Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMBINATION OF T-CELL CHECKPOINT INHIBITORS WITH INHIBITORS OF E-SELECTIN OR CXCR4, OR WITH HETEROBIFUNCTIONAL INHIBITORS OF BOTH E-SELECTIN AND CXCR4
Document Type and Number:
WIPO Patent Application WO/2018/031445
Kind Code:
A1
Abstract:
Compositions and methods for the treatment of diseases, disorders, and/or conditions associated with the increased regulatory T lymphocyte cell (Treg cell) function, comprising the administration of T-cell checkpoint inhibitors in combination with E-selectin inhibitors, CXCR4 receptor inhibitors, and/or heterobifunctional inhibitors that comprise at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor, are disclosed.

Inventors:
MAGNANI JOHN L (US)
FOGLER WILLIAM E (US)
Application Number:
PCT/US2017/045690
Publication Date:
February 15, 2018
Filing Date:
August 07, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
GLYCOMIMETICS INC (US)
International Classes:
C07K16/28; A61K31/7034; A61K39/00
Domestic Patent References:
WO2015019284A22015-02-12
WO2013096926A12013-06-27
WO2016089872A12016-06-09
Foreign References:
US9109002B22015-08-18
US8410066B22013-04-02
US20150063191W2015-12-01
Other References:
YUNCHING CHEN ET AL: "CXCR4 inhibition in tumor microenvironment facilitates anti-programmed death receptor-1 immunotherapy in sorafenib-treated hepatocellular carcinoma in mice : Hepatobiliary Malignancies", HEPATOLOGY, vol. 61, no. 5, 20 March 2015 (2015-03-20), pages 1591 - 1602, XP055399883, ISSN: 0270-9139, DOI: 10.1002/hep.27665
ARJAN W. GRIFFIOEN ET AL: "Angiostasis as a way to improve immunotherapy", THROMBOSIS AND HAEMOSTASIS, vol. 101, no. 6, 12 May 2009 (2009-05-12), DE, pages 1025 - 1031, XP055412916, ISSN: 0340-6245, DOI: 10.1160/TH08-08-0552
ABDEL KAREEM AZAB ET AL: "CXCR4 inhibitor AMD3100 disrupts the interaction of multiple myeloma cells with the bone marrow microenvironment and enhances their sensitivity to therapy", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, UNITED STATES, vol. 113, no. 18, 30 April 2009 (2009-04-30), pages 4341 - 4351, XP002663167, ISSN: 1528-0020, [retrieved on 20090112], DOI: 10.1182/BLOOD-2008-10-186668
LIU ET AL.: "Targeting Regulatory T Cells in Tumors", FEBS J., vol. 283, 2016, pages 2731 - 2748
HENDRIX ET AL., ANTIMICROB. AGENTS CHEMOTHER, vol. 44, 2000, pages 1667 - 1673
DORANZ ET AL., AIDS RESEARCH AND HUMAN RETROVIRUSES, vol. 17, 2001, pages 475 - 488
ARAKAKI ET AL., J. VIROL., vol. 73, 1999, pages 1719 - 1723
PRICE: "Dormant breast cancer micrometastases reside in specific bone marrow niches that regulate their transit to and from bone", SCIENCE TRANSLATIONAL MEDICINE, vol. 8, no. 340, 25 May 2016 (2016-05-25)
DUTTA ET AL.: "E-Selectin Inhibition Mitigates Splenic HSC Activation and Myelopoiesis in Hypercholesterolemic Mice With Myocardial Infarction", ARTERIOSCLER THROMB VASE BIOL
STEELE, MARIA M. ET AL.: "A small molecule glycomimetic antagonist of E-selectin and CXCR4 (GMI-1359) prevents pancreatic tumor metastasis and improves chemotherapy [abstract", PROCEEDINGS OF THE 106TH ANNUAL MEETING OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH, 18 April 2015 (2015-04-18)
"Cancer Res", vol. 75, 2015, AACR
GRAVINA, GIOVANNI L. ET AL.: "Dual E-selectin and CXCR4 inhibition reduces tumor growth and increases the sensitivity to docetaxe! in experimental bone metastases of prostate cancer [abstract", PROCEEDINGS OF THE 106TH ANNUAL MEETING OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH, 18 March 2015 (2015-03-18)
"Remington: The Science and Practice of Pharmacy", 2005, MACK PUB. CO.
"Transdermal Delivery of Drugs", 1987, CRL PRESS
"Percutaneous Penetration Enhancers", 1995, CRC PRESS
LENNERAS ET AL., J. PHARM. PHARMACOL., vol. 54, 2002, pages 499 - 508
KARANDE ET AL., PHARM. RES., vol. 19, 2002, pages 655 - 660
VADDI ET AL., INT. J. PHARM., vol. 91, 2002, pages 1639 - 1651
VENTURA ET AL., J. DRUG TARGET, vol. 9, 2001, pages 379 - 393
SHOKRI ET AL., INT J. PHARM., vol. 228, no. 1-2, 2001, pages 99 - 107
SUZUKI ET AL., BIOL. PHARM BULL., vol. 24, 2001, pages 698 - 700
ALBERTI ET AL., J. CONTROL RELEASE, vol. 71, 2001, pages 319 - 327
GOLDSTEIN ET AL., UROLOGY, vol. 57, 2001, pages 301 - 305
KIIJAVAINEN ET AL., EUR. J. PHARM. SCI, vol. 10, 2000, pages 97 - 102
TENJARIA ET AL., INT. J. PHARM., vol. 192, 1999, pages 147 - 158
CALLAHAN MK ET AL.: "Anti-CTLA-4 Antibody Therapy: Immune Monitoring During Clinical Development of a Novel Immunotherapy", SEMIN ONCOL., vol. 37, no. 5, October 2010 (2010-10-01), pages 473 - 484
POSTOW MA; CALLAHAN MK; WOLCHOK JD: "Immune Checkpoint Blockade in Cancer Therapy", J. CLIN. ONCOL., January 2015 (2015-01-01)
DUNN GP; OLD LJ; SCHREIBER RD.: "The Immunobiology of Cancer Immunosurveillance and Immunoediting", IMMUNITY., vol. 21, August 2004 (2004-08-01), pages 137 - 148
NAIDOO J; PAGE DB; WOLCHOK JD.: "Immune modulation for cancer therapy", BRITISH JOURNAL OF CANCER, vol. 111, September 2014 (2014-09-01), pages 2214 - 2219, XP055211569, DOI: doi:10.1038/bjc.2014.348
KIM R; EMI M; TANABE K.: "Cancer immunoediting from immune surveillance to immune escape", IMMUNOLOGY, vol. 121, no. 1, May 2007 (2007-05-01), pages 1 - 14
SCHABEL F; GRISWOLD D; LASTER W; CORBETT T; LLOYD H: "Quantitative evaluation of anticancer agent activity in experimental animals", PHARMAC. THER, A, vol. 1, 1977, pages 411 - 435, XP023855142, DOI: doi:10.1016/0362-5478(77)90004-3
CORBETT, T; GRISWOLD D; ROBERTS B.; PECKHAM J; SCHABEL F: "Evaluation of single agents and combinations of chemotherapeutic agents in mouse colon carcinomas", CANCER, vol. 40, no. 5, 1977, pages 2660 - 2690, XP000650936
SCHABEL F; GRISWOLD D; CORBETT T; LASTER R; MAYO J; LLOYD H: "Testing therapeutic hypotheses in mice and man: Observations on the therapeutic activity against advanced solid tumors of mice treated with anticancer drugs that have demonstrated or potential clinical utility for treatment of advanced solid tumors of man", METHODS IN CANCER RESEARCH, vol. 17, 1979, pages 3 - 51, XP001246662
PLOWMAN J; DYKES D; HOLLINGSHEAD M; SIMPSON-HERREN L; ALLEY M: "Teicher", 1993, HUMANA PRESS INC., article "Human tumor xenograft models in NCI drug development. In: Anticancer drug development guide: preclinical screening, clinical trials, and approval"
CORBETT T; VALERIOTE F; LORUSSO P; POLIN L; PANCHAPOR C.; PUGH S.; WHITE K; KNIGHT J; DEMCHIK L; JONES J: "Tumor models and the discovery and secondary evaluation of solid tumor active agents", INT J PHARMACOGNOSY, vol. 33, 1995, pages 102 - 122
CORBETT T; ROBERTS BJ; LAWSON AJ; LEOPOLD WR ET AL.: "Tumor Models in Cancer Research", 2011, HUMANA PRESS, article "Transplantable Syngeneic Rodent Tumors: Solid Tumors of Mice", pages: 43 - 78
Attorney, Agent or Firm:
CHAPMAN, Ernest F. et al. (US)
Download PDF:
Claims:
What is claimed is:

1. A method for treating and/or preventing at least one disease, disorder, or condition comprising administering to a subject in need thereof

(1) an effective amount of at least one T-cell checkpoint inhibitor, and

(2) an effective amount of at least one other inhibitor chosen from E-selectin inhibitors, CXCR4 receptor inhibitors, and heterobifunctional inhibitors that comprise at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor.

2. The method of claim 1 , wherein at least one of (1 ) the at least one T-cell checkpoint inhibitor and (2) the at least one other inhibitor is in the form of at least one pharmaceutical composition.

3. The method of claim 1 or 2, wherein the at least one T-cell checkpoint inhibitor is in the form of a first pharmaceutical composition and the at least one other inhibitor is in the form of a second pharmaceutical composition.

4. The method of claim 2, wherein said at least one pharmaceutical composition further comprises at least one pharmaceutically acceptable ingredient.

5. The method of claim 1 , wherein said at least one other inhibitor is chosen from E-selectin inhibitors.

6. The method of claim 1 , wherein said at least one other inhibitor is chosen from CXCR4 receptor inhibitors.

7. The method of claim 1 , wherein said at least one other inhibitor is chosen from heterobifunctional inhibitors that comprise at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor.

8. The method of any preceding claims, wherein the at least one T-cell checkpoint inhibitor targets the PD-1 receptor and/or the CTLA-4 protein on Treg cells.

9. The method of any of the preceding claims, wherein the at least one T-cell checkpoint inhibitor is chosen from nivolumab and ipilimumab.

10. The method of claim 1 , wherein the E-selectin inhibitors are chosen from sialyl Lewisx (sLex) and sLex mimetics.

11. The method of claim 1 , wherein the E-selectin inhibitors are chosen from small molecule glycomimetic antagonists of E-selectin, antibodies directed to E- selectin, aptamers to E-selectin, peptides directed to E-selectin, and peptidomimetics directed to E-selectin.

12. The method of claim 0, wherein the E-selectin inhibitors are chosen from compounds of Formula (I):

isomers of Formula (I), tautomers of Formula (I), and pharmaceutically acceptable salts of any of the foregoing, wherein:

R1 is chosen from C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C1-C8 haloalkyl, C2-C8 haloalkenyl, and C2-C8 haloalkynyl groups;

R2 is chosen from H, -M, and -L-M;

R3 is chosen from -OH, -NH2, -OC(=O)Y1, -NHC(=O)Y1, and

-NHC(=O)NHY1 groups, wherein Y1 is chosen from C1-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C1-8 haloalkyl, C2-8 haloalkenyl, C2-8 haloalkynyl, C6-18 aryl, and C1 -13 heteroaryl groups;

R4 is chosen from -OH and -NZ1Z2 groups, wherein Z1 and Z2, which may be identical or different, are each independently chosen from H, C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C1-C8 haloalkyl, C2-C8 haloalkenyl, and C2-C8 haloalkynyl groups, wherein Z1 and Z2 may together form a ring;

R5 is chosen from C3-C8 cycloalkyi groups;

R6 is chosen from -OH, C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C1-C8 haloalkyl, C2-C8 haloalkenyl, and C2-C8 haloalkynyl groups;

R7 is chosen from -CH2OH, C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C1-C8 haloalkyl, C2-C8 haloalkenyl, and C2-C8 haloalkynyl groups;

R8 is chosen from C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C1-C8 haloalkyl, C2-C8 haloalkenyl, and C2-C8 haloalkynyl groups;

L is chosen from linker groups; and

M is a non-glycomimetic moiety chosen from polyethylene glycol, thiazolyl, chromenyl, -C(=O)NH(CH2)1-4NH2, Cr8 alkyl, and -C(=O)OY, wherein Y is chosen from C1-4 alkyl, C2-4 alkenyl, and C2-4 alkynyl groups.

13. The method of claim 12, wherein the E-selectin inhibitors are chosen from compounds of Formula (la):

wherein n is chosen from integers ranging from 1 to 100.

14. The method of claim 1 , wherein the CXCR4 receptor inhibitors are chosen from peptides, diketopiperazine mimetics, bicyclams, tetrahydroquinolines, thiazolylisothiourea derivatives, and benzodiazepines.

15. The method of claim 1 , wherein the CXCR4 receptor inhibitors are chosen from AMD-3100, ALX40-4C, T134, and T22 peptide.

16. The method of claim 1 , wherein the heterobifunctional inhibitors are chosen from compounds of Formula (II):

isomers of Formula (II), tautomers of Formula (II), and pharmaceutically acceptable salts of any of the foregoing, wherein:

R1 is chosen from H, C1-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C1-8 haloalkyl, C2-8 haloalkenyl, and C2-8 haloalkynyl groups;

R2 is chosen from -OH, -NH2, -OC(=O)Y1, -NHC(=O)Y1, and

-NHC(=O)NHY1 groups, wherein Y1 is chosen from C1-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C1-8 haloalkyl, C2-8 haloalkenyl, C2-8 haloalkynyl, C6-18 aryl, and C1 -13 heteroaryl groups;

R3 is chosen from -CN, -CH2CN, and -C(=O)Y2 groups, wherein Y2 is chosen from C1-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, -OZ1, -NHOH, -NHOCH3, -NHCN, and - NZ1Z2 groups, wherein Z1 and Z2, which may be identical or different, are

independently chosen from H, C1-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C1-8 haloalkyl, C2-8 haloalkenyl, and C2-8 haloalkynyl groups, wherein Z1 and Z2 may together form a ring;

R4 is chosen from C3-8 cycloalkyl groups;

Rs is independently chosen from H, halo, C1-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C1-8 haloalkyl, C2-8 haloalkenyl, and C2-8 haloalkynyl groups;

n is chosen from integers ranging from 1 to 4; and L is chosen from linker groups.

17. The method of claim 1 , wherein the heterobifunctional inhibitors are chosen from compounds of Formula (lla):

18. The method of claim 1 , wherein the E-selectin inhibitor is GMI-127 ,

19. The method of claim 1 , wherein the heterobifunctional inhibitor is G I-1359.

20. The method of any preceding claim, wherein the at least one disease, disorder, or condition is chosen from cancers.

21. The method of claim 20, wherein the cancers are liquid cancers,

22. The method of claim 20, wherein the cancers are solid cancers.

23. The method of claim 22, wherein the subject is treated locally at a tumor of the solid cancer.

24. The method of any preceding claim, wherein the subject has cancer and has received or will receive chemotherapy and/or radiotherapy.

25. The method of claim 24, wherein the chemotherapy comprises administering an effective amount of bortezomib and/or gemcitabine.

26. The method of any of claims 1-19, wherein the at least one disease, disorder, or condition is chosen from bacterial infections, viral infections, conditions relating at least one bacterial infection, and conditions relating to at least one viral infection.

27. The method of claim 26, wherein the at least one disease, disorder, or condition is an HIV infection.

28. The method of claim 26, wherein the at least one disease, disorder, or condition is sepsis.

Description:
COMBINATION OF T-CELL CHECKPOINT INHIBITORS WITH INHIBITORS OF E-SELECTIN OR

CXCR4, OR WITH HETEROBIFUNCTIONAL INHIBITORS OF BOTH E-SELECTIN AND CXCR4.

[001] This Application claims priority to U.S. Provisional Application Nos.

62/372,116, filed August 8, 2016, 62/418,722, filed November 7, 2016, and

62/417,045, filed November 3, 2016; the disclosures of which are incorporated herein by reference.

[002] Compositions and methods for the treatment of diseases, disorders, and/or conditions associated with the increased regulatory T lymphocyte cell (T reg cell) function, comprising the administration of T-cell checkpoint inhibitors in combination with E-selectin inhibitors, CXCR4 receptor inhibitors, and/or

heterobifunctional inhibitors which comprise at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor, are disclosed. One such disease, disorder, and/or condition is cancer.

[003] It is now understood that cancer tumors are not masses of solely malignant cells, but instead comprise some malignant cells and some recruited normal cell types. Liu et al., "Targeting Regulatory T Cells in Tumors," FEBS J. 283:2731-48, at 2731 (2016). The malignant cells and normal cells both play roles in promoting tumor growth and metastasis. Id. One of the normal cells that supports cancer progression in the tumor microenvironment are Treg cells.

[004] Treg cells down-regulate other immune cells, thereby playing an important role in, for example, preventing autoimmunity. In the tumor microenvironment, malignant cells can attract T reg cells and increase the local concentration of cytokines expressed by Treg cell that down-regulate other immune cells. T reg cells are induced and maintained by immunoregulatory receptors, such as PD-1. T reg cells also respond to homing signals within the inflamed tumor microenvironment that include the endothelial ceil surface protein, E-selectin, and the CXCR4 ligand, SDF-1. Using these pathways, the cancer cells use T reg cells to prevent other immune cells from attacking the cancer. Thus, although the immune system is often able to produce a response against the malignancy, due at least in part to the influence of T^ cells, this response is often insufficient to eliminate the tumor.

[005] This realization led to the interest in blocking immunoregulatory receptors called checkpoint proteins on T cells. By blocking the checkpoint proteins on T cells, the T cells are no longer able to down-regulate the rest of the immune response, and the immune cells are able to attack the malignant cancer cells. FDA approval has already been granted to two of these immune checkpoint inhibitors, nivolumab (Opdivo®) and ipilimumab (Yervoy®). Nivolumab inhibits the activity of a protein receptor called PD-1 on T cells and ipilimumab binds to a checkpoint protein on the surface of T cells called CTLA-4. These T-cell checkpoint inhibiting drugs may be used in combination with standard cancer treatments, such as radiation therapy and chemotherapy.

[006] The T-cell checkpoint inhibitors may be useful in treating cancer, but the course of treatment is usually lengthy and there are several side-effects.

Furthermore, treatment with T-cell checkpoint inhibitors only target one aspect of the cancer cells' commandeering of the immune system - the inducement and maintenance of T reg cells - and it does not address the recruitment of the T cells to the tumor microenvironment by E-selectin and/or SDF-1.

[007] Accordingly, there is an unmet need for additional compositions and treatments for suppressing T reg cell function for the treatment of cancer, in particular, compositions and treatments that address the E-selectin and/or SDF-1 related pathways. There is also an unmet need for additional compositions and treatments for suppressing Tng cell function for the treatment of other diseases, disorders, and/or conditions related to over-active or numerous Treg cells, such as bacterial and viral infections including sepsis, septic conditions, and HIV infection.

BRIEF DESCRIPTION OF DRAWINGS

[008] Those of ordinary skill in the art will understand that the drawings, described below, are for illustrative purposes only. The drawings are not intended to limit the scope of the present teachings in any way.

[009] Figure 1 is a graph of the mean tumor growth, shown as mean tumor burden in mm 3 , in 12 groups of experimental mice (control groups included as well), with standard error.

[010] Figure 2 is a graph of the median tumor growth, shown as median tumor burden in mm 3 , in 12 groups of experimental mice (control groups included as well), with standard error.

[011 ] Figures 3A-D are graphs of the tumor growth of individual mice of group 1 (saline control group, Fig. 3A); group 2 (GMI-1359 treatment, Fig. 3B); group 3 (isotype control antibody LTF-2, Fig. 3C); and group 4 (anti-PD-L1 antibody treatment, Fig. 3D), shown as tumor burden in mm 3 and including control median and group median.

[012] Figures 4A-D are graphs of the tumor growth of individual mice of group 5 (GMI-1359 and LTF-2 antibody, Fig. 4A); group 6 (GMI-1359 and anti-PD-L1 antibody treatment, Fig. 4B); group 7 (saline control, Fig. 4C); and group 8 (GMI- 1359 treatment, Fig. 4D), shown as tumor burden in mm 3 and including control median and group median. [013] Figures 5A-D are graphs of the tumor growth of individual mice of group 9 (LTF-2 antibody, Fig. 5A); group 10 (anti-PD-L1 antibody treatment, Fig. 5B); group 11 (GMI-1359 and LTF-2 antibody, Fig. 5C); and group 12 (GMI-1359 and anti-PD- L1 antibody treatment, Fig. 5D), shown as tumor burden in mm 3 and including control median and group median.

[014] Figure 6 is a graph of the percent mean body weight change for 12 experimental groups (control groups included as well), with standard error.

[015] Figure 7 is a graph of the percentage of CD4 + /CCR7 + /CD62L + cells in tumors for the individuals in experimental groups 7 through 12.

[016] Figure 8 is a graph of the percentage of CD87CCR77CD62L+ cells in tumors for the individuals in experimental groups 7 through 12.

[017] Figure 9 is a graph of the percentage of CD11b + /GR1 + cells in tumors for the individuals in experimental groups 7 through 12.

[018] Figure 10 is a graph of the percentage of CD4 + /CD25 + /FoxP3 + cells (T reg cells) in tumors for the individuals in experimental groups 7 through 12.

[019] Figure 11 is a graph of the percentage of CD47CCR77CD62L+ cells in spleens for the individuals in experimental groups 7 through 12.

[020] Figure 12 is a graph of the percentage of CD87CCR77CD62L* cells in spleens for the individuals in experimental groups 7 through 12.

[021] Figure 13 is a graph of the percentage of CD11b7GR1 + cells in spleens for the individuals in experimental groups 7 through 12.

[022] Figure 14 is a graph of the percentage of CD4 + /CD25 + /FoxP3 + cells (T reg cells) in spleens for the individuals in experimental groups 7 through 12.

[023] Figures 15A-C are representative scatter plots showing the Tmg cells in tumors of an individual (mouse 5, group 10) treated with anti-PD-L1 antibody treatment (Fig. 15A); an individual (mouse 5, group 11) treated with GMI-1359 and LTF-2 antibody (Fig. 5B); and an individual (mouse 5, group 12) treated with GMI- 1359 and anti-PD-L1 antibody treatment (Fig. 15C).

[024] Figures 16A-B are representative scatter plots showing the

CD4 + /CCR7 + /CD62L + cells in spleens of an individual (mouse 4, group 8) treated with GMI-1359 (Fig. 16A); and an individual (mouse 1, group 12) treated with GMI- 1359 and anti-PD-L1 antibody treatment (Fig. 16B).

[025] Figures 17A-B are representative scatter plots showing the

CD8 + /CCR7 + /CD62L + cells in spleens of an individual (mouse 4, group 7) treated with saline (Fig. 17A); and an individual (mouse 3, group 10) anti-PD-L1 antibody treatment (Fig. 17B).

[026] Figures 18A-C are representative scatter plots showing the CD11 b + /GR1 + cells in tumors of an individual (mouse 3, group 9) treated with LTF-2 antibody treatment (Fig. 18A); an individual (mouse 3, group 12) treated with GMI-1359 and anti-PD-L1 antibody (Fig. 18B) and presenting with a stable disease state; and an individual (mouse 4, group 12) treated with GMI-1359 and anti-PD-L1 antibody treatment (Fig. 15C) and presenting with a progressive disease state.

[027] Figure 19 is a table of the toxicity and efficacy data for the 12 treatment groups.

[028] Figure 20 is a comparison of the complete response (CR) rate and the median days post treatment until to complete response for group 7 (anti-PD-L1 antibody treatment alone) and group 12 (GMI-1359 and anti-PD-L1 antibody treatment combined).

[029] Figure 21 is a table showing the competitive binding activity (IC50) of GMI- 1359 against E-selectin and CXCR4. [030] Figure 22 displays graphs of the percentages of CD4+, CD8+ and

Regulatory T cells in spleen and tumor tissue samples, in vivo, on study day 15, in each treatment group.

[031] Figure 23 is a table that shows the ratio of CD8/regulatory T cells in spleen and tumor tissue samples, in vivo, on study day 5, in each treatment group.

[032] Figure 24A shows a mean tumor burden group comparison and response summary table.

[033] Figure 24B is a graph that shows the number of days post tumor implant on the x-axis, and the mean tumor burden (mm 3 ) in each treatment group, on the y- axis.

[034] The terms defined below are more fully defined by reference to the specification as a whole. While the terms used herein are believed to be well understood by one of ordinary skill in the art, the definitions included in this document are set forth to facilitate explanation of the presently-disclosed subject matter.

[035] Following long-standing patent law convention, the terms "a," "an," and "the" refer to "one or more" when used in this application, including the claims. Thus, for example, reference to "a cell" includes one cell or a plurality of cells, and so forth.

[036] Throughout this disclosure, various embodiments can be presented in a range format. Numeric ranges are inclusive of the numbers defining the range. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the disclosure. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range, such as from 1 to 6, should be considered to have specifically disclosed subranges, such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3.8, 4, 5.1, 5.3, and 6. This applies regardless of the breadth of the range.

[037] E-selectin (CD62E) is a cell adhesion molecule that is expressed on activated endothelial cells and plays an important role in leukocyte recruitment to the site of injury. The terms Έ-selectin inhibitor" or Έ-selectin antagonist" and the like are used interchangeably and mean an agent that inhibits an activity of E-selectin or inhibits the binding of E-selectin to one or more E-selectin ligands (which in turn may inhibit a biological activity of E-selectin). The term Έ-selectin inhibitor" includes inhibitors of E-selectin only, as well as inhibitors of E-selectin and either P-selectin or L-selectin, and inhibitors of E-selectin, P-selectin, and L-selectin.

[038] The term "non-glycomimetic moiety" includes moieties having a structure not intended to mimic a carbohydrate molecule. A non-glycomimetic moiety may not be (and is typically not) active as an E selectin antagonist. Instead, non- glycomimetic moieties are generally moieties added to a glycomimetic moiety for purposes of altering at least one property, such as solubility, bio-availability, lipophilicity and/or other drug-like properties of the glycomimetic.

[039] "T reg cells" are a subpopulation of T cells which modulate the immune system, maintain tolerance to self-antigens, and prevent autoimmune disease. T^ cells are immunosuppressive and generally suppress or downregulate induction and proliferation of other T cells. T regs cells express the biomarkers CD4, CD25, and FoxP3 (i.e., are CD4*/CD25 + /FoxP3 + ).

[040] T-cell checkpoints are molecules that need to be activated or inactivated to start an immune response. "T-cell checkpoint inhibitors," "checkpoint inhibitors" or "immunoregulatory receptor blocks" are agents that prevent or inhibit the normal T- cell checkpoint operation and prevent an immune response. For example, PD-1 (cell death protein 1) is a checkpoint protein on T cells that is important in self-recognition. PD-1 usually prevents the T cells from attacking normal body cells because normal body cells present the PD-L1 (the ligand that binds to PD-1). When the PD-1 checkpoint protein attaches to the PD-L1 protein of a cell, the T cell does not attack that cell. As noted above, PD-L1 is present on normal cells, but some cancer cells have large amounts of PD-L1, which helps them evade immune attack. A T-cell checkpoint inhibitor may block the PD-L1 protein presented on the cancer cells (or it may block the PD-1 of the T-cells) to prevent the T-cell's PD-1 from binding to the PD-L1. If the PD-1 and PD-L1 binding is blocked, the T-cell does not recognize the cell as "self," and may attack the cell. Accordingly, a T-cell checkpoint inhibitor may assist the immune system in attacking cancer cells that present PD-L1 proteins. Examples of T-cell checkpoint inhibitors are known in the art, including anti-PD-1 antibodies, anti-PD-L1 antibodies, and anti-CTL antibodies.

[041 ] The terms "patient," "subject," "individual," and the like are used interchangeably herein, and refer to any animal, or cells thereof whether in vitro or in situ, amenable to the methods described herein. In certain non-limiting

embodiments, the patient, subject or individual is a human.

[042] The term "therapy" refers to "treating" or "treatment" of a disease or condition including inhibiting the disease (slowing or arresting its development), providing relief from the symptoms or side-effects of the disease (including palliative treatment), and relieving the disease (causing regression of the disease). "Therapy" may also refer to prophylactic treatment, which includes preventing or delaying the onset of the disease or condition from occurring in a subject that may be predisposed to the disease but does not yet experience or exhibit symptoms of the disease.

[043] As used herein, "together" is used to mean that the agents are

administered concurrently. They can be administered in the same composition or in separate compositions. In contrast to "together," "sequentially" is used herein to mean that the gap between administering one agent and the other is significant, i.e., the first administered agent may no longer be present in the bloodstream in a therapeutic amount when the second agent and/or third agent is administered. When administered sequentially, the compounds may be administered in any order (e.g., a T-cell checkpoint inhibitor administered first followed by an E-selectin inhibitor or an E-selectin inhibitor administered first followed by an E-selectin inhibitor, etc.).

[044] The term "treatment" means the slowing down, interruption, arrest, reversal or stoppage of the progression of the disease, which does not necessarily require the complete elimination of all the signs and symptoms of the disease.

Furthermore, it is not necessary for the treatment to show effectiveness in 100% of the patients treated, rather, the term "treatment" is intended to mean that a statistically significant proportion of patients can be treated effectively, in such a way that the symptoms and clinical signs show at least an improvement. The person skilled in the art can easily establish whether the proportion is statistically significant using various statistical methods (e.g. confidence intervals, determination of them p value, Student's t-test, Mann-Whitney test etc.). Confidence intervals have a confidence of at least 90%, at least 95%, at least 97%, at least 98% or at least 99%. The p values are 0.1, 0.05, 0.01, 0.005 or 0.0001.

[045] In some embodiments, a method for treatment and/or prevention of at least one disease, disorder, or condition is disclosed, the method comprising administering to a subject in need thereof (1) an effective amount of at least one T- cell checkpoint inhibitor and (2) an effective amount of at least one other inhibitor chosen from E-selectin inhibitors, CXCR4 receptor inhibitors, and heterobifunctional inhibitors that comprise at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor.

[046] In some embodiments, at least one of (1 ) the at least one T-cell checkpoint inhibitor and (2) the at least one other inhibitor is in the form of at least one pharmaceutical composition. In some embodiments, the at least one pharmaceutical composition further comprises at least one pharmaceutically acceptable ingredient. In some embodiments, the at least one T-cell checkpoint inhibitor is in the form of a first pharmaceutical composition and the at least one other inhibitor is in the form of a second pharmaceutical composition.

[047] In some embodiments, a method for treatment and/or prevention of at least one bacterial infection, viral infection, or condition relating to at least one bacterial or viral infection is disclosed, the method comprising administering to a subject in need thereof (1) an effective amount of at least one T-cell checkpoint inhibitor and an effective amount of at least one other inhibitor chosen from E- selectin inhibitors, CXCR4 receptor inhibitors, and heterobifunctional inhibitors that comprise at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor, and/or (2) an effective amount of at least one pharmaceutical composition comprising at least one T-cell checkpoint inhibitor and at least one other inhibitor chosen from E-selectin inhibitors, CXCR4 receptor inhibitors, and heterobifunctional inhibitors that comprise at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor. In some embodiments, the Infection is an HIV infection. In some embodiments, the condition relating to a bacterial or viral infection is sepsis or septic conditions.

[048] In some embodiments, a method for treatment and/or prevention of at least one cancer is disclosed, the method comprising administering to a subject in need thereof (1) an effective amount of at least one T-cell checkpoint inhibitor and an effective amount of at least one other inhibitor chosen from E-selectin inhibitors, CXCR4 receptor inhibitors, and heterobifunctional inhibitors that comprise at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor, and/or (2) an effective amount of at least one pharmaceutical composition comprising at least one T-cell checkpoint inhibitor and at least one other inhibitor chosen from E-selectin inhibitors, CXCR4 receptor inhibitors, and heterobifunctional inhibitors that comprise at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor.

[049] In some embodiments, a method for treatment and/or prevention of at least one disease, disorder, or condition in which suppression of Treg cells is desired, the method comprising administering to a subject in need thereof (1) an effective amount of at least one T-cell checkpoint inhibitor and an effective amount of at least one other inhibitor chosen from E-selectin inhibitors, CXCR4 receptor inhibitors, and heterobifunctional inhibitors that comprise at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor, and/or (2) an effective amount of at least one pharmaceutical composition comprising at least one T-cell checkpoint inhibitor and at least one other inhibitor chosen from E-selectin inhibitors, CXCR4 receptor inhibitors, and heterobifunctional inhibitors that comprise at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor.

[050] In some embodiments, a method for treatment and/or prevention of at least one disease, disorder, or condition in which suppression of T reg cells is desired, the method comprising administering to a subject in need thereof (1) an effective amount of at least one T-cell checkpoint inhibitor and an effective amount of at least one E-selectin inhibitor and/or (2) an effective amount of at least one pharmaceutical composition comprising at least one T-cell checkpoint inhibitor and at least one E- selectin inhibitor.

[051] In some embodiments, a method for treatment and/or prevention of at least one disease, disorder, or condition in which suppression of T^ cells is desired, the method comprising administering to a subject in need thereof (1) an effective amount of at least one T-cell checkpoint inhibitor and an effective amount of at least one CXCR4 receptor inhibitor and/or (2) an effective amount of at least one pharmaceutical composition comprising at least one T-cell checkpoint inhibitor and at least one CXCR4 receptor inhibitor.

[052] In some embodiments, a method for treatment and/or prevention of at least one disease, disorder, or condition in which suppression of T reg cells is desired, the method comprising administering to a subject in need thereof (1) an effective amount of at least one T-cell checkpoint inhibitor and an effective amount of at least one heterobifunctional inhibitor that comprises at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor, and/or (2) an effective amount of at least one pharmaceutical composition comprising at least one T-cell checkpoint inhibitor and at least one heterobifunctional inhibitor that comprises at least one E-selectin inhibitor linked to at least one CXCR4 receptor inhibitor.

[053] Any T-cell checkpoint inhibitors) can be used in the compositions and methods disclosed herein. In some embodiments, the at least one T-cell checkpoint inhibitor targets the PD-1 receptor and/or the CTLA-4 protein on T ree cells. In some embodiments, the at least one T-cell checkpoint inhibitor is a PD-L1 inhibitor. In some embodiments, the at least one T-cell checkpoint inhibitor is the anti-mPD-L1 antibody. In some embodiments, the at least one T-cell checkpoint inhibitor is chosen from nivolumab and ipilimumab.

[054] The E-selectin inhibitors, which include the at least one E-selectin inhibitor herein, may be chosen from glycomimetics. In some embodiments, the E-selectin inhibitors are chosen from sialyl Lewis x (sLe x ) and sLe* mimetics. In some embodiments, the E-selectin inhibitors are chosen from small molecule glycomimetic antagonists of E-selectin, antibodies directed to E-selectin, aptamers to E-selectin, peptides directed to E-selectin, and peptidomimetics directed to E-selectin.

[055] In some embodiments, the E-selectin inhibitors are chosen from compounds of Formula (I):

isomers of Formula (I), tautomers of Formula (I), and pharmaceutically acceptable salts of any of the foregoing, wherein:

R 1 is chosen from C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 1 -C 8 haloalkyl, C 2 -C 8 haloalkenyl, and C 2 -C 8 haloalkynyl groups;

R 2 is chosen from H, -M, and -L-M;

R 3 is chosen from -OH, -NH 2 , -OC(=O)Y 1 , -NHC(=O)Y 1 , and

-NHC(=O)NHY 1 groups, wherein Y 1 is chosen from C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, d-β haloalkyl, C 2-8 haloalkenyl, C 2-8 haloalkynyl, C 6-18 aryl, and C 1 -13 heteroaryl groups;

R 4 is chosen from -OH and -NZ 1 Z 2 groups, wherein Z 1 and Z 2 , which may be identical or different, are each independently chosen from H, C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 1 -C 8 haloalkyl, C 2 -C 8 haloalkenyl, and C 2 -C 8 haloalkynyl groups, wherein Z 1 and Z 2 may together form a ring;

R 5 is chosen from C 3 -C 8 cycloalkyl groups;

R 6 is chosen from -OH, C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 1-8 haloalkyl, C 2 -C 8 haloalkenyl, and C 2 -C 8 haloalkynyl groups;

R 7 is chosen from -CH 2 OH, C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2-8 alkynyl, C 1 -C 8 haloalkyl, C 2 -C 8 haloalkenyl, and C 2 -C 8 haloalkynyl groups;

R 8 is chosen from C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 1 -C 8 haloalkyl, C 2-8 haloalkenyl, and C 2 -C 8 haloalkynyl groups;

L is chosen from linker groups; and

M is a non-glycomimetic moiety chosen from polyethylene glycol, thiazolyl, chromenyl, -C(=O)NH(CH 2 ) 1-4 lMH 2 , C 1-8 alkyl, and -C(=O)OY, wherein Y is chosen from C 1-4 alkyl, C 2-4 alkenyl, and C 2-4 alkynyl groups.

[056] As would be recognized by one of ordinary skill in the art, the phrase 'isomers of Formula (I), tautomers of Formula (I), and pharmaceutically acceptable salts of any of the foregoing' includes hydrates and solvates.

[057] In some embodiments, the E-selectin inhibitors are chosen from

compounds of Formula (I), wherein the non-glycomimetic moiety comprises polyethylene glycol.

[058] As used herein, "CXCR4 receptor inhibitors," "CXCR4 chemokine receptor inhibitors," "SDF-1 inhibitor," or "SDF-1 antagonist," and the like are used interchangeably and mean an agent inhibits the binding of the chemokine SDF-I to an SDF-I ligand (e.g., prevents the binding of SDF-I to CXCR4). Such inhibitors will typically prevent the binding of stromal derived factor-1 (SDF-1) to a CXCR4 receptor. Examples of CXCR4 chemokine receptor inhibitors are AMD-3100

(Hendrix et al., Antimicrob. Agents Chemother. 44:1667-1673, 2000); ALX40-4C (Doranz et al., AIDS Research and Human Retroviruses 17:475-486, 2001); and T134 (Arakaki et al., J. Virol. 73:1719-1723, 1999). These examples include a small organic molecule and amino acid-based molecules, such as the T22 peptide.

[059] In some embodiments, the CXCR4 receptor inhibitors are chosen from petpides, diketopiperazine mimetics, bicyclams, tetrahydroquinolines,

thiazolylisothiourea derivatives, and benzodiazepines. In some embodiments, the CXCR4 receptor inhibitors are chosen from AMD-3100, ALX40-4C, T134, and T22 peptide.

[060] In some embodiments, the heterobifunctional inhibitors are chosen from compounds of Formula (II):

isomers of Formula (II), tautomers of Formula (II), and pharmaceutically acceptable salts of any of the foregoing, wherein:

R 1 is chosen from H, Ci_8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C-i-e haloalkyl, C 2 . 8 haloalkenyl, and C 2-8 haloalkynyl groups; R 2 is chosen from -OH, -NH 2 , -OC(=O)Y 1 , -NHC(=O)Y 1 , and -NHC(=O)NHY 1 groups, wherein Y 1 is chosen from alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1-8 haloalkyl, C 2-8 haloalkenyi, C 2-8 haloaikynyl, C 6-18 aryl, and C 1 -13 heteroaryl groups;

R 3 is chosen from -CN, -CH 2 CN, and -C(=O)Y 2 groups, wherein Y 2 is chosen from C 1-8 alkyl, C 2-8 alkenyl, C M alkynyl, -OZ 1 , -NHOH, -NHOCH3, -NHCN, and -NZ 1 Z 2 groups, wherein Z 1 and Z 2 , which may be identical or different, are independently chosen from H, C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1-8 haloalkyl, C 2-8 haloalkenyi, and C 2-8 haloaikynyl groups, wherein Z 1 and Z 2 may together form a ring;

R 4 is chosen from C 3-8 cycloalkyl groups;

R 5 is independently chosen from H, halo, C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1-8 haloalkyl, C 24 haloalkenyi, and C 2-8 haloaikynyl groups;

n is chosen from integers ranging from 1 to 4; and

L is chosen from linker groups.

[061] As would be recognized by one of ordinary skill in the art, the phrase 'isomers of Formula (II), tautomers of Formula (II), and pharmaceutically acceptable salts of any of the foregoing' includes hydrates and solvates.

[062] In some embodiments, the heterobifunctional inhibitors are chosen from compounds of Formula (lla):

[063] In some embodiments, the linker groups of Formula I and/or Formula II are independently chosen from groups comprising spacer groups, such spacer groups as, for example, -(CH 2 ) P - and -0(CH 2 ) P -, wherein p is chosen from integers ranging from 1 to 30. In some embodiments, p is chosen from integers ranging from 1 to 20. Other non-limiting examples of spacer groups include carbonyi groups and carbonyl- containing groups such as, for example, amide groups. A non-limiting example of a spacer group is

[064] In some embodiments, the linker groups are independently chosen from

[065] Other linker groups, such as, for example, polyethylene glycols (PEGs) and -C(=O)-NH-(CH 2 ) P -C(=O)-NH-, wherein p is chosen from integers ranging from 1 to 30, or wherein p is chosen from integers ranging from 1 to 20, will be familiar to those of ordinary skill in the art and/or those in possession of the present disclosure.

[066] In some embodiments, at least one linker group is

[067] In some embodiments, at least one linker group is

[068] In some embodiments, at least one linker group is chosen from

-C(=O)NH(CH 2 ) 2 NH-, -CH2NHCH2-, and -C(=O)NHCH 2 -. In some embodiments, at least one linker group is -C(=O)NH(CH 2 ) 2 NH-. [069] In some embodiments, the at least one E-selectin inhibitor is chosen from compounds of Formula (la):

[070] wherein n is chosen from integers ranging from 1 to 100. In some embodiments, n is chosen from 4, 8, 12, 16, 20, 24, and 28.

[071] In some embodiments, the E-selectin inhibitors are chosen from E-selectin inhibitors disclosed in U.S. Patent No. 9,109,002, which is hereby incorporated by reference. In some embodiments, the E-selectin inhibitor is GMI-1271. See, e.g., Price et al., "Dormant breast cancer micrometastases reside in specific bone marrow niches that regulate their transit to and from bone," Science Translational Medicine, Vol. 8(340), May 25, 2016, [DOI:10.1126/scitranslmed.aad4059]; Dutta et al., "E- Selectin Inhibition Mitigates Splenic HSC Activation and Myelopoiesis in

Hypercholesterolemic Mice With Myocardial Infarction", Arterioscler Thromb Vase Biol [DOI: 10.1161/ATVBAHA.116.307519]

[072] In some embodiments, the heterobifunctional inhibitors are chosen from compounds of the following Formulae:

[073] in some embodimenis, the heterobifunctional inhibitors are chosen from compounds of the following Formulae:

[074] In some embodiments, the heterobifunctional inhibitors are chosen from heterobifunctional inhibitors disclosed in U.S. Patent No. 8,410,066 and

PCT/US2015/063191, which are hereby incorporated by reference. In some embodiments, the heterobifunctional inhibitor is GMI-1359. See, e.g., Steele, Maria M. et al., "A small molecule glycomimetic antagonist of E-selectin and CXCR4 (GMI- 1359) prevents pancreatic tumor metastasis and improves chemotherapy [abstract]," Proceedings of the 106th Annual Meeting of the American Association for Cancer Research, 2015 Apr 18-22, Philadelphia, PA; Philadelphia (PA): AACR, Cancer Res 2015, 75(15 Suppl):Abstract nr 425. doi:10.1158/1538-7445. AM2015-425; Gravina, Giovanni L. et al., "Dual E-selectin and CXCR4 inhibition reduces tumor growth and increases the sensitivity to docetaxel in experimental bone metastases of prostate cancer [abstract]," Proceedings of the 106th Annual Meeting of the American Association for Cancer Research, 2015 Apr 18-22, Philadelphia, PA; Philadelphia (PA): AACR, Cancer Res 2015, 75(15 Suppl):Abstract nr 428. doi:10.1158/1538- 7445.AM2015-428, all of which are incorporated by reference.

[075] In some embodiments, the at least one disease, disorder, or condition is chosen from cancers. In some embodiments, the cancers are chosen from liquid cancers (e.g., MM, ALL, and AML) and solid cancers (e.g., prostate cancer). In some embodiments, the cancers are chosen from liquid cancers. In some embodiments, the cancers are chosen from solid cancers. In some embodiments, the subject is treated locally at a tumor of a solid cancer.

[076] In some embodiments, the rate of increase of cancerous cells (e.g., tumor growth or cancer cell proliferation) is reduced or halted. In some embodiments, the number of cancer cells is reduced. In some embodiments, the cancer cells are eliminated. In some embodiments, metastasis of cancer cells reduced. In some embodiments, the metastasis of cancer cells is halted. In some embodiments, infiltration of the cancer into bone marrow is reduced or halted.

[077] In some embodiments, the subject has cancer and has received or will receive chemotherapy and/or radiotherapy. In some embodiments, the

chemotherapy comprises administering a therapeutically effective amount of at least one compound chosen from platinum, cisplatin, carboplatin, oxaliplatin,

mechlorethamine, cyclophosphamide, chlorambucil, azathioprine, mercaptopurine, vincristine, vinblastine, vinorelbine, vindesine, etoposide, teniposide, paclitaxel, docetaxel, irinotecan, topotecan, amsacrine, etoposide, etoposide phosphate, teniposide, 5-fluorouracil (5-FU), leucovorin, methotrexate, gemcitabine, taxane, leucovorin, mitomycin C, tegafur-uracil, idarubicin, fludarabine, mitoxantrone, ifosfamide and doxorubicin.

[078] In some embodiments, the chemotherapy comprises administration of bortezomib. In some embodiments, the chemotherapy comprises administration of gemcitabine.

[079] In some embodiments, the at least one disease, disorder, or condition is chosen from a bacterial infection, a viral infection, and a condition relating to a bacterial or viral infection. In some embodiments, the viral infection is an HIV infection and/or the patient has been diagnosed with AIDS or an HIV-related illness. In some embodiments, the bacterial infection is sepsis and/or the subject has been diagnosed with a septic condition.

[080] In some embodiments, the administration to the subject of at least one T- cell checkpoint inhibitor and the at least one other inhibitor suitably overlap so that the therapeutic effect of one agent (i.e. the time period post use where a measurable benefit to the patient is observed) is concurrent, at least at some point, with the period of therapeutic effect of the second agent. In some embodiments, the at least one T-cell checkpoint inhibitor and the at least one other inhibitor are administered concurrently. In some embodiments, the at least one T-cell checkpoint inhibitor and the at least one other inhibitor are administered at the different times. In some embodiments, the at least one T-cell checkpoint inhibitor and the at least one other inhibitor are administered sequentially.

[081] In some embodiments, the at least one T-cell checkpoint inhibitor and the at least one other inhibitor are administered in a single pharmaceutical composition. [082] In some embodiments, the at least one T-cell checkpoint inhibitor and the at least one other inhibitor are administered in separate pharmaceutical

compositions.

[083] In some embodiments, the pharmaceutical composition(s) further comprises at least one additional pharmaceutically acceptable ingredient.

[084] In pharmaceutical dosage forms, any one or more of the compounds of the present disclosure may be administered in the form of a pharmaceutically acceptable derivative, such as a salt, and/or it/they may also be used alone and/or in

appropriate association, as well as in combination, with other pharmaceutically active compounds.

[085] An "effective amount" or "therapeutically effective amount" refers to an amount of a compound of the present disclosure or a composition comprising at least one such compound that, when administered to a subject, either as a single dose or as part of a series of doses, is effective to produce at least one therapeutic effect. For example, in the case of cancer, a therapeutic effect can be killing cancer cells, inducing apoptosis in cancer cells, reducing the growth rate of cancer cells, reducing the incidence or number of metastases, reducing tumor size, inhibiting tumor growth, reducing the blood supply to a tumor or cancer cells, promoting an immune response against cancer cells or a tumor, preventing or inhibiting the progression of cancer, or increasing the lifespan of a subject with cancer.

[086] Optimal doses may generally be determined using experimental models and/or clinical trials. Design and execution of pre-clinical and clinical studies for each of the therapeutics (including when administered for prophylactic benefit) described herein are well within the skill of a person of ordinary skill in the relevant art. The optimal dose of a therapeutic may depend upon the body mass, weight, and/or blood volume of the subject. The minimum dose that is sufficient to provide effective therapy may be used in some embodiments. Subjects may generally be monitored for therapeutic effectiveness using assays suitable for the disease or condition being treated or prevented, which assays will be familiar to those having ordinary skill in the art and are described herein. The level of a compound that is administered to a subject may be monitored by determining the level of the compound (or a metabolite of the compound) in a biological fluid, for example, in the blood, blood fraction (e.g., serum), and/or in the urine, and/or other biological sample from the subject. Any method practiced in the art to detect the compound, or metabolite thereof, may be used to measure the level of the compound during the course of a therapeutic regimen.

[087] The dose of a compound described herein may depend upon the subject's condition, that is, stage of the disease, severity of symptoms caused by the disease, general health status, as well as age, gender, and weight, and other factors apparent to a person of ordinary skill in the medical art. Similarly, the dose of the therapeutic for treating a disease or disorder may be determined according to parameters understood by a person of ordinary skill in the medical art.

[088] Pharmaceutical compositions may be administered in any manner appropriate to the disease or disorder to be treated as determined by persons of ordinary skill in the medical arts. An appropriate dose and a suitable duration and frequency of administration will be determined by such factors as discussed herein, including the condition of the patient, the type and severity of the patient's disease, the particular form of the active ingredient, and the method of administration. In general, an appropriate dose (or effective dose) and treatment regimen provides the pharmaceutical composition(s) as described herein in an amount sufficient to provide therapeutic and/or prophylactic benefit (for example, an improved clinical outcome, such as more frequent complete or partial remissions, or longer disease-free and/or overall survival, or a lessening of symptom severity or other benefit as described in detail above).

[089] The pharmaceutical compositions described herein may be administered to a subject in need thereof by any one of several routes that effectively delivers an effective amount of the compound. Non-limiting suitable administrative routes include topical, oral, nasal, intrathecal, enteral, buccal, sublingual, transdermal, rectal, vaginal, intraocular, subconjunctival, sublingual, and parenteral

administration, including subcutaneous, intravenous, intramuscular, intrasternal, intracavernous, intrameatal, and intraurethral injection and/or infusion.

[090] The pharmaceutical composition described herein may be sterile aqueous or sterile non-aqueous solutions, suspensions or emulsions, and may additionally comprise at least one pharmaceutically acceptable excipient (i.e., a non-toxic material that does not interfere with the activity of the active ingredient). Such compositions may be in the form of a solid, liquid, or gas (aerosol). Alternatively, the compositions described herein may be formulated as a lyophilizate, or compounds described herein may be encapsulated within liposomes using technology known in the art. The pharmaceutical compositions may further comprise at least one additional pharmaceutically acceptable ingredient, which may be biologically active or inactive. Non-limiting examples of such ingredients include buffers (e.g., neutral buffered saline or phosphate buffered saline), carbohydrates (e.g., glucose, mannose, sucrose or dextrans), mannitol, proteins, polypeptides, amino acids (e.g., glycine), antioxidants, chelating agents (e.g., EDTA and glutathione), stabilizers, dyes, flavoring agents, suspending agents, and preservatives. [091] Any suitable excipient or carrier known to those of ordinary skill in the art for use in pharmaceutical compositions may be employed in the compositions described herein. Excipients for therapeutic use are well known, and are described, for example, in Remington: The Science and Practice of Pharmacy (Gennaro, 21 et Ed. Mack Pub. Co., Easton, PA (2005)). In general, the type of excipient is selected based on the mode of administration, as well as the chemical composition of the active ingredient(s). Pharmaceutical compositions may be formulated for the particular mode of administration. For parenteral administration, pharmaceutical compositions may further comprise water, saline, alcohols, fats, waxes, and buffers. For oral administration, pharmaceutical compositions may further comprise at least one ingredient chosen, for example, from any of the aforementioned excipients, solid excipients and carriers, such as mannitol, lactose, starch, magnesium stearate, sodium saccharine, talcum, cellulose, kaolin, glycerin, starch dextrins, sodium alginate, carboxymethylcellulose, ethyl cellulose, glucose, sucrose, and magnesium carbonate.

[092] The pharmaceutical compositions (e.g., for oral administration or delivery by injection) may be in the form of a liquid. A liquid pharmaceutical composition may include, for example, at least one the following: a sterile diluent such as water for injection, saline solution, physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils that may serve as the solvent or suspending medium,

polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents; antioxidants; chelating agents; buffers and agents for the adjustment of tonicity, such as sodium chloride or dextrose. A parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. In some embodiments, the pharmaceutical composition comprises physiological saline. In some embodiments, the pharmaceutical composition an injectable pharmaceutical composition, and in some embodiments, the injectable pharmaceutical composition is sterile.

[093] For oral formulations, at least one of the compounds of the present disclosure can be used alone or in combination with at least one additive appropriate to make tablets, powders, granules and/or capsules, for example, those chosen from conventional additives, disintegrators, lubricants, diluents, buffering agents, moistening agents, preservatives, coloring agents, and flavoring agents. The pharmaceutical compositions may be formulated to include at least one buffering agent, which may provide for protection of the active ingredient from low pH of the gastric environment and/or an enteric coating. A pharmaceutical composition may be formulated for oral delivery with at least one flavoring agent, e.g., in a liquid, solid or semi-solid formulation and/or with an enteric coating.

[094] Oral formulations may be provided as gelatin capsules, which may contain the active compound or biological along with powdered carriers. Similar carriers and diluents may be used to make compressed tablets. Tablets and capsules can be manufactured as sustained release products to provide for continuous release of active ingredients over a period of time. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.

[095] A pharmaceutical composition may be formulated for sustained or slow release. Such compositions may generally be prepared using well known

technology and administered by, for example, oral, rectal or subcutaneous implantation, or by implantation at the desired target site. Sustained-release formulations may contain the active therapeutic dispersed in a carrier matrix and/or contained within a reservoir surrounded by a rate controlling membrane. Excipients for use within such formulations are biocompatible, and may also be biodegradable; the formulation may also provide a relatively constant level of active component release. The amount of active therapeutic contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release, and the nature of the condition to be treated or prevented.

[096] The pharmaceutical compositions described herein can be formulated as suppositories by mixing with a variety of bases, such as emulsifying bases or water- soluble bases. The pharmaceutical compositions may be prepared as aerosol formulations to be administered via inhalation. The compositions may be formulated into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen and the like.

[097] The compounds of the present disclosure and pharmaceutical

compositions comprising these compounds may be administered topically (e.g., by transdermal administration). Topical formulations may be in the form of a

transdermal patch, ointment, paste, lotion, cream, gel, and the like. Topical formulations may include one or more of a penetrating agent or enhancer (also call permeation enhancer), thickener, diluent, emulsifier, dispersing aid, or binder.

Physical penetration enhancers include, for example, electrophoretic techniques, such as iontophoresis, use of ultrasound (or "phonophoresis"), and the like.

Chemical penetration enhancers are agents administered either prior to, with, or immediately following administration of the therapeutic, which increase the permeability of the skin, particularly the stratum corneum, to provide for enhanced penetration of the drug through the skin. Additional chemical and physical penetration enhancers are described in, for example, Transdermal Delivery of Drugs, A. F. Kydonieus (ED) 1987 CRL Press; Percutaneous Penetration Enhancers, eds. Smith et al. (CRC Press, 1995); Lenneras et a!., J. Pharm. Pharmacol. 54:499-508 (2002); Karande et al., Pharm. Res. 19:655-60 (2002); Vaddi et al., Int. J. Pharm. 91:1639-51 (2002); Ventura et al., J. Drug Target 9:379-93 (2001); Shokri et al., Int. J. Pharm. 228(1 -2):99-107 (2001); Suzuki et al., Biol. Pharm. Bull. 24:698-700 (2001); Alberti et al., J. Control Release 71:319-27 (2001); Goldstein et al., Urology 57:301-5 (2001); Kiijavainen et al.. Eur. J. Pharm. Scl. 10:97-102 (2000); and

Tenjarla et al., Int. J. Pharm. 192:147-58 (1999).

[098] Routes of administration include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, intrathecal, and subcutaneous routes. In some embodiments, the compounds or compositions are administered locally (i.e., near a cancer tumor). In some embodiments, one or more of the compounds or compositions are administered using different routes of administration.

[099] The compounds or pharmaceutical composition(s) can be administered in one or more doses and treatment regimens, which may be the same or different. In one embodiment, each of the compounds or pharmaceutical composition(s) is administered in an amount ranging from about 1 mg/kg to about 50 mg/kg once a day. In other embodiments, the dosage may be at any dosage including, but not limited to, about 1 Mg/kg, 25 pg/kg, 50 pg/kg, 75 Mg/kg, 100 Mg/kg, 125 Mg/kg, 150 Mg/kg, 175 Mg/kg, 200 Mg/kg, 225 Mg/kg, 250 Mg/kg, 275 Mg/kg, 300 Mg/kg, 325 Mg/kg, 350 Mg/kg, 375 Mg/kg, 400 Mg/kg, 425 Mg/kg, 450 Mg/kg, 475 Mg/kg, 500 Mg/kg, 525 Mg/kg, 550 Mg/kg, 575 Mg/kg, 600 Mg/kg, 625 Mg/kg, 650 Mg/kg, 675 Mg/kg, 700 Mg/kg, 725 Mg/kg, 750 Mg/kg, 775 Mg/kg, 800 Mg/kg, 825 Mg/kg, 850 Mg/kg, 875 Mg/kg, 900 Mg/kg, 925 Mg/kg, 950 Mg/kg, 975 Mg/kg, 1 mg/kg, 2 mg/kg, 3 mg/kg, 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 8 mg/kg, 9 mg/kg, or 10 mg/kg. [0100] In some embodiments, the compounds or pharmaceutical composition(s) are administered in any of these amounts and ranges once a day, more than once a day, every other day, every two days, etc. In some embodiments, the at least one T- cell checkpoint inhibitor and the at least one E-selectin inhibitor, CXCR4 receptor inhibitor, and/or heterobifunctional inhibitor, wherein an E-selectin inhibitor is linked to a CXCR4 receptor inhibitor, are administered concurrently and/or in the same number of treatments per day. In some embodiments, the at least one T-cell checkpoint inhibitor and the at least one E-selectin inhibitor, CXCR4 receptor inhibitor, and/or heterobifunctional inhibitor, wherein an E-selectin inhibitor is linked to a CXCR4 receptor inhibitor, are administered on different schedules. One of more treatment cycles may be repeated, and any number of cycles is contemplated. The number of treatments per day and the amount per dose for each compound or pharmaceutical composition may vary during each cycle.

[0101] Kits comprising unit doses of at least one compound or pharmaceutical composition of the present disclosure, for example, in oral or injectable doses, are provided. Such kits may include a container comprising the unit dose, an

informational package insert describing the use and attendant benefits of the therapeutic in treating the pathological condition of interest, and/or optionally an appliance or device for delivery of the at least one compound and/or pharmaceutical composition comprising the same.

EXAMPLES

Overview

[0 02] The primary goal of the studies was to determine the anti-cancer activity of treatments comprising the administration of T-cell checkpoint inhibitor with an E- seiectin inhibitor and/or a CXCR4 receptor inhibitor. The T-cell checkpoint inhibitor used was an anti-mPD-L1 antibody. Heterobifunctional inhibitor GMI-1359, an inhibitor of both E-selectins and CXCR4 receptors, was used.

[0103] The anti-cancer activity of an anti-mPD-L1 antibody (10F.9G2) and GMI- 1359 was compared to a rat isotype control antibody against indeterminate subcutaneous CT26.WT carcinoma xenografts in female Balb/c mice. An immune profile of each mouse was determined by sampling tumors and spleens, staining for several markers of an active immune response, and detecting the levels of these markers via flow cytometry. Flow cytometry markers included: T-Cells (CD4 + and CD8 + ), Regulatory T-Cells (T^e). (CD4 * /CD25 + /FoxP3 + ), MDSCs (CD11b + /Gr1 + ) and co-expression of CCR7* and CD62* added to the existing CD4/CD8 panel.

Materials & Methods

[0104] GMI-1359 (MW=1115g/mol, Lot #50.279) was obtained as a

pre-weighed crystalline powder. Upon receipt, it was stored protected from light at - 20°C. GMI-1359 was formulated in sterile saline. The vehicle (sterile saline) was added to the pre-weighed compound to achieve a concentration of 0.1mg/ml. The formulation was then stirred overnight at 20°C. The final dosing solution was clear and colorless with a pH of 9.99. The dosing formulation was prepared fresh weekly and was stored protected from light at 20°C when not in use. Dose levels of GMI- 1359 were given as bulk drug substance.

[0105] InVivoMAb anti-KLH; Rat lgG2b, LTF-2 (5.8mg/ml, Lot #5535-3-6-7/0515) was obtained as a clear, colorless stock solution. Upon receipt, it was stored protected from light at 4°C. The dosing solution was prepared by diluting the stock solution with PBS (phosphate buffered saline) to a final concentration of 1mg/ml. The final dosing solution was clear and colorless with a pH of 7.29. The dosing formulation was prepared once weekly and was stored protected from light at 4°C when not in use. On each dosing day, the dosing formulations were stored on ice prior to and during dosing.

[0106] Anti-mPD-L1 (10F.9G2, 6.39mg/ml, Lot #5592-4-6/0615) was obtained as a clear, colorless stock solution. Upon receipt, it was stored protected from light at 4°C. The dosing solution was prepared by diluting the stock solution with PBS 1mg/ml. The final dosing solution was clear and colorless with a pH of 6.96. The formulation was prepared once weekly and was stored protected from light at 4°C when not in use. On each dosing day, the dosing formulations were stored on ice prior to and during dosing.

[0107] Female Harlan Balb/c mice (BALB/cAn Hsd)

were used in this study. They were 6-7 weeks old on Day 1 of the experiment. The animals were fed irradiated Harlan 2918.15 Rodent Diet and water ad libitum.

Animals were housed in static cages with Bed-O'Cobs™ bedding inside Biobubble® Clean Rooms that provide H.E.P.A filtered air into the bubble environment at 100 complete air changes per hour. All treatments, body weight determinations, and tumor measurements were carried out in the bubble environment. The environment was controlled to a temperature range of 70°±2°F and a humidity range of 30-70%. All procedures carried out in these experiments were conducted in compliance with all the laws, regulations and guidelines of the National institutes of Health (NIH) in an AAALAC accredited facility.

[0108] CT26.WT cells were grown in RPM1 1640 medium which

was modified with 1mM Na pyruvate, 10mM HEPES, 2.5g/L glucose and

supplemented with 10% nonheat-inactivated Fetal Bovine Serum (FBS) and 1% 100X Penicillin/Streptomycin/L-Glutamine (PSG). The growth environment was maintained in an incubator with a 5% C02 atmosphere at 37°C. [0109] When expansion was complete, the cells were trypsinized using 0.25% trypsin-EDTA solution. Following cell detachment, the trypsin was inactivated by dilution with complete growth medium and any clumps of cells were separated by pipetting. The cells were centrifuged at 200rcf for 8 minutes at 4°C, the supernatant was aspirated, and the pellet was re-suspended in cold Dulbecco's Phosphate Buffered Saline (DPBS) by pipetting. An aliquot of the homogeneous cell suspension was diluted in a trypan blue solution and counted using a Luna automated cell counter. The preimplantation cell viability was 92%. The cell suspension was centrifuged at 200rcf for 8 minutes at 4°C. The supernatant was aspirated and the cell pellet was re-suspended in cold serum-free medium to generate a final concentration of 2.50E+06 trypan-excluding cells/ml. The cell suspension was maintained on wet ice during implantation. Following implantation, an aliquot of the remaining cells was diluted with a trypan blue solution and counted to determine the post-impiantation cell viability (91%).

[0110] Test animals were implanted subcutaneously, high in the axilla Gust under the fore limb) on Day 0 with 5.00E+05 cells in 0.2ml of serum-free medium using a 27-gauge needle and syringe.

[0 11] The following classification was used in the following experiments. Of all the CD8 + cells the T na ive population was CD62hi CD44 " ; the T C M/SCM population was CD62hi CD44 ; and the TEM population was CD44 + CD62 " population. An increase in the TCM/SCM mouse cell population was observed (see Figures 1 through 6).

[0112] All mice were sorted into study groups based

on body weights such that the mean body weights in each group was within 10% of the overall mean. Treatment began on Day 3. [0113] Groups 1 and 7: The Vehicle Control (saline) was dosed intraperitoneal!;/, every day for 20 days (Days 3-22) and every day for 12 days (Days 3-14) for Groups 1 and 7, respectively.

[0114] Groups 2 and 8: GMI-1359 was dosed intraperitoneally at 40mg/kg, every day for 20 days (Days 3-22) and every day for 12 days (Days 3-14) for Groups 2 and 8, respectively.

[0115] Groups 3 and 9: Anti-KLH; Rat lgG2b, LTF-2 was dosed intraperitoneally at lOmg/kg, every 3 days for 2 treatments, with 3 days off for 2.5 weeks (Days 3, 6, 10, 13 and 17) for Group 3. Group 9 was dosed intraperitoneally at 10mg/kg, every 3 days for 2 treatments, with 3 days off for 2 weeks (Days 3, 6, 10 and 13).

[0116] Groups 4 and 10: Anti-mPD-L1 , 10F.9G2 was dosed intraperitoneally at lOmg/kg, every 3 days for 2 treatments, with 3 days off for 2.5 weeks (Days 3, 6, 10, 13 and 17) for Group 4.

[0117] Group 10 was dosed intraperitoneally at 10mg/kg, every 3 days for 2 treatments, with 3 days off for 2 weeks (Days 3, 6, 10 and 13).

[0118] Groups 5 and 11 : GMI-1359 was dosed in combination with anti-KLH; Rat lgG2b, LTF intraperitoneally at 40mg/kg and 10mg/kg, respectively. In Group 5, GMI- 1359 was dosed once a day for 20 days (Days 3-22) and anti-KLH; Rat lgG2b was given every 3 days for 2 treatments, with 3 days off for 2.5 weeks (Days 3, 6, 10, 13 and 17). In Group 11 , GMI-1359 was dosed once a day for 12 days (Days 3-14) and anti-KLH; Rat lgG2b was dosed every 3 days for 2 treatments, with 3 days off for 2 weeks and (Days 3, 6, 10 and 13). On days when both compounds were dosed, GMI-1359 was given first and then anti-KLH; Rat lgG2b, LTF was given within minutes thereafter. [0119] Groups 6 and 12: GMI-1359 and anti-mPD-L1 , 10F.9G2 were dosed in a combination regimen intraperitoneally at 40mg/kg and lOmg/kg, respectively. In Group 6, GMI-1359 was dosed once a day for 20 days (Days 3-22) and anti-mPD- L1 , 10F.9G2 was given every 3 days for 2 treatments, with 3 days off for 2.5 weeks and (Days 3, 6, 10, 13, and 17).

[0120] In Group 12, GMI-1359 was dosed once a day for 12 days (Days 3-14) and anti-mPD-L1 was dosed every 3 days for 2 treatments, with 3 days off for 2 weeks (Days 3, 6, 10, and 13). On days in which both compounds were given, GMI- 1359 was given first and anti-mPD-L1, 10F.9G2 was dosed within minutes thereafter.

[0121] All mice were dosed according to individual body weight on the day of treatment (0.2ml/20g).

[0122] The toxicity and efficacy data for the various treatment groups is presented in Figure 19.

[0123] At 24 hours after the final dose of GMI-1359 (Day 15), all mice

from Groups 7-12 were euthanized for tumor and spleen collection. All mice were euthanized via over exposure to carbon dioxide. The tumors and spleens were excised and placed in a labeled 5mL sampling tube filled with cold PBS and placed on ice. The tumors and spleens were provided to the Molecular Imaging's in vitro group for flow cytometric analysis.

[0124] Testing in this experiment was generally

carried out adhering to the general principles established by the groups of Schabel, Skipper, Griswold, Corbett, Leopold, Ross and the NCI (1-7). Tumor measurements were recorded three times weekly. Tumor burden (mm 3 ) was estimated from caliper measurements by the formula for the volume of a prolate ellipsoid assuming unit density as: Tumor burden (mm 3 ) = (L x W 2 )/2, where L and W are the respective orthogonal tumor length and width measurements (mm). Mean tumor burdens of each of the test groups are shown in Figure 1. Median tumor burdens of each of the test groups are shown in Figure 2. Tumor burdens for individuals in groups 1-4, as well as the group median for each of groups 1-4, are plotted in Figures 3A-D, respectively; tumor burdens for individuals in groups 5-8, as well as the group median for each of groups 5-8, are plotted in Figures 4A-D, respectively; and tumor burdens for individuals in groups 9-12, as well as the group median for each of groups 9-12, are plotted in Figures 5A-D, respectively. The median tumor burden for the control group (i.e., the median for group 1 (saline)) is also plotted in each of Figures 3A-D, 4A-D, and 5A-D for reference.

[0125] Animals with tumors in excess of 2000mm 3 were euthanized, as were those found in obvious distress or in a moribund condition. Treatment started on day 3, when there were no measurable tumors, effectively preventing analysis of tumor regressions. The nature of the therapies examined suggested that little or no on- target immune-based therapeutic effect was likely prior to Day 10. For this reason, and because all of the mice had evident tumors, Day 9 was chosen as a tumor volume benchmark for which response characteristics were determined.

[0126] The primary endpoints used to evaluate efficacy were: Tumor growth delay ("T-C"), the number of tumor-free survivors at the end of the study, and the incidences of progressive disease, stable disease and regressing disease. The mean tumor growth for each group, shown as mean tumor burden in mm 3 for the 12 experimental groups is shown in Figure 1. The median tumor growth for each group, shown as mean tumor burden in mm 3 for the 12 experimental groups is shown in Figure 2. [0127] Tumor Growth Delay (T-C) is the difference between the median times it takes the treated and control groups to reach the stated evaluation size. This is calculated from the median times to evaluation size for each animal in the group, not from interpolation of the median growth curve. Tumor growth delay results showing the statistically significant delays for this study are provided in table 1.

Table 1: Tumor Growth Delay

+ = P<0.05

NS = Nonsignificant

[0128] In this study, a therapeutic response was expected to require the activation of an immune response which takes approximately 1-2 weeks. At the time of first treatment, tumor burdens were not evident. For these reasons a Benchmark day (Day 9) was selected as the first day a response could theoretically have begun to occur. Growth and/or progression delay were calculated from the first day of dosing. Other parameters such as progression free survival and tumor doubling times were measured from the benchmark day.

[0129] An animal was coded as one with progressing disease if its tumor burden increased to≥2 fold that on the benchmark day (Day 9). [0130] The time from Day 9 to progression was used to estimate "progression- free survival." A mouse was declared to have stabilized disease if its tumor burden failed to progress (defined above) for at least 2 control group doubling times, and also failed to regress to less than 50% of the Day 9 burden.

[0131 ] Mice were coded as having regressing disease if their tumor burden decreased less than 50% of that at Day 9.

[0132] Tumor doubling times were calculated for all mice with measurable tumors starting on Day 9.

[0133] All animals were observed for clinical signs

at least once daily. Animals were weighed on each day of treatment. Individual body weights were recorded 3 times weekly. Treatment-related weight loss in excess of 20% was considered unacceptably toxic. In this report, a dosage level is described as tolerated if treatment-related weight loss (during and two weeks after treatment) is <20% and mortality during this period in the absence of potentially lethal tumor burdens is≤10%. Figure 6 provides a graph of the percent mean body weight change for the 12 experimental groups. As indicated in Figure 6 (and the Maximum Treatment Related Weight Loss column in Figure 19 indicating that all subjects gained weight), the treatments were all well-tolerated and no weight loss was observed.

[0134] Upon death or euthanasia, all animals were necropsied to provide a general assessment of potential cause of death and perhaps target organs for toxicity. The presence or absence of metastases was also noted. Remarkable observations of clinical signs and necropsy findings were tabulated as well as individual and group toxicity findings. [0135] The data were analyzed by the application of a one-way

analysis of variance (ANOVA), with post-hoc analysis by the method of Shapiro-Wilk. In cases where the data did not pass testing for either normality or equal variance, a Kruskal-Wallis ANOVA by ranks was performed with post-hoc analysis by the method of Dunn's. The statistical comparisons were performed on the time to evaluation size (750mm 3 ).

[0136] For tumor dissociation, the tumor was weighed

and <1g tissue was processed according to Miltenyi Tumor Dissociation Protocol 2.2.1, Dissociation of soft tumors, before proceeding to the red blood cell lysis.

[0137] For spleen dissociation, the plunger on a 3mL syringe was used to crush the spleen in 10mL DPBS until organ was well broken up and the DPBS became cloudy with cells. The cell suspension was then filtered with a 70μπι strainer on a 50ml_ tube and washed with 20mL DPBS. The suspension was then centrifuged for 7 minutes at 300rcf and the supernatant was discarded before proceeding the red blood cell lysis.

[0138] For the red blood cell lysis, each of the tumor and spleen samples were re- suspended in 3mL ACK Lysis Buffer and then incubated for 5 minutes at room temperature. The suspension was diluted by adding 10mL DPBS, and then the cells were collected via centrifugation for 5 minutes at 300rcf. The cells were again re- suspended in 30mL DPBS and counted. The suspension was centrifuged again, and the supernatant was discarded before proceeding to the Fc block.

[0139] For the Fc block, each of the tumor and spleen samples were re- suspended at 1E+06 cells per 100μL in Flow Cytometry Staining buffer with 1μL Fc block per 1E+06 cells. The suspensions were incubated for 5 minutes at room temperature, then centrifuged for 5 minutes at 300rcf, and the supernatant was removed before proceeding to surface staining.

[0140] For the surface staining, the following antibody dilutions were prepared,

50μL per reaction in Flow Cytometry Staining Buffer (except FoxP3):

[0141] T-Cells - Tumor: CD4 1:1000 + CD8a 1:1000 + 5μL CD197 + 5μL CD62L

[0142] T-Cells - Spleen: CD4 1 :2000 + CD8a 1 :2000 + 5μL CD197 + 5μL CD62L

[0143] CD11b & GR1 - Tumor: CD11b 1:500 + GR1 1:1000

[0144] CD11b & GR1 - Spleen: CD11b 1:500 + GR1 1:1000

[0145] PD-1 - Tumor: 1:1000

[0146] PD-1 - Spleen: 1:1000

[0147] PD-L1 - Tumor: 1 :250

[0148] PD-L1 - Spleen: 1:500

[0149] CTLA-4 - Tumor: 1 :500

[0150] CTLA-4 - Spleen: 1 :500

[0151] CD8a & Ki67 - Tumor: CD8a 1 : 1000 + Ki67 1 : 1000

[0152] CD8a & Ki67 - Spleen: CD8a 1 :2000 + Ki67 1 : 1000

[0153] CD4, CD25, & FoxP3 - Tumor: CD4 1 : 1000 + CD25 1 :500; FoxP3: 1 :500 in permeabilization buffer

[0154] CD4, CD25, & FoxP3 - Spleen: CD4 1 :2000 + CD25 1 :500; 1. FoxP3: 1:500 in permeabilization buffer

[0155] For surface staining, the tumor and spleen cells were re-suspended at 1E+06 cells per 50μL. Each of the 50μL cell suspensions containing 1E+06 cells was added to 50μL antibodies diluted in staining buffer in round-bottom 96-well plate. The plate was incubated for 30 minutes in the dark at room temperature on the orbital shaker, setting 2.5. The samples were diluted by adding 200μL Flow Cytometry Staining Buffer and then centrifuged for 5 minutes at 300rcf. The supernatant was aspirated and the cells were washed wash by re-suspending in 250μL Flow

Cytometry Staining Buffer and spinning. If staining for intracellular markers, after washing, the sample was then prepared according to the intracellular staining procedure described below. The samples were then re-suspended in 250μL Flow Cytometry Staining Buffer and were ready for Flow Cytometry Analysis.

[0156] When staining for intracellular markers, following the wash step, the cells were re-suspended in 200μL FoxP3 Fixation/Permeabilization working solution and incubated for 30 minutes in the dark at room temperature. The samples were then centrifuged at 400rcf for 5 minutes at room temperature, then the supernatant was discarded. 200μL 1X permeabilization buffer was added to each well. The samples were centrifuged at 400rcf for 5 minutes at room temperature, then the supernatant was discarded. 200μL 1X permeabilization buffer was added to each well for a second time. The samples were again centrifuged at 400rcf for 5 minutes at room temperature, then the supernatant was again discarded. The cells were then re- suspended in 50μΙ- permeabilization buffer and 50μL 1X permeabilization buffer with anti-FoxP3 or Ki67 was added. The suspension was incubated for 60 minutes in the dark at 4°C. The samples were diluted by adding 200μL 1X permeabilization buffer, and then centrifuged for 5 minutes at 400rcf. The supernatant was removed, the cells were washed with 250μL Flow Cytometry Staining Buffer per well, followed by centrifugation for 5min at 400rcf. The washing was repeated once (supernatant again removed, the cells washed with 250μL Flow Cytometry Staining Buffer per well, followed by centrifugation for 5min at 400rcf). The cells were re-suspended in 250μL Flow Cytometry Staining Buffer per well, and the samples are ready for Flow Cytometry Analysis. [0157] For the flow cytometry analysis, once the samples were prepared, the 96- well plate is loaded into the Attune Autosampler. Several samples (from which no data were generated) were required to define the instrument settings— these include unlabeled cells for voltage optimization and gating, as well as fluorescence-minus- one controls to validate the gates. The workspace was customized, beginning with a side scatter (SSC) versus forward scatter (FSC) dot plot and adding relevant "daughter" plots to display fluorescence data. This initial plot was used to gate on live cells, live tumor cells, or live lymphocytes, and further analysis was performed on only the selected populations. Once the workspace was set up, the autosampler acquired data.

[0158] In order to generate a compensation matrix, AbC mouse/rat beads were used. These beads contain two components: capture beads which bind to the heavy chain of any antibody generated in their designated species and negative beads with no antibody binding capacity. The beads provide very strong positive and negative signals which can be used to calculate the emission spillover between channels from fluorophores being used.

[0159] To begin, the AbC capture beads and negative beads were re-suspended by vortexing prior to use. For each fluorophore-conjugated antibody used, a sample tube was prepared with 1 drop of the appropriate species-specific capture beads and 50μL antibody at the determined working dilution. The solution in the sample tube was mixed well and incubated for 15 minutes are room temperature in the dark. 3ml_ Flow Cytometry Staining Buffer was added to sample tubes to dilute antibody, and then centrifuged for 5 minutes at 200rcf. The supernatant was removed and the bead pellet was re-suspended in 500μL Flow Cytometry Staining Buffer. One drop of negative beads was added to each tube and mixed well. The samples were then analyzed by flew cytometry.

[0160] Figures 7-14 provide graphs of the results of the flow cytometry analysis for various markers tested In groups 7 through 12. The graphs also categorize each of the individuals in the groups as having progressive disease, stable disease, or as a tumor-free survivor. Figure 7 graphs the percentage of CD4 + /CCR7 + /CD62L + cells in tumors; Figure 8 graphs the percentage of CD8 + /CCR7 + /CD62L + cells in tumors; Figure 9 graphs the percentage of CD11b + /GR1 + cells in tumors; Figure 10 graphs the percentage of CD4 + /CD25 + /FoxP3 + cells (T reg cells) in tumors; Figure 11 graphs the percentage of CD4 + /CCR7 + /CD62L + cells in spleens; Figure 12 graphs the percentage of CD8 + /CCR7 + /CD62L + cells in spleens; Figure 13 graphs the percentage of CD11b + /GR1 + cells in spleens; and Figure 14 graphs the percentage of CD4 + /CD25 + /FoxP3* cells (T reg cells) in spleens.

[0161] Day 0 - The day tumors are implanted into the

animals (not to be confused with the first day of treatment which is always indicated relative to Day 0).

[0162] Evaluation size - The tumor burden (mm 3 ) selected for calculation of tumor growth delay. The Evaluation Size is selected from the exponential portion of the control tumor growth curve where the error of measurement tends to be minimal (usually between 500 and 1000mm 3 ).

[0163] Progressive Disease (PD) - An animal was credited with progressive disease when within the time frame of the experiment there was a >2x increase in tumor size when compared to the benchmark day. Figure 19, PD column, discloses the percentage of the subjects within each group presenting with progressive disease. [0164] Stable Disease (SD) - An animal was credited with stable disease when within the time frame of the experiment there is a period of time during which the tumor never gets to >2x the size observed on the benchmark day or never gets to less than 50% of the size vs the benchmark day. Figure 19, SD column, discloses the percentage of the subjects within each group presenting with stable disease.

[0165] Tumor-Free Survivors (TFS) - Any animal with no measurable evidence of disease on the last day of the experiment. This value is exclusive of CRs. Figure 19, TFS column, discloses the percentage of the subjects within each group presenting as tumor free.

[0166] Rx Related Death - An animal is presumed to experience a treatment- related death if it is found dead or is euthanized in moribund condition within 2 weeks of the last treatment with a tumor burden less than half that of the smallest lethal tumor in the control group and shows no evidence of infection, mechanical dosing trauma, or other obvious causes of morbidity at necropsy. This is an individual toxicity parameter. As shown in Figure 19, Rx Related Death column, all of the treatments were well tolerated as no individuals died or were euthanized as a result of any treatment.

[0167] Tumor Doubling Time - The growth rate of the tumor expressed as the volume doubling time (days). Calculated from a log-linear least squares regression of the exponential portion of the tumor growth curve. These values are used to compute tumor cell kill, fractional effect, and surviving fraction estimates. They are also used to assess the appropriateness of the biology of the tumor in this

experiment against historical values.

[0168] Therapeutic Index - We define therapeutic index as simply the range of tolerated dosage levels that produce substantial anticancer activity. Substantial activity for this purpose is defined as a tumor growth delay that is≥ the duration of treatment and that is also statistically different from the control at the P≤0.05 level.

[0169] Time to Evaluation Size - The time (days) it takes a tumor to reach the specified Evaluation Size. Calculated from a log-linear least squares best fit of tumor burden versus time for the exponential portion of the final (post-treatment) tumor growth curve. This value is calculated for every animal in the experiment. The group medians are then used to calculate the Tumor Growth Delay. This is an individual efficacy parameter.

[0170] Tumor Burden at Last Rx - The tumor burden on the last day of treatment. This value is calculated from a log-linear least squares best fit of tumor burden versus time for the exponential portion of the final (post-treatment) tumor growth curve. (Presented to facilitate T/C comparisons.)

Example 1

[0171] A study was undertaken to evaluate anti-tumor effects, and thus, potential cancer treatments of at least one T-cell checkpoint inhibitor in combination with at least one E-selectin inhibitor and/or at least one CXCR4 receptor inhibitor. In particular, this study investigated anti-tumor effects of administering at least one anti-mPD-L1 immune checkpoint inhibitor antibody and GMI-1359 as well as of administering GMI-1359 alone. In addition, a preliminary examination of potential changes in immune cell endpoints was undertaken.

[0172] All mice were distributed into treatment groups at the time of implant prior to a tumor being established. All animals weighed≥16.6g at the initiation of therapy. Mean group body weights for all animals at first treatment were well-matched (range 17.4-18.3g). A tumor burden of 750mm 3 was chosen for evaluation of efficacy by tumor growth delay.

[0174] Between Days 21-30, all Group 1 control animals were euthanized via C02 inhalation due to tumor burden exceeding >2000mm 3 . Necropsies were performed and the findings were: enlarged spleens (9/10), and white striated tissue located on the right ventricle (1/10). One mouse did not have remarkable necropsy findings.

[0 75] The median time to evaluation size was 15.9 days from the start of dosing (Day 3) and the median post Day 9 tumor volume doubling time for Group 1 was 3.1 days. There were no spontaneous regressions in the control group and the take rate was 100%. All mice in the Control Group were identified with progressive disease. The median time to progression was on Day 11.6 and the progression free survival period was 2.6 days.

[0176] On Day 15 all Group 7 control animals were euthanized via C02 inhalation 24 hours post dose for tumor and spleen collection. Necropsies were performed and the following findings were noted: enlarged spleens, (3/5 mice), enlarged uterine horn (1/5), white striated tissue located on the right ventricle (2/5). One mouse did not have any remarkable necropsy findings.

[0177] The median time to evaluation size was 2.6 days and the median post Day 9 tumor volume doubling time for Group 7 was 2.1 days. There were no spontaneous regressions in the control group and the take rate was 100%. All mice in the group were identified with progressive disease. These findings indicate that Groups 1 and 7 were closely matched for tumor growth characteristics.

[0178] The average percentage of T-cells detected in the Group 7 tumors via flow cytometry was 7.52% CD4 + cells and 23.83% CD8 + cells. There was an average of 4.14% myeloid-derived suppressor cells (MDSCs) and 3.37% regulatory T-cells (Tregs) in the tumors. The average percentages of cells expressing PD-1, PD-L1, and CTLA-4 were 28.07%, 29.57%, and 36.51%, respectively. Neither

CD47CCR77CD62L nor CD8VCCR77CD62L " populations were detected in the tumor samples. The average percentage of CD4 + T-cells expressing both CCR7 and CD62L was 28.75% while the average percentage of CD4 + T-cells expressing CD62L but not CCR7 was 25.54%. Within the CD8 + T-cell population, the average percentage of CCR7 + /CD62L + cells was 26.47% while the percentage of CD8 + T- cells expressing CD62L but not CCR7 was 23.82%.

[0179] In the spleens of the Group 7 mice, an average of 12.62% of the cells were CD4* and 8.04% were CD8*. The average percentage of MDSCs was 2.95% and Trag was 8.29%. The average percentages of cells expressing PD-1, PD-L1, and CTLA-4 were 4.17%, 82.74%, and 7.11%, respectively. The average percentage of CD4 + T-cells expressing both CCR7 and CD62L was 3.69%, expressing CCR7 but not CD62L was 33.57%, and expressing CD62L but not CCR7 was 8.06%.

[0180] Within the CD8 + T-cell population, the average percentage of

CCR7 + /CD62L + cells was 2.10%, expressing CCR7 but not CD62L was 29.49%, and expressing CD62L but not CCR7 was 6.04%.

[0182] In Group 2, treatment with GMI-1359 was well tolerated, resulting in no treatment-related mortality. No weight loss was associated with treatment. Treated animals experienced a 16.8% mean weight gain during the treatment regimen.

Between Days 18-49, all Group 2 animals were euthanized via C02 inhalation due to tumor burden exceeding >2000mm 3 . Necropsies findings were similar to those in vehicle treated mice (Groups 1 and 7).

[0183] In Group 8, treatment with G I-1359 was well tolerated, resulting in no treatment-related mortality. No weight loss was associated with treatment. Treated animals experienced a 7.0% mean weight gain during the treatment regimen. On Day 15, all Group 8 animals were euthanized via C02 inhalation 24 hours post dose for tumor and spleen collection. Necropsies were unremarkable. These findings indicate that treatment with GMI-1359 was well tolerated and that Groups 2 and 8 were closely matched for tolerance to treatment.

[0185] In Group 3, treatment with anti-KLH; Rat lgG2b, LTF-2 was well tolerated, resulting in no treatment-related mortality. No weight loss was associated with treatment. Treated animals experienced a 9.7% mean weight gain during the treatment regimen. Between Days 18-37, all Group 3 animals were euthanized via C02 inhalation due to tumor burden exceeding >2000mm 3 . Necropsies were performed and the following findings were noted: enlarged spleens (10/10), and white striated tissue located on the right ventricle (1/10). One mouse had a non- weeping ulcerated tumor.

[0186] In Group 9, treatment with anti-KLH; Rat lgG2b, LTF-2 was also well tolerated, resulting in no treatment-related mortality. No weight loss was associated with treatment. Treated animals experienced a 9.3% mean weight gain during the treatment regimen. On Day 15, all Group 9 animals were euthanized via C02 inhalation 24 hours post dose for tumor and spleen collection. Necropsies were unremarkable. [0187] These findings indicate that treatment with anti-KLH; Rat igG2b, LTF-2 was well tolerated and that Groups 3 and 9 were well matched for tolerance to treatment with anti-KLH; Rat lgG2b, LTF-2.

[0189] In Group 4, treatment with anti-mPD-L1 , 10F.9G2 was well tolerated, resulting in no treatment-related mortality. No weight loss was associated with treatment. Treated animals experienced a 10.1% mean weight gain during the treatment regimen. Between Days 23-37, Mice 1, 2; 3, 4, 5 and 9 were euthanized via C02 inhalation due to tumor burden exceeding >2000mm 3 . Necropsies were performed and the following findings were noted: enlarged spleens (6/10), enlarged uterine horn (3/10), and one mouse had a non-weeping ulcerated tumor.

[0190] Mice 6, 7, 8 and 10 were re-implanted subcutaneously (left, high axilla) on Day 44 post initial implant. On Day 81, Mice 6, 7, 8 and 10 were euthanized via C02 inhalation per client request. Mice 8 and 10 enlarged uterine horns at necropsy. Necropsies of Mice 6 and 7 were unremarkable.

[0191] In Group 10, treatment with anti-mPD-L2, 10F.9G2 was also well tolerated, resulting in no treatment-related mortality. No weight loss was associated with treatment. Treated animals experienced a 1.6% mean weight gain during the treatment regimen. On Day 15, all Group 10 animals were euthanized via C02 inhalation 24 hours post dose for tumor and spleen collection. Necropsies were performed and the following findings were noted: enlarged uterine horn (3/5); and 1/5 mice was noted as having white striated tissue located on the right ventricle. These findings indicate that treatment with anti-mPD-L2, 10F.9G2 was well tolerated and that Groups 4 and 10 were well matched with respect to tolerance of the treatment regimen.

[0193] In Group 5, treatment with GMI-1359 + anti KLH; Rat lgG2b, LTF-2 was well tolerated, resulting in no treatment-related mortality. No weight loss was associated with treatment. Treated animals experienced a 16.7% mean weight gain during the treatment regimen. Between Days 16-28, all Group 5 animals were euthanized via C02 inhalation due to tumor burden exceeding >2000mm 3 .

Necropsies were performed and the following findings were noted: enlarged spleens (10/10); white striated tissue located on the right ventricle (1/10); enlarged uterine horn (3/10); and 2/10 mice were noted as having slightly discolored "black" intestines.

[0194] In Group 1 , treatment with GMI-1359 + anti KLH, Rat lgG2b, LTF-2 was also well tolerated, resulting in no treatment-related mortality. No weight loss was associated with treatment. Treated animals experienced a 3.5% mean weight gain during the treatment regimen. On Day 15, all Group 10 animals were euthanized via C02 inhalation 24 hours post dose for tumor and spleen collection. Necropsies were performed and the following findings were noted: enlarged spleen (1/5); enlarged uterine horn (2/5); white striated tissue located on the right ventricle (1/5); and 2/5 mice were noted as having no remarkable findings.

[0195] These findings indicate that treatment with GMI-1359 + anti KLH, Rat lgG2b, LTF-2 was well tolerated and that Groups 5 and 11 were well matched with respect to tolerance of the treatment regimen.

[0197] In Group 6, treatment with GMI-1359 + anti-mPD-L1 , 10F.9G2 was well tolerated, resulting in no treatment-related mortality. No weight loss was associated with treatment. Treated animals experienced a 9.5% mean weight gain during the treatment regimen. Between Days 18-37, Mice 1, 2, 3, 5, 7 and 10 were euthanized via C02 inhalation due to tumor burden exceeding >2000mm 3 . Necropsies were performed and the following findings were noted: enlarged spleens (6/10), alopecia on the nose/muzzle (2/10), non-weeping ulcerated tumors (2/10), and 4 mice without remarkable findings. Mice 4, 6, 8, and 9 were re-implanted subcutaneously (left, high axilla) on Day 44 post initial implant. On Day 81, Mice 4, 6, 8, and 9 were euthanized via C02 inhalation per client request. Mice 4, 6, 8, and 9 had unremarkable necropsies.

[0198] In Group 12, treatment with GMI-1359 + anti-mPD-L1 , 10F.9G2 was well tolerated, resulting in no treatment-related mortality. No weight loss was associated with treatment. Treated animals experienced a 2.5% mean weight gain during the treatment regimen. On Day 15, all Group 10 animals were euthanized via C02 inhalation 24 hours post dose for tumor and spleen collection. Necropsies were unremarkable.

[0199] The findings indicate that combination therapy with GMI-1359 + anti-mPD- L1, 10F.9G2 is well tolerated and that Groups 6 and 12 were well matched in regard to tolerance of the treatment regimen. Efficacy

[0201] In Group 2, the median time to evaluation size (750mm 3 ) was 18.4 days from the start of dosing resulting in a tumor growth delay of 2.5 days and the median post Day 9 tumor volume doubling time for Group 2 was 3.3 days, essentially identical to the control tumor doubling time. Treatment with GMI-1359 produced no regressions or tumor free survivors. All mice were identified with progressive disease (100%). Treatment with GMI-1359 produced no regressions or tumor free survivors. All mice were identified with progressive disease (100%). The median time to evaluation size was 14.3 days and the median post Day 9 tumor volume doubling time for Group 8 was 1.8 days. The median time to progression was on Day 14.5, and the progression free survival period was 5.5 days.

[0202] The average percentage of T-cells detected in the Group 8 tumors via flow cytometry was 7.02% CD4 + cells and 15.61% CD8 + cells. There was an average of 2.66% myeloid-derived suppressor cells (MDSCs) and 2.93% regulatory T-cells (Tregs) in the tumors. The average percentages of cells expressing PD-1 , PD-L1 , and CTLA-4 were 23.39%, 34.44%, and 42.80%, respectively. Neither

CD4 + /CCR7 + /CD62l_- nor CD8 + /CCR7 + /CD62L populations were detected in the tumor samples. The average percentage of CD4 + T-cells expressing both CCR7 and CD62L was 25.32% while the average percentage of cells expressing CD62L but not CCR7 was 31.43%. Within the CD8 + T-cell population, the average percentage of CCR7 + /CD62L + cells was 25.84% while the percentage of CD8 + T-cells expressing CD62L but not CCR7 was 25.65%. [0203] In the spleens of the Group 8 mice, an average of 13.43% of the cells were CD4 + and 8.77% were CD8 + . The average percentage of MDSCs was 3.70% and Trees was 10.00%. The average percentages of cells expressing PD-1, PD-L1, and CTLA-4 were 5.53%, 81.50%, and 12.31%, respectively. The average percentage of CD4 + T-cells expressing both CCR7 and CD62L was 2.05%, expressing CCR7 but not CD62L was 33.36%, and expressing CD62L but not CCR7 was 5.42%. Within the CD8 + T-cell population, the average percentage of

CCR7 + /CD62L + cells was 1.00%, expressing CCR7 + /CD62L ' was 27.95%, and expressing CD62L but not CCR7 was 2.94%.

[0204] Group 3, anti-KLH; Rat lgG2b, LTF-2, 10mg/kg, (Q3Dx2; 3off)x2.5; D3 / Group 9, anti-KLH; Rat lgG2b, LTF-2, 10mg/kg, (Q3Dx2; 3off)x2; D3

[0205] In Group 3, the median time to evaluation size (750mm 3 ) was 9.1 days from the start of dosing resulting in a tumor growth delay of 3.2 days and the median post Day 9 tumor volume doubling time for Group 3 was 3.2 days. Treatment with anti KLH; Rat lgG2b, LTF-2 produced no regressions or tumor free survivors. All mice were identified with progressive disease (100%). The median time to progression was on Day 12.4 and the progression free survival period was 3.4 days. The results in Group 9 were similar. However, one mouse never developed a tumor. A no-take was distinguishable from a response to treatment.

[0206] The average percentage of T-cells detected in the Group 9 tumors via flow cytometry was 7.18% CD4 + cells and 9.99% CD8 + cells. There was an average of 3.14% myeloid-derived suppressor cells (MDSCs) and 6.81% regulatory T-cells (Tregs) in the tumors. The average percentages of cells expressing PD-1, PD-L1, and CTLA-4 were 26.57%, 28.63%, and 38.68%, respectively. Neither

CD4 + /CCR7 + /CD62L- nor CD8 + /CCR7 + /CD62L " populations were detected in the tumor samples. The average percentage of CD4* T-cells expressing both CCR7 and CD62L was 28.09% while the average percentage of cells expressing CD62L but not CCR7 was 31.62%. Within the CD8 + T-cell population, the average percentage of CCR77CD62L* cells was 23.50% while the percentage of CD8 + T-cells expressing CD62L but not CCR7 was 26.95%.

[0207] In the spleens of the Group 9 mice, an average of 15.00% of the cells were CD4 + and 10.91% were CD8 + . The average percentage of MDSCs was 2.28% and Tregs was 10.88%. The average percentages of cells expressing PD-1 , PD-L1, and CTLA-4 were 4.80%, 84.78%, and 8.46%, respectively. The average percentage of CD4 + T-cells expressing both CCR7 and CD62L was 1.91%, expressing CCR7 but not CD62L was 37.23%, and expressing CD62L but not CCR7 was 3.52%.

Within the CD8 + T-cell population, the average percentage of CCR77CD62L* cells was 0.77%, expressing CCR7 but not CD62L was 31.59%, and expressing CD62L but not CCR7 was 1.48%.

[0208] Group 4, anti-mPD-L1 , 10F.9G2, 10mg/kg, (Q3Dx2; 3off)x2.5; D3 / Group 10, anti-mPD-L1, 10F.9G2, 10mg/kg, (Q3Dx2; 3off)x2; D3

[0209] Treatment with anti-mPD-L1 , 10F.9G2 was well-tolerated. The median time to evaluation size (750mm 3 ) was >28 days from start of dosing resulting in a tumor growth delay of 11.7 days and the median post Day 9 tumor volume doubling time for Group 4 was 4.2 days. Treatment produced a 60% incidence of progressive disease (in Mice 1, 2, 3, 4, 5 and 9) and a 40% incidence of regressing disease (Mice 6, 7, 8 and 10) which all resulted in tumor-free survivors. These mice were later re-challenged and neither primary nor re-challenged implants had any regrowth. The median time to progression was on Day 12.8 and the progression free survival period was 3.8 days. [0210] In Group 10, response status was assigned based on substantially increased tumor doubling time for respondent Euthanasia for sampling prevented an assessment of potential tumor regressions. Treatment with anti-mPD-L.1, 10F.9G2 produced a 20% incidence of progressive disease and 80% stable disease. The median time to evaluation size was not determined because the sampling group came down for analysis. The median post Day 9 tumor volume doubling time for Group 10 was 8.1 days.

[0211 ] The average percentage of T-cells detected in the Group 10 tumors via flow cytometry was 7.42% CD4 + cells and 15.52% CD8 + cells. There was an average of 2.69% myeloid-derived suppressor cells (MDSCs) and 1.69% regulatory T-cells (Tregs) in the tumors. The average percentages of cells expressing PD-1, PD-L1 , and CTLA-4 were 26.46%, 53.49%, and 43.76%, respectively. Neither

CD4 + /CCR7 + /CD62I_- nor CD8 + /CCR7 + /CD62L " populations were detected in the tumor samples. The average percentage of CD4* T-cells expressing both CCR7 and CD62L was 21.30% while the average percentage of cells expressing CD62L but not CCR7 was 34.21%. Within the CD8 + T-celi population, the average percentage of CCR7 + /CD62L + cells was 21.98% while the percentage of CD8* T-cells expressing CD62L but not CCR7 was 26.26%.

[0212] In the spleens of the Group 10 mice, an average of 12.80% of the cells were CD4 + and 6.16% were CD8 + . The average percentage of MDSCs was 1.62% and T^s was 11.24%. The average percentages of cells expressing PD-1 , PD-L1 , and CTLA-4 were 6.42%, 37.32%, and 17.15%, respectively. The average percentage of CD4 + T-cells expressing both CCR7 and CD62L was 13.00%, expressing CCR7 but not CD62L was 25.47%, and expressing CD62L but not CCR7 was 24.17%. Within the CD8 + T-cell population, the average percentage of CCR7 + /CD62L + cells was 6.69%, expressing CCR7 but not CD62L was 27.90%, and expressing CD62L but not CCR7 was 19.21%.

[0213] Group 5, GMI-1359 + anti KLH; Rat lgG2b, LTF-2, 40+1 Omg/kg,

QDx20+(Q3Dx2; 3off)x2.5; D3 / Group 11, GMI-1359 + anti KLH; Rat lgG2b, LTF-2, 40+1 Omg/kg, QDx12+(Q3Dx2; 3off)x2; D3

[0214] In Group 5, treatment with GMI-1359 + anti KLH; Rat lgG2b, LTF-2 was well-tolerated. The median time to evaluation size (750mm 3 ) was 15.8 days from the start of dosing resulting in a tumor growth delay of -0.1 days and the median post Day 9 tumor volume doubling time for Group 5 was 2.8 days. Treatment with GMI- 1359 + anti KLH; Rat lgG2b, LTF-2 produced no regressions or tumor free survivors. All mice were identified with progressive disease (100%). The median time to progression was on Day 11.2 and the progression free survival period was 2.2 days.

[0215] Findings in Group 11 were similar. There were no regressions or tumor free survivors. All mice were identified with progressive disease (100%). The median time to evaluation size was not determined because the sampling group came down for analysis. The median post Day 9 tumor volume doubling time for Group 11 was 2.2 days.

[0216] The average percentage of T-cells detected in the Group 11 tumors via flow cytometry was 5.25% CD4 + cells and 8.22% CD8 + cells. There was an average of 3.29% myeloid-derived suppressor cells (MDSCs) and 1.79% regulatory T-cells ( rees) in the tumors. The average percentages of cells expressing PD-1, PD-L1 , and CTLA-4 were 25.85%, 55.52%, and 53.54%, respectively. Neither

CD4 + /CCR7 + /CD62L- nor CD87CCR7 + /CD62L populations were detected in the tumor samples. The average percentage of CD4 + T-cells expressing both CCR7 and CD62L was 24.14% while the average percentage of cells expressing CD62L but not CCR7 was 31.16%. Within the CD8 + T-cell population, the average percentage of CCR77CD62L* cells was 17.51% while the percentage of CD8 + T-cells expressing CD62L but not CCR7 was 23.27%.

[0217] In the spleens of the Group 11 mice, an average of 13.33% of the cells were CD4 + and 9.06% were CD8 + . The average percentage of MDSCs was 3.09% and Tregg was 11.14%. The average percentages of cells expressing PD-1 , PD-L1 , and CTLA-4 were 8.18%, 84.00%, and 18.44%, respectively. The average percentage of CD4 + T-cells expressing both CCR7 and CD62L was 11.19%, expressing CCR7 but not CD62L was 31.45%, and expressing CD62L but not CCR7 was 17.33%. Within the CD8* T-cell population, the average percentage of

CCR7 + /CD62L + cells was 5.68%, expressing CCR7 but not CD62L was 35.85%, and expressing CD62L but not CCR7 was 11.18%.

[0218] Groups 6 and 12, GMI-1359 + anti-mPD-L1, 10F.9G2, 40+10mg/kg

[0219] In Group 6, the median time to evaluation size (750mm 3 ) was >29 days from the start of dosing resulting in a tumor growth delay of 13.5 days and the median post Day 9 tumor volume doubling time for Group 6 was 5.9 days. Treatment with GMI-1359 + anti-mPD-L1 , 10F.9G2 produced a 60% incidence of progressive disease, and a 30% incidence of regressing disease. Mice 4, 6, 8 and 9 were identified as tumor free survivors. Mouse 9 was unable to be identified as PD, SD, or RD because a tumor never became present and could be considered as a complete responder or in a rare incidence a no-take. The incidence of no-takes in control groups was 2% across 7 previous studies.

[0220] Response characteristics in Group 12 were similar. The median time to evaluation size was not determined because the sampling group came down for analysis. The median post Day 9 tumor volume doubling time for Group 12 was 3.7 days. The median time to progression was on Day 12.6 and the progression free survival period was 3.6 days.

[0221] The average percentage of T-cells detected in the Group 12 tumors via flow cytometry was 8.25% CD4 cells and 16.98% CD8 + cells. There was an average of 4.38% myeloid-derived suppressor cells (MDSCs) and 0.91% regulatory T-cells (Tregs) in the tumors. The average percentages of cells expressing PD-1, PD-L1, and CTLA-4 were 23.76%, 50.24%, and 45.71%, respectively. Neither

CD4 + /CCR7 + /CD62L " nor CD8 + /CCR7 + /CD62L populations were detected in the tumor samples. The average percentage of CD4 + T-cells expressing both CCR7 and CD62L was 21.70% while the average percentage of cells expressing CD62L but not CCR7 was 25.84%. Within the CD8 + T-cell population, the average percentage of CCR7 + /CD62L + cells was 20.24% while the percentage of CD8* T-cells expressing CD62L but not CCR7 was 23.32%.

[0222] In the spleens of the Group 12 mice, an average of 12.33% of the cells were CD4 + and 8.01% were CD8\ The average percentage of MDSCs was 2.22% and T^s was 11.66%. The average percentages of cells expressing PD-1 , PD-L1 , and CTLA-4 were 9.23%, 47.91%, and 17.93%, respectively. The average percentage of CD4 + T-cells expressing both CCR7 and CD62L was 7.15%, expressing CCR7 but not CD62L was 32.54%, and expressing CD62L but not CCR7 was 12.52%. Within the CD8 + T-cell population, the average percentage of

CCR7 + /CD62L + cells was 2.77%, expressing CCR7 but not CD62L was 35.65%, and expressing CD62L but not CCR7 was 6.36%.

[0223] Figure 19, Tumor Growth Delay column, provides the results, in days, that each treatment delayed tumor growth. Treatment with GMI-1359 alone did not produce a statistically significant (p>0.05) anti-tumor effect and all animals were identified with progressive disease with a minimal tumor growth delay of 2.5 days. Treatment with anti-mPD-L1 as a single agent produced a statistically significant (p<0.05) anti-tumor effect where 30% of the mice were identified as having regressing disease and 40% as tumor free survivors with a median tumor growth delay of 11.7 days.

[0224] Treatment with GMI-1359 in combination with anti-mPD-L1 also produced a statistically significant (p<0.05) anti-tumor effect vs. all groups where 30% of the mice were identified as having regressing disease and 40% as tumor free survivors with a median tumor growth delay of 13.5 days.

[0225] In Group 4 (anti-mPD-L1 alone) and Group 6 (GMI-1359 + anti-mPD-L1) four mice each were re-implanted subcutaneously (left, high axilla) on Day 44. All re- challenged mice were held out to Day 81 when they were euthanized per client request as no re-growth occurred. That is, as indicated in Figure 20, mice that achieved a complete response, even those with a quicker complete response due to the treatment combining both GMI-1359 and anti-mPD-U, rejected a subsequent challenge to CT-26.

[0226] Within the efficacy am s, there was a difference between the anti-mPD-L1 monotherapy arm and the GMI-1359 + anti-mPD-L1 combination arm. While the overall response rate was the same, the mice in the combination group

demonstrated an earlier response. Furthermore, mice in group 1 (saline control) and group 2 (with single agent GMI-1359) were all identified with progressive disease (see Figure 19). In contrast, as shown in Figures 19 and 20, treatment with anti- mPD-L1 alone (group 4) or in combination with GMI-1359 (group 6) produced a 40% complete response (CR), or tumor free survivor (TFS), rate. [0227] The median time to CR was shorter when anti-mPD-L1 was combined with GMI-1359 (group 6) compared to anti-mPD-L1 alone (group 4). As shown in Figure 20, the median time to CR for treatment group 6, treated with anti-mPD-L1 was combined with GMI-1359, versus 23 days for group 4, treated with anti-mPD-L1 alone (p<0.0471). Evaluation of tumor infiltrating cells showed that combination therapy with GMI-1359 and anti-mPD-L1 antibody (group 12) reduced the percentage of T reg compared to treatment with saline (group 7), GMI-1359 (group 8), or the anti-mPD-L1 antibody (group 10) as single treatments (0.9% vs. 3.3%, 2.9% and 1.9%, respectively (see Figure 10)). No other T cell subsets were affected (see, e.g., Figures 7 and 8 (other T cells) as compared to Figure 10 (T reg cells)). In spleens, the median percentage of T reg were unaffected by any of the treatments (see, e.g., Figure 14) and suggest that the reduction in intra-tumoral by combined treatment with anti-PD-L1 and the heterobifunctional E-selectin and CXCR4 receptor inhibitor GMI-1359 was an attenuated response to maintenance and homing signals in the tumor microenvironment.

[0228]

[0229] Within the CD4- and CD8-positive T cell population's expression of CCR7 and CD6L, two lymph node homing molecules were investigated. Co-expression of CCR7 and CD62L is found on central memory T cells (TCM). The tumor

lymphocytes were found to differ from the splenic lymphocytes in that, within the tumor, no CD4 + /CCR7 + /CD62L " or CD8 + /CCR7 + /CD62L- populations could be found whereas, within the spleen, these populations were quite abundant.

[0230] Furthermore, the combination of GMI-1359 and anti-mPD-L1 treatments resulted in the lowest levels of T regulatory (T reg ) cells and the least amount of intra- group variability in the data. [0231] In both spleens and tumors, the CTLA-4* population increased following treatment with anti-mPDL1 antibody. TIL CD4 + /CCR77CD62L + appear to decrease with anti-mPD-L1 or GMI-1359 and anti-mPD-L1 treatments. TIL PD-L1 levels increase with anti-mPD-L1 alone, GMI-1359 + isotype control and GMI-1359 + anti- mPD-L1 treatments as compared to vehicle, GMI-1359 alone, or isotype control alone, respectively. Figure 7 provides a graph of the percentage of

CD47CCR77CD62L* in tumors for the individuals in experimental groups 7-12, and Figure 11 provides a graph of the percentage of CD47CCR7 + /CD62L + in the spleens of the individuals in experimental groups 7-12.

[0232] In the tumors, the average percentage of Treg cells remained the same when the mice were treated with the anti-mPD-L1 antibody (see, e.g., exemplary scatter plot for Group 10, Figure 15A) or the GMI- 359 plus the isotype control antibody (see, e.g., exemplary scatter plot for Group 11, Figure 15B). However, when the mice were treated with a combination of GMI-1359 and anti-mPD-L1 antibody (see, e.g., exemplary scatter plot for Group 12, Figure 15C), the average percentage of Τ^ cells decreased.

[0233] More differences in markers between treatment groups were observed in the spleens compared to the tumors. Treatment with the anti-mPD-L1 antibody (Group 10), GMI-1359 + isotype control antibody (Group 11), or the combination of GMI-1359 and the anti-mPD-L1 antibody (Group 12) significantly increased the percentages of CD47CCR77CD62L* cells compared to treatment with the vehicle (Group 1), GMI-1359 (Group 2), or the isotype control antibody (Group 3), respectively.

[0234] As noted above, Figure 16A is a representative scatter plot showing the CD47CCR77CD62L cells in spleens of an individual (mouse 4, group 8) treated with GMI-1359. Figure 16B is a representative scatter plot showing the

CD4 + /CCR7 + /CD62L + cells in spleens of an individual (mouse 1, group 12) treated with GMI-1359 and anti-PD-L1 antibody treatment. These graphs indicate that GMI- 1359 in combination with the isotype control antibody affected the percentage of CD4 + /CCR7 + /CD62L + cells while GMI- 359 as a single agent did not.

[0235] Differences between groups in the percentages of cells that were

CD87CCR7 + /CD62L + were also detected in the spleens. Figure 12 provides a graph of the percentage of CD8 + /CCR77CD62L + cells in the spleens in experimental groups 7 through 12. When the mice were treated with the anti-mPD-L1 antibody (Group 10) or GMI-1359 together with the isotype control antibody (Group 11) the average percentage of CD87CCR77CD62L* cells increased compared to the vehicle control (Group 7). Figure 17A shows a representative scatter plot of data from Mouse 4 in Group 7, and Figure 17B shows a representative scatter plot of data from Mouse 3 in Group 10. When the mice were grouped according to the state of the disease and the percentages were dot-plotted in columns, the mice with stable disease in Group 12 had statistically significantly lower percentages of MDSCs than the mice with progressive disease (Figure 13, providing a graph of the percentage of CD11b + /GR1 + (MDSCs) in Spleens in groups 7-12). Representative scatter plots showing these differences are shown in Figures 18A-C. Mouse 3 from Group 9 (Figure 18A) represents all of those from Group 9, Mouse 3 from Group 12 (Figure 18B) represents those mice from Group 12 with stable disease, and Mouse 4 from Group 12 (Figure 18C) represents those mice from Group 12 with progressive disease.

Example 2 [0236] Figure 21 relates to an experiment carried out to determine the competitive binding activity (IC50) of GMI-1359 against E-selectin and CXCR4. GM 1-1359 was assessed for inhibition of sialyl Le x binding to immobilized E-selectin and a-CXCR4 antibody binding to Raji cells.

[0237] The inhibition assay to screen G I-1359 as an antagonist of E-selectin was a competitive binding assay, which allowed the determination of IC 50 values. Human E-selectin/lg chimera was immobilized by incubation at 37 °C for 2 hour in 96-well microtiter plates. To reduce nonspecific binding, BSA was added to each well and incubated at room temperature for 2 hours. After incubation with E- selectin/lg chimera, the plate was washed and serial dilutions of the test compounds were added to the wells in the presence of conjugates of biotinylated, sLe a - polyacrylamide with streptavidin/horseradish peroxidase and incubated for 2 hours at room temperature. To determine the amount of sLe a bound to immobilized E- seiectin after washing, the peroxidase substrate, 3,3',5,5' tetramethylbenzidin (TMB) was added. After 3 minutes, the enzyme reaction was stopped by the addition of H 3 P04, and the absorbance of light at a wavelength of 450 nm was determined. The absorbance of light readings were plotted as a function of increasing concentration of GMI-1359 using GraphPad Prism software, and the concentration of GMI-1359 compound required to inhibit binding by 50% was determined and reported as the IC50 value for GMI-1359.

[0238] The inhibition assay to screen GMI-1359 as an antagonist of CXCR4 was a flow based competitive binding assay, which allowed the determination of IC 5 o values. Raji cells (ATCC number TIB-152) were washed twice with Hanks Balanced Saline Solution (HBSS) containing 0.05% bovine serum albumin (BSA). After the second wash, the cells were resuspended to about 2.5 x 10 6 cells per mL and 80 μΙ of cells (approximately 2 x 10 5 cells) were added to BD 2063 tubes. Next, 10 μΙ of either GMI-1359 or HBSS/BSA (as a negative control) were added to the cells and the tube was placed at room temperature for 10 minutes. Then, 10 μΙ of a phycoerythrin-conjugated anti-CXCR4 antibody (R&D Systems, FAB170P) or as a negative control, 10 μΙ of an isotype control antibody (R&D Systems, IC003P) was added to the cells with HBSS/BSA. The antibodies were allowed to bind to the cells for 1 hour at 4°C. Next, 2 mL of cold HBSS/BSA were added to all the tubes, and the cells were pelleted by centrifugation at 250 x g for 10 minutes. The supernatants were discarded and the cell pellets were resuspended in 1 mL of HBSS/BSA. The cells were pelleted again as before, suspended in 150 μΐ of HBSS/BSA and fixed by the addition of 150 μΙ of 2% formaldehyde. Binding of the anti-CXCR4-PE antibody to the cells was assessed by flow cytometry, and the median fluorescent intensity was determined . The median fluorescent intensities were plotted as a function of increasing concentration of GMI-1359 using GraphPad Prism software, and an IC50 (defined as the concentration of GMI-1359 resulting in a 50% inhibition of

phycoerythrin-conjugated anti-CXCR4 antibody) was determined. As shown in Figure 21, the results showed that the small molecule glycomimetic, GMI-1359, inhibits ligand binding to both E-selectin and CXCR4.

Example 3

[0239] Figure 22 relates to an experiment carried out to determine percentages of CD4+, CD8+, and Regulatory T cells (CD4+, FoxP3+, and CD25+) in spleen and tumor tissue samples, in vivo, on study day 15, from each treatment group. Details of the experimental treatments for each of the groups are provided above with respect to Example 1. [0240] Twenty-four hours following the final dose of GMI-1359, five mice from each treatment group were euthanized and spleens and tumors were processed for flow cytometry. Tumors were dissociated according to Miltenyi Dissociation Protocol for soft tumors. Single cell suspensions from spleen were obtained by maceration.

[0241] The following cell determinants were assessed using fluorescent conjugated reagents for flow cytometry: rat anti-CD4 FITC conjugate, clone GK1.5, rat anti-CD8a APC-AlexaFluor 750 conjugate, rat anti-CD11b PE conjugate, clone M1 70.15, rat anti-CD25 PE conjugate, clone PC61 5.3, mouse anti-FoxP3 APC conjugate, clone 3G3, rat anti-GR1 (LY6C/G) APC conjugate, clone 1A8, rat anti- CD62L PE conjugate, clone MEL 12-H2.100, hamster anti-CD152 (CTLA-4) PE conjugate, clone UC10-4F10-11, rat anti-CD279 (PD-1) FITC conjugate, clone RMPI-30, and rat anti-CD274 (PD-L1, B7-H1) APC conjugate, clone 10F.9G2.

[0242] Once the samples were prepared, a 96-well plate was loaded into the Attune Autosampler. Several samples (from which no data were generated) were required to define the instrument settings— these include unlabeled cells for voltage optimization and gating, as well as fiuorescence-minus-one controls to validate the gates. The workspace was customized, beginning with a side scatter (SSC) versus forward scatter (FSC). This initial plot was used to gate on live cells, live tumor cells, or live lymphocytes, and further analysis was performed on only the selected cell determinants. Data was acquired from either 250,000 events or 180 μL, whichever threshold was reached first.

[0243] Figure 22 shows the results for percentage of total CD4+ and CD8+ lymphocytes and regulatory T cells.

Example 4 [0244] Figure 23 relates to an experiment carried out to determine the ratio of CD8/regulatory T cells in spleen and tumor tissue samples, in vivo, on day 15.

Details of the experimental treatments for each of the groups are provided above with respect to Example 1.

[0245] Twenty-four hours following the final dose of GMI-1359, five mice from each treatment group were euthanized and spleens and tumors were processed for flow cytometry. Tumors were dissociated according to Miltenyi Dissociation Protocol for soft tumors. Single cell suspensions from spleen were obtained by maceration.

[0246] The following cell determinants were assessed using fluorescent conjugated reagents for flow cytometry: rat anti-CD4 FITC conjugate, clone GK1.5, rat anti-CD8a APC-AlexaFluor 750 conjugate, rat anti-CD25 PE conjugate, clone PC61 5.3, and mouse anti-FoxP3 APC conjugate, clone 3G3.

[0247] Once the samples were prepared, a 96-well plate was loaded into the Attune Autosampler. Several samples (from which no data were generated) were required to define the instrument settings— these include unlabeled cells for voltage optimization and gating, as well as fluorescence-minus-one controls to validate the gates. The workspace was customized, beginning with a side scatter (SSC) versus forward scatter (FSC). This initial plot was used to gate on live cells, live tumor cells, or live lymphocytes, and further analysis was performed on only the selected cell determinants. Data was acquired from either 250,000 events or 180 μί, whichever threshold was reached first.

[0248] The results showed that combination therapy with GMI-1359 and anti-PD- L1 antibody reduced the percentage of intratumoral Tng compared to treatment with saline, GMI-1359, or the anti-mPD-L1 antibody as single treatments (0.9% vs. 3.3%, 2.9%, and 1.9%, respectively). No other T cell subsets were affected. As shown in Figure 23, reduction in intratumoral Tmg cells resulted in a more favorable increase in the ratio of total CD8 T cells to T reg cells. In spleens, the median percentage of T reg cells were unaffected by any of the treatments, which suggests that the reduction in intratumoral cells by combined treatment with anti-PD-L1 and GMI-1359 was an attenuated response to maintenance and homing signals in the tumor

microenvironment.

Example 5

[0249] Figures 24A and 24B relate to an experiment carried out to compare the mean tumor burden and the responsiveness to treatments in each group. Details of the experimental treatments for each of the groups are provided above with respect to Example 1.

[0250] Tumor volumes were estimated from caliper measurements recorded three times weekly beginning at the start of treatment. Tumor burden (mm 3 ) was calculated by the formula for the volume of a prolate ellipsoid assuming unit density as: Tumor burden (mm 3 ) = (L x W 2 )/2, where L and W are the respective orthogonal tumor length and width measurements (mm). Animals with tumors in excess of 2000 mm 3 were euthanized.

[0251] Figure 24A shows the primary endpoints used to evaluate efficacy: tumor growth delay; the number of tumor-free survivors at the end of the study; the incidences of progressive disease, stable disease, and regressing disease; and the response. Additionally, Figure 24B shows the number of days post tumor implant on the x-axis and the mean tumor burden (mm 3 ) in each treatment group on the y-axis.

[0252] As shown in Figures 24A and 24B, all treatments of tumor-bearing mice were well tolerated, resulting in no treatment-related mortality. Likewise, as shown in Figure 24A, treatment with GMI-1359 in combination with anti-PD-L1 or anti-PD-L1 alone was associated with a tumor-growth delay of 13.5 and 11.7 days, respectively, and a 40% complete response rate.

[0253] The various embodiments described above can be combined to provide further embodiments. All U.S. patents, U.S. patent application publications, U.S. patent applications, non-U. S. patents, non-U.S. patent applications, and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety. Aspects of the embodiments can be modified, if necessary, to employ concepts of the various patents, applications, and publications to provide yet further embodiments.

[0254] These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure.

Articles

[0255] Callahan MK et al. Anti-CTLA-4 Antibody Therapy: Immune Monitoring During Clinical Development of a Novel Immunotherapy. Semin Oncol. Oct 2010; 37(5):473-484.

[0256] Postow MA, Callahan MK, Wolchok JD. Immune Checkpoint Blockade in Cancer Therapy. J. Clin. Oncol. Jan 2015; Epub.

[0257] Dunn GP, Old LJ, & Schreiber RD. The Immunobiology of Cancer Immunosurveillance and Immunoediting. Immunity. Aug 2004; 21:137-148.

[0258] Naidoo J, Page DB, & Wolchok JD. Immune modulation for cancer therapy. British Journal of Cancer. Sep 2014; 111:2214-2219. [0259] Kim R, Emi M, & Tanabe K. Cancer immunoediting from immune surveillance to immune escape. Immunology. May 2007; 121(1):1-14.

[0260] Schabel F, Griswold D, Laster W, Corbett T, Lloyd H. Quantitative evaluation of anticancer agent activity in experimental animals. Pharmac. Ther. A. (1) 411-435, 1977.

[0261] Corbett, T, Griswold D, Roberts B, Peckham J, Schabel F Evaluation of single agents and combinations of chemotherapeutic agents in mouse colon carcinomas. Cancer 1977; 40(5); 2660-2690.

[0262] Schabel F, Griswold D, Corbett T, Laster R, Mayo J, Lloyd H. Testing therapeutic hypotheses in mice and man: Observations on the therapeutic activity against advanced solid tumors of mice treated with anticancer drugs that have demonstrated or potential clinical utility for treatment of advanced solid tumors of man. Methods in Cancer Research (17) 3-51, 1979.

[0263] Plowman J, Dykes D, Hollingshead M, Simpson-Herren L, and Alley M. Human tumor xenograft models in NCI drug development. In: Anticancer drug development guide: preclinical screening, clinical trials, and approval. Teicher (ed) Humana Press Inc. 1993.

[0264] Corbett T, Valeriote F, LoRusso P, Polin L, Panchapor C. Pugh S. White K, Knight J, Demchik L, Jones J, Jones L, Lowichik N, Biernat L, Foster B, Wozniak A, Lisow L, Valdivieso M, Baker L, Leopold W, Sebolt J, Bissery M, Mattes K, Dzubow J, Rake J, Perni R, Wentland M, Coughlin S, Shaw JM, Liversidge G, Liversidge E, Bruno J, Sarpotdar P, Moore R, Patterson G. Tumor models and the discovery and secondary evaluation of solid tumor active agents. Int J

Pharmacognosy 1995; 33(supplement): 102-122. [0265] Corbett T, Roberts BJ, Lawson AJ, Leopold WR, et al. Transplantable Syngeneic Rodent Tumors: Solid Tumors of Mice. In: Tumor Models in Cancer Research (BA Teicher 2nd ed). Humana Press, New York, NY. pp. 43-78, 2011.

[0266] R Development Core Team. R: A Language and Environment for

Statistical Computing. 2005, R Foundation for Statistical Computing: Vienna, Austria. R, D.C.T., R: A Language and Environment for Statistical Computing. 2005, R Foundation for Statistical Computing: Vienna, Austria. http://www.R~project.org.