing cells, as described below in Table 1. These data demonstrate strong and specific binding of the antibodies described in Table 3 to endogenous PSMA.

Table 1:

Example 2

This example illustrates in vitro data showing the assessment of anti-PSMA antibodies in a cytotoxicity assay using secondary antibody-drug conjugate technique ("Secondary Antibody-Drug Conjugates as Tools for ADC Discovery". Helen Mao, Poster, IBC 24 th Annual, 2013). This example demonstrates the potential of anti-PSMA antibodies to be used as antibody drug conjugates.

PSMA-expressing prostate cancer cells (LNCaP, ATCC CRL-1740™) were harvested with enzyme-free Cell Dissociation Buffer (GIBCO), seeded into white 96-Well Clear Bottom plates (2,000 cells/ well in 90 μΐ) and allowed to adhere overnight at 37 °C. Anti- PSMA antibodies PSGF9, PSGC9, PSGD6, PSGD3, PSGE10, PSGH3, PSGB 11, PSGD4, PSGB 12 and PSA11 were used in the experiment. Antibodies were pre-complexed with Protein G(PG)-MMAF (Monomethyl auristatin F, Concortis Biosystems) in cell culture media, at a 1:4 molar ratio. The control antibody (clg) was an isotype matched, nonspecific (i.e., does not bind to PSMA) antibody. PGMMAF alone was used as a negative control. After 10 min at room temperature, serial dilutions of the antibody-ProteinG-MMAF complex were prepared in cell culture media, incubated 10 more minutes at room temperature, and added to cells (10 μΐ/ well) in triplicate. In some experiments, plates were incubated at 37 °C for 4 days ("No wash method"). In other experiments ("2-hours-wash method"), plates were incubated at 37 °C for two hours, media was aspirated. Then cells were washed once with full media, and left in fresh full media before being incubated at 37 °C for 4 days. For all experiments, cell proliferation was then analyzed as follows: 100 μΐ of Cell Titer Glo buffer (Promega) was added to each well. Plates were incubated with shaking at room temperature for 20 min. Luminescence signal was then measured on a Flexstation 3 plate reader

(Molecular Device). Data were reported as relative Luminescent Units. Dose-response curves were generated in GraphPad prism, and IC 50 values were calculated using non-linear regression fit (Log (inhibitor) vs. response - Variable slope equation).

The results are shown in Figure 2(A-C) and below in Table 2. The results described in Figure 2A-C show that anti-PSMA ADCs comprising antibodies PSGF9, PSGC9, PSGD6, (Figure 2A); PSGD3, PSGE10, PSGH3, PSGB 11, (Figure 2B); PSGD4, PSGB 12 and PSA11 (Figure 2C), each conjugated to MMAF can induce cell killing in LNCaP cancer cells. In contrast, MMAF alone and the IgG control (see Figure 2A) had little to no effect on cell death and had high rates of cell survival. The same methods were used with PSMA-negative prostate cancer cells (PC-3, ATCC CRL-1435) to assess the non- specificity of cell killing of anti-PSMA antibodies (PSGF9, PSGC9, PSGD6, PSGD3, PSGE10, PSGH3, PSGB 11, PSGD4, PSGB 12 and PSAl iyProtein G-MMAF complexes. The results, shown in Figure 2D, indicate that anti- PSMA antibodies do not induce cell killing when complexed with a cytotoxin such as MMAF in PSMA negative cancer cells. Thus, Figures 1 and 2 show that little to no non-specific cell killing is observed on cells not overexpressing PSMA, suggesting a good selectivity index for future PSMA antibody drug conjugate (ADC). Altogether, this illustrates the potential of PSMA antibodies as antibody-drug conjugates.

IC 50 values for the anti-PSMA-MMAF ADCs were determined and are described below in Table 2. To verify that the cell killing observed for these types of secondary conjugates was due to the specifically internalized conjugates tested, unbound conjugates were washed out (2 -hours-wash method in Table 2). As described in Table 2, the IC 50 values were in the nanomolar range.

Table 2:

Example 3

In addition to the ability of the anti-PSMA antibodies described herein to bind to PSMA and also to induce cell death when conjugated to a toxin, the affinity values for antibodies PSA11 and PSGB 11 were determined on Octet. As described in Figure 3, anti- PSMA antibody PSA11 had an K D of about 6.7 x 10 M, and antibody PSA11 (described in Figure 4) had a K D of about 6.8 x 10 "10 M.

Specifically, an amine Reactive Second- Generation (AR2G) sensor was coated with antibody (15 ug/ml in acetic buffer, pH5.0) using Sulfo-N-hydroxysuccinimide/N-Ethyl-N'- (3-dimethylaminopropyl)carbodiimide hydrochloride (Sulfo-NHS/EDC) coupling methodology. The sensors were quenched with 1M pH 8.5 ethanolamine. Data was collected by moving sensors to serially diluted recombinant human PSMA/His (in PBS buffer) wells, then the data was dissociated by transferring the sensors to PBS wells. A 1: 1 binding model was used to fit the data. Results from the assay are provided in Figures 3 and 4.

Table 3. Amino Acid Heavy and Light Chain Variable Domains

Heavy chain variable domain Light chain variable domain

S LRAEDT A V Y YC AR VWD Y Y Y FTFGPGTKVDIK SEQ ID NO. 6 DSSGDAFDIWGQGTMVTVSS SEQ ID NO. 1

QVQLVQSGGGLVQPGGSLRLS C AAS GFTFS S YWMS WVRQAPG KGLEWVANIKQDGSEKYYVDS D MTQSPSSVSASVGDRVTITCR VKGRFTIS RDN AKNS LYLQMN AS QD VGTWLA W YQQKPGR APKL S LRAEDT A VYYCAR VWD YYY LIY V AS S LQS G VPS RFS GS GS GTD DSSGDAFDIWGQGTMVTVSS FTLTISSLQPEDSATYYCQQAKGIP

PSGC12 SEQ ID NO. 1 YTFGQGTKLEIK SEQ ID NO. 7

QVQLVQSGGGLVQPGGSLRLS C AAS GFTFS S YWMS WVRQAPG KGLEWVANIKQDGSEKYYVDS D MTQSPSSVSASVGDRVTITCR VKGRFTIS RDN AKNS LYLQMN AS QDINNWLA W YQQKAGKAPKL S LRAEDT A VYYCAR VWD YYY LIY V ATKLQNG VPS RFS GS GS GTD DSSGDAFDIWGQGTMVTVSS FTLS IS NLQPEDF AT Y YCQQ AKS F

PSGD3 SEQ ID NO. 1 P YTFGQGTKLEIK SEQ ID NO. 8

QVQLVQSGGGLVQPGGSLRLS C AAS GFTFS S YWMS WVRQAPG KGLEWVANIKQDGSEKYYVDS D MTQSPSSLSASVGDRVSITCR VKGRFTIS RDN AKNS LYLQMN ASQGISTWLAWYQQKPGKAPDLL S LRAEDT A VYYCAR VWD YYY IY A AS NLQS G VPS RFS GS GS GTDF DSSGDAFDIWGQGTMVTVSS TLTIS S LQPEDFAT Y YC QQ ANS FP

PSGD4 SEQ ID NO. 1 LTFGGGTKVEIK SEQ ID NO. 9

QVQLVQSGGGLVQPGGSLRLS C AAS GFTFS S YWMS WVRQAPG KGLEWVANIKQDGSEKYYVDS D MTQSPSSVSASVGDRVTITCR VKGRFTIS RDN AKNS LYLQMN ASQAISSWLAWYQQKPGKAPKLL S LRAEDT A VYYCAR VWD YYY IY A AS S LQS G VPS RFS GS GS GTDF DSSGDAFDIWGQGTMVTVSS TLTIS S LQPEDFAT YYC QQ A YS FP

PSGD6 SEQ ID NO. 1 VTFGPGTKVDIK SEQ ID NO. 10

QVQLVQSGGGLVQPGGSLRLS C AAS GFTFS S YWMS WVRQAPG KGLEWVANIKQDGSEKYYVDS DVVMTQSPSSVSASVGDRVTITC VKGRFTIS RDN AKNS LYLQMN RASQGISSWLAWYQQKPGKAPKL S LRAEDT A VYYCAR VWD YYY LIY A AS TLQS G VPS RFS GS GS GTD DSSGDAFDIWGQGTMVTVSS FTLTINNLQPEDFATYYCQQTASF

PSGE10 SEQ ID NO. 1 PINFGGGTKVEIK SEQ ID NO. 11

QVQLVQSGGGLVQPGGSLRLS C AAS GFTFS S YWMS WVRQAPG KGLEWVANIKQDGSEKYYVDS DrVMTQSPSTLSASVGDRVTITCR VKGRFTIS RDN AKNS LYLQMN AS QS IS NWLA W YQQKPGKPPKLL S LRAEDT A VYYCAR VWD YYY IY A AS S LQS G VPS RFS GS GS GTDF DSSGDAFDIWGQGTMVTVSS TLTIS S LQPEDFAT YYC QQS YRS L

PSGE11 SEQ ID NO. 1 TFAGGTKVEIK SEQ ID NO. 12 Heavy chain variable domain Light chain variable domain

QVQLVQSGGGLVQPGGSLRLS C AAS GFTFS S YWMS WVRQAPG KGLEWVANIKQDGSEKYYVDS AIQMTQSPSSVSASVGDRVTITCR VKGRFTIS RDN AKNS LYLQMN ASQGISSWLAWYQQKPGKAPKLL S LRAEDT A V Y YC AR VWD Y Y Y IY A AS S LQS G VPS RFS GS GS GTDF DSSGDAFDIWGQGTMVTVSS TLTIS S LQPEDIAT Y YC QES YS TPF

PSGF9 SEQ ID NO. 1 TFGPGTKVDIK SEQ ID NO. 13

QVQLVQSGGGLVQPGGSLRLS C AAS GFTFS S YWMS WVRQAPG KGLEWVANIKQDGSEKYYVDS DIQMTQSPSYVSASVGDRVTITCR VKGRFTIS RDN AKNS LYLQMN AS QG VS HWLA W YQQKPGKAPKL S LRAEDT A VYYCAR VWD YYY LIY A AS RLQS G VPS RFS GS GS GTD DSSGDAFDIWGQGTMVTVSS FTLTIS S LQPEDF AT Y YC QQ A YS F

PSGF11 SEQ ID NO. 1 PLTFGQGTKLEIK SEQ ID NO. 14

QVQLVQSGGGLVQPGGSLRLS C AAS GFTFS S YWMS WVRQAPG KGLEWVANIKQDGSEKYYVDS EIVLTQS PS S LS AS VGDRVTITCR A VKGRFTIS RDN AKNS LYLQMN S QGIS S YLA W YQQKPGKAPKLLI S LRAEDT A VYYCAR VWD YYY Y A AS TLQS G VPS RFS GS GS GTDFT DSSGDAFDIWGQGTMVTVSS LTIS S LQPEDF AT YYC QQLNS YPR

PSGG6 SEQ ID NO. 1 GITFGQGTKLEIK SEQ ID NO. 15

QVQLVQSGGGLVQPGGSLRLS C AAS GFTFS S YWMS WVRQAPG KGLEWVANIKQDGSEKYYVDS D MTQSPSSVSASVGDRVTITCR VKGRFTIS RDN AKNS LYLQMN ASQGISNWLAWYQQKPGKAPKL S LRAEDT A VYYCAR VWD YYY LIY V AS S LQS G VPS RFS GS GS GTD DSSGDAFDIWGQGTMVTVSS FTLTIS S LQPEDF AT YYC QQ ANS F

PSGH3 SEQ ID NO. 1 PITFGQGTRLEIK SEQ ID NO. 16

QVQLVQSGGGLVQPGGSLRLS C AAS GFTFS S YWMS WVRQAPG KGLEWVANIKQDGSEKYYVDS DrVMTQSPSSLSASVGDRVTITCR VKGRFTIS RDN AKNS LYLQMN ASQGISSWLAWYQQKPGKAPKLL S LRAEDT A VYYCAR VWD YYY IY A AS S LQS G VPS RFS GS GS GTDF DSSGDAFDIWGQGTMVTVSS TLTINSLQPEDFATYYCQQASGFP

PSGH8 SEQ ID NO. 1 FTFGPGTKVDIK SEQ ID NO. 17

Incorporation by Reference

The contents of all references, patents, pending patent applications and published patents, cited throughout this application are hereby expressly incorporated by reference.