Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR TREATMENT OF CANCERS
Document Type and Number:
WIPO Patent Application WO/2018/067819
Kind Code:
A1
Abstract:
The present application discloses a combination therapy for treatment of a cancer or a metastasis thereof. Also disclosed are methods of biomarker assessment prior to, during, and after treatment of a subject. Also disclosed are methods of identifying one or more baseline biomarkers levels in a subject and enrolling them in a clinical trial based on the one or more baseline biomarkers levels. Also disclosed are methods of identifying one or more baseline biomarkers levels in a subject and predicting whether or not the subject would be responsive to treatment with an anti-endoglin antibody and a VEGF inhibitor or a VEGF receptor inhibitor.

Inventors:
THEUER CHARLES (US)
Application Number:
PCT/US2017/055345
Publication Date:
April 12, 2018
Filing Date:
October 05, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
TRACON PHARMACEUTICALS INC (US)
International Classes:
A61K31/282; A61K31/337; A61K31/4045; A61K31/44; A61K31/69; A61K31/704
Foreign References:
US20160009811A12016-01-14
US20110142820A12011-06-16
US20120244147A12012-09-27
Other References:
ROSEN ET AL.: "Endoglin for Targeted Cancer Treatment", CURRENT ONCOLOGY REPORTS, vol. 16, no. 365, 1 February 2014 (2014-02-01), pages 1 - 9, XP055440440
CHOUEIRI ET AL.: "A Phase 1b Dose-Escalation Study of TRC105 (Anti-Endoglin Antibody) in Combination with Axitinib in Patients with Metastatic Renal Cell Carcinoma (mRCC)", POSTER, 14TH INTERNATIONAL KIDNEY CANCER SYMPOSIUM, 30 November 2015 (2015-11-30), pages 1, XP055500474, Retrieved from the Internet [retrieved on 20171120]
LIU ET AL.: "Modulation of Circulating Protein Biomarkers Following TRC105 (Anti-Endoglin Antibody) Treatment in Patients with Advanced Cancer", CANCER MEDICINE, vol. 3, 1 June 2014 (2014-06-01), pages 580 - 591, XP055500478, Retrieved from the Internet
Attorney, Agent or Firm:
JAMROZ, Megan E. (US)
Download PDF:
Claims:
CLAIMS

What is Claimed:

1. A method of treating a cancer or a metastasis thereof in a subject in need thereof, comprising

detecting whether one or more biomarkers are present in a sample from the subject by contacting the sample with one or more antibodies that bind to the one or more biomarkers and detecting binding between the one or more biomarkers and the one or more antibodies;

comparing a baseline value of the one or more biomarkers of the subject to a baseline value of a healthy subject; and

administering to the subject one or more doses of a pharmaceutical composition comprising a VEGF inhibitor and one or more doses of a pharmaceutical composition comprising an anti-endoglin antibody when the baseline value of the one or more biomarkers in the subject to be treated are above or below a median or a mean level in a subject with a cancer or a metastasis thereof.

2. A method of treating a cancer or a metastasis thereof in a subject in need thereof, comprising

detecting whether one or more biomarkers are present in a sample from the subject; diagnosing the subject with a cancer or metastasis when the presence of the one or more biomarkers in the sample are detected; and

administering to the subj ect one or more doses of a pharmaceutical composition comprising a VEGF inhibitor and one or more doses of a pharmaceutical composition comprising an anti-endoglin antibody when a baseline value of the one or more biomarkers in the subject to be treated is above or below a median or a mean level in a subject with a cancer or a metastasis thereof.

3. The method of claim 1 or 2, wherein the cancer or metastasis thereof is a carcinoma or a sarcoma.

4. The method of claim 3, comprising the carcinoma, wherein the carcinoma is a renal cell carcinoma, a hepatocellular carcinoma or a choriocarcinoma.

5. The method of claim 4, comprising the renal cell carcinoma, wherein the renal cell carcinoma is an advanced renal cell carcinoma.

6. The method of claim 4, comprising the renal cell carcinoma, wherein the renal cell carcinoma is a metastatic renal cell carcinoma.

7. The method of claim 1 or 2, wherein the cancer or metastasis thereof is a soft tissue sarcoma.

8. The method of claim 7, wherein the brain cancer is a glioblastoma, meningioma, an astrocytoma, or a malignant medulloblastoma.

9. The method of claim 1 or 2, wherein the cancer is a lung cancer or a metastasis thereof.

10. The method of claim 9, wherein the lung cancer or metastasis thereof is a non-small cell lung cancer, a lung carcinoid tumor, or a small cell lung cancer.

11. The method of claim 1 or 2, wherein the cancer or metastasis thereof is a breast cancer.

12. The method of claim 11, wherein the breast cancer is a Luminal A, a Luminal B, a Luminal B-like, a Triple negative or a HER2 type breast cancer.

13. The method of any one of claims 1-12, wherein the pharmaceutical composition comprising the VEGF inhibitor is administered orally, subcutaneously, intravenously, or in an implant.

14. The method of any one of claims 1-13, where the VEGF inhibitor is axitinib (N- methyl -2-[3-((i!r)-2-pyridin-2-yl -vinyl)- lH-indazol-6-ylsulfanyl]-benzamide).

15. The method of any one of claims 1-13, where the VEGF inhibitor is pazopanib (5- [[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2-methyl- b enzenesulfonami de) .

16. The method of any one of claims 1-12, wherein the pharmaceutical composition comprising the anti-endoglin antibody is administered subcutaneously, intravenously, or in an implant.

17. The method of any one of claims 1-16, wherein the pharmaceutical composition comprising the anti-endoglin antibody and the pharmaceutical composition comprising the VEGF inhibitor are administered simultaneously.

18. The method of any one of claims 1-16, wherein the pharmaceutical composition comprising the anti-endoglin antibody and the pharmaceutical composition comprising the VEGF inhibitor are administered sequentially.

19. The method of any one of claims 1-16, wherein the pharmaceutical composition comprising the anti-endoglin antibody and the pharmaceutical composition comprising the VEGF inhibitor are administered separately.

20. The method of any one of claims 1-19, wherein the one or more biomarkers is selected from the group consisting of angiogenin 2 (ANG-2), placental growth factor (PIGF), bone morphogenetic protein 9 (BMP-9), intercellular adhesion molecule 1 (ICAM-1), basic fibroblast growth factor (bFGF), interleukin 6 (IL-6), hepatocyte growth factor (HGF), transforming growth factor beta 1 (TGF-βΙ), osteopontin (OPN), platelet derived growth factor AA (PDGF-AA), TGF- β2, stromal cell-derived factor 1 (SDF-1), PDGF-BB, transforming growth factor beta receptor 3 (TGF -R3), tissue inhibitor of metalloproteinase-1 (TIMP-1), Thrombospondin 2 (TSP-2), vascular cell adhesion molecule 1 (VCAM-1) and a combination thereof.

21. The method of claim 20, wherein the biomarker is ANG-2.

22. The method of claim 20, wherein the biomarker is osteopontin.

23. The method of claim 20, wherein the biomarker is ICAM-1.

24. The method of claim 20, wherein the biomarker is TSP-2.

25. The method of claim 20, wherein the biomarker is TGFP-R3.

26. The method of any one of claims 20-25, comprising detecting one biomarker, two biomarkers, three biomarkers, four biomarkers, five biomarkers, six biomarkers, seven biomarkers, eight biomarkers, nine biomarkers, ten biomarkers or more.

27. The method of any one of claims 20-25, comprising detecting one or more biomarkers, wherein the one or more biomarkers are higher than a median or a mean level in a subject with a cancer or metastasis thereof.

28. The method of any one of claims 20-25, comprising detecting one or more biomarkers, wherein the one or more biomarkers are lower than a median or a mean level a subject with a cancer or metastasis thereof.

29. The method of any one of claims 20-25, comprising detecting two or more biomarkers, wherein at least one of the biomarkers is lower than a median or a mean level a subject with a cancer or metastasis thereof and at least one of the biomarkers is higher than a median or mean level a subject with a cancer or metastasis thereof.

30. The method of any one of claims 1-19, comprising (a) detecting one or more biomarkers selected from the group consisting of vascular endothelial growth factor A (VEGF- A), Endoglin, VEGF-D, vascular endothelial growth factor receptor 1 (VEGF-R1), VEGF-R2, VEGF- R3, and a combination thereof; and (b) detecting one or more biomarkers selected from the group consisting of ANG-2, PIGF, BMP -9, ICAM-1, bFGF, IL-6, HGF, TGF-βΙ, osteopontin (OPN), PDGF-AA, TGF-P2, SDF-1, PDGF-BB, TGFp-R3, TIMP-1, TSP-2, VCAM-1 and a combination thereof.

31. The method of claim 30, wherein the one or more biomarkers of (a) is VEGF-R2.

32. The method of claim 30 or 31, comprising detecting two biomarkers, three biomarkers, four biomarkers, five biomarkers, six biomarkers, seven biomarkers, eight biomarkers, nine biomarkers ten biomarkers or more.

33. The method of claim 30 or 31, comprising detecting two or more biomarkers, wherein the two or more biomarkers are higher than a median or a mean level in a subject with a cancer or a metastasis thereof.

34. The method of claim 30 or 31, comprising detecting two or more biomarkers, wherein the two or more biomarkers are lower than a median or a mean level in a subject with a cancer or a metastasis thereof.

35. The method of claim 30 or 31, comprising detecting two or more biomarkers, wherein at least one of the biomarkers is lower than a median or a mean level in a subject with a cancer or a metastasis thereof and at least one of the biomarkers is higher than a median or a mean level in a subject with a cancer or a metastasis thereof.

36. The method of claim 20 or 30, comprising detecting TGFP-R3, wherein a subject having a plasma concentration of TGF -R3 above a median or a mean level in a subject with a cancer or a metastasis thereof is selectively enrolled into a trial of a VEGF inhibitor and an anti- endoglin antibody.

37. The method of claim 20 or 30, comprising detecting osteopontin, wherein a subject having a plasma concentration of osteopontin below a median or a mean level in a subject with a cancer or a metastasis thereof at baseline is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

38. The method of claim 20 or 30, comprising detecting ICAM-1, wherein a subject having a plasma concentration of ICAM-1 below a median or mean level in a subject with a cancer or a metastasis thereof is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

39. The method of claim 20 or 30, comprising detecting TSP-2, wherein a subject having a plasma concentration of TSP-2 below a median or mean level in a subject with a cancer or a metastasis thereof at baseline is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

40. The method of claim 30, comprising detecting ANG-2, VEGF-R2, TGFp-R3, and osteopontin, wherein a subject having (i) a plasma concentration above a median or mean level in a subject with a cancer or a metastasis thereof of ANG-2, VEGF-R2, or TGFP-R3, or (ii) a plasma concentration of osteopontin below a median or mean level in a subject with a cancer or a metastasis thereof; or (iii) a combination of any of these biomarkers is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

41. The method of claim 20, wherein the cancer or metastasis thereof is a renal cell carcinoma, and wherein the range of TGF beta-R3 is from about 10 to about 250 ng/ml and the range of OPN is from about 0.1 to about 75 ng/ml

42. The method of claim 41, wherein the cancer or metastasis thereof is a renal cell carcinoma, and wherein the range of TGF beta-R3 is from about 115 to about 238 ng/ml and the range of OPN is from about 0 2 to about 59 ng/ml.

43. The method of claim 41 or 42, the median level of TGF beta-R3 is about 182 ng/mL and the medium level of OPN is about 1.2 μg/ml.

44. The method of claim 20, comprising detecting ICAM-1 and TSP-2, wherein a subject having a plasma concentration at baseline that is below the median or the mean level of ICAM-1 or TSP-2 is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

45. The method of claim 44, wherein the subject has a soft tissue sarcoma.

46. The method of claim 44 or 45, wherein the median level of ICAM-1 is from about 250 to about 2000 ng/ml and the range of TSP-2 is from about 25 to about 400 ng/ml.

47. The method of claim 44 or 45, wherein the median level of ICAM-1 is from about 293 to about 1 154 ng/ml and the range of TSP-2 is from about 58 to about 281 ng/ml.

48. The method of claim 44 or 45, wherein the median level of ICAM-1 is about 658 ng/mL and the medium level of TSP-2 is about 82 ng/ml.

49. The method of any of the preceding claims, wherein treatment reduces a size of a cancer or a metastasis thereof by about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, or more compared to administration of a placebo or compared to baseline.

50. The method of any one of claims 1-49, wherein detecting comprises an enzyme assay.

51. The method of claim 50, wherein the enzyme assay comprises an Enzyme Linked Immunosorbent Assay (ELISA).

52. The method of any one of claims 1-51, wherein the sample is a biofluid sample.

53. The method of claim 52, wherein the biofluid sample is blood, serum, plasma, urine, fluid from a tissue sample, fluid from a biopsy sample, or a combination thereof.

54. A pharmaceutical composition, comprising an anti-endoglin antibody, for use in combination therapy to treat a subj ect selected by a method of any one of claims 1 -53, wherein the anti-endoglin antibody is to be administered with a VEGF or a VEGF receptor inhibitor.

Description:
COMPOSITIONS AND METHODS FOR TREATMENT OF CANCERS

CROSS-REFERENCE

[0001] This application claims the benefit of U.S. Provisional Application No. 62/405,059, filed October 6, 2016, which application is incorporated herein by reference in its entirety.

BACKGROUND OF THE INVENTION

[0002] Endoglin, also known as, inter alia, CD105 or edg-1, is a type I homodimeric membrane glycoprotein which is expressed at high levels in proliferating vascular endothelial cells. However, there is some expression of endoglin by the vascular endothelium of normal tissues. Human endoglin is known to specifically bind transforming growth factor-β (TGF-β), and the deduced amino acid sequence of endoglin has strong homology to β-glycan, a type of TGF-β receptor.

SUMMARY OF THE INVENTION

[0003] Provided herein is a method of treating a cancer or a metastasis thereof in a subject in need thereof, comprising detecting whether one or more biomarkers are present in a sample from the subject by contacting the sample with one or more antibodies that bind to the one or more biomarkers and detecting binding between the one or more biomarkers and the one or more antibodies; comparing a baseline value of the one or more biomarkers of the subject to a baseline value of a healthy subject; and administering to the subject one or more doses of a pharmaceutical composition that comprises a vascular endothelial growth factor (VEGF) inhibitor or a VEGF receptor (VEGFR) inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody when the baseline value of the one or more biomarkers in the subject to be treated are above or below a median or mean level in a subject with the cancer or a metastasis thereof.

[0004] Provided herein is a method of selecting a subject for treatment for a cancer or a metastasis thereof, comprising: detecting whether one or more biomarkers are present in a sample from the subject by contacting the sample with one or more antibodies that bind to the one or more biomarkers and detecting binding between the one or more biomarkers and the one or more antibodies; comparing a baseline value of the one or more biomarkers of the subject to a baseline value of a healthy subject; and selecting the subject for treatment with one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor or a VEGFR inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody when the baseline value of the one or more biomarkers in the subject to be treated are above or below a median or mean level in the subject with the cancer or a metastasis thereof. [0005] Also provided herein is a method of treating a cancer or a metastasis thereof in a subject in need thereof, comprising detecting whether one or more biomarkers are present in a sample from the subject diagnosing the subject with a cancer or metastasis when the presence of the one or more biomarkers in the sample are detected; and administering to the subject one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor or a VEGFR inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody when a baseline value of the one or more biomarkers in the subject to be treated is above a median or mean level in a subject with the cancer or a metastasis thereof.

[0006] Provided herein is a method of selecting a subject for treatment for a cancer or a metastasis thereof, comprising: detecting whether one or more biomarkers are present in a sample from the subject, wherein detecting comprises contacting a sample from the subject with one or more one or more antibodies that bind to the one or more biomarkers and detecting binding between the one or more biomarkers and the one or more antibodies; and selecting the subject for treatment with one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor or a VEGFR inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody when the baseline value of the one or more biomarkers in the subject to be treated are above or below a median or mean level in the subj ect with the cancer or a metastasis thereof.

[0007] Detection in such methods can be an enzyme assay including, but not limited to, an Enzyme Linked Immunosorbent Assay (ELISA), a competitive ELISA, fluorescent activated cell sorter (FACS), etc.

[0008] Detection can occur prior to treatment, during treatment, or after completion of treatment at one or more time points.

[0009] The methods described herein can also be used, in some instances, to determine whether treatment should be continued, discontinued, increased, or decreased.

[0010] A cancer to be treated includes, but is not limited to, a breast cancer, a lung cancer, a brain cancer, a sarcoma, a carcinoma, or a metastasis of any of such cancers.

[0011] A carcinoma can be, for example, a renal cell carcinoma which can be, in some instances, an advanced renal cell carcinoma. In some instances, the carcinoma is a renal cell carcinoma which can be, in some instances, a metastatic renal cell carcinoma. In some instances, the carcinoma is a hepatocellular carcinoma or a choriocarcinoma.

[0012] In some instances, the cancer or metastasis thereof is a brain cancer such as, for example, a glioblastoma multiforme (GBM).

[0013] In some instances, the cancer or metastasis thereof is a breast cancer such as, for example, a Luminal A, a Luminal B, a Luminal B-like, a Triple negative or a HER2 type breast cancer. [0014] It will be understood that a subject can be treated for a length of time sufficient to prolong the life expectancy of the subject; or to partially or completely treat the cancer or metastasis thereof. For example, a subject can be administered treatment for about 1, about 2, about 3, about 4, about 5, about 6, about 7 or more days. A subject can be administered treatment for about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12 or more weeks. A subject can be administered treatment for about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12 or more months. A subject can be administered treatment for about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12 or more years.

[0015] The days, weeks, months or years of treatment can be consecutive. Alternatively, the days, weeks, months or years of treatment may not be consecutive. For example, if the subject enters remission, then treatment can be discontinued; if the subject shows presence of one or more abnormal biomarkers after being in remission, treatment can be reinstated. In other instances, a subject to be treated with the recited methods may have received surgery or treatment with a different anti-tumor agent.

[0016] The pharmaceutical composition comprising the anti-endoglin antibody and the

pharmaceutical composition comprising the VEGF inhibitor or the VEGFR inhibitor can be administered simultaneously. The pharmaceutical composition comprising the anti-endoglin antibody and the pharmaceutical composition comprising the VEGF inhibitor or the VEGFR inhibitor can be administered sequentially. The pharmaceutical composition comprising the anti- endoglin antibody and the pharmaceutical composition comprising the VEGF inhibitor or the VEGFR inhibitor can be administered separately.

[0017] In the described methods, a VEGF inhibitor or a VEGFR inhibitor can be, for example, Axitinib, Pazopanib, Bevacizumab (AVASTIN®), ranibizumab (LUCENTIS®), aflibercept (VEGF-Trap; EYLEA®), sunitinib (SUTENT®), brivanib (BMS-582664), sorafenib

( EXAVAR®), pegaptanib (MACUGEN®), and SU5416.

[0018] In the described methods, a VEGF inhibitor to be administered includes, but is not limited to, axitinib (N-methyl -2-[3-((7i)-2-pyridin-2-yl -vinyl)- lH-indazol-6-ylsulfanyl]-benzamide).

Axitinib can be administered by any appropriate means including, for example, orally,

subcutaneously, intravenously, or in an implant.

[0019] In the described methods, a VEGF inhibitor to be administered includes, but is not limited to, pazopanib (5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimid inyl]amino]-2- methyl-benzenesulfonamide). Pazopanib can be administered by any appropriate means including, for example, orally, subcutaneously, intravenously, or in an implant. [0020] In the described methods, an anti-endoglin antibody to be administered to a subject can be an isolated antibody that is, for example, a monoclonal antibody, a humanized antibody, a deimmunized antibody, a humanized and deimmunized antibody, a mouse-human chimeric antibody, a grafted antibody, a bi-specific antibody, a multi-specific antibody or a human antibody. The anti-endoglin antibody can be formulated in a pharmaceutical composition with one or more pharmaceutically carriers or excipients.

[0021] The anti-endoglin antibody can be administered by any appropriate means including, for example, subcutaneously, intravenously, or in an implant.

[0022] In some instances, the anti-endoglin antibody comprises an antigen-binding fragment that specifically binds to endoglin. Antigen-binding fragments include, but are not limited to, a Fab fragment, a Fab' fragment, a F(ab)2 fragment, a F(ab')2 fragment, a Fv fragment, a scFv fragment, or a single chain binding polypeptide.

[0023] The anti-endoglin antibody can be an IgG, an IgM, an IgE, an IgA, or an IgD, or is derived therefrom. When the antibody is an IgG, the antibody can be an IgGl, an IgG2, an IgG3, or an IgG4. In the case of an IgG2 antibody, the antibody can be an IgG2a antibody or an IgG2b antibody.

[0024] Anti-endoglin antibodies to be utilized in the present methods include antibodies that bind to, and inhibit and/or neutralize the activity of endoglin. An anti-endoglin antibody can be, for example, a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a humanized antibody, a deimmunized antibody, a human antibody, an affinity matured antibody or a combination thereof. Where combinations are contemplated, one would understood that, for example, a chimeric or humanized antibody could be deimmunized to reduce a T cell response when administered to a human subject.

[0025] In one example, an isolated anti-endoglin antibody comprises a light chain variable region having an amino acid sequence set forth as SEQ ID NO: 1 and a heavy chain variable region having an amino acid sequence set forth as SEQ ID NO: 3.

[0026] In one example, an isolated anti-endoglin antibody comprises a light chain variable region having an amino acid sequence set forth as SEQ ID NO: 1, a light chain constant region having an amino acid sequence set forth as SEQ ID NO: 2, a heavy chain variable region having an amino acid sequence set forth as SEQ ID NO: 3 and a gamma 1 (γΐ) constant region having an amino acid sequence set forth as SEQ ID NO: 4.

[0027] In one example, an isolated anti-CD 105 antibody comprises a V L CDR1 having an amino acid sequence set forth as SEQ ID NO: 10; a VL CDR2 having an amino acid sequence set forth as SEQ ID NO: 11 or 12; a V L CDR3 having an amino acid sequence set forth as SEQ ID NO: 13; a VH CDRl having an amino acid sequence set forth as SEQ ID NO: 5; a VH CDR2 having an amino acid sequence set forth as SEQ ID NO: 6, 7, or 8; and a VH CDR3 having an amino acid sequence set forth as SEQ ID NO: 9.

[0028] In one example, an isolated humanized, de-immunized anti-CD105 antibody can comprise a heavy chain variable region having the amino acid sequence set forth as SEQ ID NO: 14, 15, 16, 17, or 18; and a light chain variable region having the amino acid sequence set forth as SEQ ID NO: 19, 20, 21, 22, or 23.

[0029] In one example, an antibody for use in the described methods competes with an antibody described herein and, preferably, has a synergistic effect when administered with the VEGF and VEGFR inhibitors described herein.

[0030] Treatment can reduce a size of a cancer or a metastasis thereof by about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, or more compared to administration of a placebo or compared to baseline.

[0031] In some instances, the method comprises detecting one or more biomarkers, wherein the one or more biomarkers are higher than the mean or median levels of the biomarkers in a subject with that cancer type.

[0032] In some instances, the method comprises detecting two or more biomarkers, wherein the two or more biomarkers are higher than the median or mean levels of the biomarkers in a subject with that cancer type.

[0033] In some instances, the method comprises detecting one or more biomarkers, wherein the median or mean levels of one or more biomarkers are lower than the levels of the biomarkers in a subject with that cancer type.

[0034] In other instances, the method comprises detecting two or more biomarkers, wherein the levels of two or more biomarkers are lower than the median or mean levels of the biomarkers in a subject with that cancer type.

[0035] In yet other instances, the method comprises detecting two or more biomarkers, wherein at least one of the biomarkers is lower than the mean or median level of the biomarker in subjects with that cancer type and at least one of the biomarkers is higher than the median or mean level of the biomarker in a subject with that cancer type.

[0036] The one or more biomarkers to be detected in the current methods include, but are not limited to angiogenin 2 (ANG-2), placental growth factor (PIGF), bone morphogenetic protein 9 (BMP-9), intercellular adhesion molecule 1 (ICAM-1), basic fibroblast growth factor (bFGF), interleukin 6 (IL-6), hepatocyte growth factor (HGF), transforming growth factor beta 1 (TGF-β Ι), osteopontin (OPN), platelet derived growth factor AA (PDGF-AA), TGF-P2, stromal cell-derived factor 1 (SDF-1), PDGF-BB, transforming growth factor beta receptor 3 (TGFP-R3), tissue inhibitor of metalloproteinase-1 (TEVlP-1), Thrombospondin 2 (TSP-2), vascular cell adhesion molecule 1 (VCAM-1) and a combination thereof.

[0037] In some instances, the methods detect one or more biomarkers selected from the group consisting of vascular endothelial growth factor A (VEGF-A), Endoglin, VEGF-D, vascular endothelial growth factor receptor 1 (VEGF-Rl), VEGF-R2, VEGF-R3, and a combination thereof; and detect one or more biomarkers selected from the group consisting of ANG-2, PIGF, BMP-9, ICAM-1, bFGF, IL-6, HGF, TGF-βΙ, osteopontin (OPN), PDGF-AA, TGF-P2, SDF-1, PDGF-BB, TGFP-R3, ΉΜΡ-1, TSP-2, VCAM-1 and a combination thereof.

[0038] In one non-limiting example, the method comprises detecting TGFP-R3, where a subject having a plasma concentration of TGF -R3 at baseline that is above the median or mean level in a subject with the specific cancer type is selectively enrolled into a clinical trial of a VEGF inhibitor and an anti-endoglin antibody.

[0039] In one example, the method comprises detecting TGF -R3, where a subject having a plasma concentration of TGFP-R3 at baseline that is above the median or mean level in a subject with the specific cancer type is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

[0040] In one example, the method comprises detecting TGF -R3, where a subject having a plasma concentration of TGFP-R3 of above the median or a mean level in a subject with the specific cancer type is administered one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody.

[0041] In one example, the method comprises detecting osteopontin, where a subject having a plasma concentration of osteopontin at baseline that is below the median or a mean value in a subject with the specific cancer type is selectively enrolled in a clinical trial of a VEGF inhibitor and an anti-endoglin antibody.

[0042] In one example, the method comprises detecting osteopontin, where a subject having a plasma concentration of osteopontin at baseline that is below the median or mean value in a subject with the specific cancer type is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

[0043] In one example, the method comprises detecting osteopontin, where a subject having a plasma concentration of osteopontin at baseline that is below the median or mean value in a subject with the specific cancer type is administered one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody. [0044] In one example, the method comprises detecting ANG-2, VEGF-R2, TGFP-R3, and osteopontin, wherein a subject having (i) a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type of ANG-2, VEGF-R2, or TGFp-R3, or (ii) a plasma concentration of osteopontin at baseline that is above the median or mean level in a subject with the specific cancer type; or (iii) a combination of any of these biomarkers is selectively enrolled in a clinical trial of a VEGF inhibitor and an anti-endoglin antibody.

[0045] In one example, the method comprises detecting ANG-2, VEGF-R2, TGFP-R3, or osteopontin, wherein a subject having (i) a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type of ANG-2, VEGF-R2, or TGFp-R3, or (ii) a plasma concentration of osteopontin at baseline that is above the median or mean level in a subject with the specific cancer type; or (iii) a combination of any of these biomarkers is selectively enrolled in a clinical trial of a VEGF inhibitor and an anti-endoglin antibody; or is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

[0046] In one example, the method comprises detecting ANG-2, wherein a subject having a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type of ANG-2 is selectively enrolled in a clinical trial of a VEGF inhibitor and an anti- endoglin antibody; or is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

[0047] In one example, the method comprises detecting VEGF-R2, wherein a subj ect having a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type of VEGF-R2 is selectively enrolled in a clinical trial of a VEGF inhibitor and an anti-endoglin antibody; or is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

[0048] In one example, the method comprises detecting, TGFp-R3, wherein a subj ect having a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type of TGFp-R3is selectively enrolled in a clinical trial of a VEGF inhibitor and an anti-endoglin antibody; or is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

[0049] In one example, the method comprises detecting osteopontin, wherein a subject having a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type of osteopontin is selectively enrolled in a clinical trial of a VEGF inhibitor and an anti-endoglin antibody; or is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody. [0050] In one example, the method comprises detecting ANG-2, VEGF-R2, TGFP-R3, and osteopontin, wherein a subject with renal cell carcinoma having (i) a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type of ANG-2, VEGF-R2, or TGFP-R3, or (ii) a plasma concentration of osteopontin at baseline that is below the median or mean level in a subject with the specific cancer type; or (iii) a combination of any of these biomarkers is administered one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody.

[0051] In one example, the method comprises detecting ANG-2, VEGF-R2, TGFP-R3, or osteopontin, wherein a subject with renal cell carcinoma having (i) a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type of ANG-2, VEGF-R2, or TGFP-R3, or (ii) a plasma concentration of osteopontin at baseline that is below the median or mean level in a subject with the specific cancer type; or (iii) a combination of any of these biomarkers is selected for treatment with one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody.

[0052] In one example, the method comprises detecting ANG-2, wherein a subject with renal cell carcinoma having a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type of ANG-2 is selected for treatment with one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody.

[0053] In one example, the method comprises detecting VEGF-R2, wherein a subj ect with renal cell carcinoma having a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type of VEGF-R2 is selected for treatment with one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody.

[0054] In one example, the method comprises detecting TGF -R3, wherein a subject with renal cell carcinoma having a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type TGF -R3 is selected for treatment with one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody.

[0055] In one example, the method comprises detecting osteopontin, wherein a subject with renal cell carcinoma having a plasma concentration at baseline that is above the median or mean level in a subject with the specific cancer type osteopontin is selected for treatment with one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody.

[0056] In one example, a method comprises detecting ICAM-1, wherein a subject having a plasma concentration of ICAM-1 below a median or mean level in a subject with a cancer or a metastasis thereof is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

Compositions of a VEGF inhibitor and an anti-endoglin antibody can be administered as described below.

[0057] In one example, a method comprises detecting TSP-2, wherein a subject having a plasma concentration of TSP-2 below a median or mean level in a subject with a cancer or a metastasis thereof at baseline is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody. Compositions of a VEGF inhibitor and an anti-endoglin antibody can be administered as described below.

[0058] In one non-limiting aspect of the disclosed methods, where the cancer or metastasis thereof is a renal cell carcinoma, the median level of TGF beta-R3 can be from about 10 to about 250 ng/ml and the median level of OPN can be from about 0.1 to about 75 ng/ml or the median level of TGF beta-R3 can be from about 115 to about 238 ng/ml and the median level of OPN can be from about 0.2 to about 59 ng/ml. In one non-limiting instance, the median level of TGF beta-R3 can be about 182 ng/mL and the medium level of OPN can be about 1.2 μg/ml.

[0059] In one non-limiting example, the method comprises detecting ICAM-1 and TSP-2, wherein a subject having a plasma concentration at baseline that is below the median or the mean level of ICAM-1 or TSP-2 is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody. In one instance, the subject has a soft tissue carcinoma.

[0060] In one non-limiting aspect of the disclosed methods, where the cancer or metastasis thereof is a soft tissue sarcoma, the median level of ICAM-1 can be from about 250 to about 2000 ng/ml and the median level of TSP-2 can be from about 25 to about 400 ng/ml or the median level of ICAM-1 can be from about 293 to about 1 154 ng/ml and the median level of TSP-2 can be from about 58 to about 281 ng/ml. In one non-limiting instance, the median level of ICAM-1 can be about 658 ng/mL and the medium level of TSP-2 can be about 82 ng/ml.

[0061] Provided herein is a pharmaceutical composition, comprising an anti-endoglin antibody, for use in combination therapy to treat a subject selected by a method described herein, wherein the anti-endoglin antibody is to be administered with a VEGF or a VEGF receptor inhibitor. INCORPORATION BY REFERENCE

[0062] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.

SEQUENCE LISTING

[0063] The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on September 28, 2017, is named 35882-727_601_SL.txt and is 165,090 bytes in size.

BRIEF DESCRIPTION OF THE DRAWINGS

[0064] The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which are described herein below.

[0065] Figures 1A-C. OPN (Figure 1A), TGF -R3 (Figure IB) and SDF-1 (Figure 1C) were differentially expressed markers at baseline in renal cell carcinoma responders versus non- responders.

[0066] Figures 2A-B. ICAM-1 (Figure 1A) and TSP-2 (Figure IB) were differentially expressed markers at baseline in soft tissue sarcoma responders versus non-responders.

[0067] Figure 3 provides a humanized 02-VK1-39 variable (V L ) light chain having the monoclonal murine chimeric TRC105 VL CDRs (underlined) grafted between the framework regions (FRs) 1-3 of the human sequence 02-VK1-39 and a framework region 4 from the human JK4 sequence (SEQ ID NO: 52; all in bold). Variations that can be made to the human FRs are indicated at positions 1, 3, 4, 5, 36, 46, 47, 60, 70, 71, 100, and 106 of the sequence utilizing the Kabat numbering system (shown in italics beneath the humanized sequence).

[0068] Figure 4 provides a humanized VH3-15 variable (VH) heavy chain having the monoclonal murine monoclonal murine chimeric TRC105 VH CDRs (underlined) grafted between the framework regions (FRs) 1-3 of the human sequence VH3-15 and a framework region 4 from the human JH4 sequence (all in bold) (SEQ ID NO: 53). One or more variations that can be made to the human FRs are indicated at positions 49, 76, 77, 78, 82a, 89, 94, 108, 109, and 113 of the sequence utilizing the Kabat numbering system (shown in italics beneath the humanized sequence).

[0069] Figures 5A-B provide an amino acid sequence alignment of exemplary mouse and humanized VK chains (Figure 5A) and VH chains (Figure 5B). Sequence identifiers are SEQ ID NOS: 1 and 54-57, respectively, from top to bottom in Figure 5A and SEQ ID NOS: 3 and 58-60, respectively, from top to bottom in Figure 5B.

[0070] Figures 6A-B provide an amino acid sequence alignment of exemplary mouse and super- humanized VK chains (Figure 6A) and V H chains (Figure 6B). Sequence identifiers are SEQ ID NOS: 1 and 54-57, respectively, from top to bottom in Figure 6A and SEQ ID NOS: 3 and 58-60, respectively, from top to bottom in Figure 6B.

[0071] Figure 7 provides an amino acid sequence alignment and comparison of exemplary mouse and humanized and super-humanized V K chains and V H chains. The sequences are SEQ ID NOS 61-63, respectively, from top to bottom.

[0072] Figure 8 illustrates the lead humanized deimmunized heavy chain variable region with CDRs in bold and underlined (SEQ ID NO: 64). Variations that can be made are indicated at the identified positions of the sequence utilizing the Kabat numbering system (shown in italics beneath the humanized sequence). Variations may be made as a single mutation or as more than one mutation, and variations can be made with mutations in any combination.

[0073] Figure 9 illustrates the lead humanized deimmunized light chain variable region with CDRs in bold and underlined (SEQ ID NO: 65). Variations that can be made are indicated at the identified positions of the sequence utilizing the Kabat numbering system (shown in italics beneath the humanized sequence). Variations may be made as a single mutation or as more than one mutation, and variations can be made with mutations in any combination.

DETAILED DESCRIPTION OF THE INVENTION

[0074] It is to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting. Further, it is understood that a number of methods and materials similar or equivalent to those described herein can be used in the practice of the present inventions.

[0075] The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art. Such techniques are explained fully in the literature, such as, Molecular Cloning: A Laboratory Manual, second edition (Sambrook et al, 1989) Cold Spring Harbor Press; Oligonucleotide Synthesis (M. I. Gait, ed., 1984); Methods in Molecular Biology, Humana Press; Cell Biology: A Laboratory Notebook (J. E. Cellis, ed., 1998) Academic Press; Animal Cell Culture (R. I. Freshney, ed., 1987); Introduction to Cell and Tissue Culture (J. P. Mather and P. E. Roberts, 1998) Plenum Press; Cell and Tissue Culture: Laboratory Procedures (A. Doyle, J. B. Griffiths, and D. G. Newell, eds., 1993-1998) J. Wiley and Sons; Methods in Enzymology (Academic Press, Inc.); Handbook of Experimental Immunology (D. M. Weir and C. C. Blackwell, eds); Gene Transfer Vectors for Mammalian Cells (J. M. Miller and M. P. Cabs, eds., 1987); Current Protocols in Molecular Biology (F. M. Ausubel et al., eds., 1987); PCR: The Polymerase Chain Reaction, (Mullis et al, eds., 1994); Current Protocols in Immunology (J. E. Coligan et al, eds., 1991); Short Protocols in Molecular Biology (Wiley and Sons, 1999); Immunobiology (C. A. Janeway and P. Travers, 1997); Antibodies (P. Finch, 1997); Antibodies: a practical approach (D. Catty., ed., IRL Press, 1988-1989); Monoclonal antibodies: a practical approach (P. Shepherd and C. Dean, eds., Oxford University Press, 2000); Using antibodies: a laboratory manual (E. Harlow and D. Lane (Cold Spring Harbor Laboratory Press, 1999); and The Antibodies (M. Zanetti and J. D. Capra, eds., Harwood Academic Publishers, 1995).

[0076] As used in this specification and the appended claims, the singular forms "a", "an", and "the" include plural references unless the context clearly dictates otherwise. Thus for example, references to "a method" include one or more methods, and/or steps of the type described herein and/or which will become apparent to those persons skilled in the art upon reading this disclosure.

[0077] The term "about" includes equal to, and a range that takes into account experimental error in a given measurement and can refer to plus or minus 5, 4, 3, 2 or 1% or anywhere in-between such as, for example, ± 0.1%, ± 0.2%, ± 0.3%, ± 0.4%, ± 0.5%, ± 0.6%, ± 0.7%, ± 0.8%, ± 0.9%, 1.0%, ± 1.1%, ± 1.2%, ± 1.3%, ± 1.4%, ± 1.5%, ± 1.6%, ± 1.7%, ± 1.8%, ± 1.9%, ± 2.0%, 2.1%, ± 2.2%, ± 2.3%, ± 2.4%, ± 2.5%, ± 2.6%, ± 2.7%, ± 2.8%, ± 2.9%, ± 3.0%, 3.1%, ± 3.2%, ± 3.3%, ± 3.4%, ± 3.5%, ± 3.6%, ± 3.7%, ± 3.8%, ± 3.9%, ± 4.0%, 4.1%, ± 4.2%, ± 4.3%, ± 4.4%, ± 4.5%, ± 4.6%, ± 4.7%, ± 4.8%, ± 4.9% or ± 5.0% of the indicated value.

[0078] As used herein, a "baseline value" of a biomarker described herein refers to a value or range described herein.

[0079] As used herein, "substantially pure", "isolated" or "purified" refers to material which is at least 50% pure (i.e., free from contaminants), more preferably at least 90% pure, more preferably at least 95% pure, more preferably at least 98% pure, more preferably at least 99% pure. Antibodies can be isolated and purified from the culture supernatant or ascites mentioned above by saturated ammonium sulfate precipitation, euglobulin precipitation method, caproic acid method, caprylic acid method, ion exchange chromatography (DEAE or DE52), or affinity chromatography using anti-Ig column or a protein A, G or L column using art-recognized conventional methods. An antibody can be conjugated to, or recombinantly engineered with, an affinity tag (e.g., a purification tag) for purification. Affinity tags are known in the art and include, but are not limited to, a polyhistidine tag (e.g., a 6x His tag; SEQ ID NO: 26). SUBJECT SELECTION AND MONITORING

[0080] The methods described herein may be used to identify a subject having a cancer or a metastasis thereof for inclusion in a clinical trial for treatment with one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody. The methods described herein may be used to identify a subject having a cancer or a metastasis thereof for treatment with one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody. The methods may also be used to identify whether the cancer or metastasis thereof is being effectively treated with the described methods.

[0081] Provided herein is a method of treating a cancer or a metastasis thereof in a subject in need thereof, comprising detecting whether one or more biomarkers are present in a sample from the subject prior to treatment by contacting the sample with one or more antibodies that bind to the one or more biomarkers and detecting binding between the one or more biomarkers and the one or more antibodies; comparing a baseline value of the one or more biomarkers of the subject to a baseline value of a healthy subject; and administering to the subject one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody when the baseline value of the one or more biomarkers in the subject to be treated are above a median or mean level in a subject with the cancer or metastasis thereof.

[0082] Also provided herein is a method of treating a cancer or a metastasis thereof in a subject in need thereof, comprising detecting whether one or more biomarkers are present in a sample from the subject prior to treatment; diagnosing the subject with a cancer or metastasis when the presence of the one or more biomarkers in the sample are detected; and administering to the subject one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody when a baseline value of the one or more biomarkers in the subject to be treated is above or below a median or mean level in subjects with a specific cancer type.

[0083] A level of one or more biomarkers in a sample from the subject can be obtained prior to treatment and, in some instances, samples may be additionally obtained and tested at one or more time points during treatment. The methods include detection of one biomarker, two biomarkers, three biomarkers, four biomarkers, five biomarkers, six biomarkers, seven biomarkers, eight biomarkers, nine biomarkers, ten biomarkers or more at one or more times prior to, during, and/or following treatment. [0084] In some instances, the method comprises detecting one or more biomarkers, wherein the one or more baseline biomarkers are above a median or mean level in subjects with a specific cancer type.

[0085] In some instances, the method comprises detecting two or more biomarkers, wherein the two or more baseline biomarkers are above a median or mean level in subjects with a the specific cancer type.

[0086] In some instances, the method comprises detecting one or more baseline biomarkers, wherein the one or more biomarkers are lower than a median or mean level in subjects with a specific cancer type.

[0087] In other instances, the method comprises detecting two or more biomarkers, wherein the two or more biomarkers are lower than a median or mean level in subjects with a the specific cancer type.

[0088] In yet other instances, the method comprises detecting two or more biomarkers, wherein at least one of the baseline biomarkers is lower than a median or mean level in subjects with a specific cancer type and at least one of the biomarkers is at least two-fold higher than the level of the level of the biomarker in a healthy subject.

[0089] Detection of one or more biomarkers in sample obtained from a subject can be achieved using any conventionally known means such as, for example, an enzyme assay. Enzyme assays include, but are not limited to, an Enzyme Linked Immunosorbant Assay (ELISA). Detection can be achieved by exposing a sample from a subject to one or more commercially available antibodies and/or to an antibody described herein that specifically bind to a biomarker and comparing the results to a standard curve and a negative control. Assays useful in the described methods are discussed in more detail in the Examples below.

[0090] One or more biomarkers to be detected in the current methods include, but are not limited to angiogenin 2 (ANG-2), placental growth factor (PIGF), bone morphogenetic protein 9 (BMP-9), intercellular adhesion molecule 1 (ICAM-1), basic fibroblast growth factor (bFGF), interleukin 6 (IL-6), hepatocyte growth factor (HGF), transforming growth factor beta 1 (TGF-βΙ), osteopontin (OPN), platelet derived growth factor AA (PDGF-AA), TGF-P2, stromal cell-derived factor 1 (SDF-1), PDGF-BB, transforming growth factor beta receptor 3 (TGFP-R3), tissue inhibitor of metalloproteinase-1 (TIMP-1), Thrombospondin 2 (TSP-2), vascular cell adhesion molecule 1 (VCAM-1) and a combination thereof.

[0091] In some instances, the methods detect one or more biomarkers selected from the group consisting of vascular endothelial growth factor A (VEGF-A), Endoglin, VEGF-D, vascular endothelial growth factor receptor 1 (VEGF-Rl), VEGF-R2, VEGF-R3, and a combination thereof; and detect one or more biomarkers selected from the group consisting of ANG-2, PIGF, BMP-9, ICAM-1, bFGF, IL-6, HGF, TGF-βΙ, osteopontin (OPN), PDGF-AA, TGF-P2, SDF-1, PDGF-BB, TGFP-R3, ΤΊΜΡ-1, TSP-2, VCAM-1 and a combination thereof.

[0092] An amino acid sequence of human vascular endothelial growth factor A (VEGF-A) to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 49.

[0093] An amino acid sequence of human vascular endothelial growth factor A (VEGF-D) to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 50.

[0094] An amino acid sequence of human endoglin to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 28. An amino acid sequence of a human soluble endoglin to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 51.

[0095] An amino acid sequence of human vascular endothelial growth factor receptor 1 (VEGF-

Rl)to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 48.

[0096] An amino acid sequence of human VEGF-R2 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 47.

[0097] An amino acid sequence of human VEGF-R3 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 29.

[0098] An amino acid sequence of human ANG-2 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 30.

[0099] An amino acid sequence of human PIGF to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 3 1.

[0100] An amino acid sequence of human BMP-9 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 32.

[0101] An amino acid sequence of human ICAM-1 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 33.

[0102] An amino acid sequence of human bFGF to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 34.

[0103] An amino acid sequence of human IL-6 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 35.

[0104] An amino acid sequence of human HGF to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 36.

[0105] An amino acid sequence of human TGF-β Ι to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 37.

[0106] An amino acid sequence of human osteopontin (OPN) to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 38. [0107] An amino acid sequence of human PDGF-AA to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 39.

[0108] An amino acid sequence of human TGF-P2 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 40.

[0109] An amino acid sequence of human SDF-1 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 41.

[0110] An amino acid sequence of human PDGF-BB to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 42.

[0111] An amino acid sequence of human TGFP-R3 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 43.

[0112] An amino acid sequence of human TIMP-1 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 44.

[0113] An amino acid sequence of human TSP-2 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 45.

[0114] An amino acid sequence of human VCAM-1 to be detected by the methods described herein is, for example, set forth as SEQ ID NO: 46.

[0115] It will be understood that other human sequences of these proteins are known in the art and are contemplated for use herein.

[0116] In one non-limiting example, the method comprises detecting TGFP-R3, where a subject having a plasma concentration of TGFp-R3 above a median or mean level in subjects with a specific cancer type is selectively enrolled into a clinical trial of a VEGF inhibitor and an anti- endoglin antibody.

[0117] In one example, the method comprises detecting TGFp-R3, where a subject having a plasma concentration of TGFP-R3 at baseline that is above a median or mean level in subjects with a specific cancer type is administered one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody.

[0118] In one example, the method comprises detecting osteopontin, where a subject having a plasma concentration of osteopontin below a median or mean level in subjects with a specific cancer type at baseline is selectively enrolled in a clinical trial of a VEGF inhibitor and an anti- endoglin antibody.

[0119] In one example, the method comprises detecting osteopontin, where a subject having a plasma concentration of osteopontin below a median or mean level in subjects with a specific cancer type at baseline is administered one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody.

[0120] In one example, the method comprises detecting ANG-2, VEGF-R2, TGFP-R3, and osteopontin, wherein a subject having (i) a plasma concentration above a median or mean level in subjects with a specific cancer type of ANG-2, VEGF-R2, or TGFp-R3, or (ii) a plasma concentration of osteopontin below a median or mean level in subjects with a specific cancer type; or (iii) a combination of any of these biomarkers is selectively enrolled in a clinical trial of a VEGF inhibitor and an anti-endoglin antibody; or is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

[0121] In one example, the method comprises detecting ANG-2, VEGF-R2, TGFP-R3, or osteopontin, wherein a subject having (i) a plasma concentration above a median or mean level in subjects with a specific cancer type of ANG-2, VEGF-R2, or TGF -R3, or (ii) a plasma concentration of osteopontin below a median or mean level in subjects with a specific cancer type; or (iii) a combination of any of these biomarkers is selectively enrolled in a clinical trial of a VEGF inhibitor and an anti-endoglin antibody; or is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

[0122] In one example, the method comprises detecting ANG-2, VEGF-R2, TGF -R3, and osteopontin, wherein a subject having (i) a plasma concentration above a median or mean level in subjects with a specific cancer type of ANG-2, VEGF-R2, or TGFP-R3, or (ii) a plasma concentration of osteopontin below a median or mean level in subjects with a specific cancer type; or (iii) a combination of any of these biomarkers is administered one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody.

[0123] In one example, the method comprises detecting ANG-2, VEGF-R2, TGF -R3, or osteopontin, wherein a subject having (i) a plasma concentration above a median or mean level in subjects with a specific cancer type of ANG-2, VEGF-R2, or TGF -R3, or (ii) a plasma concentration of osteopontin below a median or mean level in subjects with a specific cancer type; or (iii) a combination of any of these biomarkers is administered one or more doses of a pharmaceutical composition that comprises a VEGF inhibitor and one or more doses of a pharmaceutical composition that comprises an anti-endoglin antibody; or is selected for treatment with a VEGF inhibitor and an anti-endoglin antibody.

[0124] Additional disclosure with respect to methods of administration of one or more

pharmaceutical compositions to a subject and treatment/dosing regimens are discussed in more detail below. [0125] The term "sample", "biological sample" or "test sample" as used herein denotes a sample taken or isolated from a subject. Non-limiting exemplary biological samples include, but are not limited to, a biofluid sample; blood; serum; plasma; urine; a tissue sample; and/or a biopsy sample, etc. The term also includes a mixture of the above-mentioned samples. The term "sample" also includes untreated or pretreated (or pre-processed) biological samples. In some embodiments, a sample can contain cells from a subject. As used herein, the term "biofluid" refers to any fluid obtained from a biological source and includes, but is not limited to, blood, urine, biopsies, and bodily secretions.

[0126] A sample can be obtained by removing a sample from a subject, but can also be

accomplished by using a previously isolated sample {e.g., isolated at a prior time point and isolated by the same or another person). In addition, the sample can be freshly collected or a previously collected sample.

[0127] In some embodiments, the sample can be an untreated sample. As used herein, the phrase "untreated sample" refers to a sample that has not had any prior sample pre-treatment except for dilution and/or suspension in a solution. Exemplary methods for treating a sample include, but are not limited to, centrifugation, filtration, sonication, homogenization, heating, freezing and thawing, and combinations thereof. In some embodiments, the test sample can be a frozen test sample, e.g., a frozen tissue. The frozen sample can be thawed before employing methods, assays and systems described herein. After thawing, a frozen sample can be centrifuged before being subjected to methods, assays and systems described herein. In some embodiments, the sample is a clarified test sample, for example, prepared by centrifugation and collection of a supernatant comprising the clarified sample. In some embodiments, a sample can be a pre-processed sample, for example, supernatant or filtrate resulting from a treatment selected from the group consisting of

centrifugation, filtration, thawing, purification, and any combinations thereof. In some

embodiments, the sample can be treated with a chemical and/or biological reagent. Chemical and/or biological reagents can be employed to protect and/or maintain the stability of the sample during processing. The skilled artisan is well aware of methods and processes appropriate for preprocessing of biological samples required for determination of the biomolecules/biomarkers as described herein.

[0128] In some embodiments, the methods described herein can further comprise a step of obtaining a sample from a subject. In some embodiments, the subject can be a human subject. It will be understood that a sample can be obtained from a subject utilizing any art-appropriate means.

[0129] In some embodiments, the methods described herein can comprise creating a report based on the levels as described herein. In some embodiments, the report denotes raw values the levels described herein in the test sample (plus, optionally, the level in a reference sample) or it indicates a percentage or fold increase or decrease as compared to a reference level, a base line level, and/or provides a prognosis regarding the subject's treatment responsiveness.

[0130] The one or more biomarkers described herein can be detected using any conventionally described assay including, but not limited to, an enzyme assay. Enzyme assays, for example, include, but are not limited to, an Enzyme Linked Immunosorbant Assay (ELISA), a competitive ELISA, fluorescent activated cell sorter (FACS), etc.

[0131] Antibodies that may be used to detect the one or more biomarkers include commercially available antibodies from, for example, Life Technologies, Roche, Amgen, Genentech,

Immunogen, etc.

[0132] Assay supports that may be used in the described assays are well known in the art and are contemplated herein.

ENDOGLIN, ANTIBODIES THAT SPECIFICALLY BIND TO ENDOGLIN AND PHARMACEUTICAL COMPOSITIONS THEREOF

[0133] Endoglin (CD105) is a 180 kDa homodimeric transmembrane protein. CD105 is constitutively phosphorylated in endothelial cells, mainly on serine and threonine residues, and this phosphorylation is due to the constitutively active TGF- β RII within the cell. TGF- β binding to CD 105 results in down-regulation of phosphorylation, similar to effects seen with protein kinase C inhibitors.

[0134] The sequences of human CD 105 (SEQ ID NO: 28) and murine CD 105 (SEQ ID NO: 27) are not identical. The human CD 105 amino acid sequence contains the tripeptide arginine-glycine- aspartic acid (RGD) located in an exposed region of the extracellular domain. The RGD peptide is a key recognition structure found on ECM proteins such as fibronectin, vitronectin, von Willebrand factor (vWF), type I collagen, and fibrinogen and is recognized by cell surface integrins.

[0135] As used herein, the term "antibody" refers to an immunoglobulin (Ig) whether naturally, or partly or wholly synthetically produced. The term also covers any polypeptide or protein having a binding domain which is, or is homologous to, an antigen-binding domain. The term further includes "antigen-binding fragments" and other interchangeable terms for similar binding fragments such as described below. "Antibodies" useful in the present invention encompass, but are not limited to, monoclonal antibodies, polyclonal antibodies, antibody fragments {e.g., Fab, Fab', F(ab') 2 , Fv, Fc, etc.), chimeric antibodies, bispecific antibodies, multispecific antibodies, deimmunized antibodies, heteroconjugate antibodies, single chain (ScFv), mutants thereof, fusion proteins comprising an antibody portion {e.g., a domain antibody), humanized antibodies, human antibodies, and/or any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity, including glycosylation variants of antibodies, amino acid sequence variants of antibodies, and covalently modified antibodies. In one non-limiting embodiment, an antibody for use in the methods described herein is a chimeric and deimmunized antibody. In one non-limiting embodiment, an antibody for use in the methods described herein is a humanized and deimmunized antibody.

[0136] Native antibodies and native immunoglobulins are usually heterotetrameric glycoproteins of about 150,000 Daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is typically linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain ("V H " or "VH") followed by a number of constant domains ("CH" or "CH"). Each light chain has a variable domain at one end ("VL" or "VL") and a constant domain ("CL" or "CL") at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light-chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interface between the light- and heavy-chain variable domains.

[0137] The terms "synthetic polynucleotide," "synthetic gene" or "synthetic polypeptide," as used herein, mean that the corresponding polynucleotide sequence or portion thereof, or amino acid sequence or portion thereof, is derived, from a sequence that has been designed, or synthesized de novo, or modified, compared to an equivalent naturally-occurring sequence. Synthetic

polynucleotides (antibodies or antigen binding fragments) or synthetic genes can be prepared by methods known in the art, including but not limited to, the chemical synthesis of nucleic acid or amino acid sequences. Synthetic genes are typically different from naturally-occurring genes, either at the amino acid, or polynucleotide level, (or both) and are typically located within the context of synthetic expression control sequences. Synthetic gene polynucleotide sequences, may not necessarily encode proteins with different amino acids, compared to the natural gene; for example, they can also encompass synthetic polynucleotide sequences that incorporate different codons but which encode the same amino acid {i.e. , the nucleotide changes represent silent mutations at the amino acid level).

[0138] With respect to antibodies, the term "variable domain" refers to the variable domains of antibodies that are used in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. Rather, it is concentrated in three segments called hypervariable regions (also known as CDRs) in both the light chain and the heavy chain variable domains. More highly conserved portions of variable domains are called the "framework regions" or "FRs." The variable domains of unmodified heavy and light chains each contain four FRs (FR1, FR2, FR3 and FR4), largely adopting a β -sheet configuration interspersed with three CDRs which form loops connecting and, in some cases, part of the β-sheet structure. The CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen -binding site of antibodies (see Kabat et al, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991), pages 647-669).

[0139] The terms "hypervariable region" and "CDR" when used herein, refer to the amino acid residues of an antibody which are responsible for antigen-binding. The CDRs comprise amino acid residues from three sequence regions which bind in a complementary manner to an antigen and are known as CDR1, CDR2, and CDR3 for each of the VH and VL chains. In the light chain variable domain, the CDRs typically correspond to approximately residues 24-34 (CDRLl), 50-56

(CDRL2) and 89-97 (CDRL3), and in the heavy chain variable domain the CDRs typically correspond to approximately residues 31 -35 (CDRH1), 50-65 (CDRH2) and 95-102 (CDRH3) according to Kabat et al , Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)). It is understood that the CDRs of different antibodies may contain insertions, thus the amino acid numbering may differ. The Kabat numbering system accounts for such insertions with a numbering scheme that utilizes letters attached to specific residues {e.g., 27 A, 27B, 27C, 27D, 27E, and 27F of CDRLl in the light chain) to reflect any insertions in the numberings between different antibodies. Alternatively, in the light chain variable domain, the CDRs typically correspond to approximately residues 26-32 (CDRLl), 50-52 (CDRL2) and 91 -96 (CDRL3), and in the heavy chain variable domain, the CDRs typically correspond to approximately residues 26-32 (CDRH1), 53-55 (CDRH2) and 96-101 (CDRH3) according to Chothia and Lesk, J. Mol. Biol, 196: 901-917 (1987)).

[0140] As used herein, "framework region" or "FR" refers to framework amino acid residues that form a part of the antigen binding pocket or groove. In some embodiments, the framework residues form a loop that is a part of the antigen binding pocket or groove and the amino acids residues in the loop may or may not contact the antigen. Framework regions generally comprise the regions between the CDRs. In the light chain variable domain, the FRs typically correspond to

approximately residues 0-23 (FRLl), 35-49 (FRL2), 57-88 (FRL3), and 98-109 and in the heavy chain variable domain the FRs typically correspond to approximately residues 0-30 (FRH1), 36-49 (FRH2), 66-94 (FRH3), and 103-133 according to Kabat et al, Sequences of Proteins of

Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)). As discussed above with the Kabat numbering for the light chain, the heavy chain too accounts for insertions in a similar manner (e.g. , 35 A, 35B of CDRH1 in the heavy chain).

Alternatively, in the light chain variable domain, the FRs typically correspond to approximately residues 0-25 (FRL1), 33-49 (FRL2) 53-90 (FRL3), and 97-109 (FRL4), and in the heavy chain variable domain, the FRs typically correspond to approximately residues 0-25 (FRH1), 33-52 (FRH2), 56-95 (FRH3), and 102-1 13 (FRH4) according to Chothia and Lesk, J. Mol. Biol, 196: 901-917 (1987)).

[0141] Constant domains (Fc) of antibodies are not involved directly in binding an antibody to an antigen but, rather, exhibit various effector functions, such as participation of the antibody in antibody-dependent cellular toxicity via interactions with, for example, Fc receptors (FcR). Fc domains can also increase bioavailability of an antibody in circulation following administration to a patient. Substitution of a murine Fc domain with a human Fc domain can also reduce side HAMA reactions.

[0142] Depending on the amino acid sequence of the constant domain of their heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of

immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these can be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgAl, and IgA2. The heavy-chain constant domains (Fc) that correspond to the different classes of immunoglobulins are called α, δ, ε, γ, and μ, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.

[0143] The "light chains" of antibodies from any vertebrate species can be assigned to one of two clearly distinct types, called kappa or ("κ") and lambda or ("λ"), based on the amino acid sequences of their constant domains.

[0144] The terms "antigen-binding portion of an antibody," "antigen-binding fragment," "antigen- binding domain," "antibody fragment" or a "functional fragment of an antibody" are used interchangeably herein to refer to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. Non-limiting examples of antigen-binding fragments included within such terms include, but are not limited to, a Fab fragment, a Fab' fragment, a F(ab) 2 fragment, a F(ab') 2 fragment, a Fv fragment, a scFv fragment, or a single chain binding

polypeptide. The structures of such antigen-binding fragments described herein are known in the art.

[0145] "Chimeric" forms of non-human (e.g., murine) antibodies include chimeric antibodies which contain minimal sequence derived from a non-human Ig. For the most part, chimeric antibodies are murine antibodies in which at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin, are inserted in place of the murine Fc.

[0146] The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e. , the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations, which can include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, monoclonal antibodies can be made by the hybridoma method first described by Kohler et al. , Nature 256:495 (1975), or can be made by recombinant DNA methods (see, e.g. , U. S. Pat. No. 4,816,567). In certain embodiments, the monoclonal antibodies can be isolated from phage antibody libraries using the techniques described in Clackson et al, Nature 352:624-628 (1991) and Marks et al , J. Mol. Biol. 222:581-597 (1991), for example.

[0147] As used herein, "immunoreactive" refers to binding agents, antibodies or fragments thereof that are specific to a sequence of amino acid residues (i.e., a "binding site" or an "epitope"), yet if are cross-reactive to other peptides/proteins, are not toxic at the levels at which they are formulated for administration to human use. The term "binding" refers to a direct association between two molecules, due to, for example, covalent, electrostatic, hydrophobic, and ionic and/or hydrogen- bond interactions under physiological conditions, and including interactions such as salt bridges and water bridges and any other conventional binding means.

[0148] The term "preferentially binds" means that the binding agent binds to the binding site with greater affinity than it binds unrelated amino acid sequences. Affinity can be at least 1-fold greater, at least 2-fold greater, at least 3 -fold greater, at least 4-fold greater, at least 5 -fold greater, at least 6- fold greater, at least 7-fold greater, at least 8-fold greater, at least 9-fold greater, 10-fold greater, at least 20-fold greater, at least 30-fold greater, at least 40-fold greater, at least 50-fold greater, at least 60-fold greater, at least 70-fold greater, at least 80-fold greater, at least 90-fold greater, at least 100- fold greater, or at least 1000-fold greater than the affinity of the binding agent for unrelated amino acid sequences. The terms "immunoreactive" and "preferentially binds" are used interchangeably herein. [0149] As used herein, the term "avidity" refers to the resistance of a complex of two or more agents to dissociation after dilution. Avidities can be determined by methods such as a Scatchard analysis or any other technique familiar to one of skill in the art.

[0150] As used herein, the term "affinity" refers to the equilibrium constant for the reversible binding of two agents. Apparent affinities can be determined by methods such as an enzyme linked immunosorbent assay (ELISA) or any other technique familiar to one of skill in the art.

[0151] An antibody, or antigen-binding fragment thereof, described herein may have a dissociation constant (IQ) of from about 1 femptomolar (fM) to about 500 nM, from about 1 pM to about 500 nM, from about 1 pM to about 250 nM, from about 1 picomolar (pM) to about 200 nM, from about 1 pM to about 150 nM, from about 1 pM to about 100 nM, from about 1 pM to about 50 nM, from about 1 pM to about 25 nM, from about 1 pM to about 10 nM, from about 1 pM to about 1 nM, from about 1 pM to about 70 pM, from about 1 pM to about 500 pM, from about 1 pM to about 250 pM, from about 1 pM to about 125 pM, from about 1 pM to about 100 pM, from 1 pM to about 75 pM, from 1 pM to about 50 pM, from 1 pM to about 25 pM, from 1 pM to about 15 pM, from 1 pM to about 10 pM, from about 10 pM to about 20 pM, from about 1 pM to about 29 pM, from about 30 pM to about 40 pM, from about 10 pM to about 100 pM, or from about 20 pM to about 500 pM, or any integer therebetween.

[0152] An antibody, or antigen-binding fragment thereof, described herein may have a dissociation constant (K d ) of less than about 1 nM, less than about 900 pM, less than about 800 pM, less than about 700 pM, less than about 600 pM, less than about 500 pM, less than about 400 pM, less than about 300 pM, less than about 250 pM, less than about 200 pM, less than about 100 pM, less than about 75 pM, less than about 50 pM, less than about 30 pM, less than about 25 pM, less than about 20 pM, less than about 18 pM, less than about 15 pM, less than about 10 pM, less than about 7.5 pM, less than about 5 pM, less than about 2.5 pM, less than about 1 pM, less than about 900 fM, less than about 800 fM, less than about 700 fM, less than about 600 fM, less than about 500 fM, less than about 400 fM, less than about 300 fM, less than about 250 fM, less than about 200 fM, less than about 100 fM, less than about 75 fM, less than about 50 fM, less than about 30 fM, less than about 25 fM, less than about 20 fM, less than about 18 fM, less than about 15 fM, less than about 10 fM, less than about 7.5 fM, less than about 5 fM, or less than about 2.5 fM.

[0153] An antibody, or antigen-binding fragment thereof, described herein may have an affinity (K A ) for hepcidin or a hepcidin peptide of from about 10 '9 to about 10 "14 , from about 10 '10 to about 10 "14 , from about 10 "11 to about 10 "14 , from about 10 "12 to about 10 "14 , from about 10 "13 to about 10 " 14 , from about 10 "10 to about 10 "11 , from about 10 '11 to about 10 "12 , from about 10 '12 to about 10 '13 , or 10 -13 to about 10 -14 . [0154] Also provided herein are affinity matured antibodies. For example, affinity matured antibodies can be produced by procedures known in the art (Marks et al, 1992, Bio/Technology, 10:779-783; Barbas et a!., 1994, Proc Nat. Acad. Sci, USA 91 :3809-3813; Schier et al, 1995, Gene, 169: 147-155; Ydion et al, 1995, J. Immunol, 155: 1994-2004; Jackson et al, 1995, J. Immunol, 154(7) 3310-9; Hawkins et al, 1992, J. Mol. Biol, 226:889-896; and WO2004/058184).

[0155] As used herein the term "epitope" refers to that portion of an antigen or other

macromolecule capable of forming a binding interaction with the variable region binding pocket of an antibody. Such binding interactions can be manifested as an intermolecular contact with one or more amino acid residues of one or more CDRs. Antigen binding can involve, for example, a CDR3 or a CDR3 pair or, in some cases, interactions of up to all six CDRs of the VH and VL chains. An epitope can be a linear peptide sequence (i.e., "continuous") or can be composed of noncontiguous amino acid sequences (i.e. , "conformational" or "discontinuous"). An antibody can recognize one or more amino acid sequences; therefore an epitope can define more than one distinct amino acid sequence. Epitopes recognized by antibodies can be determined by peptide mapping and sequence analysis techniques well known to one of skill in the art. Binding interactions are manifested as intermolecular contacts with one or more amino acid residues of a CDR.

[0156] The term "specific" refers to a situation in which an antibody will not show any significant binding to molecules other than the antigen containing the epitope recognized by the antibody. The term is also applicable where, for example, an antigen binding domain is specific for a particular epitope which is carried by a number of antigens, in which case the antibody will be able to bind to the various antigens carrying the epitope. The terms "preferentially binds" or "specifically binds" mean that the antibodies bind to an epitope with greater affinity than it binds unrelated amino acid sequences, and, if cross-reactive to other polypeptides containing the epitope, are not toxic at the levels at which they are formulated for administration to human use. In one aspect, such affinity is at least 1-fold greater, at least 2-fold greater, at least 3-fold greater, at least 4-fold greater, at least 5- fold greater, at least 6-fold greater, at least 7-fold greater, at least 8-fold greater, at least 9-fold greater, 10-fold greater, at least 20-fold greater, at least 30-fold greater, at least 40-fold greater, at least 50-fold greater, at least 60-fold greater, at least 70-fold greater, at least 80-fold greater, at least 90-fold greater, at least 100-fold greater, or at least 1000-fold greater than the affinity of the antibody for unrelated amino acid sequences. The terms "immunoreactive," "binds," "preferentially binds" and "specifically binds" are used interchangeably herein. The term "binding" refers to a direct association between two molecules, due to, for example, covalent, electrostatic, hydrophobic, and ionic and/or hydrogen-bond interactions under physiological conditions, and includes interactions such as salt bridges and water bridges, as well as any other conventional means of binding.

[0157] "Isolated" (used interchangeably with "substantially pure") when applied to polypeptides means a polypeptide or a portion thereof which, by virtue of its origin or manipulation: (i) is present in a host cell as the expression product of a portion of an expression vector; or (ii) is linked to a protein or other chemical moiety other than that to which it is linked in nature; or (iii) does not occur in nature, for example, a protein that is chemically manipulated by appending, or adding at least one hydrophobic moiety to the protein so that the protein is in a form not found in nature. By "isolated" it is further meant a protein that is: (i) synthesized chemically; or (ii) expressed in a host cell and purified away from associated and contaminating proteins. The term generally means a polypeptide that has been separated from other proteins and nucleic acids with which it naturally occurs. Typically, the polypeptide is also separated from substances such as antibodies or gel matrices (polyacrylamide) which are used to purify it. Antibodies can be isolated and purified from the culture supernatant or ascites mentioned above by saturated ammonium sulfate precipitation, euglobulin precipitation method, caproic acid method, caprylic acid method, ion exchange chromatography (DEAE or DE52), or affinity chromatography using anti-Ig column or a protein A, G or L column.

[0158] Methods for humanizing and deimmunizing antibody sequences are described in, for example, US Patent No. 8,221,753, issued luly 17, 2012, and such methods are incorporated by reference herein.

[0159] Antibodies and antigen-binding fragments can be constructed and produced using conventional techniques known in the art. In addition, recombinantly prepared antibodies can often be produced in large quantities, particularly when utilizing high level expression vectors. Methods for recombinantly and synthetically producing antibodies, including expressing antibodies, are described in, for example, US Patent No. 8,221,753, issued July 17, 2012, and such methods are incorporated by reference herein. The expression of antibodies and antibody fragments is well established in the art.

[0160] Provided herein are embodiments where an antibody, or antigen-binding fragment thereof, described herein are labeled with a reporter label, a therapeutic label, or a combination thereof. Methods of making immunoconjugates are known in the art and are considered herein. Such methods include those described in, for example, US Patent No. 8,221,753, issued July 17, 2012, and such methods are incorporated by reference herein. As used herein, for purposes of the specification and claims, immunoconjugates refer to conjugates comprised of the humanized anti- endoglin antibodies or fragments thereof according to the present invention and at least one therapeutic label. Such antitumor agents are known in the art and include, but not limited to, toxins, drugs, enzymes, cytokines, radionuclides, and photodynamic agents. Toxins include, but are not limited to, ricin A chain, mutant Pseudomonas exotoxins, diphtheria toxoid, streptonigrin, boamycin, saporin, gelonin, and pokeweed antiviral protein. Drugs include daunorubicin, methotrexate, and calicheamicins. Radionuclides include radiometals. Cytokines include, but are not limited to, transforming growth factor (TGF)-P, interleukins, interferons, and tumor necrosis factors. Photodynamic agents include, but are not limited to, porphyrins and their derivatives. Additional therapeutic labels will be known in the art and are also contemplated herein. The methods for complexing the anti-endoglin mAbs or a fragment thereof with at least one antitumor agent are well known to those skilled in the art {i.e., antibody conjugates as reviewed by Ghetie et ah , 1994, Pharmacol. Ther. 63 :209-34). Such methods may utilize one of several available heterobifunctional reagents used for coupling or linking molecules. Additional radionuclides are further described herein along with additional methods for linking molecules, such as therapeutic labels.

[0161] Provided herein are embodiments where antibody, or antigen-binding fragment thereof, described herein are modified to increase half-life and/or to cross the blood brain barrier. Such modification of the antibodies described herein allows for the treatment of a brain cancer.

Exemplary modifications to allow proteins such as antibodies to cross the blood-brain barrier are described in US Patent Publication 20070082380 which is hereby incorporated by reference in its entirety. Other modifications such as glycosylation, to remove fucose from complex N-glycoside- linked sugar chains, and PEGylation are known in the art and described in, for example, US Patent No. 8,221,753, issued July 17, 2012, and such methods are incorporated by reference herein. Such modifications can lead to one or more of improved circulation time, improved solubility, improved resistance to proteolysis, reduced antigenicity and immunogenicity, improved bioavailability, reduced toxicity, improve half-life and improved stability.

[0162] Chimeric antibodies that bind CD 105 are described herein that exhibit reduced

immunogenicity while maintaining and/or improving their specificity. Additionally, to address problems associated with murine antibodies, chimeric antibodies that bind CD 105 and decrease and/or inhibit angiogenesis are described herein that exhibit reduced immunogenicity while maintaining and/or improving their specificity. These anti-CD105 antibodies are useful for the diagnosis and treatment of various conditions and diseases as well as for purification and detection of CD 105. Antibodies against CD105 represent an important area for the development of therapies for the treatment of a variety of diseases and conditions which involve, are influenced by, or affected by angiogenesis. [0163] Provided herein are antibodies thereof that bind to CD105. Also provided are antibodies, (or antigen -binding fragments) thereof that bind CD 105 and inhibit (partially or fully) or manage/treat (partially or fully) a cancer or a metastasis thereof.

[0164] One can recognize that the antibodies that specifically bind CD105 generated using the methods described herein can be tested using the assays provided herein or known in the art for the ability to bind to CD 105 using conventional methods including, but not limited to, ELISA. Affinity of antibodies described herein can also be determined using conventional methods including, but not limited to, BIACORE® or surface plasmon resonance.

[0165] Provided herein are antibodies that bind CD 105. Also provided herein are isolated antibodies that specifically bind to CD 105 and inhibit a cancer or a metastasis thereof.

[0166] In one non-limiting embodiment, an isolated anti-endoglin antibody comprises chain variable region having an amino acid sequence set forth as SEQ ID NO: 1 and a heavy chain variable region having an amino acid sequence set forth as SEQ ID NO: 3.

[0167] Provided herein is an isolated anti-endoglin antibody that comprises a light chain variable region having an amino acid sequence set forth as SEQ ID NO: 1, a light chain constant region having an amino acid sequence set forth as SEQ ID NO: 2, a heavy chain variable region having an amino acid sequence set forth as SEQ ID NO: 3 and a gamma 1 (γΐ) constant region having an amino acid sequence set forth as SEQ ID NO: 4.

[0168] In one non-limiting embodiment, an isolated anti-CD105 antibody comprises a V L CDR1 having an amino acid sequence set forth as SEQ ID NO: 10; a V L CDR2 having an amino acid sequence set forth as SEQ ID NO: 11 or 12; a VL CDR3 having an amino acid sequence set forth as SEQ ID NO: 13; a VH CDRl having an amino acid sequence set forth as SEQ ID NO: 5; a VH CDR2 having an amino acid sequence set forth as SEQ ID NO: 6, 7, or 8; and a V H CDR3 having an amino acid sequence set forth as SEQ ID NO: 9.

[0169] In one non-limiting embodiment, an isolated humanized, de-immunized anti-CD105 antibody can comprise a heavy chain variable region having the amino acid sequence set forth as SEQ ID NO: 14, 15, 16, 17, or 18; and a light chain variable region having the amino acid sequence set forth as SEQ ID NO: 19, 20, 21, 22, or 23.

[0170] An isolated anti-endoglin antibody as described herein binds to human and murine endoglin and also competitively inhibits human BMP binding. However, an isolated anti-endoglin antibody as described herein does not prevent the binding of mouse BMP to mouse endoglin.

[0171] Antibodies described herein are useful in detection assays or to isolate endoglin from a sample. [0172] Antibodies or antigen-binding fragments thereof provided herein are such that they can be conjugated or linked to a therapeutic moiety, a detectable moiety, and/or an affinity tag. Methods for conjugating or linking polypeptides are well known in the art. Associations (binding) between compounds and labels include any means known in the art including, but not limited to, covalent and non-covalent interactions, chemical conjugation as well as recombinant techniques.

[0173] Pharmaceutical compositions or formulations that contain an anti-endoglin antibody described herein may be prepared with one or more optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington, The Science and Practice of Pharmacy 20th Ed. Mack Publishing (2000)), in the form of lyophilized formulations or aqueous solutions.

[0174] As used herein, "pharmaceutically acceptable carrier" or "pharmaceutical acceptable excipient" includes any material which, when combined with an active ingredient, allows the ingredient to retain biological activity and is non-reactive with the subject's immune system.

Examples include, but are not limited to, any of the standard pharmaceutical carriers such as a phosphate buffered saline solution, water, emulsions such as oil/water emulsion, and various types of wetting agents. Preferred diluents for aerosol or parenteral administration are phosphate buffered saline or normal (0.9%) saline. Compositions comprising such carriers are formulated by well- known conventional methods {see, for example, Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, Ed., Mack Publishing Co., Easton, Pa., 1990; and Remington, The Science and Practice of Pharmacy 20th Ed. Mack Publishing, 2000).

[0175] Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and may comprise buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants including ascorbic acid and methionine;

preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens, such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes {e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG).

[0176] The formulations to be used for in vivo administration may be sterilized. This may be accomplished by, for example, filtration through sterile filtration membranes, or any other art- recognized method for sterilization. Antibody compositions are generally placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle. Other methods for sterilization and filtration are known in the art and are contemplated herein.

[0177] In one embodiment of the present invention, the compositions are formulated to be free of pyrogens such that they are acceptable for administration to a subject. Testing compositions for pyrogens and preparing pharmaceutical compositions free of pyrogens are well understood to one of ordinary skill in the art.

[0178] In one embodiment, the composition is lyophilized, for example, to increase shelf-life in storage. When the compositions are considered for use in medicaments or any of the methods provided herein, it is contemplated that the composition can be substantially free of pyrogens such that the composition will not cause an inflammatory reaction or an unsafe allergic reaction when administered to a human patient. Testing compositions for pyrogens and preparing compositions substantially free of pyrogens are well understood to one or ordinary skill of the art and can be accomplished using commercially available kits.

[0179] Acceptable carriers can be a compound that stabilizes, increases or delays absorption or clearance of an anti-endoglin antibody. Such compounds include, for example, carbohydrates, such as glucose, sucrose, or dextrans; low molecular weight proteins; compositions that reduce the clearance or hydrolysis of peptides; or excipients or other stabilizers and/or buffers. Agents that delay absorption include, for example, aluminum monostearate and gelatin. Detergents can also be used to stabilize or to increase or decrease the absorption of the pharmaceutical composition, including liposomal carriers. To protect from digestion the compound can be complexed with a composition to render it resistant to acidic and enzymatic hydrolysis, or the compound can be complexed in an appropriately resistant carrier such as a liposome. Means of protecting compounds from digestion are known in the art (see, e.g., Fix (1996) Pharm Res. 13 : 1760 1764; Samanen (1996) J. Pharm. Pharmacol. 48: 1 19 135; and U.S. Pat. No. 5,391,377, describing lipid compositions for oral delivery of therapeutic agents).

[0180] The phrase "pharmaceutically acceptable" refers to molecular entities and compositions that are physiologically tolerable and do not typically produce an allergic or similar untoward reaction, such as gastric upset, dizziness and the like, when administered to a subject.

[0181] The term "unit dose" when used in reference to a therapeutic composition refers to physically discrete units suitable as unitary dosage for humans, each unit containing a

predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required diluent; i.e., carrier, or vehicle. [0182] The compositions can be administered in a manner compatible with the dosage formulation, and in a therapeutically effective amount. The quantity to be administered depends on the subject to be treated, capacity of the subject's immune system to utilize the active ingredient, and degree of binding capacity desired. Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner and are peculiar to each individual. Suitable regimes for initial administration and booster shots are also variable, but are typified by an initial administration followed by repeated doses at one or more hour intervals by a subsequent injection or other administration. Alternatively, continuous intravenous infusion sufficient to maintain concentrations in the blood are contemplated.

[0183] One embodiment of the present invention contemplates the use of any of the compositions of the present invention to make a medicament for treating a cancer. Medicaments can be formulated based on the physical characteristics of the subject needing treatment, and can be formulated in single or multiple formulations based on the disorder. Medicaments of the present invention can be packaged in a suitable pharmaceutical package with appropriate labels for the distribution to hospitals and clinics wherein the label is for the indication of treating a cancer as described herein in a subj ect. Medicaments can be packaged as a single or multiple units.

Instructions for the dosage and administration of the pharmaceutical compositions of the present invention can be included with the pharmaceutical packages.

[0184] The compositions can be administered in a manner compatible with the dosage formulation, and in a therapeutically effective amount. The quantity to be administered depends on the subject to be treated, capacity of the subject's immune system to utilize the active ingredient, and degree of binding capacity desired. Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner and are peculiar to each individual. Suitable regimes for initial administration and booster shots are also variable, but are typified by an initial administration followed by repeated doses at one or more hour intervals by a subsequent injection or other administration. Booster doses can also be given at any appropriate interval. Alternatively, continuous intravenous infusion sufficient to maintain concentrations in blood is also contemplated. VASCULAR ENDOTHELIAL GROWTH FACTORS (VEGF) AND VEGF RECEPTOR (VEGFR) INHIBITORS

[0185] VEGF and VEGFR inhibitors to be considered for use in the present compositions and methods include, but are not limited to, Axitinib, Pazopanib, Bevacizumab (AVASTIN®), ranibizumab (LUCENTIS®), aflibercept (VEGF-Trap; EYLEA®), sunitinib (SUTENT®), brivanib (BMS-582664), sorafenib (NEXAVAR®), pegaptanib (MACUGEN®), and SU5416. [0186] In some embodiments, the inhibitor is a small molecule inhibitor, such as, for example, axitinib, pazopanib, sorafenib, sunitinib, pazopanib, brivanib, etc.

[0187] In other embodiments, the inhibitor is an antagonistic anti-VEGF antibody or an antagonistic anti-VEGFR antibody such as, for example, Bevacizumab (AVASTIN®), ranibizumab (LUCENTIS®), aflibercept (VEGF-Trap; EYLEA®), etc.

Axitinib

[0188] INLYTA® (Axitinib) is a small molecule indazole derivative, kinase inhibitor of the chemical name N-methyl^-fS-i^^-pyridin^-yl-viny^-lH-indazol-e-ylsulfanylJ- benzamide and a molecular formula of C22H1 8 N4OS. The chemical structure is of Axitinib is shown in Formula I:

Formula I

[0189] Compositions/formulations of Axitinib may contain microcrystalline cellulose, lactose monohydrate, croscarmellose sodium, magnesium stearate, and a dye as inactive ingredients; and may also contain a film coating that includes lactose monohydrate, HPMC 2910/Hypromellose 15cp, titanium dioxide, triacetin (glycerol triacetate) and red iron oxide.

Pazopanib

[0190] Pazopanib (VOTRIENT®) is a potent and selective multi -targeted receptor tyrosine kinase inhibitor that blocks tumor growth and inhibits angiogenesis. Pazopanib has a chemical name of 5- [[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidiny l]amino]-2-methyl- benzenesulfonamide. The chemical structure is of Pazo anib is shown in Formula Π:

Formula II

[0191] Compositions/formulations of Pazopanib can be administered orally once daily without food (at least 13 1 hour before or 2 hours after a meal) with a maximum dosage of 800 mg daily. METHODS OF TREATMENT

[0192] Provided herein are methods of treating a cancer or a metastasis thereof in a subject selected based upon the biomarker assessment described above.

[0193] A cancer to be treated includes, but is not limited to, a breast cancer, a lung cancer, a brain cancer, a sarcoma, a carcinoma, or a metastasis of any of such cancers.

[0194] In one non-limiting example, the carcinoma is a Renal Cell Carcinoma which can be, in some instances, an advanced Renal Cell Carcinoma. In one non-limiting example, the carcinoma is a Renal Cell Carcinoma which can be, in some instances, a metastatic Renal Cell Carcinoma. In some instances, the carcinoma is a hepatocellular carcinoma or a choriocarcinoma.

[0195] In some instances, the cancer or metastasis thereof is a brain cancer such as, for example, a glioblastoma multiforme (GBM).

[0196] In some instances, the cancer or metastasis thereof is a breast cancer such as, for example, a Luminal A, a Luminal B, a Luminal B-like, a Triple negative or a HER2 type breast cancer.

[0197] In the described methods, a VEGF inhibitor to be administered includes, but is not limited to, Axitinib (N-methyl-2-[3-((£)-2-pyridin-2-yl-vinyl)-lH-indazol-6-ylsu lfanyl]-benzamide). Axitinib can be administered by any appropriate means including, for example, orally,

subcutaneously, intravenously, or in an implant.

[0198] The anti-endoglin antibody can be administered by any appropriate means including, for example, orally, subcutaneously, intravenously, or in an implant.

[0199] In some instances, the anti-endoglin antibody comprises an antigen-binding fragment that specifically binds to endoglin. Antigen-binding fragments include, but are not limited to, a Fab fragment, a Fab' fragment, a F(ab)2 fragment, a F(ab r )2 fragment, a Fv fragment, a scFv fragment, or a single chain binding polypeptide.

[0200] The anti-endoglin antibody can be an IgG, an IgM, an IgE, an IgA, or an IgD, or is derived therefrom. When the antibody is an IgG, the antibody can be an IgGl, an IgG2, an IgG3, or an IgG4. In the case of an IgG2 antibody, the antibody can be an IgG2a antibody or an IgG2b antibody.

[0201] In one non-limiting embodiment, an isolated anti-endoglin antibody comprises chain variable region having an amino acid sequence set forth as SEQ ID NO: 1 and a heavy chain variable region having an amino acid sequence set forth as SEQ ID NO: 3.

[0202] In one non-limiting embodiment, an isolated anti-endoglin antibody comprises a light chain variable region having an amino acid sequence set forth as SEQ ID NO: 1, a light chain constant region having an amino acid sequence set forth as SEQ ID NO: 2, a heavy chain variable region having an amino acid sequence set forth as SEQ ID NO: 3 and a gamma 1 (γΐ) constant region having an amino acid sequence set forth as SEQ ID NO: 4.

[0203] In one non-limiting embodiment, an isolated anti-CD105 antibody comprises a VL CDRl having an amino acid sequence set forth as SEQ ID NO: 10; a V L CDR2 having an amino acid sequence set forth as SEQ ID NO: 11 or 12; a VL CDR3 having an amino acid sequence set forth as SEQ ID NO: 13; a VH CDRl having an amino acid sequence set forth as SEQ ID NO: 5; a VH CDR2 having an amino acid sequence set forth as SEQ ID NO: 6, 7, or 8; and a V H CDR3 having an amino acid sequence set forth as SEQ ID NO: 9.

[0204] In one non-limiting embodiment, an isolated humanized, de-immunized anti-CD 105 antibody can comprise a heavy chain variable region having the amino acid sequence set forth as SEQ ID NO: 14, 15, 16, 17, or 18; and a light chain variable region having the amino acid sequence set forth as SEQ ID NO: 19, 20, 21, 22, or 23.

[0205] Treatment can reduce a size of a cancer or a metastasis thereof by about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, or more compared to administration of a placebo or compared to baseline.

[0206] An "individual" or a "subject" to be treated by a method herein may be a mammal, more preferably a human. Mammals also include, but are not limited to, farm animals, sport animals, and pets, including, but not limited to, primates, equines, bovines, alpacas, dogs, cats, rabbits, mice and rats. It will be appreciated that a subject to be treated may be suffering from a cancer, or a metastasis thereof, but may not yet be symptomatic for the disease.

[0207] "Signs or symptoms of illness" are clinically recognized manifestations or indications of disease.

[0208] As used herein, a "therapeutically effective dosage" or a "therapeutically effective amount" of a pharmaceutical composition described herein is an amount sufficient to effect beneficial or desired results. Beneficial or desired results include results such as lessening the severity or delaying the progression of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.

[0209] An "effective dosage" may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved. Accordingly, in some instances, one or more therapeutic agents may be administered to the subject. In other instances, treatment with a pharmaceutical composition described herein is conducted prior to, or after, one or more other treatment modalities described herein. [0210] By "treating" a subject suffering from a cancer or a metastasis thereof, it is meant that the subject's symptoms can be partially alleviated, totally alleviated, or remain static following treatment according to the invention. A subject that has been treated can exhibit a partial or total alleviation of tumor load. In one non-limiting example, a subject suffering from a highly metastatic cancer is treated where additional metastasis either do not occur, or are reduced in number as compared to a subject who does not receive treatment. In one non-limiting example, a subject is treated where the subject' s solid cancer either becomes reduced in size or does not increase in size as compared to a subject who does not receive treatment. In one non -limiting example, the number of cancer cells in a treated subject either does not increase or is reduced as compared to the number of cancer cells in a subj ect who does not receive treatment. Improvement can also be defined, for example, as decreased cell proliferation, decreased numbers of cells, increased apoptosis, and/or increased survival of the subject being treated.

[0211] It will be understood that a subject can be treated for a length of time sufficient to prolong the life expectancy of the subject; or to partially or completely treat the cancer or metastasis thereof. For example, a subject can be administered treatment for about 1, about 2, about 3, about 4, about 5, about 6, about 7 or more days. A subject can be administered treatment for about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12 or more weeks. A subject can be administered treatment for about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12 or more months. A subject can be administered treatment for about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12 or more years.

[0212] The days, weeks, months or years of treatment can be consecutive. Alternatively, the days, weeks, months or years of treatment may not be consecutive. For example, if the subject enters remission, then treatment can be discontinued; if the subject shows presence of one or more abnormal biomarkers after being in remission, treatment can be reinstated. In other instances, a subject to be treated with the recited methods may have received surgery or treatment with another anti-tumor agent.

[0213] For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of symptoms occurs or until sufficient therapeutic levels are achieved, for example, to reduce pain.

[0214] Non-limiting doses of an anti-endoglin antibody or antigen-binding fragment include, but are not limited to, about 0.01 mg/kg, about 0.05 mg/kg, about 0.1 mg/kg, about 0.5 mg/kg, about 1 mg/kg, about 5 mg/kg, about 10 mg/kg, about 20 mg/kg, about 30 mg/kg, about 40 mg/kg, about 50 mg/kg, about 60 mg/kg, about 70 mg/kg, about 80 mg/kg, about 90 mg/kg, about 100 mg/kg, about 125 mg/kg, about 150 mg/kg, about 175 mg/kg, about 200 mg/kg per subject, or any integer in between.

[0215] The one or more dose(s) of an anti-endoglin antibody or antigen-binding fragment can be administered twice a week, weekly, every two weeks, every three weeks, every 4 weeks, every 6 weeks, every 8 weeks, every 12 weeks, or any combination of weeks therein. Dosing cycles are also contemplated such as, for example, administering antibodies or antigen-binding fragments thereof once or twice a week for 4 weeks, followed by two weeks without therapy. Additional dosing cycles including, for example, different combinations of the doses and weekly cycles described herein are also contemplated within the invention.

[0216] An exemplary dosing regimen of an anti-endoglin antibody comprises administering an initial dose of about 2 mg/kg, followed by a weekly maintenance dose of about 1 mg/kg, or followed by a maintenance dose of about 1 mg/kg every other week. Dosing may continue for a period of about 1 month, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 12 months or more until a cancer is partially or completely eradicated. In some instances, treatment may result in stasis of the cancer, and increases the life expectancy of the subject being treated compared to a subject receiving a placebo.

[0217] Axitinib may be formulated for administration to a subject in an amount of about 0.5 mg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, about 10 mg, about 1 1 mg, about 12 mg, about 13 mg, about 14 mg, or about 15 mg dose once or twice a day.

[0218] Axitinib may be administered about every 12 hours and dose adjustments may be made based upon the subject' s safety, tolerability and/or progression of a cancer to be treated. Axitinib may be administered to a subject orally for at least 2 consecutive weeks. The dose to be administered and the timing of administration for a subject may be empirically determined for each subject depending upon the height, weight, age, the state of the cancer to be treated, and other physical characteristics of the subject to be treated in accordance with a physician's

recommendation.

[0219] In one non-limiting example, an anti-endoglin antibody can be administered at about 8 mg/kg/week and axitinib can be administered at about 5 mg twice a day (BID).

[0220] In one non-limiting example, an anti-endoglin antibody can be administered at about 10 mg/kg/week and axitinib can be administered at about 5 mg twice a day (BID).

[0221] In one non-limiting example, an anti-endoglin antibody can be administered at about 8 mg/kg/week and pazopanib can be administered at about 800 mg once a day (qD). [0222] In one non-limiting example, an anti-endoglin antibody can be administered at about 10 mg/kg/week and pazopanib can be administered at about 800 mg once a day (qD).

[0223] Other dosage regimens may be useful and are contemplated herein, depending on the pattern of pharmacokinetic decay that the treating practitioner wishes to achieve. For example, in some embodiments, dosing from one-four times a week is contemplated. The progress of this therapy is easily monitored by conventional techniques and assays. The dosing regimen can vary over time.

[0224] The appropriate dosage of an anti-endoglin antibody will depend on the anti-endoglin antibody employed, the type and stage of cancer to be treated, previous surgery and/or therapy, the subject's clinical history and response to the antibody, and the discretion of the attending physician.

[0225] Typically the clinician will administer an anti-endoglin antibody, until a dosage is reached that achieves the desired result. Dose and/or frequency can vary over course of treatment.

[0226] Empirical considerations, such as the half-life, generally will contribute to the determination of the dosage. For example, antibodies that are compatible with the human immune system, such as humanized antibodies or fully human antibodies, may be used to prolong half-life of the antibody and to prevent the antibody being attacked by the host's immune system.

[0227] Frequency of administration may be determined and adjusted over the course of therapy. Alternatively, sustained continuous release formulations of anti-endoglin antibodies may be appropriate. Various formulations and devices for achieving sustained release are known in the art. To assess efficacy of an anti-endoglin antibody, an indicator of the disease can be followed.

[0228] In some instances, the pharmaceutical composition comprising the anti-endoglin antibody and the anti-VEGF agent are administered at the same site.

[0229] In other instances, the pharmaceutical composition comprising the anti-endoglin antibody and the anti-VEGF agent are administered at the different sites.

[0230] In some instances, the pharmaceutical composition comprising the anti-endoglin antibody and the anti-VEGF agent are administered sequentially.

[0231] In some instances, the pharmaceutical composition comprising the anti-endoglin antibody and the anti-VEGF agent are administered concurrently.

[0232] As further used herein, treatment of cancer includes stasis, partial or total elimination of a cancerous growth or tumor. Treatment or partial elimination includes, for example, a fold reduction in growth or tumor size and/or volume such as about 2-fold, about 3-fold, about 4-fold, about 5- fold, about 10-fold, about 20-fold, about 50-fold, or any fold reduction in between. Similarly, treatment or partial elimination can include a percent reduction in growth or tumor size and/or volume of about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, 100%, or any percentage reduction in between.

[0233] Treatment efficacy can be assessed by methods well-known in the art.

[0234] Non-limiting examples of tumors include a carcinoma, a sarcoma, a lung cancer, a breast cancer, or a brain cancer.

[0235] The term "tumor" is used herein to refer to a cancerous tissue (as compared to expression by normal tissue of the same type). Tumors can include solid tumors and semi-solid tumors. Tumors may also, in some instances, be metastatic.

[0236] As used herein, "amelioration" "inhibition," "treatment" and "treating" refer to, for example, stasis of symptoms, prolongation of survival, partial or full amelioration of symptoms, and partial or full eradication of a condition, disease or disorder associated with a cancer or a metastasis thereof.

[0237] In one embodiment, a cancer or a metastasis thereof is inhibited by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%), or about 100%, following treatment with one or more doses of a humanized and deimmunized antibody compared to treatment with a placebo or compared to a subject that does not receive any treatment.

[0238] In one embodiment, symptoms of a cancer or a metastasis thereof may be ameliorated by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%, following treatment with one or more doses of an anti-endoglin antibody compared to treatment with a placebo or compared to a subject that does not receive any treatment.

[0239] Treatment also refers to resolution of one or more symptoms of a cancer or a metastasis thereof. Treatment also refers to stasis of symptoms where a cancer or a metastasis thereof in a subject does not progress.

[0240] Pain associated with a cancer or a metastasis thereof may be reduced by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%), about 95%), or about 100%, following treatment with one or more doses of an anti-endoglin antibody compared to treatment with a placebo or compared to a subject that does not receive any treatment. [0241] "Administering" is defined herein as a means providing the composition to the subject in a manner that results in the composition being inside the subject's body. Such an administration can be by any route including, without limitation, locally, regionally or systemically by subcutaneous, intravitreal, intradermal, intravenous, intra-arterial, intraperitoneal, or intramuscular administration {e.g., injection), or by oral administration. "Concurrent administration" means administration within a relatively short time period from each other; such time period can be less than 2 weeks, less than 7 days, less than 1 day and could even be administered simultaneously.

[0242] Compositions can be administered to a subject in a therapeutically effective amount, i.e., that are effective for producing some desired therapeutic effect by inhibiting a cancer or a metastasis thereof such as described herein which can be associated with endoglin, at a reasonable benefit risk ratio applicable to any medical treatment. A therapeutically effective amount is an amount achieves at least partially a desired therapeutic or prophylactic effect in an organ or tissue. The amount of an anti- endoglin antibody or antigen binding fragment thereof necessary to bring about prevention and/or therapeutic treatment of a cancer or a metastasis thereof is not fixed per se. The amount of anti -endoglin antibody or antigen binding fragment thereof administered may vary with the type of a cancer or a metastasis thereof, extensiveness of the cancer or a metastasis thereof, and size of the mammal suffering from the cancer or a metastasis thereof. In one embodiment, two or more anti-endoglin antibodies described herein are administered to a subject in combination. Combination includes concomitant or subsequent administration of the antibodies.

[0243] Actual dosage levels of the active ingredients in the compositions can be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular subject, composition, and mode of administration, without being toxic to the subject. The selected dosage level will depend upon a variety of factors including the activity of the particular compound employed, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular composition employed, the age, sex, weight, condition, general health and prior medical history of the subject being treated, and like factors well known in the medical arts. The antibodies and antigen-binding fragments described herein can be administered to a subject in various dosing amounts and over various time frames.

Sarcoma

[0244] A sarcoma is a cancer (malignant tumor) of connective tissue (e.g., bone, cartilage, fat) or other non-epithelial tissue resulting in mesoderm proliferation. [0245] A sarcoma to be treated with the recited methods may be a soft tissue sarcoma of endothelium (angiosarcoma), synovial tissues (tissues around joints), muscles (e.g., a

leiomyosarcoma), tendons, cartilage (e.g., a chondrosarcoma), nerves, fat, fibrous tissues, stromal tissue (e.g., a Gastrointestinal Stromal Tumor (GIST)), and/or blood vessels. In some instances, a sarcoma to be treated is a bone sarcoma (osteosarcoma).

Carcinoma

[0246] A carcinoma is any malignant cancer that arises from epithelial cells (e.g., breast, colon, pancreas, and others) of the skin or of the lining of the internal organs (e.g., liver or kidney).

Carcinomas can, in some instances, invade surrounding tissues and organs and may metastasize, or spread, to lymph nodes and other sites.

[0247] A carcinoma to be treated by the described methods includes, but is not limited to, an adenocarcinoma; a squamous cell carcinoma; a small cell carcinoma; a sinonasal undifferentiated carcinoma (S UC); an undifferentiated carcinoma; a carcinoma of the prostate; a hepatocellular carcinoma; or a renal cell carcinoma. In some embodiments, the carcinoma to be treated is a renal cell carcinoma that can be in some instances, an advanced renal cell carcinoma or a metastasized renal cell carcinoma. In some instances, a subject having a renal cell carcinoma may be symptomless (asymptomatic) until the disease is in an advanced stage.

Lung cancer

[0248] In a method described herein, a subject in need thereof having a lung cancer is administered a therapeutically effective amount of a pharmaceutical composition that comprises an antibody, or antigen-binding fragment thereof described herein.

[0249] A lung cancer to be treated with the described methods can be, for example, a non-small cell lung cancer, a lung carcinoid tumor, or a small cell lung cancer. A non-small cell lung cancer (NSCLC) can be, for example, a squamous cell carcinoma, an adenocarcinoma, or a large cell undifferentiated carcinoma.

Breast cancer

[0250] In a method described herein, a subject in need thereof having a breast cancer is administered a therapeutically effective amount of a pharmaceutical composition that comprises an antibody, or antigen-binding fragment thereof described herein. As used herein, "breast cancer" also encompasses a phenotype that displays a predisposition towards developing breast cancer in an individual.

[0251] A breast cancer to be treated using the methods described herein includes any type of breast cancer that can develop in a female subject. For example, the breast cancer may be characterized as Luminal A (ER+ and/or PR+, HER2-, low Ki67), Luminal B (ER+ and/or PR+, HER2+ (or HER2- with high Ki67), Triple negative/basal-like (ER-, PR-, HER2-) or HER2 type (ER-, PR-, HER2+). In another example, the breast cancer may be resistant to therapy or therapies such as alkylating agents, platinum agents, taxanes, vinca agents, anti-estrogen drugs, aromatase inhibitors, ovarian suppression agents, endocrine/hormonal agents, bisphophonate therapy agents or targeted biological therapy agents.

[0252] A lobular carcinoma in situ and a ductal carcinoma in situ are breast cancers that have developed in the lobules and ducts, respectively, but have not spread to the fatty tissue surrounding the breast or to other areas of the body. Infiltrating (or invasive) lobular and ductal carcinoma are cancers that have developed in the lobules and ducts, respectively, and have spread to either the breast's fatty tissue and/or other parts of the body. In one aspect, provided herein is a method of treating breast cancer, such as a ductal carcinoma in duct tissue in a mammary gland, a breast cancer that is Her2- and/or ER- and/or PR-. Other cancers of the breast that would benefit from treatment by the methods are medullary carcinomas, colloid carcinomas, tubular carcinomas, and inflammatory breast cancer.

Brain cancer

[0253] Antibodies, or antigen-binding fragments thereof, described herein can be modified so that they are able to cross the blood-brain barrier. Such modification of the antibodies or antigen- binding fragments described herein allows for the treatment of brain diseases such as glioblastoma multiforme (GBM). Exemplary modifications to allow proteins such as antibodies or antigen- binding fragments to cross the blood-brain barrier are described in US Patent Application

Publication 2007/0082380 which is hereby incorporated by reference with respect to modification of antibodies to cross the blood brain barrier.

[0254] Brain tumors to be treated using the methods described herein include, but are not limited to, a meningioma, an astrocytoma such as glioblastomas {e.g., glioblastoma multiforme (GBM)), and a malignant medulloblastoma.

EXAMPLES

[0255] The application may be better understood by reference to the following non-limiting examples, which are provided as exemplary embodiments of the application. The following examples are presented in order to more fully illustrate embodiments and should in no way be construed, however, as limiting the broad scope of the application.

EXAMPLE 1

[0256] The data presented in this application were obtained using samples from a Phase lb portion of the clinical trial "A Randomized Phase 2 Trial of Axitinib (INLYTA ® ) and TRC 105 Versus Axitinib Alone in Subjects With Advanced or Metastatic Renal Cell Carcinoma", from "A Phase lb dose-escalation study of TRC105 in combination with pazopanib in subjects with advanced soft tissue sarcoma", "An open label Phase lb dose-finding study of TRC105 in combination with capecitabine in subjects with advanced or progressive metastatic breast cancer", and "A Phase 2 evaluation of TRC 105 in combination with bevacizumab for the treatment of recurrent or progressive glioblastoma that has progressed on bevacizumab". All analyses except for BMP-9 and TGFP-R3 were performed using the CiraScan platform produced by Aushon BioSystems, Inc.

[0257] In all, 23 analytes (also referred to as markers or biomarkers) were evaluated {see, Table 1 below). The characteristics of plasma analytes were investigated using a variety of measures. Baseline and on-treatment levels were quantified and changes among analytes were determined at the desired time points (waterfall plots; data not shown).

[0258] Comparison of baseline values in renal cell carcinoma subjects with response to treatment with axitinib and TRC 105 by RECIST 1.1 versus non-responders was done to explore potential baseline characteristics that would predict a response to treatment. Baseline values were also correlated with time on treatment to explore potential baseline characteristics that would predict a longer duration of treatment without progression or death, or progression free survival (PFS).

[0259] Comparison of baseline values in soft tissue sarcoma subjects with response to treatment with pazopanib and TRC105 by Choi criteria versus non-responders was done to explore potential baseline characteristics that would predict a response to treatment. Choi criteria are as described in Choi H et al, J. Clin. One, 2007.

Table 1. List of plasma-based biomarkers.

[0260] Abbreviations of the biomarkers of Table 1 are as follows: ANG-2: Angiopoietin-2; bFGF: Basic fibroblast growth factor; HGF: hepatocyte growth factor; PDGF: Platelet-derived growth factor; TSP: Thrombospondin; PIGF: placenta growth factor; VEGF: Vascular endothelial growth factor; BMP: bone morphogenetic protein; TGF: transforming growth factor; ICAM: Intercellular Adhesion Molecule; IL: Interleukin; OPN: Osteopontin; SDF: stromal cell -derived factor; TEVIP: tissue inhibitor of metalloproteinase; and VCAM: Vascular cell adhesion protein.

Data sets:

[0261] There are 18 subjects within two treatment cohorts for the Phase lb study of TRC105 and axitinib in subjects with advanced or metastatic renal cell carcinoma ("mRCC"), three of whom received TRC105 (8 mg/kg/week) and axitinib at 5 mg twice a day and 1 of whom received TRC105 (10 mg/kg/week) and axitinib at 5 mg twice daily. Both drugs were given in combination starting at day 1.

[0262] There are 18 subjects within two treatment cohorts for the Phase lb study of TRC105 and pazopanib in subjects with soft tissue sarcoma ("Sarcoma"), three of whom received TRC105 (8 mg/kg/week) and pazopanib at 800 mg daily and 15 of whom received TRC105 (10 mg/kg/week) and pazopanib at 800 mg twice daily. Pazopanib dosing was given starting on day 1 and TRC105 treatment was started 2 to 4 weeks thereafter.

[0263] There are 19 subjects within two treatment cohorts for the Phase lb study of TRC105 and capecitabine in subjects with metastatic breast cancer ("mBC"), three of whom received TRC 105 (7.5 mg kg/week) and capecitabine at 1,000 mg/m 2 twice a day and 16 of whom received TRC 105 (10 mg/kg/week) and capecitabine at 1,000 mg/m 2 twice a day. Both drugs were given in combination starting at day 1.

[0264] There are 22 subjects within two treatment cohorts for the Phase lb study of TRC105 and bevacizumab in subjects with glioblastoma ("GBM"), three of whom received TRC105 (10 mg/kg/week) as a single agent and 16 of whom received TRC105 (10 mg/kg/week) and bevacizumab at 10 mg/kg every two weeks, starting on day 1.

[0265] Plasma samples were collected at Baseline (Day 1) prior to initiation of treatment in all studies.

OA/QC:

[0266] All samples were run in duplicate, and data points falling above or below the standard curve were imputed to provide reasonable estimates. Most analytes had Coefficient of Variations (CVs) within the range of standard ELISAs (5-15%).

Results:

1. Biomarker levels

[0267] The data are presented in Tables 2 (mRCC and Sarcoma) and 3 (mBC and GBM). Table 2. Biomarker level at baseline for metastatic

Renal Cell Carcinoma (mRCC) and Sarcoma Table 3. Biomarker level at baseline for

metastatic breast cancer (mBC) and glioblastoma multiforme (GBM)

r 22 of the 23 biomarkers, the median and range are reported. [0269] In a study of 18 subjects with advanced or metastatic renal cell carcinoma, there were 5 partial responders with more than 30% tumor size reduction. We compared these responders with the rest of the subjects. Baseline levels in two markers were significantly different (Figures 1A-B).

[0270] In a study of 18 subjects with advanced or metastatic soft tissue sarcoma, there were 6 partial responders by Choi criteria with 10% or greater tumor size reduction. We compared these responders with the rest of the subjects. Baseline levels in two markers were significantly different (Figures 2A-B).

2. Biomarker correlation with Time on Study

[0271] Baseline level and on-treatment change of each biomarker in subjects with advanced or metastatic renal cell carcinoma was correlated with time on study until disease progression or death, or progression free survival (PFS). Markers with significant correlation (p<0.05) or showing a potential trend (0.05<p<0.15) are listed in Table 4.

3. Differential biomarker features in responders;

[0272] As a systemic approach, in a trial of subjects with renal cell carcinoma, there were 5 of 18 responder subjects treated with TRC 105 and axitinib who exhibited more than 30% decrease of their tumor size, and stayed on trial for more than 9 months. We compared biomarker baseline levels in these 5 subjects to the rest of the population. OPN (Figure 1A) and TGFp-R3 (Figure IB) were differentially expressed markers at baseline in responders versus non-responders.

[0273] As a systematic approach, in a trial of subjects with renal cell carcinoma treated with TRC105 and axitinib, we correlated baseline level with time on treatment (Table 4). At baseline, high expression levels of Ang-2, VEGF-R2, and TGFP-R3 correlated with longer time on study, with hazard ratios of 0.61 (95% CI: 0.41 -0.92) for Ang-2, 0.21 (95% CI: 0.04-1.03) for VEGF-R2, and 0.13 (95% CI: 0.02-0.99) for TGF -R3, respectively. In contrast, high levels of OPN correlated with shorter time on study (HR: 1.38, 95% CI: 1.01-1.89).

Table 4. Biomarkers significantly correlated with Time on Treatment.

[0274] In a trial of subjects with soft tissue sarcoma, there were 6 of 18 responder subjects treated with TRC105 and pazopanib who had a partial response by Choi criteria as evidenced by at least a 10%) decrease of their tumor size. We compared biomarker baseline levels in these 6 subjects to the rest of the population. ICAM-1 and TSP-2 were differentially expressed markers at baseline in responders versus non-responders (Figure 2A-B)

[0275] In one non-limiting aspect, in a method described herein, a subject with advanced or metastatic renal cell carcinoma presenting to a treating physician would have a sample of blood taken from a peripheral vein, from which plasma would be obtained. The plasma would be subject to an ELISA to measure concentrations of one or more soluble proteins (e.g., such as those described in Table 1), including angiopoietin, VEGFR2, osteopontin and TGFP-R3. Based on the concentrations of the one or more soluble proteins, including but not limited to, an elevation of TGFP-R3, angiopoietin, or VEGFR2 above the median or mean level of the biomarker in subjects with the specific cancer type and/or a decrease in osteopontin below the median or mean level of the biomarker in subjects with the specific cancer type, the subject would be identified as a subject to be treated with a VEGF inhibitor (including but not limited to axitinib) and an anti-endoglin antibody (including, but not limited to, TRC 105).

[0276] In one non-limiting aspect, in a method described herein, a subject with advanced or soft tissue sarcoma presenting to a treating physician would have a sample of blood taken from a peripheral vein, from which plasma would be obtained. The plasma would be subject to an ELISA to measure concentrations of one or more soluble proteins (e.g., such as those described in Table 1), including ICAM-1 and TSP-2. Based on the concentrations of the one or more soluble proteins, including but not limited to, a decrease in ICAM-1 or TSP-2 below the median or mean level of the biomarker in subjects with the specific cancer type, the subject would be identified as a subject to be treated with a VEGF inhibitor (including but not limited to pazopanib) and an anti-endoglin antibody (including, but not limited to, TRC 105).

[0277] The described methods can be used, in one example, to identify a subject for treatment when ANG-2 is detected.

[0278] The described methods can be used, in one example, to identify a subject for treatment when ANG-2 is detected in combination with VEGF-R2, OPN and/or TGFp-R3.

[0279] The described methods can be used, in one example, to identify a subject for treatment when VEGF-R2 is detected.

[0280] The described methods can be used, in one example, to identify a subject for treatment when VEGF-R2 is detected in combination with ANG-2, OPN and/or TGFP-R3.

[0281] The described methods can be used, in one example, to identify a subject for treatment when OPN is detected.

[0282] The described methods can be used, in one example, to identify a subject for treatment when OPN is detected in combination with ANG-2, VEGF-R2 and/or TGFP-R3. [0283] The described methods can be used, in one example, to identify a subject for treatment when TGF -R3 is detected.

[0284] The described methods can be used, in one example, to identify a subj ect for treatment when TGFP-R3 is detected in combination with ANG-2, VEGF-R2 and/or OPN.

[0285] While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.