Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FUSION MOLECULES OF PSGL-1 OR TSGL ANIONIC DOMAINS TO CHECKPOINT-MODULATING ANTIBODIES AND OTHER ANTIBODIES
Document Type and Number:
WIPO Patent Application WO/2021/183685
Kind Code:
A2
Inventors:
SHAW GRAY (US)
Application Number:
PCT/US2021/021771
Publication Date:
September 16, 2021
Filing Date:
March 10, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SHAW GRAY D (US)
International Classes:
A61K38/00
Attorney, Agent or Firm:
LAZAR, Steven, R. (US)
Download PDF:
Claims:
What is claimed is:

1. A fusion molecule with a PSGL-1 anionic domain comprising amino acids 4 to 16 of SEQ ID NO:2 fused to an immune checkpoint modulating antibody, wherein the antibody retains its antigen binding activity.

2. A fusion molecule with a PSGL-1 anionic domain comprising amino acids 4 to 16 of SEQ ID NO:2 fused to an immune checkpoint modulating antibody at the N-terminus of its heavy chain or light chain, wherein the antibody retains its antigen binding activity.

3. A fusion molecule with a PSGL-1 anionic domain comprising amino acids 4 to 16 of SEQ ID NO:2 fused to an immune checkpoint modulating antibody at the C-terminus of its heavy chain, wherein the antibody retains its antigen binding activity.

4. A fusion molecule of any of claims 1-3, wherein the anionic domain does not contain a sialyl Lewis X (sLex) tetrasaccharide.

5. A fusion molecule of any of claims 1-3 that accumulates in tumors to a greater extent than the same checkpoint modulating antibody that is not fused with a PSGL-1 anionic domain.

6. A fusion molecule with tandem P-selectin glycoprotein ligand (TSGL) anionic domains wherein each of the anionic domains comprises amino acids 4 to 16 of SEQ ID NO:2 fused to an immune checkpoint modulating antibody, wherein the antibody retains antigen binding activity.

7. A fusion molecule with tandem P-selectin glycoprotein ligand (TSGL) anionic domains wherein each of the anionic domains comprises amino acids 4 to 16 of SEQ ID NO:2 and is fused to an immune checkpoint modulating antibody at the N-terminus of its heavy chain or light chain, wherein the antibody retains antigen binding activity.

8. A fusion molecule with tandem P-selectin glycoprotein ligand (TSGL) anionic domains wherein each of the anionic domains comprises amino acids 4 to 16 of SEQ ID NO:2 and is fused to an immune checkpoint modulating antibody at the C-terminus of its heavy chain, wherein the antibody retains antigen binding activity.

9. A fusion molecule of any of claims 6-8, wherein at least one of the anionic domains does not contain a sialyl Lewis X (sLex) tetrasaccharide.

10. A fusion molecule of any of claims 6-8 that accumulates in tumors to a greater extent than the same checkpoint modulating antibody that is not fused with a PSGL-1 anionic domain.

11. A method of treating a cancer, comprising administering to a subject in need thereof the fusion molecule of any of claims 1-5.

12. A method of treating a cancer, comprising administering to a subject in need thereof the fusion molecule of any of claims 6-10.

13. The method of claim 11, further comprising administration of at least one other active agent selected from the group consisting of additional checkpoint modulators, protein kinase inhibitors or ACT therapy.

14. The method of claim 12, further comprising administration of at least one other active agent selected from the group consisting of additional checkpoint modulators and protein kinase inhibitors or ACT therapy.

15. A method of preventing cancer metastasis, comprising administering to a subject in need thereof the fusion molecule of any of claims 1-5.

16. A method of preventing cancer metastasis, comprising administering to a subject in need thereof the fusion molecule of any of claims 6-10.

17. The method of claim 15, further comprising administration of at least one other active agent selected from the group consisting of additional checkpoint modulators, protein kinase inhibitors or ACT therapy.

18. The method of claim 16, further comprising administration of at least one other active agent selected from the group consisting of additional checkpoint modulators and protein kinase inhibitors or ACT therapy.

19. A fusion molecule with one or more PSGL-1 anionic domains comprising amino acids 4 to 15 of SEQ ID NO:2 fused to a therapeutic cancer antibody, wherein the antibody retains antigen binding activity and binds to human CCL21.

20. A fusion molecule with one or more PSGL-1 anionic domains comprising amino acids 4 to 15 of SEQ ID NO:2 fused to an anti-CD20 antibody, wherein the antibody retains antigen binding activity and binds to human CCL21.

21. A fusion molecule with one or more PSGL-1 anionic domains comprising amino acids 4 to 16 of SEQ ID NO:2 fused to an anti-SARS-Cov2 virus antibody, wherein the antibody retains antigen binding activity and binds to human CCL21.

22. A fusion molecule with one or more PSGL-1 anionic domains comprising amino acids 4 to 15 of SEQ ID NO:2 fused to an anti-CCR8 antibody, wherein the antibody retains its antigen binding activity and binds to human CCL21.

23. A fusion molecule with one or more PSGL-1 anionic domains comprising amino acids 4 to 16 of SEQ ID NO:2 fused to an anti-eNAMPT antibody, wherein the antibody retains its antigen binding activity and also binds to human CCL21.

Description:
Fusion Molecules of PSGL-1 or TSGL Anionic Domains to Checkpoint-Modulating Antibodies and

Other Antibodies

RELATED APPLICATION

[0001] This application claims priority from provisional patent application serial number 62/987,454, filed on March 10, 2020.

TECHNICAL FIELD

[0002] The present invention relates using the anionic region of PSGL-1 to enhance the tumor killing and anti-metastatic activities of checkpoint-modulating antibodies within the tumor microenvironment. More particularly, the present invention is directed to molecules having either a single anionic domain from human P-selectin glycoprotein ligand (also known as "PSGL") or two or more tandem selectin glycoprotein ligand (“TSGL”) anionic domains fused to an antibody that is selected from the group consisting of a checkpoint-modulating antibody, a therapeutic cancer antibody or an antiviral antibody, to create fusion proteins referred to as PSGL-Abs or TGSL-Abs, respectively.

BACKGROUND OF THE INVENTION

[0003] Immune checkpoint modulation therapy typically involves the use of antibodies to either stimulate T-cell activation or block the inhibitory signals of T-cell activation, thereby enabling tumor reactive T cells to overcome self-tolerance regulatory mechanisms and generate an anti-tumor response (see reviews by Ribas and Wolchok Science 2018; Kruger et al., Journal of Experimental & Clinical Cancer Research 2019). Examples of FDA approved checkpoint inhibitor (antagonistic) therapies include ipilimumab (anti-CTLA4) as well as antibodies that target the PD-1/PD-L1 axis such as the anti-PD-1 antibodies nivolumab, pembrolizumab and JTX-4014 as described in US Patent application US2018/0118829 Al. The immune checkpoint molecule known as V-region Immunoglobulin-containing Suppressor of T cell Activation (VISTA) has also been recently described as a potential target for checkpoint inhibitor therapy (see Xu et al., Cancer Immunol Res. 2019). Alternatively, examples of a checkpoint stimulator (agonistic) therapies are an antibody to ICOS such as vopratelimab (JTX-2011) or a therapeutic anti-4- IBB (CD 137) antibodies such as is described in US Patent Application US 2019/0194329 Al or PCT/US2018/041612.

[0004] The selectins (CD62P, CD62E, CD63L) are a family of C-type lectin cell adhesion molecules expressed, among other places, on certain types of circulating blood cells and on the activated vascular endothelium. During inflammation, leukocytes adhere to the vascular endothelium and enter subendothelial tissue, an interaction that is initially mediated by specific binding of the selectins to ligands on the surface of circulating cells. Such selectin-mediated cellular adhesion occurs during vascular inflammation, thrombotic disorders, parasitic diseases, and may be also implicated in metastatic spread of tumor cells. The selectin proteins are characterized by an N-terminal lectin-like domain, an epidermal growth factor-like domain, and regions of homology to complement binding proteins. Three human selectin proteins have been identified, E-selectin (formerly ELAM-1), L-selectin (formerly LAM-1) and P- selectin (formerly PAD GEM or GMP-140). E-selectin is induced on endothelial cells several hours after activation by cytokines, mediating the calcium-dependent interaction between neutrophils and the endothelium. L-selectin is the lymphocyte homing receptor, and P-selectin rapidly appears on the cell surface of platelets when they are activated, mediating calcium-dependent adhesion of neutrophils or monocytes to platelets. P-selectin is also found in the Weibel-Palade bodies of endothelial cells; upon its release from these vesicles P-selectin mediates early binding of neutrophils to histamine-or thrombin- stimulated endothelium. All three of the selectins bind, with varying affinity, to a ligand called PSGL (P- selectin glycoprotein ligand and also known as "PSGL-1"). Interaction of selectins with PSGL-1, which is expressed on some circulating lymphocytes and leukocytes, causes those circulating cells in the vasculature which express the active form of PSGL-1 to attach to platelets and/or the endothelium, where other adhesion molecules and chemokines then mediate extravasation into the surrounding tissues. Thus, the selectin PSGL-1 interaction has been a well-documented step in the development of inflammatory and immune responses, including vaso-occlusive crisis in sickle cell disease patients. In addition, a role for the selectin PSGL-1 interaction has been reported for the formation and maintenance of the tumor cell microenvironment (TME), involving the recruitment of myeloid cells to form a metastatic niche (see Borsig (2018) Glycobiology; 28:648-655).

[0005] The cDNA encoding human PSGL (also termed PSGL-1 or SELPLG or CD 162) has been cloned and is well-characterized as described in Larsen et ak, WO98/08949, and US 6,275,975, the disclosure and claims of which are hereby incorporated herein by reference. The application discloses polynucleotides encoding various forms of recombinant PSGL molecules, including numerous functional soluble forms of PSGL. Thus, PSGL is a well-characterized molecule, soluble forms of which are particularly amenable to administration as therapeutics to block selectin-mediated cell adhesion events (Busuttil et al. (2011) Am J Transplant, 11:786-97; Mertens et al. (2006) Am Heart J., 152:125 el-e8).

[0006] The human form of PSGL contains over 300 amino acids in its extracellular domain (See, Uniprot database accession number Q 14242). Remarkably, the principal binding site for P and L- selectin exists within a short 19 amino acid segment at the amino terminus of the mature form of PSGL.

The highest reported affinity measurements of soluble monomeric forms of PSGL demonstrate K D values of approximately 200-778nM when binding to P-selectin (Somers et al. (2000) Cell, 103 :467-79; Leppanen et al. ( 1999; J. Biol. Client., 274:24838-48). The binding affinity to E-selectin may vary according to the type and number of modified glycans present on the soluble form of PSGL. (Martinez et al. (2005) J. Biol. Chem. 280:5378-5390). PSGL-1 interaction with selectins on their respective cell type, including soluble recombinant forms of PSGL-1, has been shown to induce signaling via the selectin molecules. The extent to which the selectin molecules are cross linked or clustered on the surface of a cell may dictate the characteristics of such selectin mediated signaling events generated in a particular cell type (Yoshida at el. (1998 )J Immunol·, 161:933-941). It has been demonstrated that there is therapeutic potential for blocking cancer metastasis via the administration of a soluble agent that will bind to chemokines such as CCL21 (Lanati et al. (2010) Cancer Res; 70:8138-8149). It has also been demonstrated that the chemokines CCL27, CCL21 and CCL19 bind to the anionic domain of human PSGL-1 that contains sulfated tyrosines at its amino terminus (Hirata et al (2004; J. Biol. Chem. 279: 51775-51782). In contrast to the binding of selectins, this chemokine binding does not require the presence or the modification of the O-linked glycans found on human PSGL-1.

[0007] Recent studies describe an additional role of PSGL in regulating T cell response in the tumor microenvironment and homing-independent functions of PSGL-1 in immune checkpoint regulation and T cell effector activity. (Tinoco et al. (2016) Immunity; 44:1190-1203; Barthel and Schatton (2016) Immunity, 44:1083-1085). Figure 4D of WO2018132476 (Johnston et al.), indicates that full length human PSGL-1 molecules expressed on the surface of transfected CHO cells are capable of binding to multimers of the immune checkpoint molecule known as V-region Immunoglobulin-containing Suppressor of T cell Activation (VISTA; also known as PD-1H) under acidic pH conditions (pH 6.0). The inventors interpreted their results to suggest that PSGL-1 may be a direct counter-receptor for VISTA under acidic conditions. WO2018132476 describes the in vitro use of a commercially available recombinant PSGL-l-Fc protein modified with sLe x (see Figures 3, 4B and Table 1). However, the inventors do not contemplate the addition (fusion) of the small subset of PSGL-1 sequences that comprise the anionic domain (Sako et al 1995) directly to the sequences of an intact therapeutic antibody containing both light and heavy chains and having variable regions capable of binding to a therapeutic target. The inventors further theorized that antibodies binding to PSGL-1 and/or VISTA may be useful in the treatment of cancer in combination with checkpoint inhibitors or other immuno-oncology agents. However, proteins that bind to VISTA can serve as either agonists or antagonists (Tanbouly et al. (2021) Front Immunol. Il:e595950). Other groups have reported anti-cancer activity using antibodies that bind to VISTA (Noelle, US2018/0215826). However, antibodies that bind to VISTA ( e.g ., clone MH5A, Flies et al. (2011) J. Immunol. 187:1537-1541) can prevent graft vs host disease (GVHD), whereas in contrast, other anti-VISTA antibodies (e.g., clone 13F3, Wang et al. (2011) J. Exp \ / /;208:577-592) appear to enhance T cell responses to tumors. This has led to VISTA being described as acting as both a ligand and receptor on cells. Therefore, it has yet to be established what type of immune response will be elicited by the dosing of a soluble form of PSGL-1, in terms of VISTA signaling on a particular immune cell type. Additionally, Hmeljak et al. (2018) Cancer Discov; 8:1548-1565 discloses that VISTA is expressed on malignant pleural mesothelioma (MPM) cells. Regarding direct effects of selectins on tumor growth, it has also been reported that neuroblastoma tumors (Nolo et al. (2017) Oncotarget 8:86657-86670) and glioblastoma tumors (Ferber et al. (2017) eLife 2017:6:e25281) can be treated in vivo using molecules that specifically block P-selectin binding.

[0008] In a different approach of using soluble forms of the anionic domains of PSGL-1 as a competitive binding molecule to those binding partners that cell surface-bound PSGL-1 binds to, United States Patent 8,889,628 (Shaw) describes the production of enhanced soluble selectin ligands containing two or more sulfated glycoprotein peptide sequences from the human PSGL-1 anionic domain combined in a tandem configuration on a single peptide chain, designated as tandem selectin glycoprotein ligands, or TSGLs, and fusions of TSGLs with an immunoglobulin Fc, to form TSGL fusion proteins. A subsequent patent application WO2019/133454 (Shaw) contemplates the use of TSGL fusion molecules to both enhance the efficacy of adoptive cell therapy (ACT) and to reduce the unwanted cytokine storm side effects caused by ACT. The fusion molecules described are TSGL sequences fused only to an immunoglobulin fragment crystallizable (Fc) region. Neither patent 8,889,628 nor patent application WO2019/133454 contemplates the direct fusion of PSGL-1 or TSGL sequences to an intact therapeutic antibody that contains both light and heavy chains with variable regions capable of binding to a separate therapeutic target. Following an acute treatment, TSGL anionic domains modified with sLex and fused only to an IgG Fc has been previously shown to promote survival in a mouse model of syngeneic orthotopic liver transplantation (see Zhang et al. (2017) Am J Transplant; 17:1462-1475). Treatments with this same recombinant TSGL-Ig protein has also been shown to prevent vaso-occlusion in sickle cell disease (SCD) mice (Vats et al. (2020) Exp Hematol; 84: 1-6. el). These two studies have demonstrated the positive selectin blocking activity of fused TSGL sequences modified with sLe x in mouse preclinical models.

BRIEF SUMMARY OF THE INVENTION

[0009] The present invention describes the fusion of either a single anionic domain of PSGL-1 to an immune checkpoint-modulating antibody “PSGL-Abs” or fusion of multiple tandem domains “TSGL” anionic domains to checkpoint-modulating antibodies “TSGL-Abs” in order to enhance their anti-cancer activity. It is theorized by the inventor that this activity enhancement is due to the additional binding activity that the PSGL or TSGL sequences impart to the antibody and bring a separate new additional activity to the antigen binding activities of the antibody’s complementarity determining regions (CDRs) with in its variable domain or any binding activities of the antibody’s Fc domain (such as Fc receptor binding). The present invention also further describes methods of use for PSGL-Abs or TSGL-Abs.

[0010] It has further been shown that PSGL-1 binds to chemokines, such as CCL21. The inventor theorizes that the binding to chemokines of the PSGL or TSGL sequences in PSGL-Ab or TSGL- Ab fusion molecules may further contribute additional novel activities which may be advantageous, for example, in cancer treatment.

[0011] US Patent 8,232,252 (Larsen) describes the fusion of sLe x -modified glycopeptide segments of human PSGL-1 to the hinge region of a human IgG Fc fragment for the purpose of antagonizing selectins. These modified glycopeptide segments of PSGL-1 vary in size ranging from 47 to 360 amino acids. Concentrating on just the smaller anionic domains of PSGL-1, US Patent 8,889,628 describes the production of soluble tandem selectin glycoprotein ligand (TSGL) molecules comprising at least two short P-selectin glycoprotein ligand (PSGL) anionic domains combined in a tandem configuration along the same polypeptide chain and fused to any other polypeptide. The PSGL domains of such molecules preferably comprise sulfated tyrosines and can include at least one potential O-linked glycan addition site on either serine or threonine, also capable of being further modified to contain a sialyl Lewis x (sLe x ) structure or epitope. As described in certain aspects of WO2019/133454 (Shaw), one or more soluble PSGL domains may be missing the sLe x structure or epitope, and therefore the TSGL molecules, may be partially lacking, or even be completely devoid of sLe x structure or epitopes. In other aspects of the present invention, the threonine residue at position 16 of SEQ ID NO: 2 can be deleted as a means to enable both an sLe x -modified and sLe x unmodified domain to exist in the same fusion molecule (see Figure 11) when secreted from mammalian host cells. TSGL molecules lacking sLe x structures will not bind to selectins yet will retain their ability to bind via the amino acids contained in the anionic domain, including the sulfated tyrosine residues. The TSGL molecules may be fused to a non-TSGL polypeptide, such as an intact antibody molecule, to create TSGL-antibody fusions (TSGL-Abs). In certain preferred embodiments, the TSGL molecule comprises an amino acid sequence having at least 70%, 80% or 90% sequence identity to the amino acid sequence of SEQ ID NO: 2. More preferably, the TSGL molecule comprises the amino acid sequence of SEQ ID NO: 2, or a functional variant thereof having at least 70% sequence identity to the amino acid sequence of SEQ ID NO: 2.

[0012] In certain aspects, the present invention provides methods of treatment of a subject having a tumor or a subject having cancer, said method comprising administering an intact checkpoint- modulating antibody molecule with TSGL (TSGL-Ab fusion protein) to said subject in combination with a cellular therapy, such as administration of cells genetically modified with a chimeric antigen receptor (CAR), for the treatment of tumors or cancer. The soluble form of PSGL-1 may comprise a soluble tandem selectin glycoprotein ligand (TSGL) molecule fused to the heavy chain of an intact checkpoint-modulating antibody molecule. In certain embodiments, each of the PSGL-1 domains of the TSGL molecule comprises amino acids 4 to 16 (EYEYLDYDFLPET) of SEQ ID NO:2. In certain embodiments, the method comprises administering a TSGL molecule or a TSGL fusion protein to a subject in combination with other therapies, including adoptive cell transfer (ACT) therapy such as a cell genetically modified with a chimeric antigen receptor (CAR) for the treatment of tumors or cancer.

[0013] Without being bound to any particular theory of mechanism, it is contemplated that treatment of a subject with a molecule comprising at least one soluble PSGL-1 domain, such as a TSGL molecule or TSGL fusion protein, in combination, simultaneously with, or prior to ACT therapy, may prevent or lessen adverse immune effects of ACT, such as cytokine release syndrome (CRS), and/or other adverse effects of elevated cytokine levels. In certain embodiments, the molecule comprising at least one soluble PSGL-1 domain, such as PSGL-Abs or TSGL-Abs, and/or ACT treatment may be administered in multiple doses or cycles. In certain embodiments, the subject may be treated with one or more additional medicaments intended to prevent or lessen adverse immune effects of ACT. Such additional medicaments may inhibit the release of one or more cytokines, or block the binding of a cytokine to its receptor. Such additional medicaments include, for example, an antagonist of a cytokine or cytokine receptor, such as tocilizumab, a monoclonal antibody that binds to IL-6 receptor and blocks the binding of IL-6 to its receptor.

[0014] It is further contemplated by the inventors herein, that the PSGL domains of a molecule comprising at least one soluble PSGL-1 domain, such as PSGL-Abs or TSGL-Abs may be useful as a medicament for certain methods of treatment that are disclosed herein, for example, the molecule comprising at least one soluble PSGL-1 domain, such as a PSGL-Ab or TSGL-Ab, may be used in combination with an adoptive cell transfer (ACT) therapy, or other pro-immune therapy or agent; in methods of promoting immune activity, such as T-cell recruitment, activation or infiltration; and/or in methods of treating, preventing, lessening the incidence of, or lessening the severity of adverse effects of such ACT therapy or pro-immune therapy or agent, including treating or preventing cytokine release syndrome (CRS). Without being bound to any particular theory of mechanism, the inventor herein contemplates that the efficacy such molecules can be adjusted if one or more PSGL anionic domains of such molecules is missing one or more sLe x structures or epitopes, and therefore the molecule comprising at least one soluble PSGL-1 domain, such as a PSGL-Ab or TSGL-Ab, may be partially lacking, or completely devoid of sLe x epitopes.

[0015] In other embodiments, treatment with a soluble form of human PSGL-1, such as a PSGL- checkpoint Ab or TSGL-checkpoint Ab, is administered to a subject in combination with, simultaneously, or prior to, a therapy or agent intended to activate or promote immune activity, such as T-cell recruitment, activation or infiltration. For example, Gao et al. (2017) Nat. Med. 23:551-555, report that anti-CTLA-4 therapy may induce PD-L1 and VISTA molecules on subsets of macrophages within tumors. In the case of prostate tumors, increased VISTA expression appears to represent a compensatory inhibitor pathway that contributes to the tumor resistance of anti-CTLA-4 therapy. In such settings, a combination treatment with TSGL molecules may reduce the inhibition caused by VISTA. Pro-immune therapies include, for example, cancer vaccines, oncolytic viruses, gene therapy, and cellular therapies such as hematopoietic stem cell (HSC) and bone marrow transplantation. Pro-immune agents include, for example, checkpoint inhibitors, and cytokines. Other pro-immune therapies and agents useful in the present invention include: blinatumomab, a CD19/CD3-bispecific single chain antibody that is designed to link CD 19+ B cells with CD3+ T-cells, and induce a cytotoxic T-cell response against CD 19+ B leukemia/lymphoma. Teachey et al. (2013) Blood 121:5154-5157. Another bispecific T-cell recruiting antibody is solitomab, a fusion protein consisting of two single -chain variable fragments (scFvs) binding to T cells via the CD3 receptor and to the EpCAM tumor associated antigen. (Amarni et al. (2009) ./. Immunotherapy 32:452-464.) A third bispecific, trifunctional antibody is catumaxomab (Removab ® , Fresenius Biotech GmbFI), a trifunctional antibody that binds to CD3/EpCAM and to Fc receptors via its intact Fc region. Seimetz (2011) J. Cancer 2:309-316.

[0016] The present invention further includes methods of treating or preventing cytokine release syndrome (CRS), comprising administering to a subject in need thereof a soluble form of human PSGL-1, such as a PSGL-Ab or TSGL-Ab, in an amount effective to reduce the elevation of serum concentrations for endothelial biomarkers soluble ICAM-1 (sICAM), VWF or Ang-2, wherein one or more of the soluble form of PSGL-1 domains in such PSGL-Ab or TSGL-Ab comprises amino acids 4 to 16 of SEQ ID NO:2 and wherein one or more of the anionic domains in PSGL-Abs or TSGL-Abs either does or does not contain the sialyl Lewis X (sLe x ) tetrasaccharide. Elevation of such endothelial biomarkers have been reported for COVID-19 patients (Vassiliou et al. (2021) Cells, 10:el86). Moreover, because COVID-19 is reported as a microvascular disease (Lowenstein and Solomon (2020), Circulation; 142:1609-1611), treatments with PSGL-Ab or TSGL-Ab modified with (sLe x ) should help decrease thrombotic events. In the case of several respiratory disease settings, the anionic domains of PSGL or TSGL may be fused to an antibody that binds to extracellular nicotinamide phosphoribosyltransferase (eNAMPT) such as ALT-100 (Aqualung Therapeutics) that serves to prevent the eNAMPT -mediated activation of the toll-like receptor 4 (TLR4) pathway (Quijada et al., (2020) Eur Respir J. 57 :xx (In press)).

[0017] In certain embodiments, the method comprises administering the PSGL-Ab or TSGL-Ab and ACT or other pro-immune therapy or agent in combination, simultaneously or nearly simultaneously.

In other embodiments, the PSGL-Ab or TSGL-Ab, may be administered after administration of ACT therapy, or other pro-immune therapy or agent. In other embodiments, PSGL-Abs or TSGL-Abs may be administered prior to administration of ACT therapy, or other pro -immune therapy or agent. In other aspects, the present invention comprises a method in which a PSGL-Ab or TSGL-Ab, is administered to a subject prior to, or in combination with, an adoptive cell transfer (ACT) therapy, or other pro-immune therapy or agent, in order to treat, prevent, lessen the incidence of, or lessen the severity of adverse effects of such therapy or agent. The adverse effects may comprise cytokine release syndrome (CRS). The adverse effects may further comprise neurological effects, including seizures, headaches, delirium and edema. In certain embodiments, the PSGL-Ab or TSGL-Ab may be administered at least 20 minutes, 30 minutes, 40 minutes, 50 minutes, 60 minutes, 70 minutes, 80 minutes, 90 minutes, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours 7 horns, 8 hours, 10 horns, 12 hours, 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, or 24 hours prior to use of an ACT. In certain other embodiments, the soluble form of human PSGL-Abs or TSGL-Abs, may be administered at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days or 7 days prior to use of an ACT, or other pro-immune therapy or agent. In certain embodiments, multiple doses of the PSGL-Abs or TSGL-Abs may be administered prior to beginning ACT treatment. In other embodiments, the PSGL-Ab or TSGL-Ab and/or ACT treatment, or other pro-immune therapy or agent may be administered in multiple doses or cycles.

[0018] Additional examples of PSGL-Abs or TSGL-Abs of the present invention include that are not a checkpoint inhibitor antibodies are fusions to therapeutic antibodies for the treatment of cancer. Such antibodies useful in the invention include, for example, anti-HER2 antibodies such as trastuzumab (Herceptin ® ) for treating breast cancers or fusions to anti-CD20 antibodies such as rituximab (Rituxan ® ) for treating B cell lymphomas. Antibodies that bind to chemokine receptor 8 (anti-CCR8 antibodies) are another example of a type of anti-tumor antibody that can be fused with PSGL or TSGL anionic domains. CCR8 is selectively expressed on activated intratumoral regulatory T cells (Tregs) and exhibit anti-tumor activity (US 10,550,191 B2). Examples of therapeutic anti-CCR8 antibodies include but are not limited to JTX-1811 (Jounce Therapeutics) and FPA157 (Five Prime Therapeutics).

[0019] In certain embodiments, at least one additional active agent may be administered in combination with the ACT therapy and PSGL-Ab or TSGL-Ab. In certain embodiments, the additional active agent is selected from the group consisting of immune checkpoint modulators. Immune checkpoint modulators useful in the present invention include both immune checkpoint inhibitors and immune checkpoint stimulators. Immune checkpoint inhibitors useful in the present invention include PD-1 antagonists, PD-L1 antagonists, and CTLA-4 antagonists. In some embodiments, the immune checkpoint inhibitor is selected from the group consisting of nivolumab (Opdivo ® , Bristol-Myers Squibb), ipilimumab (Yervoy ® , Bristol-Myers Squibb); pembrolizumab (Keytmda ® , Merck); cemplimab (Libtayo ® , Regeneron); spartalizumab (PDR001, Novartis) and JTX-4014 (Jounce Therapeutics). Other immune checkpoint inhibitors that are in development and may be used in the present invention include anti-LAG3/CD223 (MK-4280 Merck), anti-HAVCR2/TfM-3 (TSR-022), anti-TREM2 (PY314 Pionyr), anti-TREMl (PY159 Pionyr), Anti-VSIG4 (VTX-1218, Verseau Therapeutics), atezolizumab (Tecentriq ® , Genentech/Roche), also known as MPDL3280A, a fully humanized engineered antibody of IgGl isotype against PD-L1; durvalumab (Imfinzi ® , Astra-Zeneca), also known as MED 14736; tremelimumab (AstraZeneca), also known as CP-675,206, which is a fully human monoclonal antibody against CTLA-4; pidilizumab (CureTech), also known as CT-011, an antibody that binds to PD-1; avelumab (Bavencio ® , Pfizer/Merck KGaA), also known as MSB0010718C), a fully human IgGl anti-PD-Ll antibody; and PDR001 (Novartis), an inhibitory antibody that binds to PD-1. Immune checkpoint stimulators useful in the present invention include agonists of the following molecules: CD27, CD28, CD40, 0X40 (CD 134), GITR, ICOS and CD 137 (4-1BB) ; for example, agonistic antibodies to ICOS (See Michaelson et al. (2016), Cancer Research Abstract #573, regarding Jounce antibody vopratelimab JTX-2011), agonistic antibodies to CD137 (e.g., Urelumab/ BMS-663513 /anti-4-lBB antibody), and agonistic antibodies to 0X40, such as MED 10562, MEDI 6469 and MED 16383 (AstraZeneca), which are 0X40 agonists and which can act as checkpoint stimulator molecules. The term “antigen” is defined as the structure that an antibody binds to via its antigen binding region. Each of these antibodies function by binding a target antigen on immune checkpoint modulating proteins.

[0020] In other embodiments, the at least one additional active agent is selected from a protein kinase inhibitor or a VEGF-R antagonist. Protein kinase inhibitors or VEGF-R antagonists useful in the present invention include axitinib (Inlyta ® , Pfizer Inc., NY, USA), sorafenib (Nexavar ® Bayer AG and Onyx); sunitinib (Sutenf ® , Pfizer, New York, US); pazopanib (Votrient ® , GlaxoSmithKline, Research Triangle Park, US); cabozanitib (Cometriq ® , Exelexis, US); regorafenib (Stivarga ® , Bayer); lenvatinib (Lenvima ® , Eisai); bevacizumab (Avastin ® , Genentech, Inc. of South San Francisco, Calif.,), an anti-VEGF monoclonal antibody; and aflibercept, also known as VEGF Trap (Zaltrap ® ; Regeneron/Sanofi). Other kinase inhibitors/ VEGF-R antagonists that are in development and may be used in the present invention include tivozanib (Aveo Pharmaceuticals, Cambridge, MA); vataianib (Bayer, Novartis, Basel,

Switzerland); lueitanib (Clovis Oncology); dovitinib (Novartis); CEP-11981 (Cepha!on, US); linifanib (Abbott Laboratories, Abbott Park, US); PTC299 (PTC Therapeutics, South Plainfield, US); CP-547,632 (Pfizer); foretinib (Exelexis, GlaxoSmithKline); and motesanib (Amgen, Takeda). [0021] The present invention also includes methods of use of PSGL-Abs or TSGL-Abs for the treatment of viruses, including viruses that cause persistent and chronic viral infections. For example, the methods of the present invention are suitable for the treatment of HIV, hepatitis (A, B and C), Herpesviruses, lymphocytic choriomeningitis viruses (LCMV), human T-lymphotrophic vims (HTLV), respiratory syncytial vims (RSV), mumps vims, measles vims, rotavimses, influenza vimses, enterovims 71and Flaviviruses, such as ZIKA, dengue, West Nile, Yellow Fever, and Japanese encephalitis vimses. PSGL-1 or TSGL antibody fusion molecules of the present invention are also contemplated with anti- SARS-Cov2 vims antibodies such as ADG-2 (Rappazzo etal. (2021) Science 371:823-829), casirivimab and imdevimab (Regeneron) or etesevimab/LY-CoV016 and bamlanivimab/LYCoV555 (Eli Lilly). The preliminary guidance for the use of these anti-SARS-Cov2 antibodies in children and adolescents has been reported (Wolf et al. (2021) J Ped Infectious Dis Soc 202LXX).

[0022] The present invention also includes methods of use of PSGL-Abs or TSGL-Abs for the treatment of pathogenic bacterial infections, such as syphilis, chlamydia, rickettsial bacteria, mycobacteria, staphylococci, streptococci, pneumonococci, meningococci and conococci, klebsiella, proteus, serratia, pseudomonas, legionella, diphtheria, salmonella, bacilli, cholera, tetanus, botulism, anthrax, plague, leptospirosis, and Lymes disease bacteria, and diseases caused by pathogenic fungi or parasites, such as: Candida (albicans, kmsei, glabrata, tropicalis, etc.), Cryptococcus neoformans, Aspergillus (fumigatus, niger, etc.), Genus Mucorales (mucor, absidia, rhizophus), Sporothrix schenkii, Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis and Histoplasma capsulatum Entamoeba histolytica, Balantidium coli, Naegleriafowleri, Acanthamoeba sp., Giardia lambia, Cryptosporidium sp., Pneumocystis carinii, Plasmodium vivax, Babesia microti, Trypanosoma brucei, Trypanosoma cruzi, Leishmania donovani, Toxoplasma gondi, and Nippostrongylus brasiliensis, Escheria coli, Clostridium dificil, Mycobacterium tuberculosis and multi-dmg resistant organisms, including vimses and bacteria.

[0023] The present invention also includes methods of use of PSGL-Abs or TSGL-Abs for the enhancement of activity of vaccines, for example, vaccines against viral infections, or other pathogenic infections. In certain embodiments, the PSGL-Abs or TSGL-Abs may be administered in a single formulation with other elements of the vaccine. In other embodiments, the PSGL-Abs or TSGL-Abs may be administered as a separate formulation, which may be co-administered with the vaccine. In other embodiments, the PSGL-Abs or TSGL-Abs may be administered as a separate formulation prior to vaccination.

BRIEF DESCRIPTION OF THE FIGURES [0024] FIGURE 1 illustrates the protein structure of various monomeric sulfated PSGL-1 gly copeptide domains within the present invention. In each of the monomeric sulfated PSGL-1 glycopeptide domains, at least one tyrosine residue is sulfated, and the threonine (alternatively a serine), is the site of an O-linked glycan bearing a sialyl Lewis x (sLe x ) epitope (illustrated by the letters (T/S). Figure la illustrates the sequence of the monomeric sulfated PSGL-1 glycopeptide domain [PSGL1-19] with tyrosine sulfation designated by asterisks, wherein the designation “[PSGL1-19]” means that the domain comprises amino acids 1 through 19 that includes the principal binding site for P and L-selectin, chemokines and VISTA found within human PSGL-1, illustrated at SEQ ID NO: 2. Figure lb illustrates the sequence of the monomeric sulfated PSGL-1 domain [PSGL4-19], which comprises amino acids 4 through 19 of SEQ ID NO:2. Figure lc illustrates the sequence of the monomeric sulfated PSGL-1 glycopeptide domain [PSGL4-19[Y5F]], which comprises amino acids 4 through 19 of SEQ ID NO:2, in which the tyrosine at amino acid residue 5 of the principal binding site for P and L-selectin found within human PSGL-1 [i.e., position 5 of SEQ ID NO: 2] has been converted to a phenylalanine. Figure Id illustrates the sequence of the monomeric sulfated PSGL-1 glycopeptide domain [PSGL9-19], which comprises amino acids 9 through 19 of SEQ ID NO: 2. Figure le illustrates the sequence of the monomeric sulfated PSGL-1 domain [PSGL4-15], which comprises amino acids 4 through 15 of SEQ ID NO:2. and lacks an O-linked glycan attachment site.

[0025] FIGURE 2 illustrates the protein structure of the tandem configuration of sulfated PSGL- 1 glycopeptide domains within the present invention [TSGLs]. Figure 2a illustrates the structure of TSGL [PSGL1-19:PSGL9-19], wherein the designation “[PSGL1-19:PSGL9-19]” means that the TSGL comprises a first soluble PSGL-1 domain that comprises amino acids 1 through 19 of the principal binding site for P and L-selectin found within human PSGL-1; fused to a second soluble PSGL-1 domain that comprises amino acids 9 through 19 of the principal binding site for P and L-selectin found within human PSGL-1. Figure 2b illustrates the structure of the TSGL [PSGL2-19:PSGL6-19]. Figure 2c illustrates the structure of the TSGL [PSGL2-19:(PSGL9-19) N :PSGL6-19] within the present invention that contains more than two sulfated PSGL-1 glycopeptide domains, wherein N is an integer one or greater and represents the number of sulfated PSGL9-19 glycopeptide domains between [PSGL2-19] and [PSGL6-19] [0026] FIGURE 3 illustrates the protein structure and configuration of TSGL fusion proteins of the present invention with optional linker sequences between the two monomeric sulfated PSGL-1 glycopeptide domains of the TSGL and between the TSGL and the heavy chain or light chain of a checkpoint modulating antibody. Figure 3a illustrates the amino acid sequence and structure of the TSGL [PSGLl-19:linker:PSGL4-16]. Figure 3b illustrates the structure of the monomeric TSGL fusion protein [PSGLl-19:linker:PSGL4-16:linker:antibody light chain]. The fusion antibody may comprise, for example, an anti-PD-1 antibody. Figure 3c illustrates the structure of the monomeric TSGL fusion protein [PSGLl-19:linker:PSGL4-16:linker:antibody heavy chain]. Figure 3d illustrates the structure of the monomeric TSGL fusion protein [antibody heavy chain:linker:PSGL4-16:linker:PSGL4-16]. Figure 3e illustrates the structure of the monomeric TSGL fusion protein [PSGLl-19:linker:antibody heavy chain:linker:PSGL4-15 lacking the O-linked glycan site]

[0027] FIGURE 4 illustrates the structure and mechanism of binding exhibited by a PSGL-1 anionic domain fusion to checkpoint-modulating antibody of the present invention. In Figure 4, each of the sulfated PSGL-1 anionic domains presents a binding site for chemokines at various pH ranges 6.0-7.5 and VISTA only under acidic conditions such as pH 6.3 or lower. [0028] FIGURE 5 illustrates the structure and mechanism of binding exhibited by a PSGL-1 anionic domain modified with sLe x fused to checkpoint-modulating antibody of the present invention. In Figure 5, each of the sulfated PSGL-1 anionic domains presents a binding site for VISTA only under acidic conditions such as pH 6.0. In addition, each monomeric sulfated PSGL-1 domain contains sLe x , which adds the ability to bind to selectins.

[0029] FIGURE 6 illustrates the structure and mechanism of binding to VISTA exhibited by a TSGL anionic domain fused to a checkpoint-modulating antagonistic antibody of the present invention. In Figure 6, each of the sulfated TSGL anionic domains presents a binding site for VISTA only under acidic conditions such as pH 6.0.

[0030] FIGURE 7 illustrates the structure and mechanism of binding to chemokines or VISTA exhibited by a TSGL anionic domain fused to checkpoint-modulating agonistic antibody of the present invention. In Figure 7, each of the sulfated TSGL anionic domain presents a binding site for chemokines at various pH ranges 6.0-7.5 and VISTA only under acidic conditions such as pH 6.0.

[0031] FIGURE 8 illustrates the structure and mechanism of binding to chemokines or VISTA exhibited by a TSGL anionic domain modified with sLe x and fused to checkpoint-modulating antibody of the present invention. In Figure 8, each monomeric sulfated TSGL glycopeptide domain contains sLe x , which adds the ability to bind to selectins. In addition, each of the sulfated TSGL anionic domain presents a binding site for chemokines at various pH ranges 6.0-7.5 and VISTA only under acidic conditions such as pH 6.3 or lower.

[0032] FIGURE 9 illustrates the structure and mechanism of binding to chemokines or VISTA exhibited by a TSGL anionic domain modified with sLe x fused at the C-terminus to checkpoint-modulating agonistic antibody of the present invention. In Figure 9, each of the sulfated TSGL anionic domain contains sLe x which adds the ability to bind to selectins. In addition, each of the sulfated TSGL anionic domain presents a binding for chemokines at various pH ranges 6.0-7.5 and VISTA only under acidic conditions such as pH 6.3 or lower.

[0033] FIGURE 10 illustrates the structure and mechanism of binding to chemokines or VISTA exhibited by a TSGL anionic domain fused at the C-terminus to checkpoint-modulating antibody and lacking sLe x modification of the present invention. In Figure 10, each of the sulfated TSGL anionic domain presents a binding site for chemokines at various pH ranges 6.0-7.5 and VISTA only under acidic conditions such as pH 6.3 or lower.

[0034] FIGURE 11 illustrates the structure and mechanism of binding to chemokines or VISTA exhibited by a PSGL anionic domain modified with sLe x fused both the N-terminus and C-terminus to checkpoint-modulating anti PD-1 antibody of the present invention. In Figure 11, only the C-terminal PSGL anionic domain contains sLe x which enables binding to selectins. The N-terminal PSGL fusion lacks threonine 16 and an O-linked glycan. Each of the sulfated PSGL anionic domain presents a binding site for chemokines at various pH ranges 6.0-7.5 and VISTA only under acidic conditions such as pH 6.3 or lower. [0035] FIGURE 12 illustrates the structure and mechanism of binding to chemokines or VISTA exhibited by a PSGL anionic domain lacking sLe x and fused at the N-terminus of the heavy chain of an anti-CD20 antibody of the present invention. In Figure 12, each of the each of the sulfated PSGL anionic domains presents a binding site for chemokines at various pH ranges 6.0-7.5 and VISTA only under acidic conditions such as pH 6.3 or lower.

[0036] FIGURE 13 shows the binding kinetics of TSGL-Ig fusion proteins to the human chemokine CCL21. The TSGL-Ig fusion protein (sample PP7100) is modified with both sulfated tyrosines and sLe x . The TSGL-Ig fusion protein (sample PP7101) is modified with sulfated tyrosines but lacks sLe x modifications. The KD values are 43nm and 36nM respectively.

[0037] FIGURE 14 shows a syngeneic tumor model of MC-38 growing inbalbCc mice after treatment with vehicle control, anti-CD 137 antibody as monotherapy, TSGL-Ig(+) modified with sLex as monotherapy, combination of anti-CD 137 mAh TSGL-Ig(+) modified with sLex, anti-CD 137 mAh TSGL- Ig(-) withour sLex modification. The combined dosing of either TSGL-Ig proteins does not impair the tumor killing of anti-CD 137 mAh treatment.

[0038] FIGURE 15 illustrates the structure and mechanism of binding to selectins, chemokines or VISTA exhibited by a PSGL anionic domain modified with sLe x fused at the N-terminus to the heavy chain of an antibody in a fusion molecule of the present invention. In Figure 15, the N-terminal PSGL anionic domain contains sLe x which enables binding to selectins, as well as presenting a binding site for chemokines at various pH ranges 6.0-7.5 and VISTA only under acidic conditions such as pH 6.3 or lower. When the heavy chain and light chain form a dimeric antibody, the dimeric antibody retains its ability to bind to its antigen, as well as presenting additional binding to selectins, chemokines or VISTA in accordance with the invention.

BRIEF DESCRIPTION OF THE SEQUENCES

[0039] SEQ ID NO: 1 is the amino acid sequence of human PSGL- 1.

[0040] SEQ ID NO: 2 is the mature N-terminal 19 amino acids of human PSGL-1 that contains the anionic domain with acidic residues and includes three tyrosines that can be post-translationally modified to enable the binding of chemokines and VISTA, as well as a threonine at position 16 that can be post-translationally modified with an O-linked glycan to enable the binding to P-selectin, E-selectin, L- selectin.

[0041] SEQ ID NO: 3 is a nucleotide sequence encoding the mature N-terminal 19 amino acids of human PSGL-1 including the anionic domain.

[0042] SEQ ID NO: 4 is an example of the amino acid sequence of the mature heavy chain of an anti-PD-1 therapeutic antibody (pembrolizumab) that is illustrative of the present invention.

[0043] SEQ ID NO: 5 is the amino acid sequence of the mature heavy chain of an anti-PD-1 therapeutic antibody (pembrolizumab) fused at its N-terminus with the anionic domain of PSGL-1 that is illustrative of the present invention. [0044] SEQ ID NO: 6 is the amino acid sequence of the mature heavy chain of an anti-PD-1 therapeutic antibody (pembrolizumab) fused at its N-terminus with the anionic domains of TSGL that is illustrative of the present invention.

[0045] SEQ ID NO: 7 is the amino acid sequence of the mature heavy chain of an anti-PD-1 therapeutic antibody (pembrolizumab) fused at its N-terminus with the anionic domains of TSGL that is illustrative of the present invention.

[0046] SEQ ID NO: 8 is the amino acid sequence of the mature heavy chain of an anti-PD-1 therapeutic antibody (pembrolizumab) fused at its C-terminus with the anionic domains of TSGL that is illustrative of the present invention.

[0047] SEQ ID NO: 9is the amino acid sequence of the mature light chain of an anti-PD-1 therapeutic antibody (pembrolizumab) that is illustrative of the present invention.

[0048] SEQ ID NO: 10 is the amino acid sequence of the mature light chain of an anti-PD-1 therapeutic antibody (pembrolizumab) fused at its N-terminus with the anionic domains of TSGL that is illustrative of the present invention.

[0049] SEQ ID NO: 11 is the amino acid sequence of the mature heavy chain of an anti-CD20 therapeutic antibody (rituximab) fused at its N-terminus with the anionic domain of PSGL-1 that is illustrative of the present invention.

[0050] SEQ ID NO: 12 is the amino acid sequence of the mature light chain of an anti-CD20 therapeutic antibody (rituximab) that is illustrative of the present invention.

[0051] SEQ ID NO: 13 is the amino acid sequence of the mature heavy chain of an anti-human PD-1 antibody that cross reacts with mouse PD-1 and fused at its N-terminus with the anionic domain of TSGL that is illustrative of the present invention.

[0052] SEQ ID NO: 14 is the amino acid sequence of the mature light chain of an anti-human PD-1 antibody that cross reacts with mouse PD-1 that is illustrative of the present invention.

[0053] SEQ ID NO: 15 is the amino acid sequence of a glycine-serine linker sequence useful in the present invention. The value of n is generally an integer from 1 to 4.

[0054] SEQ ID NO: 16 is the amino acid sequence of a second glycine-serine linker sequence useful in the present invention. The value of n is generally an integer from 1 to 4.

[0055] SEQ ID NO: 17 is the amino acid sequence of a third glycine-serine linker sequence useful in the present invention. The value of n is generally an integer from 1 to 4.

DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS

[0056] Human PSGL-1 is 412 amino acid protein (SEQ ID NO: 1) including a 17 amino acid N- terminal signal peptide (amino acids 1-17), a 24 amino acid N-terminal propeptide (amino acids 18-41) and a 371 amino acid P-selectin glycoprotein ligand 1 chain (amino acids 42-412). SEQ ID NO: 1:

MPLQLLLLLI LLGPGNSLQL WDTWADEAEK ALGPLLARDR RQATEYEYLD YDFLPETEPP EMLRNSTDTT PLTGPGTPES TTVEPAARRS TGLDAGGAVT ELTTELANMG NLSTDSAAME IQTTQPAATE AQTTQPVPTE AQTTPLAATE AQTTRLTATE AQTTPLAATE AQTTPPAATE AQTTQPTGLE AQTTAPAAME AQTTAPAAME AQTTPPAAME AQTTQTTAME AQTTAPEATE AQTTQPTATE AQTTPLAAME ALSTEPSATE ALSMEPTTKR GLFIPFSVSS VTHKGIPMAA SNLSVNYPVG APDHISVKQC LLAILILALV ATIFFVCTW LAVRLSRKGH MYPVRNYSPT EMVCISSLLP DGGEGPSATA NGGLSKAKSP GLTPEPREDR EGDDLTLHSF LP

[0057] The 19 amino acid segment at the amino terminus of the mature form of PSGL from amino acids 42 to 60 (SEQ ID NO: 2). The segment contains the principal binding sites for certain chemokines, VISTA and P, E and L-selectin.

SEQ ID NO: 2: QATEYEYLDY DFLPETEPP

[0058] PSGL-Abs or TSGL-Abs of the present invention can be made by linking the PSGL or TSGL peptide to the N-terminus and/or C-terminus of the heavy chain and or light chain of the antibody. The linkage between a PSGL and/or TSGL peptide and an antibody heavy and/or light chain may be direct (i.e., without an intervening linking sequence not derived from either protein) or through a linking sequence. In certain embodiments of the invention, the PSGL-Abs or TSGL-Abs are expressed from a recombinant DNA sequence which encodes both the PSGL-1 or TSGL anionic domain and the heavy or light chain of the antibody, joined either directly or via a DNA sequence encoding a linker sequence. Linkage can also be effected at the peptide level through chemically linking the PSGL or TSGL peptide domain to the antibody heavy chain or light chain.

[0059] PSGL-Abs or TSGL-Abs can be expressed and purified from mammalian host cells, such as a Chinese hamster ovary cells (CHO), HEK293 or COS cells. Suitable host cells contain tyrosylprotein sulfotransferase (TPST) enzymes (Moore et al. (2009) Proc Natl Acad Sci, 106: 14741- 14742) capable of modifying key PSGL-1 or TSGL tyrosine residues to form tyrosine S0 4 -sulfate esters. Suitable host cells are also capable of attaching carbohydrate side chains characteristic of functional PSGL- Abs or TSGL-Abs. Such capability may arise by virtue of the presence of a suitable glycosylating enzyme within the host cell, whether naturally occurring, induced by chemical mutagenesis, or through transfection of the host cell with a suitable expression plasmid containing a DNA sequence encoding the glycosylating enzyme. These host cells can be transfected with expression vectors to enable, via posttranslational modification, the generation of the sialyl Lewis x epitope on the N-linked and O-linked glycans of enhanced PSGL polypeptides. In the case of CHO cells, this requires the co-expression of an oc-1,3/1,4 fucosyltranseferase (Kukowska-Latallo et al. (1990) Genes Dev. 4:1288-303) and Core2 b-1,6-N- acetylglucosaminyltransferase enzymes (Kumar et al. (1996) Blood; 88:3872-79). The presence of the sialyl Lewis X epitopes on the N-linked and O-linked glycans of PSGL-Abs or TSGL-Abs enable the binding to selectins. In order to ensure processing of the mature N-terminus, these host cells may also be transfected with expression vectors with cDNA encoding a form of PACE, also known as furin, is disclosed in van den Ouweland et al. (1990) Nucl. Acids Res. 18:664, the full disclosure of which is hereby incorporated herein by reference. Other signal peptides can be utilized to generate a mature N-terminus of the heavy and light chains of PSGL-Abs and TSGL-Abs (see Haryadi et al. (2015) PLoS ONE). PSGL-Abs or TSGL-Abs without the sialyl Lewis x (sLe x ) epitope on its glycans may be produced in host cells such as CHO cells or HEK293 cells that lack appropriate modifying enzymes, such as the oc-1,3/1,4 fucosyltranseferase enzyme.

[0060] The principal binding site contains three tyrosines residues [at amino acids 5, 7 and 10 of SEQ ID NO: 2] for potential sulfation; and one threonine residue [at amino acid residue 16 of SEQ ID NO: 2] for an O-linked glycan bearing a sialyl Lewis x (sLe x ) epitope. Accordingly, in a preferred embodiment, each monomeric sulfated PSGL-1 glycopeptide domain contained within the PSGL-Abs or TSGL-Abs of the present invention may comprise at least amino acids residues 4 to 16 of SEQ ID NO:2 (EYEYLDYDFLPET). In alternative embodiments, the monomeric sulfated PSGL-1 glycopeptide domain may each independently comprise one or more additional amino acids from the N-terminal end [e.g., amino acids 1-16; 2-16; 3-16; 4-16; 5-16; 6-16; 7-16; 8-16; or 9-16]; one or more additional amino acids from the C-terminal end [e.g., amino acids 10-17; 10-18; 10-19]; or one or more amino additional amino acids from both the N-terminal and C-terminal ends of SEQ ID NO: 2: [e.g. amino acids: 1-17; 2-17; 3-17; 4-17; 5-17; 6-17; 7-17; 8-17; and 9-17; 1-18; 2-18; 3-18; 4-18; 5-18; 6-18; 7-18; 8-18; and 9-18; or 1-19; 2-19; 3-19; 4- 19; 5-19; 6-19; 7-19; 8-19; and 9-19] In certain embodiments, the PSGL-Abs or TSGL-Abs of the present invention comprise at least two sulfated PSGL-1 glycopeptide domains. In other embodiments, the TSGL- Abs of the present invention may comprise only amino acids 4-15 without inclusion of the threonine at position 16 that serves as the addition site for an O-linked glycan. This allows for the fusion of two anionic domain along the same polypeptide chain, one devoid of selectin binding activity and one having selectin binding activity (see figure 3e and fig 11). In other embodiments, the PSGL-Abs or TSGL-Abs of the present invention may comprise at least one additional monomeric sulfated PSGL-1 glycopeptide domain, that is, the soluble forms of PSGL-Abs or TSGL-Abs comprises three or more sulfated PSGL-1 glycopeptide domains, with each PSGL-1 glycopeptide domain independently comprising at least amino acids 10 to 16 of SEQ ID NO: 2. PSGL-Abs or TSGL-Abs containing multiple sulfated residues increases the amount of negative (anionic) charge on the protein. PSGL-Abs or TSGL-Abs containing multiple sulfated residues can be purified from proteins having fewer sulfated residues (hyposulfated TSGL proteins) using methods similar to those described in US patent 6,933,370.

[0061] PSGL-Abs or TSGL-Abs of the present invention may be fused to amino acid sequences derived from one or more other proteins (e.g., a fragment of a protein that exhibits a desired activity), forming a PSGL-Ab fusion protein or a TSGL-Ab fusion protein, and the PSGL-1 fusion proteins or TSGL fusion proteins thereby formed constitute another aspect of the present invention. In any fusion protein incorporating a soluble PSGL-1 domain or TSGL protein, the amino acid sequence derived from one or more proteins other than P-selectin ligand can be linked to either the C-terminus or N-terminus of the enhanced PSGL-1 or TSGL sequence, or both. The linkage may be direct (i.e., without an intervening linking sequence not derived from either protein) or through a linking sequence. In certain embodiments of the invention, the PSGL-1 or TSGL fusion antibodies are expressed from a recombinant DNA sequence which encodes both the PSGL-1 or TSGL anionic domain and the heavy or light chain of the antibody, joined either directly or via a DNA sequence encoding a linker sequence.

[0062] Suitable linker sequences are known in the art and include glycine-serine polymers (including, for example, (GS)n, (GSGGS)n [SEQ ID NO:15], (GGGGS)n [SEQ ID NO:16] and (GGGS)n [SEQ ID NO: 17], where n is an integer of at least one, e.g., one, two, three, or four), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers. Other examples include peptide linkers described in US Patent 5,073,627, the disclosure of which is hereby incorporated by reference.

ADOPTIVE CELLULAR THERAPIES (ACT)

[0063] Adoptive cellular therapies, or ACT, have been employed in a number of applications, primarily to increase the efficacy of the immune system to fight off disease such as a wide range of cancers. ACT may involve the enrichment, or expansion, of an immune cell population, such as autologous or allogeneic (donor) T-cells, natural killer (NK) cells, and/or hematopoietic stem cells (HSC), in order to provide larger doses of activated immune cells, such as tumor-infiltrating lymphocytes. See Besser et al. (2010) Clin. Cancer Res. 16:2646-2655. Other types of ACT involve genetic manipulation of immune cells, such as chimeric antigen receptor (CAR) therapy, in which cells are modified by the addition of chimeric antigen receptors in order to confer antigen recognition for tumor-associated antigens. CAR- modified T cells have been used in order to fight various forms of solid tumors, as well as CD 19-expressing hematologic malignancies and other tumors and cancers. See Kalos et al. (2011) Science Translational Medicine 3:95ra73. At least two CAR-T therapies have been approved by the FDA, such as tisagenlecleucel (Kymriah®), and axicabtagene ciloleucel (Yescarta®), both CD-19-adopted CAR therapies used for B-cell acute lymphoblastic leukemia; and large B-cell lymphoma.

[0064] Both tisagenlecleucel and axicabtagene ciloleucel have black box warnings of the significant adverse side effects, primarily cytokine release syndrome or CRS, in which the immune system essentially kicks into overdrive and neurological problems including seizures, headaches, delirium and edema, and pose serious risks, including death. In order to prevent or lessen the risk of such adverse side effects, researchers have employed various approaches, including administration of tocilizumab, an IL-6 receptor antagonistic monoclonal antibody, to help block the binding of the cytokine IL-6 to its receptor.

See Maude et al. (2014) Cancer J. 20:119-122; Bonifant et al. (2016) Molecular Therapy Oncolytics 3:16011.

PSGL-1 and TSGL COMPOSITIONS AND FORMULATIONS:

[0065] In certain embodiments, the composition comprising PSGL-Abs or TSGL-Abs further comprises one or more surfactants. Exemplary surfactants include, but are not limited to, natural emulsifiers (e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and Veegum [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g. stearyl alcohol, cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g. polyoxyethylene sorbitan monolaurate [Tween 20], polyoxyethylene sorbitan [Tween 60], polyoxyethylene sorbitan monooleate [Tween 80], sorbitan monopalmitate [Span 40], sorbitan monostearate [Span 60], sorbitan tristearate [Span 65], glyceryl monooleate, sorbitan monooleate [Span 80]), polyoxyethylene esters (e.g. polyoxyethylene monostearate [Myq 45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and Solutol), sucrose fatty acid esters, polyethylene glycol fatty acid esters (e.g. Cremophor), polyoxyethylene ethers, (e.g. polyoxyethylene lauryl ether [Brij 30]), poly(vinyl- pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, Pluronic F 68, Poloxamer 188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, etc. and/or combinations thereof. In certain embodiments, the surfactant is a Tween surfactant (e.g., Tween 60, Tween 80, etc.).

[0066] In certain embodiments, the composition further comprises one or more preservatives. Exemplary preservatives may include antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and other preservatives.

[0067] In certain embodiments, the one or more preservative comprises an antioxidant. Exemplary antioxidants include, but are not limited to, phosphites, dibutyl phosphite, alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, sodium sulfite, cysteine hydrochloride, thioglycerol, sodium mercaptoacetate, sodium formaldehyde sulfoxylate (SFS), lecithin, and alpha-tocopherol. In certain embodiments, the antioxidant is dibutyl phosphite or sodium bisulfite (NaHS0 3 ).

[0068] In certain embodiments, the one or more preservative comprises a chelating agent. Exemplary chelating agents include, but are not limited to, ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and trisodium edetate.

[0069] In certain embodiments, the one or more preservative comprises an antimicrobial preservative. Exemplary antimicrobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol, and thimerosal. [0070] In certain embodiments, the one or more preservative comprises an antifungal preservative. Exemplary antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and sorbic acid.

[0071] In certain embodiments, the one or more preservative comprises an alcohol preservative. Exemplary alcohol preservatives include, but are not limited to, ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and phenylethyl alcohol.

[0072] In certain embodiments, the one or more preservative comprises an acidic preservative. Exemplary acidic preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta- carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and phytic acid.

[0073] Other preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, Glydant Plus, Phenonip, methylparaben, Germall 115, Germaben II, Neolone, Kathon, and Euxyl.

[0074] In certain embodiments, the composition further comprises one or more diluents. Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc. , and combinations thereof.

[0075] In certain embodiments, the composition further comprises one or more granulating and/or dispersing agents. Exemplary granulating and or dispersing agents include, but are not limited to, potato starch, com starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation-exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked poly(vinyl-pyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcry stalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (Veegum), sodium lauryl sulfate, quaternary ammonium compounds, etc., and combinations thereof.

[0076] In certain embodiments, the composition further comprises one or more binding agents. Exemplary binding agents include, but are not limited to, starch (e.g. cornstarch and starch paste); gelatin; sugars (e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol, etc.); natural and synthetic gums (e.g. acacia, sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate (Veegum), and larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol; inorganic calcium salts; silicic acid; polymethacrylates; waxes; water; alcohol; etc. ; and combinations thereof.

[0077] In certain embodiments, the composition further comprises one or more buffering agents. Exemplary buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, D-gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water, isotonic saline, Ringer’s solution, ethyl alcohol, etc. , and combinations thereof.

[0078] In certain embodiments, the composition further comprises one or more lubricating agents. Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, etc., and combinations thereof.

[0079] In certain embodiments, the composition further comprises one or more solubilizing or suspending agents. Exemplary solubilizing or suspending agents include, but are not limited to, water, organic solvents, oils, and mixtures thereof. Exemplary oils include, but are not limited to, almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, camauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, com, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquana, savoury, sea buckthorn, sesame, shea butter, silicone, soybean, sunflower, tea tree, thistle, tsubaki, vetiver, walnut, and wheat germ oils, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and combinations thereof. In certain embodiments, the oil is mineral oil.

[0080] Protein formulation is a well-known field and the skilled practitioner is readily able to design liquid formulations for administration via oral, injectable, intravenous, intrathecal, intramuscular and other routes, as well as stable lyophilized protein formulations, which may be administered orally, for example via capsule form, as well as other routes. See Carpenter et al. (1997) Pharmaceutical Research, 14:969-975; Manning et al. (2010) Pharmaceutical Research, 27:544-575; and Chang and Hershenson (2002) “Practical Approaches to Protein Formulation Development; in Rational Design of Stable Protein Formulations, Carpenter and Manning (eds), Volume 13 of the series Pharmaceutical Biotechnology (Springer US).

[0081] In some embodiments, the pharmaceutically acceptable excipient is at least 95%, 96%, 97%, 98%, 99%, or 100% pure. In some embodiments, the excipient is approved for use in humans and for veterinary use. In some embodiments, the excipient is approved by United States Food and Drug Administration. In some embodiments, the excipient is pharmaceutical grade. In some embodiments, the excipient meets the standards of the United States Pharmacopoeia (U SP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.

[0082] The formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient or variant (e.g., a glycosylated variant) into association with a carrier and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping and/or packaging the product into a desired single- or multi-dose unit.

[0083] A pharmaceutical composition of the invention may be prepared, packaged, and or sold in bulk, as a single unit dose, and or as a plurality of single unit doses. As used herein, a “unit dose” is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.

[0084] The relative amounts of the active ingredient, the pharmaceutically acceptable carrier, and/or any additional ingredients in a pharmaceutical composition of the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1% and 100% (w/w) of the active ingredient.

[0085] Preferred dosage forms include oral and parenteral dosage forms. Liquid dosage forms for oral and parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredients, the liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, com, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. In certain embodiments for parenteral administration, the conjugates of the invention are mixed with solubilizing agents such as Cremophor, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and combinations thereof. [0086] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.

[0087] The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.

[0088] In order to prolong the effect of a drug, it is often desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.

[0089] Compositions for rectal or vaginal administration are typically suppositories which can be prepared by mixing the conjugates of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.

[0090] Compositions for oral administration are typically liquid or in solid dosage forms. Compositions for oral administration may include protease inhibitors, including organic acids such as citric acid, in order to inhibit pancreatic and brush border proteases. Compositions for oral administration may additionally include absorption enhancers, such as acylcamitine and lauroylcamitine, to facilitate the uptake of the peptide through the lumen of the intestine into the systemic circulation by a paracellular transport mechanism. Compositions for oral administration may additionally include detergents to improve the solubility of the peptides and excipients and to decrease interactions with intestinal mucus. Solid form compositions for oral administration, such as tablets or capsules, may typically comprise an enteric coating which further protects the peptides from stomach proteases and permits passage of the tablet or capsule into the small intestine. The solid form composition may additionally comprise a subcoat such as a non-ionic polymer. Examples of preparation of such orally available formulations are disclosed in US Patent 5,912,014, US Patent 6,086,918 and US Patent 6,673,574. The disclosure of each of these documents is hereby incorporated herein by reference.

[0091] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may comprise buffering agents.

[0092] Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.

[0093] The active ingredients can be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active ingredient may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may comprise buffering agents. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.

[0094] A pharmaceutical composition of the invention may be prepared, packaged, and/or sold in a formulation suitable for pulmonary administration via the buccal cavity . Such a formulation may comprise dry particles which comprise the active ingredient and which have a diameter in the range from about 0.5 to about 7 nanometers or from about 1 to about 6 nanometers. Such compositions are conveniently in the form of dry powders for administration using a device comprising a dry powder reservoir to which a stream of propellant may be directed to disperse the powder and/or using a self-propelling solvent/powder dispensing container such as a device comprising the active ingredient dissolved and/or suspended in a low-boiling propellant in a sealed container. Such powders comprise particles wherein at least 98% of the particles by weight have a diameter greater than 0.5 nanometers and at least 95% of the particles by number have a diameter less than 7 nanometers. Alternatively, at least 95% of the particles by weight have a diameter greater than 1 nanometer and at least 90% of the particles by number have a diameter less than 6 nanometers. Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.

[0095] Low boiling propellants generally include liquid propellants having a boiling point of below 65°F at atmospheric pressure. Generally, the propellant may constitute 50 to 99.9% (w/w) of the composition, and the active ingredient may constitute 0.1 to 20% (w/w) of the composition. The propellant may further comprise additional ingredients such as a liquid non-ionic and/or solid anionic surfactant and/or a solid diluent (which may have a particle size of the same order as particles comprising the active ingredient).

[0096] Pharmaceutical compositions of the invention formulated for pulmonary delivery may provide the active ingredient in the form of droplets of a solution and/or suspension. Such formulations may be prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions and/or suspensions, optionally sterile, comprising the active ingredient, and may conveniently be administered using any nebulization and/or atomization device. Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface active agent, and or a preservative such as methylhydroxybenzoate. The droplets provided by this route of administration may have an average diameter in the range from about 0.1 to about 200 nanometers.

[0097] The formulations described herein as being useful for pulmonary delivery are useful for intranasal delivery of a pharmaceutical composition of the invention. Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 to 500 micrometers. Such a formulation is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nares.

[0098] Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of the active ingredient, and may comprise one or more of the additional ingredients described herein. A pharmaceutical composition of the invention may be prepared, packaged, and/or sold in a formulation suitable for buccal administration. Such formulations may, for example, be in the form of tablets and/or lozenges made using conventional methods, and may, for example, comprise 0.1 to 20% (w/w) active ingredient, the balance comprising an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein. Alternately, formulations suitable for buccal administration may comprise a powder and or an aerosolized and/or atomized solution and/or suspension comprising the active ingredient. Such powdered, aerosolized, and/or aerosolized formulations, when dispersed, may have an average particle and or droplet size in the range from about 0.1 to about 200 nanometers, and may further comprise one or more of the additional ingredients described herein. [0099] A pharmaceutical composition of the invention may be prepared, packaged, and/or sold in a formulation suitable for ophthalmic administration. Such formulations may, for example, be in the form of eye drops including, for example, a 0.1/1.0% (w/w) solution and/or suspension of the active ingredient in an aqueous or oily liquid carrier. Such drops may further comprise buffering agents, salts, and/or one or more other of the additional ingredients described herein. Other opthalmically-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form and/or in a liposomal preparation. Ear drops and/or eye drops are contemplated as being within the scope of this invention.

[0100] General considerations in the formulation and or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21 st ed., Lippincott Williams & Wilkins, 2005.

[0101] The skilled clinician will be able to determine the appropriate dosage amount and number of doses of an agent to be administered to subject, dependent upon both the age and weight of the subject, the underlying condition, and the response of an individual patient to the treatment. In addition, the clinician will be able to determine the appropriate timing for delivery of the agent in a manner effective to treat the subject.

[0102] Preferably, the agent is delivered within 48 horns prior to exposure of the patient to an amount of a thrombosis or thrombocytopenia provoking stimulus effective to induce thrombosis or thrombocytopenia, and more preferably, within 36 horns, and more preferably within 24 hours, and more preferably within 12 hours, and more preferably within 6 hours, 5 hours, 4 hours, 3 hours, 2 hours, or 1 hour prior to exposure of the patient to an amount of thrombosis or thrombocytopenia provoking stimulus effective to induce thrombosis or thrombocytopenia. In one embodiment, the agent is administered as soon as it is recognized (i.e., immediately) by the subject or clinician that the subject has been exposed or is about to be exposed to a thrombosis or thrombocytopenia provoking stimulus, and especially a thrombosis or thrombocytopenia provoking stimulus to which the subject is sensitized. In another embodiment, the agent is administered upon the first sign of development of thrombosis or thrombocytopenia, and preferably, within at least 2 hours of the development of symptoms of thrombosis or thrombocytopenia, and more preferably, within at least 1 hour, and more preferably within at least 30 minutes, and more preferably within at least 10 minutes, and more preferably within at least 5 minutes of development of symptoms of thrombosis or thrombocytopenia. Symptoms of thrombosis or thrombocytopenia and methods for measuring or detecting such symptoms have been described and are well known in the art. Preferably, such administrations are given until signs of reduction of thrombosis or thrombocytopenia appear, and then as needed until the symptoms of thrombosis or thrombocytopenia are gone.

[0103] Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to animals of all sorts. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation.

[0104] Still further encompassed by the invention are kits that comprise one or more inventive complexes and/or compositions. Kits are typically provided in a suitable container (e.g., for example, a glass, foil, plastic, or cardboard package). In certain embodiments, an inventive kit may include one or more pharmaceutical excipients, pharmaceutical additives, therapeutically active agents, and the like, as described herein. In certain embodiments, an inventive kit may include means for proper administration, such as, for example, graduated cups, syringes, needles, cleaning aids, and the like. In certain embodiments, an inventive kit may include instructions for proper administration and/or preparation for proper administration.

Methods of Treatment:

[0105] The methods of the present invention may be useful in treating tumors and cancers. The methods may also help prevent or reduce the occurrence of side effects, such as CRS and other forms of inflammation or destruction of normal tissue. The methods of the present invention may be further useful in preventing undesired inflammation due, for example, to the production of cytokines, such as in cytokine release syndrome (CRS). Thus, the methods of the present invention include treatments of inflammatory disorders, as well as the moderation or prevention of side effects in pro-inflammatory and anti-cancer or anti-tumor treatments.

[0106] The compositions and kits of the present invention may be useful in treating conditions characterized by P-, E- or L-selectin mediated intercellular adhesion. Such conditions include, without limitation, myocardial infarction, bacterial or viral infection, metastatic conditions, inflammatory disorders such as arthritis, gout, uveitis, acute respiratory distress syndrome, asthma, emphysema, delayed type hypersensitivity reaction, systemic lupus erythematosus, thermal injury such as bums or frostbite, autoimmune thyroiditis, experimental allergic encephalomyelitis, multiple sclerosis, multiple organ injury syndrome secondary to trauma, diabetes, Reynaud's syndrome, neutrophilic dermatosis (Sweet's syndrome), inflammatory bowel disease, Grave's disease, glomerulonephritis, gingivitis, periodontitis, hemolytic uremic syndrome, ulcerative colitis, Crohn's disease, necrotizing enterocolitis, granulocyte transfusion associated syndrome, cytokine-induced toxicity, and the like.

[0107] The compositions and kits of the present invention may be used as an antimetastatic agent, for example in the treatment of many types of metastatic cancers, (see Borsig Glycobiology v28, 2018) as well as multiple myeloma. The compositions and kits of the present invention may be used itself as an inhibitor of P-, E- or L-selectin-mediated intercellular adhesion or to design inhibitors of selectin- mediated intercellular adhesion. The present invention encompasses both pharmaceutical compositions and kits of the present invention and therapeutic methods of treatment or use that employ the compositions and kits of the present invention.

[0108] Additional uses of the compositions and kits of the present invention include treatment of ischemia and reperfusion, bacterial sepsis and disseminated intravascular coagulation, adult respiratory distress syndrome and related pulmonary disorders, tumor metastasis, rheumatoid arthritis and atherosclerosis. Reperfusion injury is a major problem in clinical cardiology. Therapeutic agents that reduce leukocyte adherence in ischemic myocardium can significantly enhance the therapeutic efficacy of thrombolytic agents. Thrombolytic therapy with agents such as tissue plasminogen activator or streptokinase can relieve coronary artery obstruction in many patients with severe myocardial ischemia prior to irreversible myocardial cell death. However, many such patients still suffer myocardial neurosis despite restoration of blood flow. This "reperfusion injury" is known to be associated with adherence of leukocytes to vascular endothelium in the ischemic zone, presumably in part because of activation of platelets and endothelium by thrombin and cytokines that makes them adhesive for leukocytes (Romson et al., Circulation 67: 1016-1023, 1983). These adherent leukocytes can migrate through the endothelium and destroy ischemic myocardium just as it is being rescued by restoration of blood flow.

[0109] Bacterial sepsis and disseminated intravascular coagulation often exist concurrently in critically ill patients. They are associated with generation of thrombin, cytokines, and other inflammatory mediators, activation of platelets and endothelium, and adherence of leukocytes and aggregation of platelets throughout the vascular system. Leukocyte-dependent organ damage is an important feature of these conditions.

[0110] Tumor cells from many malignancies (including carcinomas, lymphomas, and sarcomas) can metastasize to distant sites through the vasculature. The mechanisms for adhesion of tumor cells to endothelium and their subsequent migration are not well understood, but may be similar to those of leukocytes in at least some cases. Specifically, certain carcinoma cells have been demonstrated to bind to both E-selectin, as reported by Rice and Bevilacqua. Science 246:1303-1306 (1991), and P-selectin, as reported by Aruffo, et al., Proc. Natl. Acad. Sci. USA 89:2292-2296 (1992). The association of platelets with metastasizing tumor cells has been well described, suggestion a role for platelets in the spread of some cancers. Since P-selectin is expressed on activated platelets, it is believed to be involved in association of platelets with at least some malignant tumors. (Borsig (2018) Glycobiology; 28:648-655). Specific cancers where the methods of the present invention may be helpful include malignant pleural mesothelioma, neuroblastoma, and glioblastoma. Other cancers wherein the methods of the present invention may be useful include renal cell and kidney cancer, pancreatic cancer, lung cancer, liver cancer, bile duct cancer, breast cancer, ovarian cancer, testicular and prostate cancer, head and neck cancer, gastrointestinal and stomach cancer, endometrial cancer, bladder cancer, colon, rectal, colorectal, and anal cancer, thyroid cancer, non-melanoma skin cancer, melanoma, lymphoma and leukemia.

[0111] The compositions, materials and kits of the present invention may also be useful in methods of treating subjects having a tumor or cancer, and include methods using PSGL-Abs or TSGL-Abs in combination with other antitumor and anticancer therapeutic molecules for enhanced antitumor and antitumor therapies, also termed immunotherapies. PSGL-Abs or TSGL-Abs may be combined with other therapeutics known to modulate checkpoint molecules on T cells such as anti-PD-1 antibodies, anti-PD-Ll antibodies; anti-CTLA-4 antibodies, anti-ICOS antibodies, anti-CD 137 antibodies, as well as other therapies and agents developed for such purposes. Such molecules may include, for example, inhibitors of adenosine A2A receptor; B7-H3 (CD276); B7-H4 (VTCN1); BTLA; CTLA-4; IDO; KIR; LAG3; PD-1; PD-L1; PD-L2; TIM-3; TREMM2; and VISTA. In the case of glioblastoma, TSGL molecules may be used in combination with peptidomimetics of thrombospondin- 1 (TSP-1 PM) to inhibit angiogenesis.

[0112] The compositions, materials and kits of the present invention may also be useful in methods of treating subjects having pathogenic infections, whether viral, bacterial, fungal or parasitic in origin, and include methods using antibodies fused with human PSGL-1 anionic domains or TSGL anionic domains in combination with other antiviral, antibacterial, antifungal or anti-pathogenic and therapeutic molecules or treatments for enhanced anti-pathogenic therapies. In such indications, the PSGL-Abs or TSGL-Abs may also be used in conjunction with therapeutics known to modulate checkpoint molecules. See, Velu et al. (2009) Nature, 458:7235; and Ha et al. (2008) J. Experimental Medicine, 205:543-555.

[0113] For the anti-cancer and anti-pathogenic uses of the present invention, the PSGL-Abs or TSGL-Abs may not require sLe x be present. In these cases, the PSGL-Abs or TSGL-Abs may, for example, be made in cells, such as CHO or HEK293, which lack the appropriate glycosylation enzymes, resulting in a PSGL-1 or TSGL fusion antibody that primarily binds via sulfated tyrosine residues within the anionic domain. PSGL-1 or TSGL fusion antibodies made in this manner would be expected to promote a more anti-tumor responce, but would likely not block selectin-mediated events of T cell, and myeloid cells. See Veerman et al. 2012, J. Immunology, 188: 1638-1646; Ley and Kansas 2004, Nature Reviews, 4:1-11. The present inventors theorize that PSGL-1 or TSGL fusion antibodies, whether partially or fully lacking the sLe x epitope, yet retaining sulfated tyrosines, may be especially useful in anti-cancer and anti-pathogenic uses, since they will presumably stimulate T cells in the tumor or pathogen microenvironment, without adversely affecting the normal interaction between PSGL-1 and selectin molecules.

[0114] Additionally, PSGL-1 or TSGL fusion antibodies of the invention may be used in vaccines in order to promote, or enhance, immunity, such as to pathogenic viruses, bacteria, fungi and parasites. The PSGL-1 or TSGL fusion antibodies may be administered, along with other immune- boosting and/or antigenic treatments in order to enhance immune responses to pathogenic infections. See, Velu et al., and Ha et al.

[0115] Platelet-leukocyte interactions are believed to be important in atherosclerosis. Platelets might have a role in recruitment of monocytes into atherosclerotic plaques; the accumulation of monocytes is known to be one of the earliest detectable events during atherogenesis. Rupture of a fully developed plaque may not only lead to platelet deposition and activation and the promotion of thrombus formation, but also the early recruitment of neutrophils to an area of ischemia.

[0116] Another area of potential application is in the treatment of rheumatoid arthritis. In these clinical applications, the glycoprotein ligand, or fragments thereof, can be administered to block selectin- dependent interactions by binding competitively to P-selectin expressed on activated cells. In particular, carbohydrate components of the ligand, which play a key role in recognition by P-selectin, can be administered. PSGL-1 or TSGL domains could be fused with anti-TNF antibodies such as adalimumab (HUMIRA; Abbvie). The antibodies are preferably of human origin or modified to delete those portions most likely to cause an immunogenic reaction. Carbohydrate components of the ligand or the antibodies, in an appropriate pharmaceutical carrier; are preferably administered intravenously where immediate relief is required. The carbohydrate(s) can also be administered intramuscularly, intraperitoneally, subcutaneously, orally, as the carbohydrate, conjugated to a carrier molecule, or in a drug delivery device. The carbohydrate can be modified chemically to increase its in vivo half-life. See US Patent 6,506,382 and US Patent 8,232,252, the complete disclosures of which are hereby incorporated herein by reference.

[0117] Practice of the invention is illustrated in the following, non-limiting examples. The skilled artisan, having read the present specification, will recognize that many modifications, variations and extensions are possible without deviating from the teachings of the present specification. Those modifications, variations and extensions form a part of the invention, and will be recognized to constitute subject matter that may be embodied within the claims.

EXAMPLE 1

[0118] PSGL Fusion proteins. A PSGL-Ab with a novel amino acid sequence can be constructed in accordance with the following procedure:

[0119] A cDNA is constructed encoding a suitable signal peptide and the 19 amino acid sulfated PSGL sequence fused to the heavy chain of an anti-human PD-1 antibody (pembrolizumab) SEQ ID NO:4 and is constructed into a pcDNA3.1 or similar mammalian expression vector to produce the amino acid sequence shown in SEQ ID NO:5. The sequence of the DNA that encodes the amino acids of SEQ ID NO:2 is reported as SEQ ID NO:3. A cDNA encoding a suitable signal peptide and the light chain of an antihuman PD-1 antibody (pembrolizumab) SEQ ID NO:9 is constructed into a pcDNA3.1 or similar mammalian expression vector. The two expression vectors are co -transfected into a CHO host cell engineered to express in stable fashion the enzymes Core2 | >- 1 6-N-accty Igl ucosam i ny I transferase and oc- 1,3/1, 4 fucosyltranseferase in order to modify the O-linked glycan at the Thrl6 residue of the mature PSGL-Ab fusion with the sialyl Lewis x (sLe x ) epitope. The version of this PSGL-Ab without sLe x - modified glycans is produced in CHO host cells that lack the glycan modifying enzymes. Secreted PSGL- Ab is then purified from the conditioned cell culture medium.

EXAMPLE 2

[0120] TSGL Fusion proteins. A TSGL-Ab with a novel amino acid sequence can be constructed in accordance with the following procedure:

[0121] A cDNA is constructed encoding a suitable signal peptide and the 38 amino acid sulfated TSGL sequence fused to the heavy chain of an anti-human PD-1 antibody (pembrolizumab) SEQ ID NO:4 and is constructed into a pcDNA3.1 or similar mammalian expression vector to produce the amino acid sequence shown in SEQ ID NO:7. A cDNA encoding a suitable signal peptide and the light chain of an anti-human PD-1 antibody (pembrolizumab) SEQ ID NO:9 is constructed into a pcDNA3.1 or similar mammalian expression vector. The two expression vectors then are co-transfected into a CHO host cell engineered to express in stable fashion the enzymes Core2 | >- 1 6-N-accty Igl ucosam i ny I transferase and oc- 1,3/1, 4 fucosyltranseferase in order to modify the O-linked glycans at the Thrl6 and Thr37 residues of the mature TSGL-Ab fusion with the sialyl Lewis x (sLe x ) epitope. The version of this TSGL-Ab without sLe x - modified glycans is produced in CHO host cells that lack the two glycan modifying enzymes. Secreted TSGL-Ab is then purified from the conditioned cell culture medium.

[0122] For evaluation in preclinical mouse syngeneic tumor models, the 28 amino acid sulfated TSGL sequence fused to the heavy chain of an anti-human PD-1 antibody (heavy chain sequence 13407 from US patent 10,654,929 B2; SEQ ID NO: 13 herein) and is constructed into apcDNA3.1 or similar mammalian expression vector to produce the amino acid sequence shown in SEQ ID NO: 13. A cDNA encoding a suitable signal peptide and the light chain of an anti-human PD-1 antibody (light chain sequences 13407 from US patent 10,654,929 B2; SEQ ID NO: 14 herein) is constructed into a pcDNA3.1 or similar mammalian expression vector. This intact anti-human PD-1 antibody is capable of cross reacting with the mouse PD-1 homologue protein (see Table 1 from US patent 10,654,929 B2). Similar constructs and methods can be employed to create PSGL-1 or TSGL-fusions to other therapeutic antibodies such as the example given for an anti-CD20 heavy chain fusion (SEQ ID NO: 11) and its co-expression with an anti-CD20 light chain (SEQ ID NO: 12). These fusions can be evaluated in transgenic mice models that express human CD20 as described in US patent 7,402,728 (Chan). The disclosure of each of these publications is hereby incorporated herein by reference for the disclosure cited herein.

EXAMPLE 3

[0123] Binding analysis of anionic domains to chemokines or VISTA. Purified PSGL-1 or TSGL-fusion proteins, including fusions to intact antibody heavy or light chains can be constructed and produced as described above. A PSGL-1 or TSGL-fusion glycoproteins is captured using an immobilized Anti-Hu Fc (AHC biosensors from ForteBio). The recombinant human CCL21 (Sino Biologicals Inc. CAT#10477-HNAB) is then added in solution at various diluted concentrations and the binding kinetics are recorded using an Octet HTX system (ForteBio) and conditions recommended by the manufacturer (see Figure 13). The format can be reversed by immobilizing the recombinant human CCL21 or biotinylated human CCL21 and titrating PSGL-1 or TSGL-1 fusion proteins. Typically, the KD values of these interactions are recorded in the range of 40-400nM. ELISA-based binding assays to recombinant hVISTA- Fc (R&D Systems Catalogue 7126-B7) for either PSGL-1 or TSGL-fusion antibodies are performed essentially as is described in Mehta et al., Sci Rep. 2020.

EXAMPLE 4

[0124] In vivo effect of soluble TSGL anionic domains on T cell tumor killing.

The impact of recombinant fusion proteins, such as TSGL-Ig, on tumor growth in mouse models was examined by injecting approximately 1 X10 6 MC38 cells subcutaneously into the flanks of C57BL6/J mice. Mice were randomized into groups with an approximate 125 mm 3 average tumor size on day 11 and injected intraperitoneally with either 100 pg of anti-CD 137 antibody (cat# MAB9371 R&D Systems), lOOug recombinant TSGL-Ig fusion proteins, or vehicle on day 0. Repeat doses of the recombinant fusion proteins were administered on days 7 and 14. Tumor sizes were measured twice weekly and the experiment was terminated after 21 days. Tumor weights at the end of the experiment are shown in FIG.

14 with P values comparing the various treatment groups to vehicle group 1. Results indicate that the single dose of the effector stimulatory anti-CD 137 antibody significantly reduced tumor growth in mice (p =0.0003) and the additional dosing on days 0,7 and 14 of either TSGL-Ig modified with sLex or unmodified with sLex does not significantly impair the actions of cytotoxic effector cells on tumors. This indicates that the systemic dosing of soluble forms of the PSGL-1 anionic domain does not prevent the homing and trafficking of cytotoxic T cells into tumors.

[0125] Testing of the efficacy of the PSGL-Abs or TSGL-Abs in antitumor and anticancer indications can be accomplished, for example, using methods such as those described in US Patent 9,073,994, for cytotoxicity, effects on tumor growth and proliferation, survival rates, interferon production, PDL-1 and PDL-2 expression, ICAM-1 expression, and other relevant assays. Testing the efficacy of PSGL-Abs or TSGL-Abs to reduce cytokine release syndrome and neurotoxicity can be accomplished using preclinical xenograft models such as described by Sterner RM et al. (2018) Blood: blood-2018-10- 881722.

[0126] All patents, patent applications and scientific literature references cited in the disclosure are hereby incorporated herein by reference for the cited teachings, as if fully set forth in the specification.