Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTIBODIES TO TIP-1 AND GRP78
Document Type and Number:
WIPO Patent Application WO/2015/116653
Kind Code:
A1
Abstract:
The present invention is directed towards isolated antibodies that bind to GRP78 and TIP-1.

Inventors:
HALLAHAN DENNIS E (US)
YAN HEPING (US)
Application Number:
PCT/US2015/013241
Publication Date:
August 06, 2015
Filing Date:
January 28, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV WASHINGTON (US)
UNIV VANDERBILT (US)
International Classes:
C07K16/28; A61K39/395; A61P35/00; G01N33/534; G01N33/574; G01N33/577
Domestic Patent References:
WO2013019730A12013-02-07
Other References:
DATABASE GENBANK 17 May 2011 (2011-05-17), XP055218017, accession no. EC23014.1
DATABASE GENBANK 22 June 1999 (1999-06-22), XP055218019, accession no. AD40244.1
GREGORY.A WEISS ET AL.: "Rapid mapping of protein functional epitopes by combinatorial alanine scanning", PNAS, vol. 97, 2000, pages 8950 - 8954, XP002161102
Attorney, Agent or Firm:
RILEY-VARGAS, Rebecca et al. (Mark Twain Plaza III105 West Vandalia, Suite 40, Edwardsville IL, US)
Download PDF:
Claims:
IN THE CLAIMS

What is claimed is:

1 . An isolated antibody that binds to TIP-1 , wherein the antibody comprises a heavy chain variable domain comprising a CDR1 , CDR2, and a CDR3, wherein the CDR3 comprises at least five contiguous amino acids of SEQ ID NO:30.

2. The antibody of claim 1 wherein the heavy chain variable domain CDR2 comprises at least five contiguous amino acids of SEQ ID NO:29.

3. The antibody of claim 1 , wherein the heavy chain variable domain CDR1 comprises at least five contiguous amino acids of SEQ ID NO:28.

4. The antibody of claim 1 , wherein the heavy chain variable domain CDR1 comprises at least five contiguous amino acids of SEQ ID NO:28 and the heavy chain variable domain CDR2 comprises at least five contiguous amino acids of SEQ ID NO:29.

5. The antibody of claim 1 , wherein the heavy chain variable domain CDR1 comprises SEQ ID NO:28, the heavy chain variable region domain CDR2 comprises SEQ ID NO:29, and the heavy chain variable region domain CDR3 comprises SEQ ID NO:30.

6. The antibody of claim 1 , wherein the antibody further comprises a light chain

variable domain comprising a CDR1 , CDR2, and a CDR3, wherein the CDR3 comprises at least five contiguous amino acids of SEQ ID NO:33.

7. The antibody of claim 6, wherein the light chain variable domain CDR2 comprises at least five contiguous amino acids of SEQ ID NO:32.

8. The antibody of claim 6, wherein the light chain variable domain CDR1 comprises at least five contiguous amino acids of SEQ ID NO:31 .

9. The antibody of claim 6, wherein the light chain variable domain CDR1 comprises at least five contiguous amino acids of SEQ ID NO:31 and the light chain variable domain CDR2 comprises at least five contiguous amino acids of SEQ ID NO:32.

10. The antibody of claim 6, wherein the light chain variable domain CDR1 comprises SEQ ID NO:31 , the light chain variable region domain CDR2 comprises SEQ ID NO:32, and the light chain variable region domain CDR3 comprises SEQ ID NO:33.

1 1 .The antibody of claim 1 , wherein the antibody recognizes an epitope within an amino acid sequence selected from the group consisting of SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:1 1 , SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, or SEQ ID NO:21 .

12. An antibody of any of the preceding claims, wherein the antibody is selected from the group consisting of a humanized antibody, a single chain variable fragment (scFv) antibody, an antibody fragment, or a chimeric antibody.

13. An antibody of any of the preceding claims, wherein the antibody is conjugated

directly or indirectly to a payload selected from the group consisting of a therapeutic agent, an imaging agent, or a combination thereof.

14. A method of enhancing radiotherapy in a subject, the method comprising

administering a pharmacologically effective amount of an antibody of claim 13 to the subject, such that radiotherapy is enhanced.

15. The method of claim 14, further comprising administering ionizing radiation to the subject.

16. The method of claim 14, further comprising imaging the subject.

17. A method of detecting a tumor in a subject, the method comprising:

(a) exposing a target area of the subject where the presence of tumor is suspected to ionizing radiation;

(b) administering to the subject a composition comprising an antibody of claim 13, wherein the antibody is conjugated to an imaging agent; and

(c) detecting the imaging agent, wherein the presence of the imaging agent indicates the presence of a tumor in the target area of the subject.

18. The method of claim 17, wherein the exposing comprises exposing the tumor to less than about 2 Gy ionizing radiation.

19. The method of claim 18, wherein the exposing comprise exposing the tumor to at least about 2 Gy radiation.

20. The method of claim 19, wherein the exposing comprises exposing the tumor to about 10 Gy to about 20 Gy ionizing radiation.

21 . The method of claim 17, wherein the administering comprises administering the antibody subsequent to radiation exposure.

22. The method of claim 21 , wherein the administering comprises administering the antibody 0 hour to about 24 hours following radiation exposure.

23. The method of claim 22, wherein the administering comprises administering the antibody about 4 hours to about 24 hours following radiation exposure.

Description:
ANTIBODIES TO TIP-1 AND GRP78

GOVERNMENTAL RIGHTS

[0001 ] This invention was made with government support under grants R01 -CA125757, R21 -CA128456-01 , R01 -CA1 12385-01 , and R01 -CA88076, awarded by the National Institutes of Health (NIH). The government has certain rights in the invention.

FIELD OF INVENTION

[0002] The invention encompasses antibodies useful in the recognition of tumor cells and tumor specific delivery of drugs and therapies.

BACKGROUND OF THE INVENTION

[0003] In the United States, the probability that an individual, over the course of a lifetime, will develop or die from cancer is 1 in 2 for men and 1 in 3 for women. Tumor-specific drug delivery and therapy methods have the potential to reduce or prevent tumor growth in organisms allowing them to lead longer, healthier lives. Many anti-tumor drugs, however, are also toxic to non-tumor cells, resulting in hard to tolerate side-effects. Hence, there is a need in the art for a way to deliver anti-tumor agents specifically to tumor cells to reduce tumor cell growth.

SUMMARY OF THE INVENTION

[0004] One aspect of the present invention encompasses an isolated antibody that binds to GRP78. The antibody comprises a heavy chain variable domain comprising a CDR1 , CDR2, and a CDR3, wherein the CDR3 comprises at least five contiguous amino acids of SEQ ID NO:17.

[0005] Another aspect of the present invention encompasses an isolated antibody that binds to TIP-1 . The antibody comprises a heavy chain variable domain comprising a CDR1 , CDR2, and a CDR3, wherein the CDR3 comprises at least five contiguous amino acids of SEQ ID NO:30. [0006] Yet another aspect of the present invention encompasses a method of enhancing radiotherapy in a subject. The method comprising administering a pharmacologically effective amount of an antibody of the invention to a subject, such that radiotherapy is enhanced.

BRIEF DESCRIPTION OF THE FIGURES

[0007] The application file contains at least one photograph executed in color. Copies of this patent application publication with color photographs will be provided by the Office upon request and payment of the necessary fee.

[0008] FIG. 1 depicts dotblots showing that anti-GRP78 antibody 2D6F9 is of the lgG1 isotype. (A) lgG1 dotblot. (B) lgG2a dotblot. (C) lgG2b dotblot. (D) lgG3 dotblot.

[0009] FIG. 2 depicts a graph showing the stability of the anti-GRP78 antibody 2B6F9. Antibody stocks stored at -20°C and tested over six months are stable when tested by ELISA. Dilutions are shown on the right hand legend.

[0010] FIG. 3A depicts images of an irradiated GL261 tumor bearing mouse treated with anti-GRP78 antibody 2B6F9. FIG. 3B depicts images of a second irradiated GL261 tumor bearing mouse treated with anti-GRP78 antibody 2B6F9. FIG. 3C depicts images of a third irradiated GL261 tumor bearing mouse treated with anti- GRP78 antibody 2B6F9. Each mouse has tumors on their right hind limbs and no tumors on their left hind limb. Each mouse was exposed to three separate 3Gy doses of radiation, separated by approximately 6 hours. Following radiation exposure, each mouse was administered antibody at 50 g/mouse via i.v. Images were taken at 18, 24, 48, 72, 96, 120, 144, 168, 192, 216, 240, 264, 288, and 312 hours. The antiGRP78 antibody 2B6F9 was conjugated with Alexa Fluor 750, and the images show

accumulation of the antibody at the site of tumor only.

[001 1 ] FIG. 4 depicts images of an irradiated GL261 tumor bearing mouse treated with anti-GRP78 antibody 2B6F9. The mouse was exposed to a single dose of 3Gy radiation on the hind left limb while there was no radiation exposure on the hind right limb. Following radiation exposure, each mouse was administered antibody at 50 g/mouse via i.v. Images were taken at 2 hours and 1 , 2, 3, 5, 6, 7, 8, 9, 10, and 1 1 days. The anti-GRP78 antibody 2B6F9 was conjugated with Alexa Fluor 750, and the images show accumulation of the antibody on the irradiated side.

[0012] FIG. 5A depicts control images of a GL261 tumor bearing mouse treated with normal mouse IgG. FIG. 5B depicts control images of a second GL261 tumor bearing mouse treated with normal mouse IgG. Each non-irradiated mouse bears tumors on the right hind limb. Each mouse was administered antibody at 50 g/mouse via i.v. Images were taken at 18, 24, 48, 72, 96, 120, and 144 hours. The antiGRP78 antibody 2B6F9 was conjugated with Alexa Fluor 750, and the images show no accumulation of antibody. FIG. 5C depicts an irradiated mouse bearing tumors on the right hind limb. FIG. 5D depicts a second irradiated mouse bearing tumors on the right hind limb. Each mouse was exposed to a single dose of 3Gy radiation. Following exposure, each mouse was administered antibody at 50 g/mouse via i.v. Images were taken at 18, 24, 48, 72, 96, 120, and 144 hours. The antiGRP78 antibody 2B6F9 was conjugated with Alexa Fluor 750, and the images show no accumulation of antibody. FIG. 5E depicts an irradiated mouse bearing tumors on the right hind limb. The mouse was exposed to three separate 3Gy doses of radiation, separated by approximately 6 hours. Following exposure, the mouse was administered antibody at 50 g/mouse via i.v. Images were taken at 18, 24, 48, 72, 96, 120, 144, and 168 hours. The anti-GRP78 antibody 2B6F9 was conjugated with Alexa Fluor 750, and the images show no accumulation of antibody.

[0013] FIG. 6A depicts an image showing distribution of 64 Cu-anti-GRP78 antibody 2D6F9 in mice. FIG. 6B depicts an image showing distribution of 64 Cu-anti- GRP78 antibody 2D6F9 in mice. Mice were imaged 24 hours after administration of 50 pg 64 Cu-anti-GRP78 antibody 2D6F9. The left hindlimb was irradiated with 3Gy and the right hindlimb was not irradiated. FIG. 6C depicts mice 48 hours after administration of 50 pg 64 Cu-anti-GRP78 antibody 2D6F9. The left hindlimb was irradiated with 3Gy and the right hindlimb was not irradiated. FIG. 6D depicts a graph showing the ex-vivo biodistribution of 64 Cu-NOTA-anti-GRP78 2D6F9 antibody. [0014] FIG. 7 depicts the variable region sequences for anti-GRP78 antibody 2B6F9.

[0015] FIG. 8 depicts a graph showing the radiance emitted from the anti- GRP78 antibody 2B6F9 conjugated to Alexa Fluor 750 over time.

[0016] FIG. 9A depicts images of an irradiated D54 tumor bearing mouse treated with anti-GRP78 antibody 2B6F9. FIG. 9B depicts images of an irradiated D54 tumor bearing mouse treated with anti-GRP78 antibody 2B6F9. The mice were exposed to a single dose of 3Gy radiation on the hind left limb while there was no radiation exposure on the hind right limb. Following radiation exposure, each mouse was administered antibody at 50 g/mouse via i.v. Images were taken at 2 hours and 1 , 2, 3, 4, 5, 6, and 7 days. The antiGRP78 antibody 2B6F9 was conjugated with Alexa Fluor 750.

[0017] FIG. 10A depicts a graph showing urine excretion data plotted as a function of time. FIG. 10B depicts a graph showing feces excretion data plotted as a function of time. The heavy dash lines represent fit with an uptake function used to calculate the total amount of activity excreted expressed in units of hours. The light dash lines show the fit accounting for radio-active decay.

[0018] FIG. 11 depicts a plot showing blood clearance.

[0019] FIG. 12A depicts a graph showing urine excretion data plotted as a function of time. FIG. 12B depicts a graph showing feces excretion data plotted as a function of time. The heavy dash lines represent fit with an uptake function used to calculate the total amount of activity excreted expressed in units of hours. The light dash lines show the fit accounting for radioactive decay.

[0020] FIG. 13 depicts an image of a Western blot of GRP78 expression in XRT-treated and untreated (No Tx) MDA-MB-231 breast carcinoma tumor sections showing that GRP78 is upregulated to the membrane in response to XRT.

[0021 ] FIG. 14 depicts micrograph images showing GRP78 is induced in HUVECs grown in coculture with lung cancer cells after XRT treatment. Lung cancer cells (3 x 10 5 ) and HUVECs (1 10 4 ) were cocultured for 24 hours before treatment with 3 Gy XRT. Three hours posttreatment, AlexaFluor594-labeled GRP78 antibodies were added to the culture plates.

[0022] FIG. 15 depicts a plot showing that 2D6F3 enhances radiation induced cytotoxicity. Cancer cells were plated on 10 cm dishes and treated with Control IgG, 2D6F9 5pg, or no antibody. 12 hours later cells were treated with 2 Gy or 0 Gy. The bar graph shows the percentage of MDA-MB231 glioma cells forming colonies after 7 days of incubation normalized to the untreated control. Shown is the mean and SEM of 3 experiments * p<0.01 .

[0023] FIG. 16 depicts images of mice and a graph showing 1 D6B2, 2D6F3 and scFv binding in irradiated NSCLC in mice. Shown are near infrared images of mice obtained 48 hours after administration. Tumors were implanted into the hind limb of mice and treated with 3 Gy. (A) 1 D6B2 Ab was labeled with near infrared fluorophore ALX750 and injected immediately after irradiation. (B) 2D6F3 antibody labeled with ALX750 and injected immediately after treatment of tumors with 3Gy (left tumor) or 0 Gy (right tumor). (C) scFv antibody K13 labeled with ALX750 and injected immediately after irradiation of tumors. (D) Graph showing the time course of radiance (photons/sec/cm 2 ) from NIR images acquired daily after administration of 2D6F3.

[0024] FIG. 17 depicts a graph showing specificity and binding activity to TIP-1 antigen using TE1 1 anti TIP-1 scFv antibody produced and prepared in two individual batches (TE1 1 (1 ) and TE1 1 (2)).

[0025] FIG. 18 depicts a graph showing a test of DOTA-conjugate TE1 1 anti TIP-1 scFv antibody.

[0026] FIG. 19 depicts images of irradiated tumor bearing mice treated with anti-TIP-1 scFv antibody TE1 1 . The mice were exposed to a single dose of 3Gy radiation on the hind left limb while there was no radiation exposure on the hind right limb. Following radiation exposure, each mouse was administered antibody. Images were taken at 2, 16, 24, 46, and 72 hours.

[0027] FIG. 20 depicts images of irradiated tumor bearing mice treated with control scFv antibody. The mice were exposed to a single dose of 3Gy radiation on the hind left limb while there was no radiation exposure on the hind right limb. Following radiation exposure, each mouse was administered antibody. Images were taken at 2, 16, 24, 46, and 72 hours.

[0028] FIG. 21 depicts a graph showing the radiance emitted from the anti- TIP-1 scFv antibody TE1 1 over time.

[0029] FIG. 22 depicts sequences of two anti-TIP-1 scFv antibody clones (SEQ ID NO:22 - upper panel; SEQ ID NO:23 - lower panel). Blue=leader sequence, Green=epitope tag.

[0030] FIG. 23A depicts flow cytometry analysis of the human

glioblastoma D54 (WHO grade IV) cell population with the gated cells encircled. The top panel is sham treated cells, the middle panel is 3Gy x 3 treated cells at 24h, the bottom panel is 3Gy x 3 treated cells at 48 h. FIG. 23B depicts the human glioblastoma D54 (WHO grade IV) cell population stained with the secondary antibody. The top panel is sham treated cells, the middle panel is 3Gy x 3 treated cells at 24h, the bottom panel is 3Gy x 3 treated cells at 48 h. FIG. 23C depicts the human glioblastoma D54 (WHO grade IV) cell population stained with a commercial Tip-1 mAb. The top panel is sham treated cells, the middle panel is 3Gy x 3 treated cells at 24h, the bottom panel is 3Gy x 3 treated cells at 48 h. FIG. 23D depicts the human glioblastoma D54 (WHO grade IV) cell population stained with the anti-TIP-1 monoclonal antibody, 2C6F3 Ab. The top panel is sham treated cells, the middle panel is 3Gy x 3 treated cells at 24h, the bottom panel is 3Gy x 3 treated cells at 48 h.

[0031 ] FIG. 24A depicts images of an irradiated GL261 tumor bearing mouse treated with anti-TIP-1 antibody 2C6F3. FIG. 24B depicts images of a second irradiated GL261 tumor bearing mouse treated with anti-TIP-1 antibody 2C6F3. Each mouse was exposed to three separate 3Gy doses of radiation, separated by

approximately 6 hours, on the hind right limb while there was no radiation exposure on the hind left limb. Following radiation exposure, each mouse was administered antibody via i.v. Images were taken at 18, 24, 48, 72, 96, 120, 144, 168, 192 and 216 hours. The anti-TIP-1 antibody 2C6F3 was conjugated with Alexa Fluor 750, and the images show accumulation of the antibody on the irradiated side. [0032] FIG. 25 depicts a graph showing labeling of DTPA chelator on antibody 2C6F3 tested by ELISA.

[0033] FIG. 26A depicts CT/SPECT images showing distribution of 111 In- DTPA-2C6F3 in a LLC tumor bearing mouse. FIG. 26B depicts CT/SPECT images showing distribution of 111 ln-DTPA-2C6F3 in a second LLC tumor bearing mouse. FIG. 26C depicts CT/SPECT images showing distribution of 111 ln-DTPA-2C6F3 in a third LLC tumor bearing mouse. Mice were irradiated with 3Gy x 3 over a 24 hour period or sham non-irradiated. Images are of three mice 48 hours after administration of 111 ln-DTPA- 2C6F3.

[0034] FIG. 27A depicts CT/SPECT images showing distribution of 111 In- DTPA-2C6F3 in a LLC tumor bearing mouse. FIG. 27B depicts CT/SPECT images showing distribution of 111 ln-DTPA-2C6F3 in a second LLC tumor bearing mouse. FIG. 27C depicts CT/SPECT images showing distribution of 111 ln-DTPA-2C6F3 in a third LLC tumor bearing mouse. Mice were irradiated with 3Gy x 3 over a 24 hour period or sham non-irradiated. Images are of three mice 78 hours after administration of 111 ln-DTPA- 2C6F3.

DETAILED DESCRIPTION OF THE INVENTION

[0035] The present invention encompasses antibodies that recognize tumor cells. The antibodies may be used to provide tumor specific delivery, for instance, of drugs or therapeutic agents, as well as enhancing the efficacy of radiotherapy. In particular, the present invention provides for antibodies that bind to GRP78 and TIP-1 . Advantageously, these antibodies specifically bind tumor cells and not normal cells.

[0036] In an exemplary embodiment, antibodies of the invention

specifically bind to epitopes exposed on irradiated tumor related cells. For instance, antibodies of the invention may bind to extracellular, transmembrane or intracellular epitopes on irradiated tumor related cells. I. Antibodies

[0037] The present invention encompasses antibodies that recognize tumor cells. In an aspect, antibodies useful herein include those antibodies which have been isolated, characterized, purified, are functional and have been recovered

(obtained) for use in a functional therapeutic composition which is administered to a living patient.

[0038] The term "antibody" refers to an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, or

polypeptide through at least one antigen recognition site. As used herein, an antibody encompasses not only intact polyclonal or monoclonal antibodies, but also fragments thereof (such as Fab, Fab', F(ab')2. Fv), single chain (scFv), mutants thereof, fusion proteins comprising an antibody portion, humanized antibodies, chimeric antibodies, diabodies, linear antibodies, single chain antibodies, multispecific antibodies (e.g., bispecific antibodies) and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity.

[0039] An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), or the antibody need not be of any particular class. As long as the protein retains the ability specifically to bind its intended target, it is included within the term "antibody."

[0040] Also included within the definition "antibody" for example are single chain forms, generally designated Fv or scFv, regions, of antibodies with this specificity. These scFvs comprise heavy and light chain variable regions connected by a linker. In most instances, but not all, the linker may be a peptide. A linker peptide is preferably from about 10 to 25 amino acids in length. Preferably, a linker peptide is rich in glycine, as well as serine or threonine. ScFvs can be used to facilitate phage display or can be used for flow cytometry, immunohistochemistry, or as targeting domains. Methods of making and using scFvs are known in the art.

[0041 ] The basic structural unit of an antibody useful herein comprises a tetramer. Each tetramer is 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 1 10 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.

[0042] Depending on the antibody amino acid sequence of the constant domain of its heavy chains, immunoglobulins can be assigned to different classes. In mammals, heavy-chains are classified as alpha, delta, epsilon, gamma, or mu, and define the antibody's isotype as IgA, IgD, IgE, IgG, and IgM, respectively. Several of these isotypes may be further divided into subclasses (isotypes), e.g., lgG1 , lgG2, lgG3, lgG4, lgA1 and lgA2. Light chains are classified as kappa and lambda.

[0043] The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. 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. The variable regions of each light/heavy chain pair form the antibody binding site. Thus, an intact antibody has two binding sites. The chains exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarily determining regions (hereinafter referred to as "CDRs.") The CDRs from the two chains are aligned by the framework regions, enabling binding to a specific epitope. From N-terminal to C-terminal, both light and heavy chains comprise the domains FR1 , CDR1 , FR2, CDR2, FR3, CDR3 and FR4 respectively. The assignment of amino acids to each domain is in accordance with known conventions (See, Kabat "Sequences of Proteins of Immunological Interest" National Institutes of Health, Bethesda, Md., 1987 and 1991 ; Chothia, et al, J. Mol. Bio. (1987) 196:901 -917; Chothia, et al., Nature (1989) 342:878-883).

[0044] In some embodiments, the antibodies of the invention may be monoclonal antibodies. "Monoclonal antibody" refers to an antibody that is derived from a single copy or clone. A monoclonal antibody is not limited to antibodies produced through hybridoma technology. Monoclonal antibodies may be produced using e.g., hybridoma techniques well known in the art, as well as recombinant technologies, phage display technologies, synthetic technologies or combinations of such technologies and other technologies readily known in the art. A monoclonal antibody may encompass not only intact monoclonal antibodies and full-length monoclonal antibodies, but also fragments thereof (such as Fab, Fab', F(ab')2, Fv), single chain (scFv), mutants thereof, fusion proteins comprising an antibody portion, humanized monoclonal antibodies, chimeric monoclonal antibodies, and any other modified configuration of the

immunoglobulin molecule that comprises an antigen recognition site of the required specificity and the ability to bind to an antigen. Furthermore, the monoclonal antibody may be labeled with a detectable label, immobilized on a solid phase and/or conjugated with a heterologous compound (e.g., an enzyme or toxin) according to methods known in the art.

[0045] Preferably, but not necessarily, the antibodies useful in the discovery are produced recombinantly, as manipulation of the typically murine or other non-human antibodies with the appropriate specificity is required in order to convert them to humanized form. Antibodies may or may not be glycosylated, though

glycosylated antibodies are preferred. Antibodies are properly cross-linked via disulfide bonds, as is known.

[0046] Antibodies useful herein include those which are isolated, characterized, purified, functional and have been recovered (obtained) from a process for their preparation and thus available for use herein in a useful form in a

therapeutically and medicinally sufficient amount.

[0047] In an aspect, antibodies of the invention are generated with appropriate specificity by standard techniques of immunization of mammals, forming hybridomas from the antibody-producing cells of said mammals or otherwise

immortalizing them, and culturing the hybridomas or immortalized cells to assess them for the appropriate specificity. In the present case, such antibodies may be generated by immunizing a human, rabbit, rat or mouse, for example, with a peptide representing an epitope encompassing a region of the GRP78 or TIP-1 protein coding sequences or an appropriate subregion thereof. Materials for recombinant manipulation may be obtained by retrieving the nucleotide sequences encoding the desired antibody from the hybridoma or other cell that produces it. These nucleotide sequences may then be manipulated and isolated, characterized, purified and recovered to provide them in humanized form, if desired.

(a) antibodies that bind to GRP78

[0048] One aspect of the present invention encompasses an antibody that binds to GRP78.

(i) heavy chain sequence

[0049] In one embodiment, such an antibody is encoded by a heavy chain variable region nucleic acid sequence that comprises at least about 60% homology to SEQ ID NO:5. In some embodiments, an antibody comprises at least 60, 65, 70, 75, 80, 85, 90, or 95% homology with SEQ ID NO: 5. In one embodiment, the heavy chain variable region nucleic acid sequence has at least about 95, 96, 97, 98, or 99% sequence homology with SEQ ID NO:5. In a preferred embodiment, the heavy chain variable region nucleic acid sequence encodes the amino acid sequence of SEQ ID NO:6.

[0050] In other embodiments, an isolated antibody of the present invention comprises a heavy chain variable region amino acid sequence with at least about 60% homology to SEQ ID NO: 6. In some embodiments, an antibody comprises at least 60, 65, 70, 75, 80, 85, 90, or 95% homology with SEQ ID NO: 6. In one embodiment, the heavy chain variable region nucleic acid sequence has at least about 95, 96, 97, 98, or 99% sequence homology with SEQ ID NO:6.

[0051 ] In certain embodiments, an antibody that binds to GRP78 is an isolated antibody that comprises at least one heavy chain variable domain

complementary determining region (CDR) sequence. Typically, a heavy chain variable domain comprises three CDR sequences (CDR1 , CDR2, and CDR3), separated by framework regions. In one embodiment, an antibody of the invention comprises a heavy chain CDR1 region that comprises at least five contiguous amino acids of SEQ ID NO: 15 (SFTGYFMN). For instance, the heavy chain CDR1 region may comprise 6, 7, or 8 contiguous amino acids of SEQ ID NO: 15. In another embodiment, an antibody of the invention comprises a heavy chain CDR1 region comprising SEQ ID NO:15.

[0052] In other embodiments, an antibody of the invention comprises a heavy chain CDR2 region that comprises at least five contiguous amino acids of SEQ ID NO: 16 (IGRIDPYNGNIFYNQ). For instance, the heavy chain CDR2 region may comprise 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, or 15 contiguous amino acids of SEQ ID NO: 16. In another embodiment, an antibody of the invention comprises a heavy chain CDR2 region comprising SEQ ID NO:16.

[0053] In yet another embodiment, an antibody of the invention comprises a heavy chain CDR3 region that comprises at least five contiguous amino acids of SEQ ID NO: 17 (AVYYCGRSYGNY). For instance, the heavy chain CDR3 region may comprise 6, 7, 8, 9, 10, 1 1 , or 12 contiguous amino acids of SEQ ID NO: 17. In another embodiment, an antibody of the invention comprises a heavy chain CDR3 region comprising SEQ ID NO:17.

[0054] A heavy chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO: 15 and a CDR2 of SEQ ID NO:16. In another embodiment, a heavy chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO:15 and a CDR3 of SEQ ID NO: 17. In yet another embodiment, a heavy chain variable region of an antibody of the present invention may comprise a CDR2 of SEQ ID NO: 16 and a CDR3 of SEQ ID NO:17. In still another embodiment, a heavy chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO:15, a CDR2 of SEQ ID NO:16, and a CDR3 of SEQ ID NO:17.

[0055] In each of the above embodiments, a CDR sequence may have one, two, or three amino acid substitutions. These substitutions may be conservative or non-conservative, providing the antibody specifically recognizes GRP78.

(ii) light chain sequence [0056] In an embodiment, such an antibody is encoded by a light chain variable region nucleic acid sequence that comprises at least about 60% homology to SEQ ID NO:7. In some embodiments, an antibody comprises at least 60, 65, 70, 75, 80, 85, 90, or 95% homology with SEQ ID NO: 7. In one embodiment, the light chain variable region nucleic acid sequence has at least about 95, 96, 97, 98, or 99% sequence homology with SEQ ID NO:7. In a preferred embodiment, the light chain variable region nucleic acid sequence encodes the amino acid sequence of SEQ ID NO:8.

[0057] In other embodiments, an isolated antibody of the present invention comprises a light chain variable region amino acid sequence with at least about 60% homology to SEQ ID NO: 8. In some embodiments, an antibody comprises at least 60, 65, 70, 75, 80, 85, 90, or 95% homology with SEQ ID NO: 8. In one embodiment, the heavy chain variable region nucleic acid sequence has at least about 95, 96, 97, 98, or 99% sequence homology with SEQ ID NO:8.

[0058] In certain embodiments, an antibody that binds to GRP78 is an isolated antibody that comprises at least one light chain variable domain complementary determining region (CDR) sequence. Typically, a light chain variable domain comprises three CDR sequences (CDR1 , CDR2, and CDR3), separated by framework regions. In one embodiment, an antibody of the invention comprises a light chain CDR1 region that comprises at least five contiguous amino acids of SEQ ID NO: 18 (GETITINCRA). For instance, the light chain CDR1 region may comprise 6, 7, 8, 9, or 10 contiguous amino acids of SEQ ID NO: 18. In another embodiment, an antibody of the invention

comprises a light chain CDR1 region that comprises SEQ ID NO:18.

[0059] In an alternative embodiment, an antibody of the invention comprises a light chain CDR2 region that comprises at least five contiguous amino acids of SEQ ID NO: 19 (KPGKTNKLLIYF). For instance, the light chain CDR2 region may comprise 6, 7, 8, 9, 10, 1 1 , or 12 contiguous amino acids of SEQ ID NO: 19. In another embodiment, an antibody of the invention comprises a light chain CDR2 region that comprises SEQ ID NO:19. [0060] In yet another embodiment, an antibody of the invention comprises a light chain CDR3 region that comprises at least five contiguous amino acids of SEQ ID NO: 20 (EPEDFAMYFCQQHNE). For instance, the light chain CDR3 region may comprise 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, or 15 contiguous amino acids of SEQ ID NO: 20. In another embodiment, an antibody of the invention comprises a light chain CDR3 region that comprises SEQ ID NO:20.

[0061 ] A light chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO: 18 and a CDR2 of SEQ ID NO:19. In another embodiment, a light chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO:18 and a CDR3 of SEQ ID NO: 20. In yet another embodiment, a light chain variable region of an antibody of the present invention may comprise a CDR2 of SEQ ID NO: 19 and a CDR3 of SEQ ID NO:20. In still another embodiment, a light chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO:18, a CDR2 of SEQ ID NO:19, and a CDR3 of SEQ ID NO:20.

[0062] In each of the above embodiments, a CDR sequence may have one, two, or three amino acid substitutions. These substitutions may be conservative or non-conservative, providing the antibody specifically recognizes GRP78.

(Hi) preferred embodiments

[0063] In preferred embodiments, an antibody of the invention is encoded by a heavy chain variable region nucleic acid sequence of SEQ ID NO:5 and a light chain variable region nucleic acid sequence of SEQ ID NO:7. In another preferred embodiment, an antibody of the invention is encoded by a heavy chain variable region nucleic acid that encodes SEQ ID NO:6 and a light chain variable region nucleic acid sequence that encodes SEQ ID NO:8. In still other preferred embodiments, an antibody of the invention comprises a heavy chain variable region of SEQ ID NO:6 and a light chain variable region of SEQ ID NO:8.

[0064] In an exemplary embodiment, an antibody of the invention may comprise a combination of CDR sequences listed in Table A below. Table A: CDR combinations comprising antibodies that recognize GRP78

43 16 18 20

44 16 18 19

45 16 20

46 16 19

47 16 18

48 16

49 17 18 19 20

50 17 19 20

51 17 18 20

52 17 18 19

53 17 20

54 17 19

55 17 18

56 17

57 18 19 20

58 19 20

59 18 20

60 18 19

61 20

62 19

63 18

(iv) GRP78 epitopes

[0065] In one embodiment, an isolated antibody of the present invention that binds to GPR78 recognizes an epitope within the amino acid sequence of SEQ ID NO:1 . For instance, a GRP78 antibody of the invention may recognize an epitope with 5, 6, 7, 8, or 9 contiguous amino acids of SEQ ID NO:1 .

[0066] In yet another embodiment, an isolated antibody of the present invention that binds to GPR78 recognizes an epitope within the amino acid sequence of SEQ ID NO:2. For instance, a GRP78 antibody of the invention may recognize an epitope with 5, 6, 7, 8, or 9 contiguous amino acids of SEQ ID NO: 2.

[0067] In yet a further embodiment, an isolated antibody of the present invention that binds to GPR78 recognizes an epitope within the amino acid sequence of SEQ ID NO:3. For instance, a GRP78 antibody of the invention may recognize an epitope with 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, or 15 contiguous amino acids of SEQ ID NO:3. [0068] In an additional embodiment, an isolated antibody of the present invention that binds to GPR78 recognizes an epitope within the amino acid sequence of SEQ ID NO:4. For instance, a GRP78 antibody of the invention may recognize an epitope with 5, 6, 7, 8, or 9 contiguous amino acids of SEQ ID NO: 4.

(b) antibodies that bind to TIP-1

[0069] One aspect of the present invention encompasses an antibody that binds to TIP-1 .

(i) heavy chain sequence

[0070] In one embodiment, such an antibody is encoded by a heavy chain variable region nucleic acid sequence that comprises at least about 60% homology to SEQ ID NO:24. In some embodiments, an antibody comprises at least 60, 65, 70, 75, 80, 85, 90, or 95% homology with SEQ ID NO: 24. In one embodiment, the heavy chain variable region nucleic acid sequence has at least about 95, 96, 97, 98, or 99% sequence homology with SEQ ID NO:24. In a preferred embodiment, the heavy chain variable region nucleic acid sequence encodes the amino acid sequence of SEQ ID NO:25.

[0071 ] In other embodiments, an isolated antibody of the present invention comprises a heavy chain variable region amino acid sequence with at least about 60% homology to SEQ ID NO: 25. In some embodiments, an antibody comprises at least 60, 65, 70, 75, 80, 85, 90, or 95% homology with SEQ ID NO: 25. In one embodiment, the heavy chain variable region nucleic acid sequence has at least about 95, 96, 97, 98, or 99% sequence homology with SEQ ID NO:25.

[0072] In certain embodiments, an antibody that binds to TIP-1 is an isolated antibody that comprises at least one heavy chain variable domain

complementary determining region (CDR) sequence. Typically, a heavy chain variable domain comprises three CDR sequences (CDR1 , CDR2, and CDR3), separated by framework regions. The assignment of amino acids to each domain is in accordance with known conventions (See, Kabat "Sequences of Proteins of Immunological Interest" National Institutes of Health, Bethesda, Md., 1987 and 1991 ; Chothia, et al, J. Mol. Bio. (1987) 196:901 -917; Chothia, et al., Nature (1989) 342:878-883).

[0073] In one embodiment, an antibody of the invention comprises a heavy chain CDR1 region that comprises at least five contiguous amino acids of SEQ ID NO: 28 (SNYWMN). For instance, the heavy chain CDR1 region may comprise 6 contiguous amino acids of SEQ ID NO: 28. In another embodiment, an antibody of the invention comprises a heavy chain CDR1 region comprising SEQ ID NO:28.

[0074] In other embodiments, an antibody of the invention comprises a heavy chain CDR2 region that comprises at least five contiguous amino acids of SEQ ID NO: 29 (QIRLKSDNYATHY). For instance, the heavy chain CDR2 region may comprise 6, 7, 8, 9, 10, 1 1 , 12, or 13 contiguous amino acids of SEQ ID NO: 29. In another embodiment, an antibody of the invention comprises a heavy chain CDR2 region comprising SEQ ID NO:29.

[0075] In yet another embodiment, an antibody of the invention comprises a heavy chain CDR3 region that comprises at least five contiguous amino acids of SEQ ID NO: 30 (GIYYCLLYY). For instance, the heavy chain CDR3 region may comprise 6, 7, 8, or 9 contiguous amino acids of SEQ ID NO: 30. In another embodiment, an antibody of the invention comprises a heavy chain CDR3 region comprising SEQ ID NO:30.

[0076] A heavy chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO:28 and a CDR2 of SEQ ID NO:29. In another embodiment, a heavy chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO:28 and a CDR3 of SEQ ID NO:30. In yet another embodiment, a heavy chain variable region of an antibody of the present invention may comprise a CDR2 of SEQ ID NO:29 and a CDR3 of SEQ ID NO:30. In still another embodiment, a heavy chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO:28, a CDR2 of SEQ ID NO:29, and a CDR3 of SEQ ID NO:30. [0077] In each of the above embodiments, a CDR sequence may have one, two, or three amino acid substitutions. These substitutions may be conservative or non-conservative, providing the antibody specifically recognizes TIP-1 .

(ii) light chain sequence

[0078] In an embodiment, such an antibody is encoded by a light chain variable region nucleic acid sequence that comprises at least about 60% homology to SEQ ID NO:26. In some embodiments, an antibody comprises at least 60, 65, 70, 75, 80, 85, 90, or 95% homology with SEQ ID NO:26. In one embodiment, the light chain variable region nucleic acid sequence has at least about 95, 96, 97, 98, or 99% sequence homology with SEQ ID NO:26. In a preferred embodiment, the light chain variable region nucleic acid sequence encodes the amino acid sequence of SEQ ID NO:27.

[0079] In other embodiments, an isolated antibody of the present invention comprises a light chain variable region amino acid sequence with at least about 60% homology to SEQ ID NO:27. In some embodiments, an antibody comprises at least 60, 65, 70, 75, 80, 85, 90, or 95% homology with SEQ ID NO:27. In one embodiment, the heavy chain variable region nucleic acid sequence has at least about 95, 96, 97, 98, or 99% sequence homology with SEQ ID NO:27.

[0080] In certain embodiments, an antibody that binds to GRP78 is an isolated antibody that comprises at least one light chain variable domain complementary determining region (CDR) sequence. Typically, a light chain variable domain comprises three CDR sequences (CDR1 , CDR2, and CDR3), separated by framework regions. The assignment of amino acids to each domain is in accordance with known

conventions (See, Kabat "Sequences of Proteins of Immunological Interest" National Institutes of Health, Bethesda, Md., 1987 and 1991 ; Chothia, et al, J. Mol. Bio. (1987) 196:901 -917; Chothia, et al., Nature (1989) 342:878-883).

[0081 ] In one embodiment, an antibody of the invention comprises a light chain CDR1 region that comprises at least five contiguous amino acids of SEQ ID NO: 31 (SQSLVHSNG). For instance, the light chain CDR1 region may comprise 6, 7, 8, or 9 contiguous amino acids of SEQ ID NO:31 . In another embodiment, an antibody of the invention comprises a light chain CDR1 region that comprises SEQ ID NO:31 .

[0082] In an alternative embodiment, an antibody of the invention comprises a light chain CDR2 region that comprises at least five contiguous amino acids of SEQ ID NO:32 (KLLIYKVSNRF). For instance, the light chain CDR2 region may comprise 6, 7, 8, 9, 10, or 1 1 contiguous amino acids of SEQ ID NO:32. In another embodiment, an antibody of the invention comprises a light chain CDR2 region that comprises SEQ ID NO:32.

[0083] In yet another embodiment, an antibody of the invention comprises a light chain CDR3 region that comprises at least five contiguous amino acids of SEQ ID NO:33 (GVYFCSQST). For instance, the light chain CDR3 region may comprise 6, 7, 8, or 9 contiguous amino acids of SEQ ID NO:33. In another embodiment, an antibody of the invention comprises a light chain CDR3 region that comprises SEQ ID NO:33.

[0084] A light chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO:31 and a CDR2 of SEQ ID NO:32. In another embodiment, a light chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO:31 and a CDR3 of SEQ ID NO:33. In yet another embodiment, a light chain variable region of an antibody of the present invention may comprise a CDR2 of SEQ ID NO:32 and a CDR3 of SEQ ID NO:33. In still another embodiment, a light chain variable region of an antibody of the present invention may comprise a CDR1 of SEQ ID NO:31 , a CDR2 of SEQ ID NO:32, and a CDR3 of SEQ ID NO:33.

[0085] In each of the above embodiments, a CDR sequence may have one, two, or three amino acid substitutions. These substitutions may be conservative or non-conservative, providing the antibody specifically recognizes TIP-1 .

(Hi) specific embodiments

[0086] In specific embodiments, an antibody of the invention is encoded by a heavy chain variable region nucleic acid sequence of SEQ ID NO:24 and a light chain variable region nucleic acid sequence of SEQ ID NO:26. In another specific embodiment, an antibody of the invention is encoded by a heavy chain variable region nucleic acid that encodes SEQ ID NO:25 and a light chain variable region nucleic acid sequence that encodes SEQ ID NO:27. In still other specific embodiments, an antibody of the invention comprises a heavy chain variable region of SEQ ID NO:25 and a light chain variable region of SEQ ID NO:27. In an exemplary embodiment, an antibody of the invention may comprise a combination of CDR sequences listed in Table B below.

Table B: CDR combinations comprising antibodies that recognize TIP-1

33 28 31 32 33

34 28 32 33

35 28 31 33

36 28 31 32

37 28 33

38 28 32

39 28 31

40 28

41 29 31 32 33

42 29 32 33

43 29 31 33

44 29 31 32

45 29 33

46 29 32

47 29 31

48 29

49 30 31 32 33

50 30 32 33

51 30 31 33

52 30 31 32

53 30 33

54 30 32

55 30 31

56 30

57 31 32 33

58 32 33

59 31 33

60 31 32

61 33

62 32

63 31

(iv) TIP-1 epitopes

[0087] In one embodiment, the isolated antibody of the present invention that binds to TIP-1 recognizes an epitope within the amino acid sequence of SEQ ID NO:9. For instance, a TIP-1 antibody of the invention may recognize an epitope with 5, 6, 7, 8, or 9 contiguous amino acids of SEQ ID NO: 9.

[0088] In yet another embodiment, the isolated antibody of the present invention that binds to TIP-1 recognizes an epitope within the amino acid sequence of SEQ ID NO:10. For instance, a TIP-1 antibody of the invention may recognize an epitope with 5, 6, 7, 8, or 9 contiguous amino acids of SEQ ID NO: 10.

[0089] In yet a further embodiment, the isolated antibody of the present invention that binds to TIP-1 recognizes an epitope within the amino acid sequence of SEQ ID NO:1 1 . For instance, a TIP-1 antibody of the invention may recognize an epitope with 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, or 15 contiguous amino acids of SEQ ID NO:1 1 .

[0090] In an additional embodiment, the isolated antibody of the present invention that binds to TIP-1 recognizes an epitope within the amino acid sequence of SEQ ID NO:12. For instance, a TIP-1 antibody of the invention may recognize an epitope with 5, 6, 7, 8, or 9 contiguous amino acids of SEQ ID NO: 12.

[0091 ] Yet another embodiment provides for the isolated antibody of the present invention that binds to TIP-1 , wherein the antibody recognizes an epitope within the amino acid sequence of SEQ ID NO: 13. For instance, a TIP-1 antibody of the invention may recognize an epitope with 5, 6, 7, 8, 9, or 10 contiguous amino acids of SEQ ID NO: 13.

[0092] In still another embodiment, the isolated antibody of the present invention that binds to TIP-1 recognizes an epitope within the amino acid sequence of SEQ ID NO:14. For instance, a TIP-1 antibody of the invention may recognize an epitope with 5, 6, 7, 8, 9, 10, 1 1 , or 12 contiguous amino acids of SEQ ID NO:14.

[0093] In another embodiment, the isolated antibody of the present invention that binds to TIP-1 recognizes an epitope within the amino acid sequence of SEQ ID NO:21 . For instance, a TIP-1 antibody of the invention may recognize an epitope with 5, 6, 7, 8, or 9 contiguous amino acids of SEQ ID NO: 21 .

[0094] In yet another embodiment, an isolated antibody of the invention that binds to TIP-1 is an scFv antibody encoded by a nucleic acid sequence that encodes SEQ ID NO:22 (Fig. 22).

[0095] In another embodiment, an isolated antibody of the invention that binds to TIP-1 is an scFv antibody encoded by a nucleic acid sequence that encodes SEQ ID NO:23 (Fig. 22). [0096] Each of the sequences referred to above may be found in Table C below.

KPGKTNKLLIYF

EPEDFAMYFCQQHNE

QPVTAWQRV

FFGDFQREKIIIRNSFSCSFLCGPAGHGPGETAAVWGQGTTVTV

SSGGGGSGGGGSGGGGSDIELTQSPSTMTASPGEKVTITCRAS

SSVSYMHWYQQKPGASPKPWIYDTSKLASGVPDRFSGSGSGT

SYSLTINNMEAEDAATYYCQQWNYPSTFGAGTKLEIKPAAAGAP

VPYPDPLEPRAATVESCLAKPHTENSFTNVWKDDKTLDRYANY

EGCLWNATGVWCTGDETQCYGTWVPIGLAIPEN

FQREKIIIRNSFSCSFLCGPAGHGPGETAAVWGQGTTVTVSSGG

GGSGGGGSGGGGSDIELTQSPSTMTASPGEKVTITCRASSSVS

YMHWYQQKPGASPKPWIYDTSKLASGVPDRFSGSGSGTSYSL

TINNMEAEDAATYYCQQWNYPSTFGAGTKLEIKPAAAGAPVPYP

DPLEPRAATVESCLAKPHTENSFTNVWKDDKTLDRYANYEGCL

WNATGVWCTGDETQCYGTWVPI

GAAGTGAAGCTTGAGGAGTCTGGAGGAGGCTTGGTGCAACC TG G AG G ATCCATG AAACTCTCCTGTGTTGCCTCTG G ATTCAC I I I CAGTAACTACTG G ATG AACTG G GTCCGCCAGTCTCCAG A G AAG G G GCTTG AGTG G GTTGCTCAAATTAG ATTG AAATCTG A TAATTATGCAACACATTATGCG G AGTCTGTG AAAG G G AG GTT CACCATCTCAAGAGATGATTCCAAAAGTAGTGTCTACCTGCAA ATGAACAACTTAAGGGCTGAAGACACTGGAA I I I ATTACTGCT TAC I I I ACTACGGTCCTAGCGGGACTGCTTACTGGGGCCAAG G G ACTCTG GTCACTGTCTCTGCA

EVKLEESGGGLVQPGGSMKLSCVASGFTFSNYWMNWVRQSPE KGLEWVAQIRLKSDNYATHYAESVKGRFTISRDDSKSSVYLQMN NLRAEDTGIYYCLLYYGPSGTAYWGQGTLVTVSA

GATGTTGTGATGACCCAAACTCCACTCTCCCTGCCTGTCAGT CTTGGAGATCAAGCCTCCATCTCTTGCAGATCTAGTCAGAGC CTTGTACACAGTAATGGAAACACCTA I I I ACATTGGTACCTGC AGAAGCCAGGCCAGTCTCCAAAGCTCCTGATCTACAAAG I I I CCAACCGA I I I I CTGGGGTCCCAGACAGGTTCAGTGGCAGTG GATCAGGGACAGA I I I CACACTCAAGATCAGCAG AGTG GAG G CTG AG G ATCTG G G AGTTTATTTCTGCTCTCAAAGTACAC ATGT TCCTCGGACGTTCGGTGGAGGCACCAAGCTGGAAATCAAAC GG

DWMTQTPLSLPVSLGDQASISCRSSQSLVHSNGNTYLHWYLQ KPGQSPKLLIYKVSNRFSGVPDRFSGSGSGTDFTLKISRVEAED LGVYFCSQSTHVPRTFGGGTKLEIKR

SNYWMN

QIRLKSDNYATHY

GIYYCLLYY

SQSLVHSNG

KLLIYKVSNRF 33 GVYFCSQST

(c) humanized antibodies

[0097] The antibodies of the present invention may also be chimeric antibodies. Preferably, these chimeric antibodies involve the merging of a portion of a monoclonal antibody with antibody-producing DNA in living cells to produce a monoclonal antibody that has material from more than one species of animal. This procedure is well known in the art and any known method to produce chimeric antibodies is suitable for purposes of the present invention. In a preferred embodiment, the chimeric antibody comprises mouse elements conjugated to the genetic material of another species. In a particularly preferred embodiment, the chimeric antibody comprises mouse and human elements to form humanized antibodies. The process of humanization decreases the potential for the antibody to induce an immune response in a human host.

[0098] As used herein "humanized antibody" includes an anti-GRP78 or anti-TIP-1 antibody that is composed partially or fully of amino acid sequences derived from a human antibody germline by altering the sequence of an antibody having non- human complementarity determining regions ("CDR"). The simplest such alteration may consist simply of substituting the constant region of a human antibody for the murine constant region, thus resulting in a human/murine chimera which may have sufficiently low immunogenicity to be acceptable for pharmaceutical use. Preferably, however, the variable region of the antibody and even the CDR is also humanized by techniques that are by now well known in the art. The framework regions of the variable regions are substituted by the corresponding human framework regions leaving the non-human CDR substantially intact, or even replacing the CDR with sequences derived from a human genome. CDRs may also be randomly mutated such that binding activity and affinity for GRP78 or TIP-1 is maintained or enhanced in the context of fully human germline framework regions or framework regions that are substantially human.

Substantially human frameworks have at least 90%, 95%, or 99% sequence identity with a known human framework sequence. Fully useful human antibodies are produced in genetically modified mice whose immune systems have been altered to correspond to human immune systems. As mentioned above, it is sufficient for use in the methods of this discovery, to employ an immunologically specific fragment of the antibody, including fragments representing single chain forms.

[0100] If desired, the design of humanized immunoglobulins may be carried out as follows. When an amino acid falls under the following category, the framework amino acid of a human immunoglobulin to be used (acceptor immunoglobulin) is replaced by a framework amino acid from a CDR-providing nonhuman immunoglobulin (donor immunoglobulin): (a) the amino acid in the human framework region of the acceptor immunoglobulin is unusual for human immunoglobulin at that position, whereas the corresponding amino acid in the donor immunoglobulin is typical for human

immunoglobulin at that position; (b) the position of the amino acid is immediately adjacent to one of the CDRs; or (c) any side chain atom of a framework amino acid is within about 5-6 angstroms (center-to-center) of any atom of a CDR amino acid in a three dimensional immunoglobulin model (Queen, et al., op. cit, and Co, ct al, Proc. Natl. Acad. Sci. USA (1991 ) 88:2869). When each of the amino acids in the human framework region of the acceptor immunoglobulin and a corresponding amino acid in the donor immunoglobulin is unusual for human immunoglobulin at that position, such an amino acid is replaced by an amino acid typical for human immunoglobulin at that position.

[0101 ] One embodiment of the present invention encompasses a humanized antibody that binds to GRP78. In particular, an embodiment of the invention encompasses a humanized antibody where amino acids in the framework region of either the heavy or light chain variable regions are humanized, leaving the CDRs intact. For instance, in one embodiment, an antibody may comprise a combination of CDRs listed in Table A, and a humanized framework region. In another embodiment, the sequence of one or more CDR regions may be humanized as well as the framework region.

[0102] In an exemplary embodiment, a humanized GRP78 antibody recognizes at least one epitope from the group consisting of SEQ ID NO:1 , 2, 3, or 4. [0103] In an alternative embodiment, the present invention encompasses a humanized antibody that binds to TIP-1 . In particular, an embodiment of the invention encompasses a humanized antibody where amino acids in the framework region of either the heavy or light chain variable regions are humanized, leaving the CDRs intact. For instance, in one embodiment, an antibody may comprise a combination of CDRs listed in Table B, and a humanized framework region. In another embodiment, the sequence of one or more CDR regions may be humanized as well as the framework region.

[0104] In an exemplary embodiment, a humanized TIP-1 antibody recognizes at least one epitope from the group consisting of SEQ ID NO:9, 10, 1 1 , 12, 13, 14, or 21 .

[0105] In another alternative embodiment, the present invention

encompasses a scFV antibody that binds TIP-1 .

(d) antibody conjugates

[0106] In another aspect, an antibody of the present invention is conjugated to a therapeutic agent. In some embodiments, a scFv of the present invention is conjugated to a therapeutic agent. The therapeutic agent preferably reduces or interferes with tumor growth or otherwise reduces the effect of the tumor within the body or organism. A therapeutic agent that reduces the symptoms produced by the tumor or reduces tumor growth is suitable for the present invention.

[0107] Additionally, any therapeutic agent that reduces the symptoms associated with tumor cell growth will work for purposes of the present invention. Non- limiting examples of therapeutic agents may include drugs, therapeutic compounds, genetic materials, metals (such as radioactive isotopes), proteins, peptides,

carbohydrates, lipids, steroids, nucleic acid based materials, or derivatives, analogues, or combinations thereof in their native form or derivatized with hydrophobic or charged moieties to enhance incorporation or adsorption into a cell. Such therapeutic agents may be water soluble or may be hydrophobic. Non-limiting examples of therapeutic agents may include immune-related agents, thyroid agents, respiratory products, antineoplastic agents, anti-helmintics, anti-malarials, mitotic inhibitors, hormones, anti- protozoans, anti-tuberculars, cardiovascular products, blood products, biological response modifiers, anti-fungal agents, vitamins, peptides, anti-allergic agents, anticoagulation agents, circulatory drugs, metabolic potentiators, anti-virals, anti-anginals, antibiotics, anti-inflammatories, anti-rheumatics, narcotics, cardiac glycosides, neuromuscular blockers, sedatives, local anesthetics, general anesthetics, or radioactive atoms or ions. Non-limiting examples of therapeutic agents are included in Table D below. An isolated antibody of the present invention may be conjugated to one, two, three, four, or five therapeutic agents. Methods of conjugating an antibody to a therapeutic agent are known in the art. Generally speaking, the conjugation should not interfere with the antibody recognizing its target, and should not interfere with the active site of the target. In some instances, a scFv may be generated with a cleavable linkage between the scFv and therapeutic agent. Such a linker may allow release of the therapeutic agent at a specific cellular location.

daunorubicin hydrochloride, doxorubicin

hydrochloride, paclitaxel and other taxenes, rapamycin, manumycin A, TNP-470, plicamycin (mithramycin), aminoglutethimide, estramustine phosphate sodium, flutamide, leuprolide acetate, megestrol acetate, tamoxifen citrate, testolactone, trilostane, amsacrine (m-AMSA), asparaginase (L- asparaginase) Erwina asparaginase, interferon a-2a, interferon a-2b, teniposide (VM-26), vinblastine sulfate (VLB), vincristine sulfate, bleomycin sulfate, hydroxyurea, procarbazine, and dacarbazine anti-helmintics pyrantel pamoate, piperazine, tetrachloroethylene, thiabendazole, niclosamide

antimalarials Chloroquine, amodiaquine, antifolate drugs,

proguanil (chloroguanide), mefloquine, quinine, halofantrine, artemesinin and derivaties, primaquine, doxycycline, tetracycline, and clindamycin

mitotic inhibitors etoposide, colchicine, and the vinca alkaloids hormones androgens, progestins, estrogens and antiestrogens, growth hormone, melanocyte stimulating hormone, estradiol, beclomethasone dipropionate,

betamethasone, betamethasone acetate and betamethasone sodium phosphate, vetamethasone disodium phosphate, vetamethasone sodium phosphate, cortisone acetate, dexamethasone, dexamethasone acetate, dexamethasone sodium phosphate, flunisolide, hydrocortisone,

hydrocortisone acetate, hydrocortisone cypionate, hydrocortisone sodium phosphate, hydrocortisone sodium succinate, methylprednisolone,

methylprednisolone acetate, methylprednisolone sodium succinate, paramethasone acetate, prednisolone, prednisolone acetate, prednisolone sodium phosphate, prednisolone tebutate, prednisone, triamcinolone, triamcinolone acetonide, triamcinolone diacetate, triamcinolone hexacetonide, fludrocortisone acetate, oxytocin, vassopressin, glucagon and their derivatives

antiprotozoans chloroquine, hydroxychloroquine, metronidazole, quinine and meglumine antimonite

antituberculars para-aminosalicylic acid, isoniazid, capreomycin sulfate cycloserine, ethambutol hydrochloride ethionamide, pyrazinamide, rifampin, and

streptomycin sulfate

cardiovascular products chelating agents and mercurial diuretics and cardiac glycosides

blood products parenteral iron, hemin, hematoporphyrins and their derivatives

biological response muramyldipeptide, muramyltripeptide, microbial cell modifiers wall components, lymphokines (e.g., bacterial

endotoxin such as lipopolysaccharide, macrophage activation factor), sub-units of bacteria (such as Mycobacteria, Corynebacteria), the synthetic dipeptide N-acetyl-muramyl-L-alanyl-D-isoglutamine anti-fungal agents ketoconazole, nystatin, griseofulvin, flucytosine (5- fc), miconazole, amphotericin B, ricin, cyclosporins, and β-lactam antibiotics (e.g., sulfazecin)

vitamins cyanocobalamin neinoic acid, retinoids and derivatives such as retinol palmitate, and a- tocopherol

peptides manganese super oxide dismutase; enzymes such as alkaline phosphatase

anti-allergic agents Amelexanox

anti-coagulation agents phenprocoumon and heparin

circulatory drugs Propranolol

metabolic potentiators Glutathione

antivirals acyclovir, amantadine azidothymidine (AZT, DDI,

Foscarnet, or Zidovudine), ribavirin and vidarabine monohydrate (adenine arabinoside, ara-A) antianginals diltiazem, nifedipine, verapamil, erythritol tetranitrate, isosorbide dinitrate, nitroglycerin (glyceryl trinitrate) and pentaerythritol tetranitrate

antibiotics dapsone, chloramphenicol, neomycin, cefaclor,

cefadroxil, cephalexin, cephradine erythromycin, clindamycin, lincomycin, amoxicillin, ampicillin, bacampicillin, carbenicillin, dicloxacillin, cyclacillin, picloxacillin, hetacillin, methicillin, nafcillin, oxacillin, penicillin including penicillin G and penicillin V, ticarcillin rifampin, aminoglycosides and tetracycline antiinflammatories diflunisal, ibuprofen, indomethacin, meclofenamate, mefenamic acid, naproxen, oxyphenbutazone, phenylbutazone, piroxicam, sulindac, tolmetin, aspirin and salicylates

antirheumatics Adalimumab, azathioprine, chloroquine and

hydroxychloroquine (antimalarials),

cyclosporine (Cyclosporin A), D-penicillamine, etanercept, gold salts (sodium aurothiomalate, auranofin), infliximab, leflunomide, methotrexate, minocycline (a tetracycline antibiotic), sulfasalazine narcotics Paregoric, opiates, codeine, heroin, methadone, morphine and opium

cardiac glycosides deslanoside, digitoxin, digoxin, digitalin and digitalis neuromuscular blockers atracurium mesylate, gallamine triethiodide,

hexafluorenium bromide, metocurine iodide, pancuronium bromide, succinylcholine chloride (suxamethonium chloride), tubocurarine chloride and vecuronium bromide

sedatives (hypnotics) amobarbital, amobarbital sodium, aprobarbital,

butabarbital sodium, chloral hydrate, ethchlorvynol, ethinamate, flurazepam hydrochloride, glutethimide, methotrimeprazine hydrochloride, methyprylon, midazolam hydrochloride, paraldehyde,

pentobarbital, pentobarbital sodium, phenobarbital sodium, secobarbital sodium, talbutal, temazepam and triazolam

local anesthetics bupivacaine hydrochloride, chloroprocaine

hydrochloride, etidocaine hydrochloride, lidocaine hydrochloride, mepivacaine hydrochloride, procaine hydrochloride and tetracaine hydrochloride

general anesthetics droperidol, etomidate, fentanyl citrate with droperidol, ketamine hydrochloride, methohexital sodium and thiopental sodium

radioactive particles or strontium, iodide rhenium, yttrium, and

ions radiopharmaceuticals, such as radioactive iodine, copper and phosphorus product II. Method of Use

[0108] In another aspect, a composition of the present invention, as described above, may be used in treating, stabilizing and preventing cancer and associated diseases in a subject. By "treating, stabilizing, or preventing cancer" is meant causing a reduction in the size of a tumor or in the number of cancer cells, slowing or preventing an increase in the size of a tumor or cancer cell proliferation, increasing the disease-free survival time between the disappearance of a tumor or other cancer and its reappearance, preventing an initial or subsequent occurrence of a tumor or other cancer, or reducing an adverse symptom associated with a tumor or other cancer. In a desired embodiment, the percent of tumor or cancerous cells surviving the treatment is at least 20, 40, 60, 80, or 100% lower than the initial number of tumor or cancerous cells, as measured using any standard assay (e.g., caspase assays, TUNEL and DNA fragmentation assays, cell permeability assays, and Annexin V assays).

Desirably, the decrease in the number of tumor or cancerous cells induced by

administration of a peptide of the invention is at least 2, 5, 10, 20, or 50-fold greater than the decrease in the number of non-tumor or non-cancerous cells. Desirably, the methods of the present invention result in a decrease of 20, 40, 60, 80, or 100% in the size of a tumor or in the number of cancerous cells, as determined using standard methods. Desirably, at least 20, 40, 60, 80, 90, or 95% of the treated subjects have a complete remission in which all evidence of the tumor or cancer disappears. Desirably, the tumor or cancer does not reappear or reappears after at least 5, 10, 15, or 20 years.

[0109] The antibodies of the present invention may be conjugated to radioisotopes or chemotherapeutic compounds in order to provide specific delivery of radiation and chemotherapy to the site of a tumor. Further, the antibodies of the present invention may be part of a combination therapy. Preferably, a combination therapy would include the use of the antibody of the present invention along with a radiation therapy or chemotherapy course of treatment. It has also been suggested that monoclonal antibodies, such as those described herein, may increase the susceptibility of tumor cells to the effects of chemotherapy or radiation. In preferred embodiments, the antibodies of the invention may be used to enhance the efficacy of cancer radiotherapy. [01 10] In yet another aspect, the present invention provides a method of imaging a cancer or detecting a tumor in a subject. The method comprises exposing a target area of the subject where the presence of a tumor is suspected to ionizing radiation, administering to the subject a composition comprising an antibody that specifically binds a protein exposed on an irradiated cell, wherein the antibody is conjugated to an imaging agent, and detecting the imaging agent to detect binding of the antibody to a cell in the subject, wherein the presence of the imaging agent indicates the presence of a tumor in the target area of the subject. In specific

embodiments, the method may be used to diagnose or image a cancer in a subject. In some embodiments, a method for detecting a tumor can comprise (a) exposing a suspected tumor to ionizing radiation; (b) biopsing a suspected tumor; (c) contacting a peptide of the invention with the suspected tumor in vitro, wherein the peptide is directly or indirectly coupled to a detectable label via its N-terminus such that the C-terminus is free; and (d) detecting the detectable label, whereby a tumor is diagnosed.

[01 1 1 ] As described above, an antibody of the invention may be conjugated to an imaging agent. For instance, an scFv may be conjugated to an imaging agent. Suitable imaging agents may include, but are not limited to,

imaging/tracking agents that may be used for microscopy, e.g. fluorescent microscopy, confocal microscopy, or electron microscopy, magnetic resonance imaging,

tomography, such as gamma (SPECT/CT, planar) and positron emission tomography (PET/CT), radiography, or ultrasound. Imaging/tracking agents may be detectable in situ, in vivo, ex vivo, and in vitro. In general, imaging/tracking agents may include luminescent molecules, chemiluminescent molecules, fluorochromes, fluorescent quenching agents, colored molecules, radioisotopes, scintillants, massive labels (for detection via mass changes), biotin, avidin, streptavidin, protein A, protein G, antibodies or fragments thereof, Grb2, polyhistidine, Ni2+, Flag tags, myc tags, heavy metals, enzymes, alkaline phosphatase, peroxidase, luciferase, electron donors/acceptors, acridinium esters, and colorimetric substrates. The skilled artisan would readily recognize other useful labels that are not mentioned above, which may be employed in the operation of the present invention. [01 12] The antibodies are as described in Section I above. The subject, the cancer, and the administration of the antibodies are described below.

(a) subject

[01 13] A method of the invention may be used to detect or treat a tumor in a subject that is a human, a livestock animal, a companion animal, a lab animal, or a zoological animal. In one embodiment, the subject may be a rodent, e.g. a mouse, a rat, a guinea pig, etc. In another embodiment, the subject may be a livestock animal. Non- limiting examples of suitable livestock animals may include pigs, cows, horses, goats, sheep, llamas and alpacas. In yet another embodiment, the subject may be a

companion animal. Non-limiting examples of companion animals may include pets such as dogs, cats, rabbits, and birds. In yet another embodiment, the subject may be a zoological animal. As used herein, a "zoological animal" refers to an animal that may be found in a zoo. Such animals may include non-human primates, large cats, wolves, and bears. In preferred embodiments, the animal is a laboratory animal. Non-limiting examples of a laboratory animal may include rodents, canines, felines, and non-human primates. In certain embodiments, the animal is a rodent. Non-limiting examples of rodents may include mice, rats, guinea pigs, etc. The genotype of the sterile animal can and may vary depending on the intended use of the animal. In embodiments where the animal is a mouse, the mouse may be a C57BL/6 mouse, a Balb/c mouse, a 129sv mouse, a GL261 tumor bearing mouse, a D54 tumor bearing mouse, or any other laboratory strain.

(b) tumor

[01 14] An antibody of the invention may be used to treat or recognize tumor derived from a neoplasm or a cancer. The neoplasm may be malignant or benign, the cancer may be primary or metastatic; the neoplasm or cancer may be early stage or late stage. Non-limiting examples of neoplasms or cancers that may be treated include acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, anal cancer, appendix cancer, astrocytomas (childhood cerebellar or cerebral), basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brainstem glioma, brain tumors (cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic gliomas), breast cancer, bronchial adenomas/carcinoids, Burkitt lymphoma, carcinoid tumors (childhood, gastrointestinal), carcinoma of unknown primary, central nervous system lymphoma (primary), cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, cervical cancer, childhood cancers, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon cancer, cutaneous T-cell lymphoma, desmoplastic small round cell tumor, endometrial cancer,

ependymoma, esophageal cancer, Ewing's sarcoma in the Ewing family of tumors, extracranial germ cell tumor (childhood), extragonadal germ cell tumor, extrahepatic bile duct cancer, eye cancers (intraocular melanoma, retinoblastoma), gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, germ cell tumors (childhood extracranial, extragonadal, ovarian), gestational trophoblastic tumor, gliomas (adult, childhood brain stem, childhood cerebral astrocytoma, childhood visual pathway and hypothalamic), gastric carcinoid, hairy cell leukemia, head and neck cancer, hepatocellular (liver) cancer, Hodgkin lymphoma, hypopharyngeal cancer, hypothalamic and visual pathway glioma (childhood), intraocular melanoma, islet cell carcinoma, Kaposi sarcoma, kidney cancer (renal cell cancer), laryngeal cancer, leukemias (acute lymphoblastic, acute myeloid, chronic lymphocytic, chronic myelogenous, hairy cell), lip and oral cavity cancer, liver cancer (primary), lung cancers (non-small cell, small cell), lymphomas (AIDS-related, Burkitt, cutaneous T-cell, Hodgkin, non-Hodgkin, primary central nervous system),

macroglobulinemia (Waldenstrom), malignant fibrous histiocytoma of

bone/osteosarcoma, medulloblastoma (childhood), melanoma, intraocular melanoma, Merkel cell carcinoma, mesotheliomas (adult malignant, childhood), metastatic squamous neck cancer with occult primary, mouth cancer, multiple endocrine neoplasia syndrome (childhood), multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative diseases, myelogenous leukemia (chronic), myeloid leukemias (adult acute, childhood acute), multiple myeloma, myeloproliferative disorders (chronic), nasal cavity and paranasal sinus cancer, nasopharyngeal carcinoma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, osteosarcoma/malignant fibrous histiocytoma of bone, ovarian cancer, ovarian epithelial cancer (surface epithelial- stromal tumor), ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, pancreatic cancer (islet cell), paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pineal astrocytoma, pineal germinoma, pineoblastoma and supratentorial primitive neuroectodermal tumors (childhood), pituitary adenoma, plasma cell neoplasia, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell carcinoma (kidney cancer), renal pelvis and ureter transitional cell cancer, retinoblastoma, rhabdomyosarcoma (childhood), salivary gland cancer, sarcoma (Ewing family of tumors, Kaposi, soft tissue, uterine), Sezary syndrome, skin cancers (nonmelanoma, melanoma), skin carcinoma (Merkel cell), small cell lung cancer, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, squamous neck cancer with occult primary (metastatic), stomach cancer, supratentorial primitive neuroectodermal tumor (childhood), T-Cell lymphoma (cutaneous), testicular cancer, throat cancer, thymoma (childhood), thymoma and thymic carcinoma, thyroid cancer, thyroid cancer (childhood), transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor (gestational), unknown primary site (adult, childhood), ureter and renal pelvis transitional cell cancer, urethral cancer, uterine cancer (endometrial), uterine sarcoma, vaginal cancer, visual pathway and hypothalamic glioma (childhood), vulvar cancer, Waldenstrom macroglobulinemia, and Wilms tumor (childhood). In specific embodiments, the neoplasm or cancer is non-small cell lung carcinoma.

(c) administration

[01 15] In certain aspects, a pharmacologically effective amount of an antibody of the invention, including immunologically reactive fragments, may be administered to a subject. Administration is performed using standard effective techniques, including peripherally (i.e. not by administration into the central nervous system) or locally to the central nervous system. Peripheral administration includes but is not limited to intravenous, intraperitoneal, subcutaneous, pulmonary, transdermal, intramuscular, intranasal, buccal, sublingual, or suppository administration. Local administration, including directly into the central nervous system (CNS) includes but is not limited to via a lumbar, intraventricular or intraparenchymal catheter or using a surgically implanted controlled release formulation.

[01 16] Pharmaceutical compositions for effective administration are deliberately designed to be appropriate for the selected mode of administration, and pharmaceutically acceptable excipients such as compatible dispersing agents, buffers, surfactants, preservatives, solubilizing agents, isotonicity agents, stabilizing agents and the like are used as appropriate. Remington's Pharmaceutical Sciences, Mack

Publishing Co., Easton Pa., 16Ed ISBN: 0-912734-04-3, latest edition, incorporated herein by reference in its entirety, provides a compendium of formulation techniques as are generally known to practitioners. It may be particularly useful to alter the solubility characteristics of the antibodies useful in this discovery, making them more lipophilic, for example, by encapsulating them in liposomes or by blocking polar groups.

[01 17] Effective peripheral systemic delivery by intravenous or

intraperitoneal or subcutaneous injection is a preferred method of administration to a living patient. Suitable vehicles for such injections are straightforward. In addition, however, administration may also be effected through the mucosal membranes by means of nasal aerosols or suppositories. Suitable formulations for such modes of administration are well known and typically include surfactants that facilitate cross- membrane transfer. Such surfactants are often derived from steroids or are cationic lipids, such as N-[1 -(2,3-dioleoyl)propyl]-N,N,N-trimethyl ammonium chloride (DOTMA) or various compounds such as cholesterol hemisuccinate, phosphatidyl glycerols and the like.

[01 18] The concentration of antibody in formulations to be administered is an effective amount and ranges from as low as about 0.1 % by weight to as much as about 15 or about 20% by weight and will be selected primarily based on fluid volumes, viscosities, and so forth, in accordance with the particular mode of administration selected if desired. A typical composition for injection to a living patient could be made up to contain 1 mL sterile buffered water of phosphate buffered saline and about 1 -1000 mg of any one of or a combination of the humanized antibody of the present discovery. The formulation could be sterile filtered after making the formulation, or otherwise made microbiologically acceptable. A typical composition for intravenous infusion could have volumes between 1 -250 mL of fluid, such as sterile Ringer's solution, and 1 -100 mg per ml, or more in anti-GRP78 or anti-TIP-1 antibody concentration. Therapeutic agents of the discovery can be frozen or lyophilized for storage and reconstituted in a suitable sterile carrier prior to use. Lyophilization and reconstitution may lead to varying degrees of antibody activity loss (e.g. with conventional immune globulins, IgM antibodies tend to have greater activity loss than IgG antibodies). Dosages administered are effective dosages and may have to be adjusted to compensate. The pH of the formulations generally pharmaceutical grade quality, will be selected to balance antibody stability (chemical and physical) and comfort to the patient when administered. Generally, a pH between 4 and 8 is tolerated. Doses will vary from individual to individual based on size, weight, and other physiobiological characteristics of the individual receiving the successful administration.

[01 19] As used herein, the term "effective amount" means an amount of a substance such as a compound that leads to measurable and beneficial effects for the subject administered the substance, i.e., significant efficacy. The effective amount or dose of compound administered according to this discovery will be determined by the circumstances surrounding the case, including the compound administered, the route of administration, the status of the symptoms being treated and similar patient and administration situation considerations among other considerations.

[0120] In some embodiments, when the antibody is an anti-TIP-1 antibody labeled with 64 Cu, the dose administered may be about 0.01 , 0.02, 0.03, 0.04, 0.05 0.06, 0.07, 0.08, 0.09, 0.1 , 0.01 1 , 0.012, 0.013, 0.013, 0.014, 0.015, 0.016, 0.017, 0.018, 0.019, 0.02, 0.021 , 0.022, 0.023, 0.023, 0.024, 0.025, 0.026, 0.027, 0.028, 0.029, 0.03, 0.031 , 0.032, 0.033, 0.033, 0.034, 0.035, 0.036, 0.037, 0.038, 0.039, 0.04, 0.041 , 0.042, 0.043, 0.043, 0.044, 0.045, 0.046, 0.047, 0.048, 0.049, 0.05, 0.051 , 0.052, 0.053, 0.053, 0.054, 0.055, 0.056, 0.057, 0.058, 0.059, 0.06, 0.061 , 0.062, 0.063, 0.063, 0.064, 0.065, 0.066, 0.067, 0.068, 0.069, 0.07, 0.071 , 0.072, 0.073, 0.073, 0.074, 0.075, 0.076, 0.077, 0.078, 0.079, 0.08, 0.081 , 0.082, 0.083, 0.083, 0.084, 0.085, 0.086, 0.087, 0.088, 0.089, 0.09, 0.091 , 0.092, 0.093, 0.093, 0.094, 0.095, 0.096, 0.097, 0.098, 0.099, or about 0.1 rem/mCi.

[0121 ] In some embodiments, when the antibody is an anti-GRP78 labeled with 64 Cu, the dose administered may be about 0.01 , 0.02, 0.03, 0.04, 0.05 0.06, 0.07, 0.08, 0.09, 0.1 , 0.01 1 , 0.012, 0.013, 0.013, 0.014, 0.015, 0.016, 0.017, 0.018, 0.019, 0.02, 0.021 , 0.022, 0.023, 0.023, 0.024, 0.025, 0.026, 0.027, 0.028, 0.029, 0.03, 0.031 , 0.032, 0.033, 0.033, 0.034, 0.035, 0.036, 0.037, 0.038, 0.039, 0.04, 0.041 , 0.042, 0.043, 0.043, 0.044, 0.045, 0.046, 0.047, 0.048, 0.049, 0.05, 0.051 , 0.052, 0.053, 0.053, 0.054, 0.055, 0.056, 0.057, 0.058, 0.059, 0.06, 0.061 , 0.062, 0.063, 0.063, 0.064, 0.065, 0.066, 0.067, 0.068, 0.069, 0.07, 0.071 , 0.072, 0.073, 0.073, 0.074, 0.075, 0.076, 0.077, 0.078, 0.079, 0.08, 0.081 , 0.082, 0.083, 0.083, 0.084, 0.085, 0.086, 0.087, 0.088, 0.089, 0.09, 0.091 , 0.092, 0.093, 0.093, 0.094, 0.095, 0.096, 0.097, 0.098, 0.099, or about 0.1 rem/mCi.

[0122] The frequency of dosing may be daily or once, twice, three times or more per week or per month, as needed as to effectively treat the symptoms. The timing of administration of the treatment relative to the disease itself and duration of treatment will be determined by the circumstances surrounding the case. Treatment could begin immediately, such as at the site of the injury as administered by emergency medical personnel. Treatment could begin in a hospital or clinic itself, or at a later time after discharge from the hospital or after being seen in an outpatient clinic. Duration of treatment could range from a single dose administered on a one-time basis to a life-long course of therapeutic treatments.

[0123] Although the foregoing methods appear the most convenient and most appropriate and effective for administration of proteins such as antibodies, by suitable adaptation, other effective techniques for administration, such as intraventricular administration, transdermal administration and oral administration may be employed provided proper formulation is utilized herein.

[0124] In addition, it may be desirable to employ controlled release formulations using biodegradable films and matrices, or osmotic mini-pumps, or delivery systems based on dextran beads, alginate, or collagen.

[0125] Typical dosage levels can be determined and optimized using standard clinical techniques and will be dependent on the mode of administration.

(d) exposing to ionizing radiation - Radiotherapy

[0126] In an aspect, the method comprises exposing a target area of a subject where the presence of a tumor is suspected to ionizing radiation. Low doses of radiation can be used for selective targeting using the antibody compositions disclosed herein. In some embodiments, the dose of radiation comprises up to about 2 Gy ionizing radiation. Higher radiation doses can also be used, especially in the case of local radiation treatment as described hereinbelow.

[0127] Radiation can be localized to a tumor using conformal irradiation, brachytherapy, or stereotactic irradiation. The threshold dose for inductive changes can thereby be exceeded in the target tissue but avoided in surrounding normal tissues. A "target tissue" as used herein refers to an intended site for accumulation of an atiboedy following administration to a subject. For example, the methods disclosed herein can employ a target tissue comprising an irradiated tumor. A "control tissue" as used herein refers to a site suspected to substantially lack binding and/or accumulation of an administered antibody. For example, in accordance with the methods of the presently disclosed subject matter, a non-irradiated tumor and a non-cancerous tissue are control tissues. In some embodiments, doses of at least about 2 Gy ionizing radiation can be used, and in some embodiments a dose of about 10 Gy to about 20 Gy ionizing radiation can be used. For treatment of a subject having two or more tumors, local irradiation enables differential drug administration and/or dose at each of the two or more tumors. Alternatively, whole body irradiation can be used, as permitted by the low doses of radiation required for targeting of peptides disclosed herein. Radiotherapy methods suitable for use in the practice of the presently disclosed subject matter can be found in Leibel & Phillips, 1998, among other sources.

[0128] In an embodiment, the radiation treatment comprises administration of less than about 2 Gy ionizing radiation. In another embodiment, the radiation treatment comprises at least about 2 Gy ionizing radiation, in some embodiments about 2 Gy to about 3 Gy ionizing radiation, and in some embodiments about 2 Gy to about 6 Gy ionizing radiation. In other embodiments, radiation treatment comprises about 10 Gy to about 20 Gy ionizing radiation.

[0129] Administration of a composition to a subject can be performed by irradiating the tumor prior to, concurrent with, or subsequent to administration of a composition of the invention. Accordingly, the tumor is irradiated in some embodiments 0 hours to about 24 hours before administration of the composition, and in some embodiments about 4 hours to about 24 hours before administration of the composition.

DEFINITIONS

[0130] As used herein, "antibody" refers to an immunoglobulin derived molecule that specifically recognizes either GRP78 or TIP-1 . An antibody of the invention may be a full length antibody (IgM, IgG, IgA, IgE) or may be an antibody fragment (Fab, F(ab')2, scFv). An antibody may be chimeric or may be humanized.

[0131 ] As used herein, "CDR" means "complementary determining region." CDRs may also be referred to as hypervariable regions.

[0132] As used herein, "light chain" is the small polypeptide subunit of the antibody. A typical antibody comprises two light chains and two heavy chains.

[0133] As used herein, the "heavy chain" is the large polypeptide subunit of the antibody. The heavy chain of an antibody contains a series of immunoglobulin domains, with at least one variable domain and at least one constant domain.

[0134] "Humanized", as used herein, refers to the process where monoclonal antibodies are produced using recombinant DNA to create constructs capable of expression in human cell culture. Any known techniques for producing these constructs will work for purposes of the present invention. [0135] As used herein, "single chain variable fragments" or "scFv" or "scFvs", refer to fusion proteins of the variable regions of the heavy and light chains of immunoglobulins connected via a linker. In some embodiment, the linker is a peptide of about 10 to 25 amino acids.

[0136] A "therapeutic agent" for purposes of the present invention, refers to an agent that reduces tumor growth, any related cancer growth, or reduces the symptoms associated with cancerous cell growth. The therapeutic agent that is preferably conjugated to the antibody of the present invention is preferably a biologic, pharmaceutical or chemical agent. A non-limiting list of therapeutic agents that may be suitable for use in the present invention is described above.

[0137] As used herein, "imaging agent" refers to any agent that can be used to locate and produce an image of cancerous cell growth or tumors. A non-limiting list of imagining agents that may be suitable for use in the present invention is described above.

EXAMPLES

[0138] The following examples illustrate various iterations of the invention.

Materials and Methods

Tumor model

[0139] GL261 murine glioma cancer cell line was purchased from

American Type Culture Collection. D54 cells were similarly purchased commercially. Heterotopic tumor models were developed by s.c. inoculating cell suspensions (6 10 6 cells) into nude or C57/BI6 mice.

Near IR imaging

[0140] Tumor-bearing mice were treated with anywhere from one to three once-daily doses of 3 Gy XRT or sham XRT (three per group) and injected with peptide or antibody 3 hours after the last XRT treatment. Mice were then injected with an antibody to 78-kDa glucose regulated protein (GRP78) conjugated with AlexaFluor750 and tumors were removed 7 days after labeled antibody injection; polyclonal serum IgG antibody was used as a control. Near IR images were taken using the MS imaging system with an ICG filter setting at various time points after the injection.

Anti-TIP-1 and anti-GRP78 monoclonal antibody production and purification.

[0141 ] BALB/C mice were initially immunized with 50 mg of TIP-1 or

GRP78 antigen mixed with equivalent amounts of Titermax adjuvant (CytRx

Corporation) for each mouse. One month after initial immunization, mice were boosted with equivalent amounts of antigen without adjuvant two to three times at two week intervals. The mouse polyclonal antibody titer was evaluated by ELISA and Western blot methods. Mice exhibiting high immune response to either antigen were chosen as B cell donors. Spleens were removed and homogenized in RPMI 1640 culture medium free of serum and other additives. Spleen cells were combined with Sp2/O mouse myeloma (2 x 10 7 per spleen). Mixed cells were washed twice and centrifuged at 1200 rpm for 8 minutes at room temperature. Supernatant was removed, and the cell pellet was lightly agitated to loosen the cells. Approximately 1 ml PEG (polyethylene glycol 1500)

(Roche) was added to fuse the cells. The fused cells were washed once with plain medium and finally re-suspended in RPMI 1640 medium supplemented with 10% fetal bovine serum (Gemini Bioproducts, CA), L-glutamine, antibiotics and HAT (Sigma), plated into 24 well tissue culture plates and incubated at 37°C in a humidified CO2 incubator. Fifteen days after fusion, hybridoma culture supernatants were removed from individual wells and transferred to separate 96 well microtiter plates for ELISA, Western blot and antibody printing assays against TIP-1 antigen. Hybridomas that produced antibodies positive by immunoassay were chosen for sub-cloning by using limiting dilution. Resulting single-cell clones were retested by the aforementioned methods to detect antigen-positive monoclonal antibody-producing hybridomas. Positive hybridoma clones were transferred to individual flasks to expand cell number from one cell clone, and incubated with serum-free medium for antibody production. The antibodies produced by positive clones in serum-free medium were harvested twice a week for further monoclonal antibody purification. Filtered monoclonal antibody collected from serum-free medium was purified by protein A and protein G columns. The concentrated purified monoclonal antibody was assayed and stored at -20°C. Example 1. GRP78

[0142] A monoclonal antibody against GRP78 was created, and named 2D6F9. The antibody comprised a heavy chain variable region amino acid sequence of SEQ ID NO:6 (minus the leader sequence) and a light chain variable region amino acid sequence of SEQ ID NO:8 (minus the leader sequence) (Fig. 7). As shown in Fig. 1 , the antibody was an lgG1 isotype, and as shown in Fig. 2 was stable at -20°C for at least six months. Epitope mapping was performed to determine that 2D6F9 recognized SEQ ID NO. 1 , 2, 3, and 4 derived from human GRP78.

[0143] Fig. 8 depicts a graph showing the radiance emitted from the anti- GRP78 antibody 2B6F9 conjugated to Alexa Fluor 750 over time. The antibody 2D6F9 (conjugated to Alexa Fluor 750) recognized tumor cells from the GL261 tumor model that were exposed to either 3 separate 3Gy doses of radiation (Fig. 3) or a single dose of 3Gy radiation (Fig. 4), but did not recognize non-irradiated tumor tissue or normal tissue (either irradiated or non-irradiated). For comparison purposes a control nonspecific IgG antibody was used in Fig. 5. Additionally, 2D6F9 (conjugated to Alexa Fluor 750) recognized tumor cells from the D54 tumor model that were exposed to a single dose of 3Gy radiation (Fig. 9),

[0144] The distribution of 64 Cu-anti-GRP78 antibody 2D6F9 was

measured. Mice were administered 50 g of 64 Cu-anti-GRP78 antibody 2D6F9 and imaged at 24 (Fig. 6A,B) and 48 hours (Fig. 6C). The left hindlimb was irradiated with 3Gy and the right hindlimb was not irradiated. Animal biodistribution data was obtained. Animals were euthanized and organs were harvested at 24 and 48 hours post injection. The following organs were harvested, weighed and counted for radio-activity in a gamma counter: blood, lungs, liver, spleen, kidneys, muscle, heart, bone, pancreas, tumor(r), tumor(l), stomach, small intestines, and large intestines. The biodistribution data are presented in Fig. 6D.

Example 2. TIP-1

[0145] At least three monoclonal antibodies against TIP-1 were created, and named 1A6H14, 3A415, and 2C6F3. Epitope mapping was performed to determine that 1A6H14 recognized SEQ ID NO: 9, 10, and 1 1 derived from human TIP-1 , that 3A415 recognized SEQ ID NO: 12, 13, and 14 derived from human TIP-1 , and that 2C6F3 recognized SEQ ID NO:21 derived from human TIP-1 (see Table C).

[0146] The antibodies were shown to recognize irradiated tumor cells compared to non-irradiated tumor tissue or normal tissue (either irradiated or non- irradiated). Specifically, anti-TIP-1 monoclonal antibody, 2C6F3, showed binding by flow cytometry to human glioblastoma D54 cells (WHO grade IV) (Fig. 23). 2C6F3

(conjugated to Alexa Fluor 750) recognized tumor cells from the GL261 tumor model that were exposed to radiation (Fig. 24), but did not recognize non-irradiated tumor tissue or normal tissue (either irradiated or non-irradiated).

[0147] CT/SPECT imaging with 111 ln-DTPA was performed with the anti- TIP-1 2C6F3 monoclonal antibody. LLC (Lewis lung carcinoma) tumors were grown to approximately 1 cm 3 in C57/BL mice. Two groups of mice each containing 3 mice were utilized. Group 1 mice were irradiated with 3Gy x 3 over 24 hours. Group 2 mice were sham non-irradiated. 111 In was labeled with DTPA-2C6F3 TIP-1 antibody with a final specific activity of 0.7 pCi/ g. Fig. 25 shows the labeling of DTPA chelator on 2C6F3. Mice were subjected to tail vein injection of -250 Ci in 150 μΙ volume for SPECT imaging. Mice were imaged using nano-SPECT scan at 48 hours (Fig. 26) and 72 hours (Fig. 27) post IV injection. Fig. 26 and Fig. 27 show the distribution of 111 ln-DTPA- 2C6F3 anti-TIP-1 antibody.

Example 3. Dosimetry of labeled anti-GRP78 2D6F9 antibody.

Biodistribution in healthy mice

[0148] Human radiation dosimetry estimates were calculated from animal biodistribution data obtained by standard method of organ dissection and using the standard MIRD methodology. Animal biodistribution data was obtained using 30 C57 mature male mice injected with 16 Ci/100 L of 64 Cu-DOTA-2D6F9 antibody divided in 6 groups of 5 animals. The average animal mouse weight was 25.0 g. Animals were euthanized and organs were harvested in groups of five at the following time points: 1 , 2, 6, 12, 24 and 48 hours post injection. The following organs were harvested, weighed and counted for radio-activity in a gamma counter: blood, lungs, liver, spleen, kidneys, bladder, muscle, fat, heart, brain, bone, red marrow, testes, adrenals, thyroid, pancreas, stomach, small intestines, and upper and lower large intestines. The animals were maintained in metabolic cages where urine and feces excretion were collected, weighed and counted for radio-activity. The biodistribution data are presented in Table 1.

Residence times

[0149] The residence time represents the cumulative presence time of radioactivity in any given organ and is expressed in units of time (either seconds, minutes or hours). Multiplying these values by unit of radioactivity leads to the number of radio-active decays occurring in any given organ and is therefore proportional to radiation dose. Using the above animal biodistribution data, the organ residence times (in hour) for each harvested organ were calculated by numerical integration of the time activity data expressed in percent injected dose per gram of tissue. The following initial organ activity content immediately after injection was assumed to be: 5.9% in the lungs, 2.9% in the liver, 1 .5% in the spleen and kidneys, 3.7% in the bone, and 72% in the blood. It was assumed that no biological excretion occurred beyond the last measured time point and that radio-activity only decreased due to physical decay. The animal organ residence times were then scaled to human organ weight by the "relative organ mass scaling" method. Organ residence times are presented in Table 2. The cumulative urine and feces activity (in percent injected) were plotted as a function of time and an uptake function was fitted to the data (F(t) = AO (1 -exp(-A1 t) ), see Fig. 10A and B). Analytical integration, accounting for radio-active decay, yielded to an excreted residence time of 1 .62 hr in the feces. Analytical integration of the excreted urine data resulted in a urine residence times of 7.75 hr. The filling fraction of 73% and the filling half-life of 9.23 hr (=ln(2)/0.075hr-1 ) was used in the MIRD voiding model along with a voiding interval of 2hr to yield a bladder residence time of 0.425 hr. The amount of excreted activity is therefore equal to 9.4 hr. The remainder of the body residence time was calculated from the maximum theoretical residence time minus the excreted residence time minus the sum of all residence times measured in the organ above at the exception of blood and fat. This resulted in a residence time associated to the remainder of the body of 0.78 hr. The blood and bone mass was assumed to be 8% and 15% of the human body mass, respectively. The errors bars on the measured residence times were determined from the standard deviation of the biodistribution data points. The largest residence times are observed in the liver, muscle and bone. The residence time in the blood is also very high with a value of 4.58 hr and is dependent on the relative slow clearance of the activity from the blood (Fig. 11 ). The blood clearance was observed to clearing component with a biological half-life of 7.3 hr using a mono- exponential model.

Table 2. Organ residence times extrapolated to

Radiation dosimetry

[0150] The residence times of Table 2 were entered in the program OLINDA EXM for 64 Cu and using the standard MIRD adult male model. The following additional assumptions were made: 10% of the blood residence time was assigned to the heart left ventricle; the bone activity was assigned in equal part to the cortical and trabecular bone source organs. Organ radiation dose estimates for the adult male model are presented in Table 3. The error bars on the dose estimates were assumed to be in proportion of the organ residence times uncertainties.

[0151 ] Although the calculated doses are for the human male model, dose to female organs (breasts, uterus and ovaries) are also provided in italic. The radiation doses above include contribution from beta (both minus and plus) and gamma rays emitted from 64 Cu, and include contribution from activity within one organ to itself and from neighboring organs. Due to the nature and energy of the beta particles, the self- organ dose contribution from the beta particles dominates the dose contributions. The largest radiation dose is observed in the liver and urinary bladder wall with values of 0.295 and 0.321 rad per mCi injected. The effective dose is calculated at 0.083 rem/mCi. Based on RDRC limit of 5 rem to any organ, this indicates a 16 mCi maximum injection. Example 4. Dosimetry of labeled anti-TIP1 2C6F3 antibody.

Biodistribution in Healthy Mice

[0152] Human radiation dosimetry estimates were calculated from animal biodistribution data obtained by standard method of organ dissection and using the standard MIRD methodology. Animal biodistribution data was obtained using 30 C57 mature male mice injected with 24 μΟί/100μΙ_ of [ 64 Cu]-DOTA-2C6F3 antibody divided in 6 groups of 5 animals. The average animal mouse weight was 23.3 g. Animals were euthanized and organs were harvested in groups of five at the following time points: 1 , 2, 6, 12, 24 and 48 hours post injection. The following organs were harvested, weighed and counted for radio-activity in a gamma counter: blood, lungs, liver, spleen, kidneys, bladder, muscle, fat, heart, brain, bone, red marrow, testes, adrenals, thyroid, pancreas, stomach, small intestines, upper and lower large intestines. The animals were

maintained in metabolic cages where urine and feces excretion were collected, weighed and counted for radioactivity. The biodistribution data are presented in Table 4.

Small

1 .43 ± 0.28 1 .17 ± 0.24 1 .25 ± 0.12 1 .24 ± 0.16 1 .20 ± 0.22 1 .79 ± 0.39 Intestines

Upper L.

1 .21 ± 0.29 1 .01 ± 0.1 1 1 .10 ± 0.18 1 .16 ± 0.20 1 .21 ± 0.15 1 .76 ± 0.27 Intestines

Lower L.

0.67 ± 0.10 1 .22 ± 0.09 1 .25 ± 0.17 1 .38 ± 0.27 1 .51 ± 0.31 1 .98 ± 0.34 Intestines

Residence Times

[0153] The residence times represent the cumulative presence time of radioactivity in any given organ and are expressed in units of second or hours (hr). Multiplying these values by unit of radioactivity leads to the number of radioactive decays occurring in any given organ and is therefore proportional to radiation dose. Using the above animal biodistribution data, the organ residence times (in hours) for each harvested organ were calculated by numerical integration of the time activity data expressed in percent injected dose per gram of tissue. The following initial organ activity content immediately after injection was assumed to be: 5.9% in the lungs, 2.9% in the liver, 1 .5% in the spleen and kidneys, 3.7% in the bone, and 72% in the blood. It was assumed that no biological excretion occur beyond the last measured time point and that radioactivity only decreased due to physical decay. The animal organ residence times were then scaled to human organ weight by the "relative organ mass scaling" method. Organ residence times are presented in Table 5. The cumulative urine and feces activity (in percent injected) were plotted as a function of time and an uptake function was fitted to the data (F(t) = AO (1 -exp(-A1 t) ), see Fig. 12A and B). Analytical integration, accounting for radioactive decay, yielded to an excreted residence time of 1 .62 hr in the feces. Analytical integration of the excreted urine data resulted in a urine residence times of 9.36 hr. The filling fraction of 65% and the filling half-life of 3.47 hr (=ln(2)/0.020hr-1 ) was used in the MIRD voiding model along with a voiding interval of 2hr to yield a bladder residence time of 0.533 hr, and a modeled amount of activity excreted in urine of 8.83 hr. The amount of excreted activity is therefore equal to 10.5 hr. The remainder of the body residence time was calculated from the maximum theoretical residence time minus the excreted residence time minus the sum of all residence times measured in the organ above at the exception of blood and fat. This resulted in a residence time associated to the remainder of the body of 0.27 hr. The blood and bone mass was assumed to be 8% and 15% of the human body mass respectively. The errors bars on the measured residence times were determined from the standard deviation of the biodistribution data points. The largest residence times are observed in the liver, muscle and bone. The residence time in the blood is also very high with a value of 2.64 hr and is dependent on the relative slow clearance of the activity from the blood. The blood clearance half-life was measured at 36 hr using a mono-exponential model with an initial concentration of 1 1 .3% of the injected dose.

Radiation Dosimetry

[0154] The residence times of Table 5 were entered in the program OLINDA EXM for 64 Cu and using the standard MIRD adult male model. The following additional assumptions were made: 10% of the blood residence time was assigned to the heart left ventricle; the bone activity was assigned in equal part to the cortical and trabecular bone source organs. The error bars on the dose estimates were assumed to be in proportion of the organ residence times uncertainties.

[0155] Although the calculated doses are for the human male model, dose to female organs (breasts, uterus and ovaries) are also provided in italics. The radiation doses above include contribution from beta (both minus and plus) and gamma rays emitted from 64 Cu, and include contribution from activity within one organ to itself and from neighboring organs. Due to the nature and energy of the beta particles, the self- organ dose contribution from the beta particles dominates the dose contributions. The largest radiation dose is observed in the liver and spleen with values of 0.62 and 0.6 rad per mCi injected. The effective dose is calculated at 0.088 rem/mCi.

Example 5. GRP78 surface expression increases on irradiated NSCLC.

[0156] Lung cancer cell membrane preparations were studied on Western immunoblot. Antibodies to GRP78 showed an increase in this protein on the cell membrane in response to 3 Gy of irradiation in lung cancer cells (Fig. 13). GRP78 not only remained within the membrane preparations, but was also secreted into the medium of irradiated NSCLC cells. Fig. 14 shows that HUVEC cells irradiated together with lung cancer cells show an increase in GRP78 surface staining. Radiation induction of GRP78 on the surface of endothelial cells was not accomplished when endothelial cells were irradiated alone.

Example 6. Anti-GRP78 antibodies reduce NSCLC cell survival and enhance cytotoxicity of radiotherapy.

[0157] To determine whether anti-GRP78 monoclonal antibodies enhance the efficacy of radiotherapy in NSCLC, the colony forming assay was utilized (Fig. 15). 2D6F9 antibody, 5 g/ml was added to lung cancer cells at 6 hours after plating. Cells were then treated with 2 Gy or sham irradiation (0 Gy). The antibody alone reduced plating efficiency to 49% and reduced survival to 6% after 2 Gy irradiation. In

comparison, 2Gy alone or with control antibody reduced survival by less than 20% (p<0.01 ). Example 7. Anti-GRP78 antibodies achieve specific binding to NSCLC in the mouse models.

[0158] To determine whether antibodies against the C- and N-terminal domains of GRP78 achieve cancer specific binding, lung cancer tumors in mice (Fig. 16) were studied. Lung cancer tumors were grown both within the hind limb of mice. Antibodies were labeled with ALX750 and imaged by NIR imaging. Each of the antibodies 1 D6B2, 2D6F3 and scFv K13 achieve specific binding to the irradiated NSCLC. The kinetic curves show that the antibodies rapidly bind to the irradiated tumor as they are cleared from the circulation through enterohepatic clearance. Binding within tumors persist for over one week for IgG antibodies and 3 days for scFv. All antibodies also bind specifically to human NSCLCs H460 and LLC in irradiated nude mice.

[0159] TE1 1 anti TIP-1 scFv antibody and a DOTA-conjugated TE1 1 anti- TIP-1 scFv antibody were generated. The sequence of two TE1 1 clones are shown in Fig. 22.

[0160] The TE1 1 antibody was tested using ELISA (Fig. 17 and 18). In short, ELISA plates were coated with TIP-1 antigen at 10 μg ml in PBS, washed 3 times, and blocked with 2% BSA. TE1 1 or DOTA-conjugated TE1 1 in PBS was added and, after incubation, washed 3 x before adding anti-myc antibody at 5 μg ml, followed by goat anti-mouse IgG-HRP, washed 3 x before adding ABTS substrate and reading at 405 nm. The reaction volume in each step is 50 μΙ / well.

[0161 ] The anti-TIP-1 scFv antibody TE1 1 (Fig. 19) recognized tumor cells that were exposed to radiation, but a control scFv antibody did not (Fig. 20). See also Fig. 21.

Example 8. Humanizing anti-TIP1 antibody 2C6F3

[0162] The heavy chain (HC) and light chain (LC) sequences of 2C6F3, a murine antibody to TIP-1 , were synthesized. The HC nucleic acid sequence is SEQ ID NO:24 and the amino acid sequence is SEQ ID NO:25 (Table C). The LC nucleic acid sequence is SEQ ID NO:26 and the amino acid sequence is SEQ ID NO:27 (Table C). According to the sequences, two plasmids for expressing murine HC and LC separately were constructed. The HC and LC genes were cloned into the plasmid under the control of PCMV promoter. The endotoxin free plasmids were isolated and transiently

transfected into 293 cells. The antibody present in the supernatant was then tested by ELISA. Results showed the recombinant murine 2C6F3 antibody (2C6F3 rmlgG) binds to antigen as well as the original 2C6F3 antibody (2C6F3 mAb) (Table 6). The CDRs of the murine antibody are then cloned to a selected matching human lgG1 backbone.