NOVEL AGENTS AND USES THEREOF

Information

  • Patent Application
  • 20230092356
  • Publication Number
    20230092356
  • Date Filed
    December 07, 2020
    3 years ago
  • Date Published
    March 23, 2023
    a year ago
Abstract
The present invention provides agents comprising or consisting of a binding moiety with specificity for Signaling Lymphocytic Activating Molecule Family Member 6 (SLAMF6) for use in inducing cell death and/or inhibiting the growth and/or proliferation of pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder, wherein the cells express SLAMF6 and/or modulating their interactions with immune cells that may also express SLAMF6. A related aspect of the invention provides agents comprising or consisting of a binding moiety with specificity for SLAMF6 for use in detecting pathological stem cells, progenitor cells and/or immune cells associated with a neoplastic hematologic disorder, wherein the cells express SLAMF6. Further provided are pharmacological compositions comprising the agents of the invention and methods of using the same.
Description
FIELD OF INVENTION

The present invention relates to agents for use in the treatment and diagnosis of neoplastic hematologic disorders, such as acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), myelodysplastic syndrome (MDS), myeloproliferative disorders (MPD)/myeloproliferative neoplasia (MPN) and chronic myeloid leukemia (CML).


BACKGROUND

Acute myeloid leukemia (AML) is, despite recent advancements in targeted therapies, still associated with an overall dismal prognosis. Newly introduced drugs such as inhibitors of FLT3, IDH1, IDH2, and BCL-2 show promise but their effects on disease outcome are still not fully discerned and most likely will not provide a cure for most genetic subgroups of AML.1-3 Patients carrying a TP53 mutation in their leukemic cells have a poor prognosis and rarely show long term survival despite allogenic stem cell transplantation.4-6 Recent evidence also suggest that patients that relapse with TP53 mutated AML sometimes carry the mutation at low levels already at the time of diagnosis, further emphasizing the need for specific and novel therapeutic approaches to improve the outcome for this subtype of AML.7


Antibody based targeted therapies have long been available in clinical practice, however, reliable and specific targets in AML have proven difficult to identify. Cell surface markers explored for therapeutic purposes in AML using recombinant antibodies include CD123, IL1RAP, CD47, TIM-3, CLL-1 and CD33.8-13 Apart from the drug-conjugated antibody gemtuzumab ozogamicin targeting CD33 that has been approved for low or intermediate risk AML patients, these markers are still under investigation both in pre-clinical and clinical studies.14 However, none has been specifically evaluated in the TP53 mutated setting. A therapeutic target in AML should ideally be expressed on the leukemic stem cells (LSC) (also referred to as leukemia stem cells), as these cells have the capacity to regenerate the leukemia and cause relapse but are not effectively targeted by current treatments. The precise phenotype of LSCs is still an unresolved topic but these cells are generally agreed to be enriched in the CD34+CD38 compartment, making this a preferred population for studying LSCs in AML.15


In other malignancies, different members of the Signaling Lymphocytic Activating Molecule (SLAM) family have been shown to be upregulated.16 SLAMF6, also known as NTB-A, Ly108 or CD352 is known to be expressed on human B, T and NK cells and to play a role in immune modulation and NK cell activation.17 It is also expressed on eosinophils but not basophils or neutrophils.18 SLAMF6 has been shown to be upregulated in myeloma, some lymphomas as well as chronic lymphocytic leukemia, however it has not been studied in the context of AML.19-21


For example, WO 2014/100740 A1 (Seattle Genetics, Inc.) demonstrates the expression of NTB-A on multiple myeloma cell lines and investigates antibodies directed to this target. However, WO 2014/100740 A1 provides no data to support a role for NTB-A in AML LSCs or any primary AML cells and only tests on cell lines that are known to have different surface marker expression compared with primary patient cells. The cell lines used are adapted to in vitro conditions and form homogenous cell populations that fail to recapitulate the hierarchy with a small leukemia stem cell population giving rise to a large population of more mature leukemic cells, which is characteristic of AML and other hematopoietic malignancies. Therefore, the data of WO 2014/100740 A1 fail to demonstrate the expression of SLAMF6 on AML LSCs and its potential as a therapeutic target on this critical cell population.


Furthermore, WO 2008/027739 A2 (Nuvelo, Inc.) contrarily shows all three AML or CML cell lines investigated are NTB-A (SLAMF6) negative (see FIG. 1D). Indeed, the HL-60 AML cell line is used as a negative control for SLAMF6 expression, and SLAMF6 expression is only demonstrated in lymphoid cells. Therefore, the data of WO 2008/027739 A2 does not in any way demonstrate expression of SLAMF6 in myeloid malignancies (including but not limited to AML, MDS, MPN and CML) or on the critical leukemia stem cells.


SLAMF6 is a self-ligand and thus binds to other SLAMF6 molecules, which are expressed on immune cells such as NK, T and B cells, hence its other name NTB-A. It was recently shown that targeting SLAMF6 on exhausted T cells could reactivate them and thus induce killing of leukemia cells (Yigit et al., 2019, Cancer Immunology Research). It has also been shown that SLAMF6 mediates NK cell activity (Wu et al., 2016, Nature Immunology).


SUMMARY OF INVENTION

The invention provides agents for use in the treatment and/or diagnosis of neoplastic hematologic disorders and evolved directly from the discovery by the inventors that stem cells and/or progenitor cells associated with neoplastic hematologic disorders (for example, acute myeloid leukemia (AML)) exhibit an upregulation of Signaling Lymphocytic Activating Molecule Family Member 6 (also known as SLAMF6, NTB-A, Ly108 or CD352) on their surface. In contrast, normal healthy hematopoietic stem cells (as well as progenitor cells) do not express, or show very low expression levels, of SLAMF6. Thus, the invention provides agents for use in the treatment and/or diagnosis of neoplastic hematologic disorders, such as AML, associated with upregulation of SLAMF6 on the surface of stem cells and/or progenitor cells.


Stem cells can be assessed based on the expression of particular markers, indicative of maturity. For example, immature populations can be characterized by being CD34+CD38+ or CD34+CD38low/−. CD34+CD38 or CD34+CD38low as used herein refer to the same potential stem cell population.


In the present study, a flowcytometry based arrayed screening assay was performed of 362 cell surface markers on diagnostic bone marrow samples from AML patients carrying a TP53 mutation and showed that SLAMF6 is specifically upregulated on immature CD3CD19CD34+CD38 cells in TP53 mutated AML but not corresponding cells from normal bone marrow. Antibodies against SLAMF6 are also shown that can target and kill AML cells by antibody dependent cellular cytotoxicity (ADCC). This demonstrates that SLAMF6 is an interesting target for therapies in AML. Until this study, ADCC had not been demonstrated with antibodies against SLAMF6. In view of SLAMF6 being discovered as a novel target of immature CD34+CD38 cells from AML patients and not corresponding healthy cells, it follows that cell death mechanisms of action other than ADCC would be workable when directed to these cells based on SLAMF6 expression. One such mechanism would be modulation of SLAMF6-expressing immune cells (e.g. T, B and NK cells) by interference with SLAMF6 function. In the present study, it was observed that disruption of SLAMF6 expression on AML cells increases T cell-mediated killing of said AML cells. It was also demonstrated that a SLAMF6 antibody activates T cells and promotes T cell-mediated killing of leukemia cells. SLAMF6 expression discriminates between healthy stem cells and those that are pathological, thereby providing a previously unknown therapeutic window for direct targeting of SLAMF6-expressing AML stem cells, as well as attracting immune cells for cell killing (e.g. by ADCC), activating SLAMF6-expressing immune cells (e.g. T, B or NK cells), or a combination thereof.


A first aspect of the invention provides an agent comprising or consisting of a binding moiety with specificity for Signaling Lymphocytic Activating Molecule Family Member 6 (SLAMF6) for use in inducing cell death (either directly or indirectly via triggering of the immune system) and/or inhibiting the growth (i.e. size) and/or proliferation (i.e. number) of pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder, wherein the stem and/or progenitor cells express SLAMF6. Thus, the agent may be for use in inhibiting the growth and/or proliferation of pathological stem cells alone, of progenitor cells alone, or of both pathological stem cells and progenitor cells.


The agent may also be for use in inducing differentiation of pathological stem and/or progenitor cells which express SLAMF6.


A second, related aspect of the invention provides an agent comprising or consisting of a binding moiety with specificity for Signaling Lymphocytic Activating Molecule Family Member 6 (SLAMF6) for use in detecting pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder, wherein the stem cells express SLAMF6. Thus, the agent may be for use in detecting pathological stem cells alone, progenitor cells alone, or both pathological stem cells and progenitor cells.


SLAMF6 may also be an attractive target for identifying subjects susceptible to cancer relapse, and/or in the treatment, prophylaxis or prevention of relapse in subjects. Relapse of cancer may be attributed to a failure of current therapies to target and remove/reduce cancer stem cells, which can often be resistant to known therapies. Cancer stem cells are the only cells with the capacity to regenerate neoplastic hematological disorders, and incomplete eradication of this population can lead to relapse, which is the major cause of death in many such diseases. Thus, having a cancer stem cell-specific marker can be beneficial for detecting and/or preventing cancer relapse (or risk thereof). Therefore, any of the aspects and embodiments described herein may be suitable for a patient subgroup that is at higher risk of cancer relapse. In one embodiment, an agent comprising or consisting of a binding moiety with specificity for Signaling Lymphocytic Activating Molecule Family Member 6 (SLAMF6) is for use in preventing or reducing the risk of relapse of a neoplastic hematologic disorder, for example relapse that develops from pathological stem cells and/or progenitor cells associated with the neoplastic hematologic disorder, wherein the stem and/or progenitor cells express SLAMF6.


By “Signaling Lymphocytic Activating Molecule Family Member 6” and “SLAMF6” we specifically include the human SLAMF6 protein, for example as described in UniProtKB/Swiss-Prot Accession No. Q96DU3. SLAMF6 is also known in the scientific literature as Activating NK Receptor; NK-T-B-Antigen; NTB-A; KALI; Natural Killer-, T- And B-Cell Antigen; NTBA Receptor; CD352 Antigen; SF2000; CD352; KALIb; Ly108; and NTBA.


By “binding moiety” we include all types of chemical entity (for example, oligonucleotides, polynucleotide, polypeptides, peptidomimetics and small compounds/molecules) which are capable of binding to SLAMF6. Advantageously, the binding moiety is capable of binding selectively (i.e. preferentially) to SLAMF6 under physiological conditions. The binding moiety preferably has specificity for human SLAMF6, which may be localised on the surface of a cell (e.g. the pathological stem cell or progenitor cell).


By “pathological stem cells” associated with a neoplastic hematologic disorder we include stem cells which are responsible for the development of a neoplastic hematologic disorder in an individual, i.e. neoplastic stem cells. In particular, the pathological stem cells may be leukemic stem cells (for example, as described in Guo et al., 2008, Nature 453(7194):529-33). Such stem cell may be distinguished from normal hematopoietic stem cells by their expression of the cell surface protein, SLAMF6 (see the examples below).


In one embodiment, the pathological stem cells are CD34+CD38 cells.


In one embodiment the pathological stem cells are CD3CD19CD34+CD38 cells.


By “progenitor cells” associated with a neoplastic hematologic disorder we include cells derived from pathological stem cells which are responsible for the development of a neoplastic hematologic disorder in an individual. In particular, the progenitor cells may be leukemic progenitor cells (for example, as described in Example 1 below). Such progenitor cells may be distinguished from normal hematopoietic progenitor cells by their higher expression of the cell surface protein, SLAMF6 (see Example 1 below). In one embodiment, the pathological progenitor cells are CD34+CD38+ cells.


By “neoplastic hematologic disorder” we specifically include hematologic cancers such as leukemias, as well as leukemia-like diseases such as myeloproliferative disorders (MPD) (also referred to as myeloproliferative neoplasia (MPN)) and myelodysplastic syndromes (MDS).


Thus, in one embodiment of the first aspect of the invention, the neoplastic hematologic disorder is a leukemic disease or disorder, i.e. a cancer of the blood or bone marrow, which may be acute or chronic.


More specifically, the neoplastic hematologic disorder may be selected from the group consisting of chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). In one particularly preferred embodiment, the neoplastic hematologic disorder is acute myeloid leukemia (AML).


In a further embodiment, the neoplastic hematologic disorder is associated with cells comprising a mutation in the TP53 gene. For example, the pathological stem cells and/or progenitor cells may comprise a TP53 mutation, such as CD34+CD38 cells having a TP53 mutation.


TP53 mutations define a distinct subtype in AML according to the World Health Organisation (WHO) classification. Mutations of TP53 can occur throughout the gene and abrogate the function of the p53 protein in multiple ways, for example by amino acid substitution, truncation, deletion or altered splicing. Thus, TP53 mutations may refer to any mutations that alter the amino acid sequence of the p53 protein. Further, p53 function can also be disrupted by other mechanisms than TP53 mutation. For example, suitable patients may have observed or predicted loss of p53 function by other means, including but not limited to full or partial loss of chromosome 17, epigenetic silencing and alterations in p53 signaling pathways.


Thus, by “TP53 mutation” we include any observed or predicted loss or reduction of p53 function.


This may be as a result of either one or more structural mutations (i.e. wherein the amino acid sequence of the P53 protein is altered) and/or by a functional alteration (i.e. wherein the function of the p53 mutation is disrupted by other means). In a functional alteration there is an observed or predicted loss of p53 function by other means, including but not limited to full or partial loss of chromosome 17, epigenetic silencing and alterations in p53 signalling pathways, as outlined above.


Thus, in one embodiment, the neoplastic hematologic disorder is TP53 mutated AML.


In relation to the diagnostic aspects of the invention, it is sufficient that the agent is merely capable of binding to SLAMF6 present on the surface of the pathological stem cells and/or progenitor cells (without having any functional impact upon those cells).


In relation to the therapeutic and prophylactic aspects of the invention, it will be appreciated by persons skilled in the art that binding of the agent to SLAMF6 present on the surface of the pathological stem cells and/or progenitor cells may lead to a modulation (i.e. an increase or decrease) of a biological activity of SLAMF6. Modulation can be an increase or decrease in inhibition or activation of biological activity. For example, modulation can mean an increase in inhibition of a biological activity or a decrease in inhibition of a biological activity. However, such modulatory effects are not essential; for example, the agents of the invention may elicit a therapeutic and prophylactic effect simply by virtue of binding to SLAMF6 on the surface of the pathological stem cells and/or progenitor cells, which in turn may trigger the immune system to induce cell death (e.g. by ADCC).


Accordingly, in some embodiments, the therapeutic and/or prophylactic aspects of the invention may be through use of a SLAMF6 binding agent that induces cell death by ADCC; via action of a conjugated moiety, such as a moiety that is cytotoxic or radioactive, i.e. an antibody drug conjugate (ADC); and/or death receptor ligation (for example, a bispecific antibody with specificity to SLAMF6 and to said death receptor).


Additionally, modulating interactions between leukemic stem cells and immune cells and/or leukemic cells and immune cells could modulate immune activity against the leukemia, which could have strong therapeutic potential.


In one embodiment an agent targeting SLAMF6 could bring leukemia stem cells in close proximity to immunological effector cells, activate these effector cells, and enhance killing of the leukemia stem cells by effector cells.


In one embodiment an agent targeting SLAMF6 could bring leukemic cells in close proximity to immunological effector cells, activate these effector cells, and enhance killing of the leukemic cells by effector cells.


In some embodiments, the therapeutic and/or prophylactic aspects of the invention may be through use of a SLAMF6 binding agent that induces cell death by a T cell mediated mechanism. For example, the SLAMF6 binding agent may recruit T cells to target cells (e.g. the pathological stem cells and/or progenitor cells), activate T cells, and induce T cell-mediated apoptosis in the target cells via mechanisms known in the art (e.g. release of cytolytic granules, release of cytokines that recruit other effector cells, etc). Alternatively, or additionally, the SLAMF6 binding agent may prevent a SLAMF6-mediated response that would otherwise prevent T cell function and/or activation, for example by masking or blocking the interaction between SLAMF6 expressing pathological stem cells and/or progenitor cells and T cells. Similarly, the SLAMF6 binding agent may recruit, activate or otherwise stimulate immune cells, such as NK cells, for increased anti-leukemic effects.


By “biological activity of SLAMF6” we include any interaction or signalling event which involves SLAMF6 on pathological stem cells and/or progenitor cells.


Such inhibition of the biological activity of SLAMF6 by an agent of the invention may be in whole or in part. For example, the agent may inhibit the biological activity of SLAMF6 by at least 10%, preferably at least 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90%, and most preferably by 100% compared to the biological activity of SLAMF6 in pathological stem cells and/or progenitor cells which have not been exposed to the agent. In a preferred embodiment, the agent is capable of inhibiting the biological activity of SLAMF6 by 50% or more compared to the biological activity of SLAMF6 in pathological stem cells and/or progenitor cells which have not been exposed to the agent. The biological activity of SLAMF6 that is inhibited could be, for example, its self-ligand activity (SLAMF6 interacting with other SLAMF6) and/or downstream signalling. Examples of downstream signalling include, but are not limited to, recruitment and/or phosphorylation of mediators such as SAP, Fyn, EAT-2 and SHP-1.2 (reviewed in Yigit et al., 2018, Clinical Immunology).


Likewise, it will be appreciated that inhibition of growth and/or proliferation of the pathological stem cells and/or progenitor cells may be in whole or in part. For example, the agent may inhibit the growth and/or proliferation of the pathological stem cells and/or progenitor cells by at least 10%, preferably at least 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90%, and most preferably by 100% compared to the growth and/or proliferation of the pathological stem cells and/or progenitor cells which have not been exposed to the agent.


Similarly, it will be appreciated that the induction of differentiation of pathological stem cells and/or progenitor cells may be to any extent. For example, the agent may induce differentiation of the pathological stem cells and/or progenitor cells by at least 10%, preferably at least 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90%, and most preferably by 100% compared to the differentiation of the pathological stem cells and/or progenitor cells which have not been exposed to the agent.


Additionally, in one embodiment the agent is capable of modulating the interaction between an immune cell and leukemic stem cells. By immune cell, we include B cells, T cells and/or NK cells. In one embodiment, the immune cell expresses SLAMF6.


Additionally, in one embodiment the agent is capable of modulating the interaction between an immune cell and leukemic cells. By immune cell, we include B cells, T cells and/or NK cells. In one embodiment, the immune cell expresses SLAMF6.


Thus, in one embodiment, the agent is capable of recruiting and/or activating (which includes enhancing an ongoing function) immune cells, such as B cells, T cells and/or NK cells that express SLAMF6.


In a further preferred embodiment, the agent is capable of killing the pathological stem cells and/or progenitor cells. In particular, the agent may be capable of inducing stem cell and/or progenitor cell death by apoptosis or autophagy. For example, the agent may induce apoptosis by antibody-dependent cell-mediated cytotoxicity (ADCC). In certain embodiments, the killing of pathogenic stem cells and/or progenitor cells may be enhanced by the agent modulating the interaction between an immune cell and leukemic stem cells. In one embodiment, the immune cells express SLAMF6.


In a further preferred embodiment, the agent is capable of killing the leukemic cells. In particular, the agent may be capable of inducing leukemic cell death by apoptosis or autophagy. For example, the agent may induce apoptosis by antibody-dependent cell-mediated cytotoxicity (ADCC). In certain embodiments, the killing of leukemic cells may be enhanced by the agent modulating the interaction between an immune cell and leukemic cells. In one embodiment, the immune cells express SLAMF6.


In one embodiment, the killing of pathogenic stem cells and/or progenitor cells may be enhanced by the agent recruiting and/or activating immune cells, such as B cells, T cells and/or NK cells, preferably wherein the immune cells recruited are also SLAMF6 positive.


As indicated above, the agents of the invention may comprise or consist of any suitable chemical entity constituting a binding moiety with specificity for SLAMF6.


Methods for detecting interactions between a test chemical entity and SLAMF6 are well known in the art. For example, ultrafiltration with ion spray mass spectroscopy/HPLC methods or other physical and analytical methods may be used. In addition, Fluorescence Energy Resonance Transfer (FRET) methods may be used, in which binding of two fluorescent labelled entities may be observed by measuring the interaction of the fluorescent labels when in close proximity to each other.


Alternative methods of detecting binding of SLAMF6 to macromolecules, for example DNA, RNA, proteins and phospholipids, include a surface plasmon resonance assay, for example as described in Plant et al., 1995, Analyt Biochem 226(2), 342-348. Such methods may make use of a polypeptide that is labelled, for example with a radioactive or fluorescent label.


A further method of identifying a chemical entity that is capable of binding to SLAMF6 is one where the protein is exposed to the compound and any binding of the compound to the said protein is detected and/or measured. The binding constant for the binding of the compound to the polypeptide may be determined. Suitable methods for detecting and/or measuring (quantifying) the binding of a compound to a polypeptide are well known to those skilled in the art and may be performed, for example, using a method capable of high throughput operation, for example a chip-based method. Technology called VLSIPS™ has enabled the production of extremely small chips that contain hundreds of thousands or more of different molecular probes. These biological chips have probes arranged in arrays, each probe assigned a specific location. Biological chips have been produced in which each location has a scale of, for example, ten microns. The chips can be used to determine whether target molecules interact with any of the probes on the chip. After exposing the array to target molecules under selected test conditions, scanning devices can examine each location in the array and determine whether a target molecule has interacted with the probe at that location.


Another method of identifying compounds with binding affinity for SLAMF6 is the yeast two-hybrid system, where the polypeptides of the invention can be used to “capture” proteins that bind SLAMF6. The yeast two-hybrid system is described in Fields & Song, Nature 340:245-246 (1989).


In one preferred embodiment, the agent comprises or consists of a polypeptide.


For example, the agent may comprise or consist of an antibody or an antigen-binding fragment thereof with binding specificity for SLAMF6, or a variant, fusion or derivative of said antibody or antigen-binding fragment, or a fusion of a said variant or derivative thereof, which retains the binding specificity for SLAMF6.


By “antibody” we include substantially intact antibody molecules, as well as chimaeric antibodies, humanised antibodies, human antibodies (wherein at least one amino acid is mutated relative to the naturally occurring human antibodies), single chain antibodies, bispecific antibodies, antibody heavy chains, antibody light chains, homodimers and heterodimers of antibody heavy and/or light chains, and antigen binding fragments and derivatives of the same.


By “antigen-binding fragment” we mean a functional fragment of an antibody that is capable of binding to SLAMF6.


Preferably, the antigen-binding fragment is selected from the group consisting of Fv fragments (e.g. single chain Fv and disulphide-bonded Fv), Fab-like fragments (e.g. Fab fragments, Fab′ fragments and F(ab)2 fragments), single variable domains (e.g. VH and VL domains) and domain antibodies (dAbs, including single and dual formats [i.e. dAb-linker-dAb]).


The advantages of using antibody fragments, rather than whole antibodies, are several-fold. The smaller size of the fragments may lead to improved pharmacological properties, such as better penetration of solid tissue. Moreover, antigen-binding fragments such as Fab, Fv, ScFv and dAb antibody fragments can be expressed in and secreted from E. coli, thus allowing the production of large amounts of the said fragments.


Also included within the scope of the invention are modified versions of antibodies and antigen-binding fragments thereof, e.g. modified by the covalent attachment of polyethylene glycol or other suitable polymers (see below).


Methods of generating antibodies and antibody fragments are well known in the art. For example, antibodies may be generated via any one of several methods which employ induction of in vivo production of antibody molecules, screening of immunoglobulin libraries (Orlandi et al, 1989. Proc. Natl. Acad. Sci. U.S.A. 86:3833-3837; Winter et al., 1991, Nature 349:293-299) or generation of monoclonal antibody molecules by cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the Epstein-Barr virus (EBV)-hybridoma technique (Kohler et al., 1975. Nature 256:4950497; Kozbor et al., 1985. J. Immunol. Methods 81:31-42; Cote et al., 1983. Proc. Natl. Acad. Sci. USA 80:2026-2030; Cole et al., 1984. Mol. Cell. Biol. 62:109-120).


Suitable monoclonal antibodies to selected antigens may be prepared by known techniques, for example those disclosed in “Monoclonal Antibodies: A manual of techniques”, H Zola (CRC Press, 1988) and in “Monoclonal Hybridoma Antibodies: Techniques and Applications”, J G R Hurrell (CRC Press, 1982).


Likewise, antibody fragments can be obtained using methods well known in the art (see, for example, Harlow & Lane, 1988, “Antibodies: A Laboratory Manual”, Cold Spring Harbor Laboratory, New York). For example, antibody fragments according to the present invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli or mammalian cells (e.g. Chinese hamster ovary cell culture or other protein expression systems) of DNA encoding the fragment. Alternatively, antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods.


It will be appreciated by persons skilled in the art that for human therapy or diagnostics, human or humanised antibodies are preferably used. Humanised forms of non-human (e.g. murine) antibodies are genetically engineered chimaeric antibodies or antibody fragments having preferably minimal-portions derived from non-human antibodies. Humanised antibodies include antibodies in which complementary determining regions of a human antibody (recipient antibody) are replaced by residues from a complementary determining region of a non-human species (donor antibody) such as mouse, rat of rabbit having the desired functionality. In some instances, Fv framework residues of the human antibody are replaced by corresponding non-human residues. Humanised antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported complementarity determining region or framework sequences. In general, the humanised antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the complementarity determining regions correspond to those of a non-human antibody and all, or substantially all, of the framework regions correspond to those of a relevant human consensus sequence. Humanised antibodies optimally also include at least a portion of an antibody constant region, such as an Fc region, typically derived from a human antibody (see, for example, Jones et al., 1986. Nature 321:522-525; Riechmann et al., 1988, Nature 332:323-329; Presta, 1992, Curr. Op. Struct. Biol. 2:593-596).


Methods for humanising non-human antibodies are well known in the art. Generally, the humanised antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues, often referred to as imported residues, are typically taken from an imported variable domain. Humanisation can be essentially performed as described (see, for example, Jones et al., 1986, Nature 321:522-525; Reichmann et al., 1988. Nature 332:323-327; Verhoeyen et al., 1988, Science 239:1534-15361; U.S. Pat. No. 4,816,567) by substituting human complementarity determining regions with corresponding rodent complementarity determining regions. Accordingly, such humanised antibodies are chimaeric antibodies, wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanised antibodies may be typically human antibodies in which some complementarity determining region residues and possibly some framework residues are substituted by residues from analogous sites in rodent antibodies.


Human antibodies can also be identified using various techniques known in the art, including phage display libraries (see, for example, Hoogenboom & Winter, 1991, J. Mol. Biol. 227:381; Marks et al., 1991, J. Mol. Biol. 222:581; Cole et al., 1985, In: Monoclonal antibodies and Cancer Therapy, Alan R. Liss, pp. 77; Boerner et al., 1991. J. Immunol. 147:86-95).


Once suitable antibodies are obtained, they may be tested for activity, for example by ELISA.


In an alternative embodiment of the first aspect of the invention, the agent comprises or consists of a non-immunoglobulin binding moiety, for example as described in Skerra, Curr Opin Biotechnol. 2007 August; 18(4):295-304.


In a further alternative embodiment, the agent comprises or consists of an aptamer. For example, the agent may comprise or consist of a peptide aptamer or a nucleic acid aptamer (see Hoppe-Seyler & Butz, 2000, J Mol Med. 78 (8): 426-30; Bunka D H & Stockley P G, 2006, Nat Rev Microbiol. 4 (8): 588-96 and Drabovich et al., 2006, Anal Chem. 78 (9): 3171-8).


In a still further alternative embodiment, the agent comprises or consists of a small chemical entity (i.e. small molecules). Such entities with SLAMF6 binding properties may be identified by screening commercial libraries of small compounds/molecules (for example, as available from ChemBridge Corporation, San Diego, USA)


In addition to the binding moiety, the agents of the invention may further comprise a moiety for increasing the in vivo half-life of the agent, such as but not limited to polyethylene glycol (PEG), human serum albumin, glycosylation groups, fatty acids and dextran. Such further moieties may be conjugated or otherwise combined with the binding moiety using methods well known in the art.


Likewise, it will be appreciated that the agents of the invention may further comprise a cytotoxic moiety. For example, the cytotoxic moiety may comprise or consist of a radioisotope, such as astatine-211, bismuth-212, bismuth-213, iodine-131, yttrium-90, lutetium-177, samarium-153 and palladium-109. Alternatively, the cytotoxic moiety may comprise or consist of a toxin (such as saporin or calicheamicin). In a further alternative, the cytotoxic moiety may comprise or consist of a chemotherapeutic agent (such as an antimetabolite).


Likewise, it will be appreciated that the agents of the invention may further comprise a detectable moiety. For example, the detectable moiety may comprise or consist of a radioisotope, such as technetium-99m, indium-111, gallium-67, gallium-68, arsenic-72, zirconium-89, iodine-12 or thallium-201. Alternatively, the detectable moiety comprises or consists of a paramagnetic isotope, such as gadolinium-157, manganese-55, dysprosium-162, chromium-52 or iron-56.


Cytotoxic and detectable moieties may be conjugated or otherwise combined with the binding moiety using methods well known in the art (for example, the existing immunoconjugate therapy, gemtuzumab ozogamicin [tradename: Mylotarg®], comprises a monoclonal antibody linked to the cytotoxin calicheamicin).


A third aspect of the invention provides a pharmaceutical composition comprising an effective amount of an agent as defined in relation to the first or second aspects of the invention together with a pharmaceutically acceptable buffer, diluent, carrier, adjuvant or excipient.


Additional compounds may also be included in the compositions, including, chelating agents such as EDTA, citrate, EGTA or glutathione.


The pharmaceutical compositions may be prepared in a manner known in the art that is sufficiently storage stable and suitable for administration to humans and animals. For example, the pharmaceutical compositions may be lyophilised, e.g. through freeze drying, spray drying, spray cooling, or through use of particle formation from supercritical particle formation.


By “pharmaceutically acceptable” we mean a non-toxic material that does not decrease the effectiveness of the SLAMF6-binding activity of the agent of the invention. Such pharmaceutically acceptable buffers, carriers or excipients are well-known in the art (see Remington's Pharmaceutical Sciences, 18th edition, A. R Gennaro, Ed., Mack Publishing Company (1990) and handbook of Pharmaceutical Excipients, 3rd edition, A. Kibbe, Ed., Pharmaceutical Press (2000), the disclosures of which are incorporated by reference).


The term “buffer” is intended to mean an aqueous solution containing an acid-base mixture with the purpose of stabilising pH. Examples of buffers are Trizma, Bicine, Tricine, MOPS, MOPSO, MOBS, Tris, Hepes, HEPBS, MES, phosphate, carbonate, acetate, citrate, glycolate, lactate, borate, ACES, ADA, tartrate, AMP, AMPD, AMPSO, BES, CABS, cacodylate, CHES, DIPSO, EPPS, ethanolamine, glycine, HEPPSO, imidazole, imidazolelacetic acid, PIPES, SSC, SSPE, POPSO, TAPS, TABS, TAPSO and TES.


The term “diluent” is intended to mean an aqueous or non-aqueous solution with the purpose of diluting the agent in the pharmaceutical preparation. The diluent may be one or more of saline, water, polyethylene glycol, propylene glycol, ethanol or oils (such as safflower oil, corn oil, peanut oil, cottonseed oil or sesame oil).


The term “adjuvant” is intended to mean any compound added to the formulation to increase the biological effect of the agent of the invention. The adjuvant may be one or more of zinc, copper or silver salts with different anions, for example, but not limited to fluoride, chloride, bromide, iodide, thiocyanate, sulfite, hydroxide, phosphate, carbonate, lactate, glycolate, citrate, borate, tartrate, and acetates of different acyl composition. The adjuvant may also be cationic polymers such as cationic cellulose ethers, cationic cellulose esters, deacetylated hyaluronic acid, chitosan, cationic dendrimers, cationic synthetic polymers such as poly(vinyl imidazole), and cationic polypeptides such as polyhistidine, polylysine, polyarginine, and peptides containing these amino acids.


The excipient may be one or more of carbohydrates, polymers, lipids and minerals. Examples of carbohydrates include lactose, glucose, sucrose, mannitol, and cyclodextrines, which are added to the composition, e.g. for facilitating lyophilisation. Examples of polymers are starch, cellulose ethers, cellulose carboxymethylcellulose, hydroxypropylmethyl cellulose, hydroxyethyl cellulose, ethylhydroxyethyl cellulose, alginates, carrageenans, hyaluronic acid and derivatives thereof, polyacrylic acid, polysulphonate, polyethylene glycol/polyethylene oxide, polyethylene oxide/polypropylene oxide copolymers, polyvinylalcohol/polyvinylacetate of different degree of hydrolysis, and polyvinylpyrrolidone, all of different molecular weight, which are added to the composition, e.g. for viscosity control, for achieving bioadhesion, or for protecting the lipid from chemical and proteolytic degradation. Examples of lipids are fatty acids, phospholipids, mono-, di-, and triglycerides, ceramides, sphingolipids and glycolipids, all of the different acyl chain length and saturation, egg lecithin, soy lecithin, hydrogenated egg and soy lecithin, which are added to the composition for reasons similar to those for polymers. Examples of minerals are talc, magnesium oxide, zinc oxide and titanium oxide, which are added to the composition to obtain benefits such as reduction of liquid accumulation or advantageous pigment properties.


The agents of the invention may be formulated into any type of pharmaceutical composition known in the art to be suitable for the delivery thereof.


In one embodiment, the pharmaceutical compositions of the invention may be in the form of a liposome, in which the agent is combined, in addition to other pharmaceutically acceptable carriers, with amphipathic agents such as lipids, which exist in aggregated forms as micelles, insoluble monolayers and liquid crystals. Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like. Suitable lipids also include the lipids above modified by poly(ethylene glycol) in the polar headgroup for prolonging bloodstream circulation time. Preparation of such liposomal formulations is can be found in for example U.S. Pat. No. 4,235,871, the disclosures of which are incorporated herein by reference.


The pharmaceutical compositions of the invention may also be in the form of biodegradable microspheres. Aliphatic polyesters, such as poly(lactic acid) (PLA), poly(glycolic acid) (PGA), copolymers of PLA and PGA (PLGA) or poly(caprolactone) (PCL), and polyanhydrides have been widely used as biodegradable polymers in the production of microspheres. Preparations of such microspheres can be found in U.S. Pat. No. 5,851,451 and in EP 0 213 303, the disclosures of which are incorporated herein by reference.


In a further embodiment, the pharmaceutical compositions of the invention are provided in the form of polymer gels, where polymers such as starch, cellulose ethers, cellulose carboxymethylcellulose, hydroxypropylmethyl cellulose, hydroxyethyl cellulose, ethylhydroxyethyl cellulose, alginates, carrageenans, hyaluronic acid and derivatives thereof, polyacrylic acid, polyvinyl imidazole, polysulphonate, polyethylene glycol/polyethylene oxide, polyethylene oxide/polypropylene oxide copolymers, polyvinylalcohol/polyvinylacetate of different degree of hydrolysis, and polyvinylpyrrolidone are used for thickening of the solution containing the agent. The polymers may also comprise gelatin or collagen.


Alternatively, the agents may simply be dissolved in saline, water, polyethylene glycol, propylene glycol, ethanol or oils (such as safflower oil, corn oil, peanut oil, cottonseed oil or sesame oil), tragacanth gum, and/or various buffers.


It will be appreciated that the pharmaceutical compositions of the invention may include ions and a defined pH for potentiation of action of the active agent. Additionally, the compositions may be subjected to conventional pharmaceutical operations such as sterilisation and/or may contain conventional adjuvants such as preservatives, stabilisers, wetting agents, emulsifiers, buffers, fillers, etc.


The pharmaceutical compositions according to the invention may be administered via any suitable route known to those skilled in the art. Thus, possible routes of administration include parenteral (intravenous, subcutaneous, and intramuscular), topical, ocular, nasal, pulmonary, buccal, oral, parenteral, vaginal and rectal. Also, administration from implants is possible.


In one preferred embodiment, the pharmaceutical compositions are administered parenterally, for example, intravenously, intracerebroventricularly, intraarticularly, intra-arterially, intraperitoneally, intrathecally, intraventricularly, intrasternally, intracranially, intramuscularly or subcutaneously, or they may be administered by infusion techniques. They are conveniently used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood. The aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary. The preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.


Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.


Thus, the pharmaceutical compositions of the invention are particularly suitable for parenteral, e.g. intravenous, administration.


Alternatively, the pharmaceutical compositions may be administered intranasally or by inhalation (for example, in the form of an aerosol spray presentation from a pressurised container, pump, spray or nebuliser with the use of a suitable propellant, such as dichlorodifluoromethane, trichlorofluoro-methane, dichlorotetrafluoro-ethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA 134A3 or 1,1,1,2,3,3,3-heptafluoropropane (HFA 227EA3), carbon dioxide or other suitable gas). In the case of a pressurised aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. The pressurised container, pump, spray or nebuliser may contain a solution or suspension of the active polypeptide, e.g. using a mixture of ethanol and the propellant as the solvent, which may additionally contain a lubricant, e.g. sorbitan trioleate. Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of a compound of the invention and a suitable powder base such as lactose or starch.


The pharmaceutical compositions will be administered to a patient in a pharmaceutically effective dose. A ‘therapeutically effective amount’, or ‘effective amount’, or ‘therapeutically effective’, as used herein, refers to that amount which provides a therapeutic effect for a given condition and administration regimen. This is a predetermined quantity of active material calculated to produce a desired therapeutic effect in association with the required additive and diluent, i.e. a carrier or administration vehicle. Further, it is intended to mean an amount sufficient to reduce and most preferably prevent, a clinically significant deficit in the activity, function and response of the host. Alternatively, a therapeutically effective amount is sufficient to cause an improvement in a clinically significant condition in a host. As is appreciated by those skilled in the art, the amount of a compound may vary depending on its specific activity. Suitable dosage amounts may contain a predetermined quantity of active composition calculated to produce the desired therapeutic effect in association with the required diluent. In the methods and use for manufacture of compositions of the invention, a therapeutically effective amount of the active component is provided. A therapeutically effective amount can be determined by the ordinary skilled medical or veterinary worker based on patient characteristics, such as age, weight, sex, condition, complications, other diseases, etc., as is well known in the art. The administration of the pharmaceutically effective dose can be carried out both by single administration in the form of an individual dose unit or else several smaller dose units and also by multiple administrations of subdivided doses at specific intervals. Alternatively, the does may be provided as a continuous infusion over a prolonged period.


The polypeptides can be formulated at various concentrations, depending on the efficacy/toxicity of the compound being used. Preferably, the formulation comprises the active agent at a concentration of between 0.1 μM and 1 mM, more preferably between 1 μM and 500 μM, between 500 μM and 1 mM, between 300 μM and 700 μM, between 1 μM and 100 μM, between 100 μM and 200 μM, between 200 μM and 300 μM, between 300 μM and 400 μM, between 400 μM and 500 μM and most preferably about 500 μM.


It will be appreciated by persons skilled in the art that the pharmaceutical compositions of the invention may be administered alone or in combination with other therapeutic agents used in the treatment of a neoplastic hematologic disorder, such as inhibitors of tyrosine kinase (e.g. imatinib mesylate [Glivec®], dasatinib, nilotinib), omacetaxine, antimetabolites (e.g. cytarabine, hydroxyurea), alkylating agents, Interferon alpha-2b and/or steroids.


A fourth aspect of the invention provides a kit comprising an agent as defined in relation to the first or second aspects of the invention or a pharmaceutical composition according to the third aspect of the invention.


A fifth aspect of the invention provides the use of an agent as defined in relation to the first or second aspects of the invention in the preparation of a medicament for inducing cell death and/or inhibiting the growth and/or proliferation of pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder, wherein the stem cells and/or progenitor cells express SLAMF6.


The agent may also be for use in inducing differentiation of pathological stem and/or progenitor cells which express SLAMF6.


A related sixth aspect of the invention provides the use of an agent as defined in relation to the first or second aspects of the invention in the preparation of a diagnostic agent for detecting pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder, wherein the stem cells and/or progenitor cells express SLAMF6. Another related aspect of the invention may be the diagnosis of a patient population that is at risk of relapse; such as relapse may be caused by the persistence of cancer stem cells.


A related seventh aspect of the invention provides the use of an agent as defined in relation to the first or second aspects of the invention for detecting pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder, wherein the stem cells and/or progenitor cells express SLAMF6.


In one embodiment of the above use aspects of the invention, the neoplastic hematologic disorder is a leukemia. In a further embodiment, the neoplastic hematologic disorder may be associated with cells comprising a TP53 mutation. Mutations of TP53 can occur throughout the gene and abrogate the function of the p53 protein in multiple ways, for example by amino acid substitution, truncation, deletion or altered splicing. Thus, TP53 mutations may refer to any mutations that alter the protein sequence of the p53 protein. Further, p53 function can also be disrupted by other mechanisms than TP53 mutation. For example, suitable patients may have observed or predicted loss of p53 function by other means, including but not limited to full or partial loss of chromosome 17, epigenetic silencing and alterations in p53 signaling pathways.


Thus, by “TP53 mutation” we include any observed or predicted loss or reduction of p53 function.


This may be as a result of either one or more structural mutations (i.e. wherein the amino acid sequence of the P53 protein is altered) and/or by a functional alteration (i.e. wherein the function of the p53 mutation is disrupted by other means). In a functional alteration there is an observed or predicted loss of p53 function by other means, including but not limited to full or partial loss of chromosome 17, epigenetic silencing and alterations in p53 signaling pathways, as outlined above.


In another further embodiment, the neoplastic hematologic disorder may be associated with cells expressing CD34+CD38. In yet another further embodiment, the neoplastic hematologic disorder may be associated with cells comprising a TP53 mutation and expressing CD34+CD38.


More specifically, the neoplastic hematologic disorder may be selected from the group consisting of chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). In one particularly preferred embodiment, the neoplastic hematologic disorder is acute myeloid leukemia (AML).


An eighth aspect of the invention provides a method for inducing cell death and/or inhibiting the growth and/or proliferation of pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder in an individual, comprising the step of administering to the individual an effective amount of an agent as defined in relation to the first or second aspects of the invention, or a pharmaceutical composition according to the third aspect of the invention, wherein the stem cells and/or progenitor cells express SLAMF6. In a further embodiment, the neoplastic hematologic disorder may be associated with cells comprising a TP53 mutation. In another further embodiment, the neoplastic hematologic disorder may be associated with cells expressing CD34+CD38. In yet another further embodiment, the neoplastic hematologic disorder may be associated with cells comprising a TP53 mutation and expressing CD34+CD38.


The method may also be for inducing differentiation of pathological stem and/or progenitor cells which express SLAMF6.


Thus, the invention provides methods for the treatment of neoplastic hematologic disorders. By ‘treatment’ we include both therapeutic and prophylactic treatment of the patient. The term ‘prophylactic’ is used to encompass the use of a polypeptide or formulation described herein, which either prevents or reduces the likelihood of a neoplastic hematologic disorder in a patient or subject.


As above, the neoplastic hematologic disorder may be selected from the group consisting of chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). In one particularly preferred embodiment, the neoplastic hematologic disorder is acute myeloid leukemia (AML).


A ninth aspect of the invention provides a method for detecting pathological stem cells and/or progenitor cells associated with neoplastic hematologic disorder in an individual, comprising the step of administering to the individual an effective amount of an agent as defined in relation to the first or second aspects of the invention, or a pharmaceutical composition according to the third aspect of the invention, wherein the stem cells and/or progenitor cells express SLAMF6. In a further embodiment, the neoplastic hematologic disorder may be associated with cells comprising a TP53 mutation. In another further embodiment, the neoplastic hematologic disorder may be associated with cells expressing CD34+CD38. In yet another further embodiment, the neoplastic hematologic disorder may be associated with cells comprising a TP53 mutation and expressing CD34+CD38.


As above, the neoplastic hematologic disorder may be selected from the group consisting of chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). In one particularly preferred embodiment, the neoplastic hematologic disorder is acute myeloid leukemia (AML).


A tenth aspect of the invention provides an in vitro method for diagnosing or prognosing a neoplastic hematologic disorder.


In one embodiment, the method comprises:

    • (a) providing a bone marrow or peripheral blood sample of haematopoietic cells from an individual to be tested;
    • (b) isolating a subpopulation of CD34+, CD38 cells from the haematopoietic cells; and
    • (c) determining whether stem cells, contained within the CD34+, CD38 cells, express the cell surface markers SLAMF6;
      • wherein stem cells that exhibit the cell surface marker profile CD34+, CD38 and SLAMF6+ are indicative of the individual having or developing leukemia.


In one embodiment, the method comprises:

    • (a) isolating a subpopulation of CD34+, CD38 cells from haematopoietic cells in a sample; and
    • (b) determining whether stem cells, contained within the CD34+, CD38 cells, express the cell surface markers SLAMF6;
      • wherein stem cells that exhibit the cell surface marker profile CD34+, CD38 and SLAMF6+ are indicative of the individual having or developing leukemia. The sample may optionally be a bone marrow sample or peripheral blood sample.


In an alternative embodiment, the in vitro method for diagnosing or prognosing a neoplastic hematologic disorder comprises the following steps:

    • (a) providing a bone marrow or peripheral blood sample of haematopoietic cells from an individual to be tested; and
    • (b) isolating a subpopulation of SLAMF6+, CD34+, CD38 cells from the haematopoietic cells;
      • wherein stem cells that exhibit the cell surface marker profile CD34+, CD38 and SLAMF6+ are indicative of the individual having or developing leukemia.


In one embodiment, the method comprises: isolating a subpopulation of SLAMF6+, CD34+, CD38 cells from haematopoietic cells in a sample; wherein stem cells that exhibit the cell surface marker profile CD34+, CD38 and SLAMF6+ are indicative of the individual having or developing leukemia. The sample may optionally be a bone marrow sample or peripheral blood sample.


In all embodiments of the tenth aspect, the provision of a sample is not necessarily to be construed as involving a surgical step. The provision of a sample could be a pre-isolated and stored frozen sample, for example. Further, the term “isolating” is to be construed as meaning the same as “detecting” and “determining”. For example, step (b) could be the step of “detecting a subpopulation” or “determining whether a subpopulation exists”.


In one embodiment of the above in vitro method, it is used to identify patients that may be at a risk (or increased risk) of cancer relapse. Thus, potentially in addition to diagnosing or prognosing a neoplastic hematologic disorder (such as in a sample derived from a patient), the method may also diagnose or prognose an increased risk of relapse of the neoplastic hematologic disorder. For example, the in vitro method may be used to identify patients with leukemic stem cells (LSC), which are cells that have the capacity to regenerate the leukemia and cause relapse but are not effectively targeted by current treatments. This may be achieved by detecting LSCs that are generally enriched in the CD34+CD38 compartment.


In one optional embodiment, the individual has also been tested for the presence of certain immune cells. For example, the in vitro method may be used to quantify the number of immune cells (such as B cells, T cells and NK cells) that express SLAMF6.


For example, in one embodiment the method may further comprise an additional step performing FACS on the bone marrow or peripheral blood sample to identify B cell, T cell and/or NK cell markers with SLAMF6 co-expression, e.g. using CD19 expression as a marker to identify B cells or CD3 expression as a marker to identify T cells. The skilled person would be aware of FACS panels for these cell subsets.


In one embodiment, the method further comprises the step of treating a patient diagnosed as having a neoplastic haematologic disorder with an effective therapy therefor, for example chemotherapy, biological therapy (e.g. immunotherapy), targeted therapy, radiation therapy and/or stem cell or bone marrow transplant.


In one embodiment of the above method aspects of the invention, the neoplastic hematologic disorder is a leukemia. More specifically, the neoplastic hematologic disorder may be selected from the group consisting of chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). In one particularly preferred embodiment, the neoplastic hematologic disorder is acute myeloid leukemia (AML).


In a further embodiment, the neoplastic hematologic disorder may be associated with cells comprising a TP53 mutation.


However, it will be appreciated by persons skilled in the art that the agents of the invention may also be used in the treatment and/or diagnosis of neoplastic hematologic disorders which are not associated with cells comprising a TP53 mutation (but nevertheless show upregulation of SLAMF6). Such neoplastic hematologic disorders which are associated with cells which do not comprise a TP53 mutation may include the myelodysplastic syndromes (MDS) and myeloproliferative disorders (MPD) such as polycythemia vera (PV), essential thrombocytosis (ET) and myelofibrosis (MF).





Preferred, non-limiting examples which embody certain aspects of the invention will now be described, with reference to the following figures:



FIG. 1: Arrayed antibody screen shows SLAMF6 to be specifically expressed in TP53 mutated AML cells.


A: Gates used to define immature, viable, single cells with a CD3CD19CD34+CD38 phenotype. B: Waterfall plot of median difference in MFI between AML and NBM within the CD3CD19CD34+CD38 compartment. Three TP53 mutated AML and three NBM samples were analyzed. Markers with high expression in NBM are excluded from this plot. C: Spearman correlation of MFI between biological replicates from antibody screen for NBM #2 and NBM #3. 14 values not depicted due to value of zero or negative values and logarithmic axes. D: Spearman correlation of MFI between biological replicates from antibody screen for AML #83 and AML #80. 42 values not depicted due to value of zero or negative values. E: Top ranked cell surface markers based on difference in AML MFI and NBMMFI from three separate analyses of TP53 mutated AML and NBM samples. Median MFI is plotted for AML (black bars) and NBM (white bars).



FIG. 2: Flow cytometric validation of SLAMF6 overexpression in TP53 mutated AML cells.


Eight novel cell surface receptors with high ranking in the screen were analyzed in separate experiments. Representative histograms of an AML (far left), an NBM sample (middle left), percent positive cells (middle right), and MFI (far right) for each marker within the CD34+CD38 population are shown. Mean and standard deviation are shown.



FIG. 3: SLAMF6 is upregulated in immature TP53 mutated AML cells.


A: AML SLAMF6 expression is higher in phenotypically immature CD34+CD38+ and CD34+CD38 populations. Histograms from a representative sample (AML 48) shown. B: NBM SLAMF6 expression is detected in mature CD34CD38 and CD34CD38+ NBM population but low or absent in more immature CD34+CD38+ and CD34+CD38 respectively. Histograms from one representative sample shown. C: SLAMF6 gene expression levels are higher in TP53 mutated AML MNC cells than in stem and progenitor populations of normal bone marrow. CMP: common myeloid progenitor, GMP: granulocyte-macrophage progenitor, MEP: megakaryocyte-erythroid progenitor, LMPP: lymphoid-primed multipotent progenitor, MPP: multipotent progenitor, HSC: hematopoietic stem cells. D: CD3+ T cells and CD19+ B cells in AML samples retain their SLAMF6 expression in all tested genetic subgroups of AML. AML 66 is shown as a representative example.



FIG. 4: SLAMF6 expression levels in genetic subtypes of AML.


A: SLAMF6 gene expression levels are significantly higher in TP53 mutated AML samples (n=14) compared to AML samples of other genetic subtypes (n=151) in the publicly available TCGA dataset. B: SLAMF6 is expressed in the leukemic CD19CD3 population in a TP53 mutated CD34 negative AML sample. C: SLAMF6 is expressed on leukemic cells in AML 33, AML 34 and on a subpopulation of MDS144. D: SLAMF6 is possibly expressed at low levels on leukemic cells in AML 32, MDS35 and AML 66. E: No expression of SLAMF6 could be detected on the leukemic cells in AML 21, AML 28, MDS70 or AML 94.



FIG. 5: SLAMF6 expression is retained in AML patient derived xenografts and anti-SLAMF6 antibodies induce ADCC killing of KG1 cells.


A: Histograms showing SLAM6 expression in the AML cell lines KG1 and K562 but not OCI-AML 3. B: Specific SLAMF6 mediated ADCC killing of KG1 cells (black, solid line) compared to isotype control (grey, dashed line). Mean effect and standard deviation of three different NK donors are shown, results are normalization to base line killing by the NK cells. C: Xenografts of TP53 mutated AML show high levels of SLAMF6 expression within the human CD45+CD34+CD38 compartment. Spleen from secondary an AML 48 xenograft is shown as a representative example with 99% CD45+ human cells, 45% CD34+CD38 cells and a high SLAMF6 expression.



FIG. 6: A SLAMF6 antibody mediates killing of AML patient cells by ADCC.


A. Specific killing of AML patient cells ex vivo by a SLAMF6 antibody eliciting NK cell recruitment and killing through ADCC. Values indicate the average number of remaining viable target cells+/−SEM after treatment with a SLAMF6 antibody (black, solid line) or an isotype control (grey, dashed line), normalized to a control without antibody. For AML-83, cells were passaged in vivo for two generations for LSC enrichment before being subjected to ADCC. One NK cell donor was used for PDX-83 and four for AML-61. B. Protein expression of SLAMF6 on the AML patient cells.



FIG. 7: SLAMF6 expression on AML samples.


SLAMF6 as determined by flow cytometry is expressed on a majority of AML samples, in particular in the CD34+CD38low leukemic stem cell containing fraction. Samples were classified as “High” when containing >50% SLAMF6 positive cells, “Intermediate” when containing 10-50% SLAMF6 positive cells and “Negative” when containing <10% SLAMF6 positive cells.



FIG. 8: SLAMF6 expression on leukemic stem cells from AML samples.


SLAMF6 as determined by flow cytometry is expressed on leukemic stem cells from AML samples with a large variety of mutational backgrounds.



FIG. 9: SLAMF6 expression on cell lines.


SLAMF6 as determined by flow cytometry is expressed on the AML cell lines KG1 (DSMZ #ACC14), HNT-34 (DSMZ #ACC600), TF-1 (DSMZ #ACC334), CMK (DSMZ #ACC392), and K562 (DSMZ #ACC10) but not on THP-1 (DSMZ #ACC16), OCI-AML 3 (DSMZ #ACC582), NB4 (DSMZ #ACC207), or MonoMac6 (DSMZ #ACC124).



FIG. 10. Antibody-mediated activation of SLAMF6 on T cells promotes killing of AML cells.


T cell-mediated killing of HNT-34 cells, measured as the total number of target cells 72 h after seeding of T cells and HNT-34 target cells at a 4:1 ratio, with the addition of an activating SLAMF6 antibody or an isotype control.



FIG. 11. SLAMF6 mediates resistance to T cell killing.


T cell-mediated killing of SLAMF6 knockout cells, measured as the total number of target cells (A), T cells (B) and activated T cells (C) three days after seeding of T cells and KG-1 target cells at a 4:1 ratio. Dashed line indicates the number of cells seeded. (D) Validation of SLAMF6 knockout by FACS.





EXAMPLE 1

SLAMF6 is a Cell Surface Biomarker for Acute Myeloid Leukemia Stem Cells


Summary


Therapeutic strategies for acute myeloid leukemia (AML) aiming at achieving a permanent cure of the disorder, will require a full eradication of the AML stem cells. The AML stem cells, sharing the capacity to self-renew with normal hematopoietic stem cells (HSCs), represent a small population of leukemic cells that so far have been indistinguishable from normal (HSCs) using cell surface markers. One strategy to target the AML stem cell would be to identify a cell surface biomarker for AML stem cells, to which future therapeutic antibodies could be directed. In this study, SLAMF6 was identified in a surface marker screen of TP53-mutated AML as commonly expressed on primitive CD34+CD38 AML cells but not on corresponding normal cells. Furthermore, targeting of SLAMF6 was shown to induce killing of AML cells through antibody-dependent cellular cytotoxicity (ADCC).


This study thus identifies SLAMF6 as a novel cell surface biomarker distinguishing AML stem cells from normal HSC and opens up new avenues for therapeutic and diagnostic strategies in AML. In addition to direct effects on the leukemia cell, targeting of SLAMF6 may also alter its interaction with immune cells to induce an anti-tumor response. Further, as SLAMF6 is also expressed on immune cells (B cells, T cells and NK cells), it is a suitable target for dual targeting on both pathological stem cells (e.g. leukemic stem cells) and immune cells (e.g. B cells, T cells and/or NK cells), whereby the immune cells may be activated to enhance elimination of the pathological stem cells. For example, the agent may be used to target pathological stem cells and B cells; or pathological stem cells, B cells and T cells; or pathological stem cells, NK cells and T cells, etc.


Introduction


To identify a cell surface biomarker for AML stem cells, the inventors performed an antibody screen and identified SLAMF6 as a novel candidate, being upregulated in primitive AML patient cells.


Materials and Methods


Patient Samples


Bone marrow and peripheral blood samples were collected after written informed consent in accordance to the Declaration of Helsinki. Samples were collected from patients with AML, myelodysplastic syndrome (MDS) or healthy controls. Mononuclear cells (MNC) were isolated using Lymphoprep (GE Healthcare Bio-Sciences AB, Sweden) and subsequently viably frozen. Patients included in the study and their clinical characteristics are shown in Table 1. The study was approved by a regional ethics committee in Lund (Dnr2011/289).


Cell Surface Marker Screen


Arrayed antibody libraries were prepared based on the LEGENDScreen system (BioLegend, USA), containing 362 PE-conjugated antibodies. Two different iterations of the LEGENDScreen (BioLegend, USA) were used, containing slightly different antibodies (#700001 had 34 antibodies not included in #700007 which contained 61 antibodies not included in #700001; for a complete list, see Table 2). Antibodies targeting IL1RAP, previously shown to be upregulated on AML stem cells,9, 22, 23 and CD177, suggested to be upregulated on the mRNA level in AML (data not shown), were added to the arrays. Antibodies against CD3, CD19, CD34, CD38 and the viability marker 7AAD were added to each well of the 96-well U-shaped plates. All antibodies and reagents used are listed in Table 3. Bone marrow mononuclear cells from TP53 mutated AML samples (n=3) or healthy normal bone marrow (NBM) donors (n=3) were added and incubated at 4° C. for 20 minutes and subsequently washed and resuspended. Flow cytometry read out was performed using an LSR Fortessa with an HTS (BD Bioscience, USA). Median fluorescent intensity (MFI) for each marker within the 7AADCD3CD19CD34+CD38 fraction were used to compare expression between AML and NBM (FIG. 1A). First, markers with a high expression within the 7AADCD3CD19CD34+CD38 fraction of NBM samples (>10% positive or MFI>10000) were excluded. Next, marker NBMMFI were subtracted from paired AML MFI and markers were ranked according to median MFI from three separate experiments. The corresponding analyses and ranking were performed using the quota of AML MFI/NBMMFI. The two ranking systems were combined to produce a top list of specifically upregulated cell surface markers (Table 4).









TABLE 1







AML Patient Characteristics

























SLAMF6











in CD34+


Pat#
Gender
Disease
FAB
ELN
Karyotype
Mutations
CD34
SLAMF6
CD38−



















21
M
AML-D
M2
IR
46, XY
IDH2, STAG2, BCOR
pos
neg
neg


28
M
SAML-D
M2
IR
46, XY
NPM1, FLT3-ITD, TET2
neg
neg



32
M
AML-D
M2
HR
47, XY, +13
RUNX1, ASXL1,
pos
low
low


33
F
SAML-R
M4
IR
46, XX
DNMT3A, NPM1, FLT3-
neg
pos









ITD, IDH1, CEBPA


34
F
AML-D
M5
IR
46, XX
DNMT3A, IDH1, NRAS,
pos
pos
pos








CEBPA


35
F
MDS


46, XX
RUNX1, ASXL1,
pos
low
low




RAEB-2



DNMT3A, IDH2


48
F
tAML-D
M2
HR
Complex1
TP53
pos
pos
pos


66
M
SAML-D
M2
LR
46, XY
IDH1, NPM1, JAK2,
neg
low









CEBPA


70
M
MPN/MDS


46, XY
RUNX1, ASXL1, TET2,
neg
neg



80
M
AML-D
M2
HR
Complex2
TP53
pos
pos
pos


83
M
SAML-D
M2
HR
Complex3
TP53, TET2, NF1
pos
pos
pos


94
M
AML-D
M0
HR
47, XY, +10
DNMT3A, IDH1, FLT3-
pos
neg
neg








ITD, RUNX1, BCOR,








STAG2


144
F
MPN/MDS


47, XX, +8
RUNX1, ASXL1, TET2,
neg
pos









BCOR


155
M
AML-D
M5
HR
Complex4
TP53, FLT3
neg
pos






Abbreviations:


D: diagnosis,


R: relapse,


sAML: secondary AML,


tAML: therapy-related AML,


ELN: European Leukemia net risk classification 2017,


IR: intermediate risk,


HR: high risk,


CD34 pos: ≥15% CD34+ cells


Complex1: 43-47, XX, del(5)(q13q33), der(8)t(8;12)(p22;p13), add(11)(p15), −12, add(13)(p11), −18, del(20)(q11), add(22)(q13), +mar


Complex2: 48, XXYc, +i(12)(p10), der(17)t(13;17)(q1?4;p1?3)


Complex3: 45, XY, der(13;14)(q10;q10)/42-44, idem, −5, −7, −9, −10, −11, ?hsr(11)(q23), −14, −16, −20, +4mar


Complex4: 47, XY, +8, t(9;11)(p21;q23)/46-47, idem, der(9)del(9)(p12)del(9)(q12), der(17)t(9;17)(q31;p13)/46-47, idem, der(7;10)(q10;q10), +add(8)(p11)













TABLE 2





Antibodies included in the screen







Screen Iteration 70001












Plate
Well ID
Specificity
Clone
Isotype
Cat#





Plate 1
A01
Blank


Plate 1
A02
CD1a
HI149
Mouse IgG1, κ
300106


Plate 1
A03
CD1b
SN13 (K5- 1B8)
Mouse IgG1, κ
329108


Plate 1
A04
CD1c
L161
Mouse IgG1, κ
331506


Plate 1
A05
CD1d
51.1
Mouse IgG2b, κ
350306


Plate 1
A06
CD2
RPA-2.10
Mouse IgG1, κ
300208


Plate 1
A07
CD3
HIT3a
Mouse IgG2a, κ
300308


Plate 1
A08
CD4
RPA-T4
Mouse IgG1, κ
300508


Plate 1
A09
CD5
UCHT2
Mouse IgG1, κ
300608


Plate 1
A10
CD6
BL-CD6
Mouse IgG1, κ
313906


Plate 1
A11
CD7
CD7-6B7
Mouse IgG2a, κ
343106


Plate 1
A12
CD8a
HIT8a
Mouse IgG1, κ
300908


Plate 1
B01
CD9
HI9a
Mouse IgG1, κ
312106


Plate 1
B02
CD10
HI10a
Mouse IgG1, κ
312204


Plate 1
B03
CD11a
HI111
Mouse IgG1, κ
301208


Plate 1
B04
CD11b
ICRF44
Mouse IgG1, κ
301306


Plate 1
B05
CD11b (activated)
CBRM1/5
Mouse IgG1, κ
301406


Plate 1
B06
CD11c
3.9
Mouse IgG1, κ
301606


Plate 1
B07
CD13
WM15
Mouse IgG1, κ
301704


Plate 1
B08
CD14
M5E2
Mouse IgG2a, κ
301806


Plate 1
B09
CD15 (SSEA-1)
W6D3
Mouse IgG1, κ
323006


Plate 1
B10
CD16
3G8
Mouse IgG1, κ
302008


Plate 1
B11
CD18
TS1/18
Mouse IgG1, κ
302108


Plate 1
B12
CD19
HIB19
Mouse IgG1, κ
302208


Plate 1
C01
CD20
2H7
Mouse IgG2b, κ
302306


Plate 1
C02
CD21
Bu32
Mouse IgG1, κ
354904


Plate 1
C03
CD22
HIB22
Mouse IgG1, κ
302506


Plate 1
C04
CD23
EBVCS-5
Mouse IgG1, κ
338508


Plate 1
C05
CD24
ML5
Mouse IgG2a, κ
311106


Plate 1
C06
CD25
BC96
Mouse IgG1, κ
302606


Plate 1
C07
CD26
BA5b
Mouse IgG2a, κ
302706


Plate 1
C08
CD27
O323
Mouse IgG1, κ
302808


Plate 1
C09
CD28
CD28.2
Mouse IgG1, κ
302908


Plate 1
C10
CD29
TS2/16
Mouse IgG1, κ
303004


Plate 1
C11
CD30
BY88
Mouse IgG1, κ
333906


Plate 1
C12
CD31
WM59
Mouse IgG1, κ
303106


Plate 1
D01
CD32
FUN-2
Mouse IgG2b, κ
303206


Plate 1
D02
CD33
WM53
Mouse IgG1, κ
303404


Plate 1
D03
CD34
581
Mouse IgG1, κ
343506


Plate 1
D04
CD35
E11
Mouse IgG1, κ
333406


Plate 1
D05
CD36
5-271
Mouse IgG2a, κ
336206


Plate 1
D06
CD38
HIT2
Mouse IgG1, κ
303506


Plate 1
D07
CD39
A1
Mouse IgG1, κ
328208


Plate 1
D08
CD40
HB14
Mouse IgG1, κ
313006


Plate 1
D09
CD41
HIP8
Mouse IgG1, κ
303706


Plate 1
D10
CD42b
HIP1
Mouse IgG1, κ
303906


Plate 1
D11
CD43
CD43-10G7
Mouse IgG1, κ
343204


Plate 1
D12
CD44
BJ18
Mouse IgG1, κ
338808


Plate 1
E01
CD45
HI30
Mouse IgG1, κ
304008


Plate 1
E02
CD45RA
HI100
Mouse IgG2b, κ
304108


Plate 1
E03
CD45RB
MEM-55
Mouse IgG2b, κ
310204


Plate 1
E04
CD45RO
UCHL1
Mouse IgG2a, κ
304206


Plate 1
E05
CD46
TRA-2-10
Mouse IgG1
352402


Plate 1
E06
CD47
CC2C6
Mouse IgG1, κ
323108


Plate 1
E07
CD48
BJ40
Mouse IgG1, κ
336708


Plate 1
E08
CD49a
TS2/7
Mouse IgG1, κ
328304


Plate 1
E09
CD49c
ASC-1
Mouse IgG1, κ
343804


Plate 1
E10
CD49d
9F10
Mouse IgG1, κ
304304


Plate 1
E11
CD49e
NKI-SAM-1
Mouse IgG2b, κ
328010


Plate 1
E12
CD49f
GoH3
Rat IgG2a, κ
313612


Plate 1
F01
CD50 (ICAM-3)
CBR-IC3/1
Mouse IgG1, κ
330005


Plate 1
F02
CD51
NKI-M9
Mouse IgG2a, κ
327910


Plate 1
F03
CD51/61
23C6
Mouse IgG1, κ
304406


Plate 1
F04
CD52
HI186
Mouse IgG2b, κ
316006


Plate 1
F05
CD53
HI29
Mouse IgG1, κ
325406


Plate 1
F06
CD54
HA58
Mouse IgG1, κ
353106


Plate 1
F07
CD55
JS11
Mouse IgG1, κ
311308


Plate 1
F08
CD56 (NCAM)
HCD56
Mouse IgG1, κ
318306


Plate 1
F09
CD57
HCD57
Mouse IgM, κ
322312


Plate 1
F10
CD58
TS2/9
Mouse IgG1, κ
330905


Plate 1
F11
CD59
p282(H19)
Mouse IgG2a, κ
304708


Plate 1
F12
CD61
VI-PL2
Mouse IgG1, κ
336406


Plate 1
G01
CD62E
HAE-1f
Mouse IgG1, κ
336008


Plate 1
G02
CD62L
DREG-56
Mouse IgG1, κ
304806


Plate 1
G03
CD62P (P-Selectin)
AK4
Mouse IgG1, κ
304906


Plate 1
G04
CD63
H5C6
Mouse IgG1, κ
353004


Plate 1
G05
CD64
10.1
Mouse IgG1, κ
305008


Plate 1
G06
CD66a/c/e
ASL-32
Mouse IgG2b, κ
342304


Plate 1
G07
CD66b
G10F5
Mouse IgM, κ
305106


Plate 1
G08
CD69
FN50
Mouse IgG1, κ
310906


Plate 1
G09
CD70
113-16
Mouse IgG1, κ
355104


Plate 1
G10
CD71
CY1G4
Mouse IgG2a, κ
334106


Plate 1
G11
CD73
AD2
Mouse IgG1, κ
344004


Plate 1
G12
CD74
LN2
Mouse IgG1, κ
326808


Plate 1
H01
CD79b
CB3-1
Mouse IgG1, κ
341404


Plate 1
H02
CD80
2D10
Mouse IgG1, κ
305208


Plate 1
H03
CD81
5A6
Mouse IgG1, κ
349506


Plate 1
H04
CD82
ASL-24
Mouse IgG1, κ
342104


Plate 1
H05
CD83
HB15e
Mouse IgG1, κ
305308


Plate 1
H06
CD84
CD84.1.21
Mouse IgG2a, κ
326008


Plate 1
H07
CD85a (ILT5)
MKT5.1
Rat IgG2a, κ
337704


Plate 1
H08
CD85d (ILT4)
42D1
Rat IgG2a, κ
338706


Plate 1
H09
CD85g (ILT7)
17G10.2
Mouse IgG1, κ
326408


Plate 1
H10
CD85h (ILT1)
24
Mouse IgG2b, κ
337904


Plate 1
H11
CD85j (ILT2)
GHI/75
Mouse IgG2b, κ
333708


Plate 1
H12
CD85k (ILT3)
ZM4.1
Mouse IgG1, κ
333008


Plate 2
A01
Blank


Plate 2
A02
CD86
IT2.2
Mouse IgG2b, κ
305406


Plate 2
A03
CD87
VIM5
Mouse IgG1, κ
336906


Plate 2
A04
CD88
S5/1
Mouse IgG2a, κ
344304


Plate 2
A05
CD89
A59
Mouse IgG1, κ
354104


Plate 2
A06
CD90 (Thy1)
50000000000
Mouse IgG1, κ
328110


Plate 2
A07
CD93
VIMD2
Mouse IgG1, κ
336108


Plate 2
A08
CD94
DX22
Mouse IgG1, κ
305506


Plate 2
A09
CD95
DX2
Mouse IgG1, κ
305608


Plate 2
A10
CD96
NK92.39
Mouse IgG1, κ
338406


Plate 2
A11
CD97
VIM3b
Mouse IgG1, κ
336308


Plate 2
A12
CD99
HCD99
Mouse IgG2a, κ
318008


Plate 2
B01
CD100
A8
Mouse IgG1, κ
328408


Plate 2
B02
CD101 (BB27)
BB27
Mouse IgG1, κ
331006


Plate 2
B03
CD102
CBR-IC2/2
Mouse IgG2a, κ
328506


Plate 2
B04
CD103
Ber-ACT8
Mouse IgG1, κ
350206


Plate 2
B05
CD104
58XB4
Mouse IgG2a, κ
327808


Plate 2
B06
CD105
43A3
Mouse IgG1, κ
323206


Plate 2
B07
CD106
STA
Mouse IgG1, κ
305806


Plate 2
B08
CD107a (LAMP-1)
H4A3
Mouse IgG1, κ
328608


Plate 2
B09
CD108
MEM-150
Mouse IgM, κ
315704


Plate 2
B10
CD109
W7C5
Mouse IgG1, κ
323306


Plate 2
B11
CD111
R1.302
Mouse IgG1, κ
340404


Plate 2
B12
CD112 (Nectin-2)
TX31
Mouse IgG1, κ
337410


Plate 2
C01
CD114
LMM741
Mouse IgG1, κ
346106


Plate 2
C02
CD115
9-4D2-1E4
Rat IgG1, κ
347304


Plate 2
C03
CD116
4H1
Mouse IgG1, κ
305908


Plate 2
C04
CD117 (c-kit)
104D2
Mouse IgG1, κ
313204


Plate 2
C05
CD119 (IFN-g Rd
GIR-208
Mouse IgG1, κ
308606




chain)


Plate 2
C06
CD122
TU27
Mouse IgG1, κ
339006


Plate 2
C07
CD123
6H6
Mouse IgG1, κ
306006


Plate 2
C08
CD124
G077F6
Mouse IgG2a, κ
355004


Plate 2
C09
CD126 (IL-6Rα)
UV4
Mouse IgG1, κ
352804


Plate 2
C10
CD127 (IL-7Rα)
A019D5
Mouse IgG1, κ
351304


Plate 2
C11
CD129 (IL-9 R)
AH9R7
Mouse IgG2b, κ
310404


Plate 2
C12
CD131
1C1
Mouse IgG1, κ
306104


Plate 2
D01
CD132
TUGh4
Rat IgG2b, κ
338606


Plate 2
D02
CD134
Ber-ACT35 (ACT35)
Mouse IgG1, κ
350004


Plate 2
D03
CD135
BV10A4H2
Mouse IgG1, κ
313306


Plate 2
D04
CD137 (4-1BB)
4B4-1
Mouse IgG1, κ
309804


Plate 2
D05
CD137L (4-1BB
5F4
Mouse IgG1, κ
311504




Ligand)


Plate 2
D06
CD138
DL-101
Mouse IgG1, κ
352306


Plate 2
D07
CD140a
16A1
Mouse IgG1, κ
323506


Plate 2
D08
CD140b
18A2
Mouse IgG1, κ
323606


Plate 2
D09
CD141
M80
Mouse IgG1, κ
344104


Plate 2
D10
CD143
5-369
Mouse IgG1, κ
344204


Plate 2
D11
CD144
BV9
Mouse IgG2a, κ
348506


Plate 2
D12
CD146
SHM-57
Mouse IgG2a, κ
342004


Plate 2
E01
CD148
A3
Mouse IgG1, κ
328708


Plate 2
E02
CD150 (SLAM)
A12 (7D4)
Mouse IgG1, κ
306308


Plate 2
E03
CD152
L3D10
Mouse IgG1, κ
349906


Plate 2
E04
CD154
24-31
Mouse IgG1, κ
310806


Plate 2
E05
CD155 (PVR)
SKII.4
Mouse IgG1, κ
337610


Plate 2
E06
CD156c (ADAM10)
SHM14
Mouse IgG1, κ
352704


Plate 2
E07
CD158a/h
HP-MA4
Mouse IgG2b, κ
339506


Plate 2
E08
CD158b (KIR2DL2/L3)
DX27
Mouse IgG2a, κ
312606


Plate 2
E09
CD158d
mAb 33 (33)
Mouse IgG1, κ
347006


Plate 2
E10
CD158e1 (KIR3DL1)
DX9
Mouse IgG1, κ
312708


Plate 2
E11
CD158f
UP-R1
Mouse IgG1, κ
341304


Plate 2
E12
CD161
HP-3G10
Mouse IgG1, κ
339904


Plate 2
F01
CD162
KPL-1
Mouse IgG1, κ
328806


Plate 2
F02
CD163
GHI/61
Mouse IgG1, κ
333606


Plate 2
F03
CD164
67D2
Mouse IgG1, κ
324808


Plate 2
F04
CD165
SN2 (N6- D11)
Mouse IgG1, κ
329010


Plate 2
F05
CD166
3A6
Mouse IgG1, κ
343904


Plate 2
F06
CD167a (DDR1)
51D6
Mouse IgG3, κ
334006


Plate 2
F07
CD169
7-239
Mouse IgG1, κ
346004


Plate 2
F08
CD170 (Siglec-5)
1A5
Mouse IgG1, κ
352004


Plate 2
F09
CD172a (SIRPa)
SE5A5
Mouse IgG1, κ
323806


Plate 2
F10
CD172b (SIRPb)
B4B6
Mouse IgG1, κ
323906


Plate 2
F11
CD172g (SIRPg)
LSB2.20
Mouse IgG1, κ
336606


Plate 2
F12
CD178 (Fas-L)
NOK-1
Mouse IgG1, κ
306407


Plate 2
G01
CD179a
HSL96
Mouse IgG1, κ
347404


Plate 2
G02
CD179b
HSL11
Mouse IgG1, κ
349804


Plate 2
G03
CD180 (RP105)
MHR73-11
Mouse IgG1, κ
312906


Plate 2
G04
CD181 (CXCR1)
8F1/CXCR1
Mouse IgG2b, κ
320608


Plate 2
G05
CD182 (CXCR2)
5E8/CXCR2
Mouse IgG1, κ
320706


Plate 2
G06
CD183
G025H7
Mouse IgG1, κ
353706


Plate 2
G07
CD184 (CXCR4)
12G5
Mouse IgG2a, κ
306506


Plate 2
G08
CD193 (CCR3)
500000000
Mouse IgG2b, κ
310706


Plate 2
G09
CD195 (CCR5)
T21/8
Mouse IgG1, κ
321406


Plate 2
G10
CD196
G034E3
Mouse IgG2b, κ
353410


Plate 2
G11
CD197 (CCR7)
G043H7
Mouse IgG2a, κ
353204


Plate 2
G12
CD200 (OX2)
OX-104
Mouse IgG1, κ
329206


Plate 2
H01
CD200 R
OX-108
Mouse IgG1, κ
329306


Plate 2
H02
CD201 (EPCR)
RCR-401
Rat IgG1, κ
351904


Plate 2
H03
CD202b (Tie2/Tek)
33.1 (Ab33)
Mouse IgG1, κ
334206


Plate 2
H04
CD203c (E-NPP3)
NP4D6
Mouse IgG1, κ
324606


Plate 2
H05
CD205 (DEC- 205)
HD30
Mouse IgG1, κ
342204


Plate 2
H06
CD206 (MMR)
15-2
Mouse IgG1, κ
321106


Plate 2
H07
CD207 (Langerin)
10E2
Mouse IgG1, κ
352204


Plate 2
H08
CD209 (DC- SIGN)
9E9A8
Mouse IgG2a, κ
330106


Plate 2
H09
CD210 (IL- 10 R)
3F9
Rat IgG2a, κ
308804


Plate 2
H10
CD213a2
SHM38
Mouse IgG1, κ
354404


Plate 2
H11
CD215 (IL- 15Rα)
JM7A4
Mouse IgG2b, κ
330208


Plate 2
H12
CD218a (IL-18Rα)
H44
Mouse IgG1, κ
313808


Plate 3
A01
Blank


Plate 3
A02
CD220
B6.220
Mouse IgG2b,
352604


Plate 3
A03
CD221 (IGF-1R)
1H7/CD221
Mouse IgG1,
351806


Plate 3
A04
CD226 (DNAM-1)
11A8
Mouse IgG1,
338306


Plate 3
A05
CD229 (Ly-9)
HLy-9.1.25
Mouse IgG1,
326108


Plate 3
A06
CD231 (TALLA)
SN1a (M3-3D9)
Mouse IgG1,
329406


Plate 3
A07
CD235ab
HIR2
Mouse IgG2b,
306604


Plate 3
A08
CD243
UIC2
Mouse IgG2a,
348606


Plate 3
A09
CD244 (2B4)
C1.7
Mouse IgG1,
329508


Plate 3
A10
CD245 (p220/240)
DY12
Mouse IgG1,
Inquire


Plate 3
A11
CD252 (OX40L)
11C3.1
Mouse IgG1,
326308


Plate 3
A12
CD253 (Trail)
RIK-2
Mouse IgG1,
308206


Plate 3
B01
CD254
MIH24
Mouse IgG1,
347504


Plate 3
B02
CD255 (TWEAK)
CARL-1
Mouse IgG3,
308305


Plate 3
B03
CD257 (BAFF, BLYS)
T7-241
Mouse IgG1,
318606


Plate 3
B04
CD258 (LIGHT)
T5-39
Mouse IgG2a,
318706


Plate 3
B05
CD261 (DR4, TRAIL-R1)
DJR1
Mouse IgG1,
307206


Plate 3
B06
CD262 (DR5, TRAIL-R2)
DJR2-4 (7-8)
Mouse IgG1,
307406


Plate 3
B07
CD263 (DcR1, TRAIL-R3)
DJR3
Mouse IgG1,
307006


Plate 3
B08
CD266 (Fn14)
ITEM-1
Mouse IgG1,
314004


Plate 3
B09
CD267 (TACI)
1A1
Rat IgG2a,
311906


Plate 3
B10
CD268 (BAFF-R, BAFFR)
11C1
Mouse IgG1,
316906


Plate 3
B11
CD270 (HVEM)
122
Mouse IgG1,
318806


Plate 3
B12
CD271
ME20.4
Mouse IgG1,
345106


Plate 3
C01
CD273 (B7- DC, PD-L2)
24F.10C12
Mouse IgG2a,
329606


Plate 3
C02
CD274 (B7- H1, PD-L1)
29E.2A3
Mouse IgG2b,
329706


Plate 3
C03
CD275 (B7- H2, B7-RP1)
9F.8A4
Mouse IgG1,
329806


Plate 3
C04
CD276
MIH42
Mouse IgG1,
351004


Plate 3
C05
CD277
BT3.1
Mouse IgG1,
342704


Plate 3
C06
CD278 (ICOS)
C398.4A
Arm. Hamster IgG
313508


Plate 3
C07
CD279 (PD-1)
EH12.2H7
Mouse IgG1,
329906


Plate 3
C08
CD282 (TLR2)
TL2.1
Mouse IgG2a,
309708


Plate 3
C09
CD284 (TLR4)
HTA125
Mouse IgG2a,
312806


Plate 3
C10
CD286 (TLR6)
TLR6.127
Mouse IgG1,
334708


Plate 3
C11
CD290
3C10C5
Mouse IgG1,
354604


Plate 3
C12
CD294
BM16
Rat IgG2a,
350106


Plate 3
D01
CD298
LNH-94
Mouse IgG1,
341704


Plate 3
D02
CD300e (IREM-2)
UP-H2
Mouse IgG1,
339704


Plate 3
D03
CD300F
UP-D2
Mouse IgG1,
340604


Plate 3
D04
CD301
H037G3
Mouse IgG2a,
354704


Plate 3
D05
CD303
201A
Mouse IgG2a,
354204


Plate 3
D06
CD304
12C2
Mouse IgG2a,
354504


Plate 3
D07
CD307
509f6
Mouse IgG2a,
340304


Plate 3
D08
CD307d (FcRL4)
413D12
Mouse IgG2b,
340204


Plate 3
D09
CD314 (NKG2D)
1D11
Mouse IgG1,
320806


Plate 3
D10
CD317
RS38E
Mouse IgG1,
348406


Plate 3
D11
CD318 (CDCP1)
CUB1
Mouse IgG2b,
324006


Plate 3
D12
CD319 (CRACC)
162.1
Mouse IgG2b,
331806


Plate 3
E01
CD324 (E-Cadherin)
67A4
Mouse IgG1,
324106


Plate 3
E02
CD325
8C11
Mouse IgG1,
350805


Plate 3
E03
CD326 (Ep- CAM)
9C4
Mouse IgG2b,
324206


Plate 3
E04
CD328 (Siglec-7)
6-434
Mouse IgG1,
339204


Plate 3
E05
CD334 (FGFR4)
4FR6D3
Mouse IgG1,
324306


Plate 3
E06
CD335 (NKp46)
9.00E+02
Mouse IgG1,
331908


Plate 3
E07
CD336 (NKp44)
P44-8
Mouse IgG1,
325108


Plate 3
E08
CD337 (NKp30)
P30-15
Mouse IgG1,
325208


Plate 3
E09
CD338 (ABCG2)
5D3
Mouse IgG2b,
332008


Plate 3
E10
CD340 (erbB2/HER-2)
24D2
Mouse IgG1,
324406


Plate 3
E11
CD344 (Frizzled-4)
CH3A4A7
Mouse IgG1,
326606


Plate 3
E12
CD351
TX61
Mouse IgG1,
137306


Plate 3
F01
CD352 (NTB-A)
NT-7
Mouse IgG1,
317208


Plate 3
F02
CD354 (TREM-1)
TREM-26
Mouse IgG1,
314906


Plate 3
F03
CD355 (CRTAM)
Cr24.1
Mouse IgG2a,
339106


Plate 3
F04
CD357 (GITR)
621
Mouse IgG1,
311604


Plate 3
F05
CD360 (IL- 21R)
2G1-K12
Mouse IgG1,
347806


Plate 3
F06
_2- micro- globulin
2M2
Mouse IgG1,
316306


Plate 3
F07
BTLA
MIH26
Mouse IgG2a,
344506


Plate 3
F08
C3AR
hC3aRZ8
Mouse IgG2b
345804


Plate 3
F09
C5L2
1D9-M12
Mouse IgG2a,
342404


Plate 3
F10
CCR10
May-88
Arm. hamster IgG
341504


Plate 3
F11
CLEC12A
50C1
Mouse IgG2a,
353604


Plate 3
F12
CLEC9A
8F9
Mouse IgG2a,
353804


Plate 3
G01
CX3CR1
2A9-1
Rat IgG2b,
341604


Plate 3
G02
CXCR7
8F11-M16
Mouse IgG2b,
331104


Plate 3
G03
_-Opioid Receptor
DOR7D2A4
Mouse IgG2b,
327206


Plate 3
G04
DLL1
MHD1-314
Mouse IgG1,
346404


Plate 3
G05
DLL4
MHD4-46
Mouse IgG1,
346506


Plate 3
G06
DR3 (TRAMP)
JD3
Mouse IgG1,
307106


Plate 3
G07
EGFR
AY13
Mouse IgG1,
352904


Plate 3
G08
erbB3/HER-3
1B4C3
Mouse IgG2a,
324706


Plate 3
G09
Fc_Rl
AER-37 (CRA-1)
Mouse IgG2b,
334610


Plate 3
G10
FcRL6
2H3
Mouse IgG2b,
Inquire


Plate 3
G11
Galectin-9
9M1-3
Mouse IgG1,
348906


Plate 3
G12
GARP (LRRC32)
7B11
Mouse IgG2b,
352504


Plate 3
H01
HLA-A, B, C
W6/32
Mouse IgG2a,
311406


Plate 3
H02
HLA-A2
BB7.2
Mouse IgG2b,
343306


Plate 3
H03
HLA-DQ
HLADQ1
Mouse IgG1,
318106


Plate 3
H04
HLA-DR
L243
Mouse IgG2a,
307606


Plate 3
H05
HLA-E
3D12
Mouse IgG1,
342604


Plate 3
H06
HLA-G
87G
Mouse IgG2a,
335906


Plate 3
H07
IFN-g R b chain
2HUB-159
Hamster IgG
308504


Plate 3
H08
Ig light chain k
MHK-49
Mouse IgG1,
316508


Plate 3
H09
Ig light chain
MHL-38
Mouse IgG2a,
316608


Plate 3
H10
IgD
IA6-2
Mouse IgG2a,
348204


Plate 3
H11
IgM
MHM-88
Mouse IgG1,
314508


Plate 3
H12
IL-28RA
MHLICR2a
Mouse IgG2a,
337804


Plate 4
A01
Blank


Plate 4
A02
Integrin α9β1
Y9A2
Mouse IgG1, κ
351606


Plate 4
A03
integrin β5
AST-3T
Mouse IgG2a, κ
345204


Plate 4
A04
integrin β7
FIB504
Rat IgG2a, κ
321204


Plate 4
A05
Jagged 2
MHJ2-523
Mouse IgG1, κ
346904


Plate 4
A06
LAP
TW4-6H10
Mouse IgG1, κ
349704


Plate 4
A07
Lymphotoxin b Receptor
31G4D8
Mouse IgG2b, κ
322008


Plate 4
A08
Mac-2 (Ga- lectin-3)
Gal397
Mouse IgG1, κ
126705


Plate 4
A09
MAIR-II
TX45
Mouse IgG1, κ
334804


Plate 4
A10
MICA/MICB
6D4
Mouse IgG2a, κ
320906


Plate 4
A11
MSC (W3D5)
W3D5
Mouse IgG2a, κ
327506


Plate 4
A12
MSC (W5C5)
W5C5
Mouse IgG1, κ
327406


Plate 4
B01
MSC (W7C6)
W7C6
Mouse IgG1, κ
327606


Plate 4
B02
MSC and NPC (W4A5)
W4A5
Mouse IgG1, κ
330806


Plate 4
B03
MSCA-1 (MSC, W8B2)
W8B2
Mouse IgG1, κ
327306


Plate 4
B04
NKp80
5D12
Mouse IgG1, κ
346706


Plate 4
B05
Notch 1
MHN1-519
Mouse IgG1, κ
352106


Plate 4
B06
Notch 2
MHN2-25
Mouse IgG2a, κ
348304


Plate 4
B07
Notch 3
MHN3-21
Mouse IgG1, κ
345406


Plate 4
B08
Notch 4
MHN4-2
Mouse IgG1, κ
349004


Plate 4
B09
NPC (57D2)
57D2
Mouse IgG1, κ
327706


Plate 4
B10
Podoplanin
NC-08
Rat IgG2a, λ
337004


Plate 4
B11
Pre-BCR
HSL2
Mouse IgG1, κ
347904


Plate 4
B12
PSMA
LNI-17
Mouse IgG1, κ
342504


Plate 4
C01
Siglec-10
5G6
Mouse IgG1, κ
347604


Plate 4
C02
Siglec-8
7C9
Mouse IgG1, κ
347104


Plate 4
C03
Siglec-9
K8
Mouse IgG1, κ
351504


Plate 4
C04
SSEA-1
MC-480
Mouse IgM, κ
125606


Plate 4
C05
SSEA-3
MC-631
Rat IgM, κ
330312


Plate 4
C06
SSEA-4
MC-813-70
Mouse IgG3, κ
330406


Plate 4
C07
SSEA-5
8E+11
Mouse IgG1, κ
355204


Plate 4
C08
TCR g/d
B1
Mouse IgG1, κ
331210


Plate 4
C09
TCR Vβ13.2
H132
Mouse IgG1, κ
333108


Plate 4
C10
TCR Vβ23
αHUT7
Mouse IgG1, κ
349406


Plate 4
C11
TCR Vβ8
JR2 (JR.2)
Mouse IgG2b, κ
348104


Plate 4
C12
TCR Vβ9
MKB1
Mouse IgG2b, κ
349204


Plate 4
D01
TCR Vδ2
B6
Mouse IgG1, κ
331408


Plate 4
D02
TCR Vg9
B3
Mouse IgG1, κ
331308


Plate 4
D03
TCR Vα24- Jα18
6B11
Mouse IgG1, κ
342904


Plate 4
D04
TCR Vα7.2
3C10
Mouse IgG1, κ
351706


Plate 4
D05
TCR α/β
IP26
Mouse IgG1, κ
306708


Plate 4
D06
Tim-1
1D12
Mouse IgG1, κ
353904


Plate 4
D07
Tim-3
F38-2E2
Mouse IgG1, κ
345006


Plate 4
D08
Tim-4
9F4
Mouse IgG1, κ
354004


Plate 4
D09
TLT-2
MIH61
Mouse IgG1, κ
351104


Plate 4
D10
TRA-1-60-R
TRA-1-60-R
Mouse IgM, κ
330610


Plate 4
D11
TRA-1-81
TRA-1-81
Mouse IgM, κ
330708


Plate 4
D12
TSLPR (TSLP-R)
1B4
Mouse IgG1, κ
322806


Plate 4
E01
Ms IgG1, κ ITCL
MOPC-21
Mouse IgG1, κ
400112


Plate 4
E02
Ms IgG2a, κ ITCL
MOPC-173
Mouse IgG2a, κ
400212


Plate 4
E03
Ms IgG2b, κ ITCL
MPC-11
Mouse IgG2b, κ
400314


Plate 4
E04
Ms IgG3, κ ITCL
MG3-35
Mouse IgG3, κ
401320


Plate 4
E05
Ms IgM, κ ITCL
MM-30
Mouse IgM, κ
401609


Plate 4
E06
Rat IgG1, κ ITCL
RTK2071
Rat IgG1, κ
400408


Plate 4
E07
Rat IgG2a, κ ITCL
RTK2758
Rat IgG2a, κ
400508


Plate 4
E08
Rat IgG2b, κ ITCL
RTK4530
Rat IgG2b, κ
400636


Plate 4
E09
Rat IgM, κ ITCL
RTK2118
Rat IgM, κ
400808


Plate 4
E10
AH IgG, ITCL
HTK888
Arm. Hamster IgG
400907


Plate 4
E11
CD177
MEM-166
mouse IgG1, k
315806


Plate 4
E12
IL1RAP
89412
mouse IgG1, k
FAB676P










Screen Iteration 70007











Plate
WELL ID
Specificity
Clone
Isotype





Plate 1
A01
Blank


Plate 1
A02
IgG Isotype Ctrl
HTK888
Arm. Hamster IgG


Plate 1
A03
CCR10
6588-5
Arm. Hamster IgG


Plate 1
A04
CD278
C398.4A
Arm. Hamster IgG


Plate 1
A05
IFN-γ R b chain
2HUB-159
Hamster IgG


Plate 1
A06
IgG1, κ Isotype Ctrl
MOPC-21
Mouse IgG1, k


Plate 1
A07
CD46
TRA-2-10
Mouse IgG1


Plate 1
A08
CD70
113-16
Mouse IgG1


Plate 1
A09
CD1a
HI149
mouse IgG1, k


Plate 1
A10
CD2
RPA-2.10
mouse IgG1, k


Plate 1
A11
β2-microglobulin
2M2
mouse IgG1, k


Plate 1
A12
B7-H4
MIH43
mouse IgG1, k


Plate 1
B01
Cadherin 11
16G5
mouse IgG1, k


Plate 1
B02
CD10
HI10a
mouse IgG1, k


Plate 1
B03
CD100
A8
mouse IgG1, k


Plate 1
B04
CD103
Ber-ACT8
mouse IgG1, k


Plate 1
B05
CD105 (Endoglin)
SN6h
mouse IgG1, k


Plate 1
B06
CD106
STA
mouse IgG1, k


Plate 1
B07
CD107a
H4A3
mouse IgG1, k


Plate 1
B08
CD107b
H4B4
mouse IgG1, k


Plate 1
B09
CD109
W7C5
mouse IgG1, k


Plate 1
B10
CD111
R1.302
mouse IgG1, k


Plate 1
B11
CD112
TX31
mouse IgG1, k


Plate 1
B12
CD114
LMM741
mouse IgG1, k


Plate 1
C01
CD116
4H1
mouse IgG1, k


Plate 1
C02
CD117
104D2
mouse IgG1, k


Plate 1
C03
CD119
GIR-208
mouse IgG1, k


Plate 1
C04
CD11a
HI111
mouse IgG1, k


Plate 1
C05
CD11b
ICRF44
mouse IgG1, k


Plate 1
C06
CD122
TU27
mouse IgG1, k


Plate 1
C07
CD123
6H6
mouse IgG1, k


Plate 1
C08
CD126
UV4
mouse IgG1, k


Plate 1
C09
CD127
A019D5
mouse IgG1, k


Plate 1
C10
CD13
WM15
mouse IgG1, k


Plate 1
C11
CD131
1C1
mouse IgG1, k


Plate 1
C12
CD134
Ber-ACT35 (ACT35)
mouse IgG1, k


Plate 1
D01
CD135
BV10A4H2
mouse IgG1, k


Plate 1
D02
CD137
4B4-1
mouse IgG1, k


Plate 1
D03
4-1BB Ligand
5F4
mouse IgG1, k


Plate 1
D04
CD138
MI15
mouse IgG1, k


Plate 1
D05
CD14
63D3
mouse IgG1, k


Plate 1
D06
CD140a
16A1
mouse IgG1, k


Plate 1
D07
CD140b
18A2
mouse IgG1, k


Plate 1
D08
CD141
M80
mouse IgG1, k


Plate 1
D09
CD142
NY2
mouse IgG1, k


Plate 1
D10
CD143
5-369
mouse IgG1, k


Plate 1
D11
CD146
P1H12
mouse IgG1, k


Plate 1
D12
CD148
A3
mouse IgG1, k


Plate 1
E01
CD15
W6D3
mouse IgG1, k


Plate 1
E02
CD150
A12 (7D4)
mouse IgG1, k


Plate 1
E03
CD151
50-6
mouse IgG1, k


Plate 1
E04
CD154
24-31
mouse IgG1, k


Plate 1
E05
CD156c
SHM14
mouse IgG1, k


Plate 1
E06
CD158e1
DX9
mouse IgG1, k


Plate 1
E07
CD16
3G8
mouse IgG1, k


Plate 1
E08
CD161
HP-3G10
mouse IgG1, k


Plate 1
E09
CD162
KPL-1
mouse IgG1, k


Plate 1
E10
CD163
GHI/61
mouse IgG1, k


Plate 1
E11
CD164
67D2
mouse IgG1, k


Plate 1
E12
CD165
SN2 (N6- D11)
mouse IgG1, k


Plate 1
F01
CD166
3A6
mouse IgG1, k


Plate 1
F02
CD169
7-239
mouse IgG1, k


Plate 1
F03
CD170
1A5
mouse IgG1, k


Plate 1
F04
CD172a/b (SIRPα/β)
SE5A5
mouse IgG1, k


Plate 1
F05
CD172g (SIRPy)
LSB2.20
mouse IgG1, k


Plate 1
F06
CD178
NOK-1
mouse IgG1, k


Plate 1
F07
CD179a
HSL96
mouse IgG1, k


Plate 1
F08
CD179b
HSL11
mouse IgG1, k


Plate 1
F09
CD18
TS1/18
mouse IgG1, k


Plate 1
F10
CD180
MHR73-11
mouse IgG1, k


Plate 1
F11
CD182
5E8/CXCR2
mouse IgG1, k


Plate 1
F12
CD183
G025H7
mouse IgG1, k


Plate 1
G01
CD185
J252D4
mouse IgG1, k


Plate 1
G02
CD19
HIB19
mouse IgG1, k


Plate 1
G03
CD191
5F10B29
mouse IgG1, k


Plate 1
G04
CD194
L291H4
mouse IgG1, k


Plate 1
G05
CD1b
SN13 (K5- 1B8)
mouse IgG1, k


Plate 1
G06
CD1c
L161
mouse IgG1, k


Plate 1
G07
CD200
OX-104
mouse IgG1, k


Plate 1
G08
CD200R
OX-108
mouse IgG1, k


Plate 1
G09
CD202b
33.1 (Ab33)
mouse IgG1, k


Plate 1
G10
CD203c
NP4D6
mouse IgG1, k


Plate 1
G11
CD205
HD83
mouse IgG1, k


Plate 1
G12
CD206
15-2
mouse IgG1, k


Plate 1
H01
CD207
1E3
mouse IgG1, k


Plate 1
H02
CD21
Bu32
mouse IgG1, k


Plate 1
H03
CD213a1
SS12B
mouse IgG1, k


Plate 1
H04
CD213a2
SHM38
mouse IgG1, k


Plate 1
H05
CD218a
H44
mouse IgG1, k


Plate 1
H06
CD221
1H7/CD221
mouse IgG1, k


Plate 1
H07
CD223 (LAG-3)
11C3C65
mouse IgG1, k


Plate 1
H08
CD226
11A8
mouse IgG1, k


Plate 1
H09
CD227
16A
mouse IgG1, k


Plate 1
H10
CD229
HLy-9.1.25
mouse IgG1, k


Plate 1
H11
CD23
EBVCS-5
mouse IgG1, k


Plate 1
H12
CD231
SN1a (M3- 3D9)
mouse IgG1, k


Plate 2
A01
Blank


Plate 2
A02
CD244 (2B4)
C1.7
mouse IgG1, k


Plate 2
A03
CD245
DY12
mouse IgG1, k


Plate 2
A04
CD25
M-A251
mouse IgG1, k


Plate 2
A05
CD252
11C3.1
mouse IgG1, k


Plate 2
A06
CD261
DJR1
mouse IgG1, k


Plate 2
A07
CD262
DJR2-4 (7-8)
mouse IgG1, k


Plate 2
A08
CD263
DJR3
mouse IgG1, k


Plate 2
A09
CD266
ITEM-1
mouse IgG1, k


Plate 2
A10
CD268
11C1
mouse IgG1, k


Plate 2
A11
CD27
M-T271
mouse IgG1, k


Plate 2
A12
CD271
ME20.4
mouse IgG1, k


Plate 2
B01
CD275
9F.8A4
mouse IgG1, k


Plate 2
B02
CD276
MIH42
mouse IgG1, k


Plate 2
B03
CD277
BT3.1
mouse IgG1, k


Plate 2
B04
CD279
EH12.2H7
mouse IgG1, k


Plate 2
B05
CD28
CD28.2
mouse IgG1, k


Plate 2
B06
CD29
TS2/16
mouse IgG1, k


Plate 2
B07
CD290
3C10C5
mouse IgG1, k


Plate 2
B08
CD298
LNH-94
mouse IgG1, k


Plate 2
B09
CD3
UCHT1
mouse IgG1, k


Plate 2
B10
CD30
BY88
mouse IgG1, k


Plate 2
B11
CD300c
TX45
mouse IgG1, k


Plate 2
B12
CD309
7D4-6
mouse IgG1, k


Plate 2
C01
CD31
WM59
mouse IgG1, k


Plate 2
C02
CD314
1D11
mouse IgG1, k


Plate 2
C03
CD317
RS38E
mouse IgG1, k


Plate 2
C04
CD324
67A4
mouse IgG1, k


Plate 2
C05
CD325
8C11
mouse IgG1, k


Plate 2
C06
CD328
6-434
mouse IgG1, k


Plate 2
C07
CD33
WM53
mouse IgG1, k


Plate 2
C08
CD334
4FR6D3
mouse IgG1, k


Plate 2
C09
CD335
9E2
mouse IgG1, k


Plate 2
C10
CD336
P44-8
mouse IgG1, k


Plate 2
C11
CD337
P30-15
mouse IgG1, k


Plate 2
C12
CD34
581
mouse IgG1, k


Plate 2
D01
CD340
24D2
mouse IgG1, k


Plate 2
D02
CD344
CH3A4A7
mouse IgG1, k


Plate 2
D03
CD35
E11
mouse IgG1, k


Plate 2
D04
CD354
TREM-26
mouse IgG1, k


Plate 2
D05
CD360
17A12
mouse IgG1, k


Plate 2
D06
CD365
1D12
mouse IgG1, k


Plate 2
D07
CD366
F38-2E2
mouse IgG1, k


Plate 2
D08
CLEC4A
9E8
mouse IgG1, k


Plate 2
D09
CD36L1
m1b9
mouse IgG1, k


Plate 2
D10
CD38
HIT2
mouse IgG1, k


Plate 2
D11
CD39
A1
mouse IgG1, k


Plate 2
D12
CD4
RPA-T4
mouse IgG1, k


Plate 2
E01
CD40
5C3
mouse IgG1, k


Plate 2
E02
CD41
HIP8
mouse IgG1, k


Plate 2
E03
CD42b
HIP1
mouse IgG1, k


Plate 2
E04
CD43
CD43-10G7
mouse IgG1, k


Plate 2
E05
CD44
BJ18
mouse IgG1, k


Plate 2
E06
CD45
HI30
mouse IgG1, k


Plate 2
E07
CD47
CC2C6
mouse IgG1, k


Plate 2
E08
CD48
BJ40
mouse IgG1, k


Plate 2
E09
CD49a
TS2/7
mouse IgG1, k


Plate 2
E10
CD49b
P1E6-C5
mouse IgG1, k


Plate 2
E11
CD49c
ASC-1
mouse IgG1, k


Plate 2
E12
CD49d
9F10
mouse IgG1, k


Plate 2
F01
CD5
UCHT2
mouse IgG1, k


Plate 2
F02
CD50
CBR-IC3/1
mouse IgG1, k


Plate 2
F03
CD54
HA58
mouse IgG1, k


Plate 2
F04
CD55
JS11
mouse IgG1, k


Plate 2
F05
CD56 (NCAM)
5.1H11
mouse IgG1, k


Plate 2
F06
CD58
TS2/9
mouse IgG1, k


Plate 2
F07
CD6
BL-CD6
mouse IgG1, k


Plate 2
F08
CD61
VI-PL2
mouse IgG1, k


Plate 2
F09
CD62E
HAE-1f
mouse IgG1, k


Plate 2
F10
CD62L
DREG-56
mouse IgG1, k


Plate 2
F11
CD62P
AK4
mouse IgG1, k


Plate 2
F12
CD63
H5C6
mouse IgG1, k


Plate 2
G01
CD64
10.1
mouse IgG1, k


Plate 2
G02
CD69
FN50
mouse IgG1, k


Plate 2
G03
CD73
AD2
mouse IgG1, k


Plate 2
G04
CD74
LN2
mouse IgG1, k


Plate 2
G05
CD79b
CB3-1
mouse IgG1, k


Plate 2
G06
CD8a
SK1
mouse IgG1, k


Plate 2
G07
CD80
2D10
mouse IgG1, k


Plate 2
G08
CD81
5A6
mouse IgG1, k


Plate 2
G09
CD82
ASL-24
mouse IgG1, k


Plate 2
G10
CD83
HB15e
mouse IgG1, k


Plate 2
G11
CD85
17G10.2
mouse IgG1, k


Plate 2
G12
CD85k
ZM4.1
mouse IgG1, k


Plate 2
H01
CD87
VIM5
mouse IgG1, k


Plate 2
H02
CD89
A59
mouse IgG1, k


Plate 2
H03
CD8a
RPA-T8
mouse IgG1, k


Plate 2
H04
CD9
HI9a
mouse IgG1, k


Plate 2
H05
CD90
5E10
mouse IgG1, k


Plate 2
H06
CD93
VIMD2
mouse IgG1, k


Plate 2
H07
CD94
DX22
mouse IgG1, k


Plate 2
H08
CD95
DX2
mouse IgG1, k


Plate 2
H09
CD96
NK92.39
mouse IgG1, k


Plate 2
H10
CD97
VIM3b
mouse IgG1, k


Plate 2
H11
CD99
3B2/TA8
mouse IgG1, k


Plate 2
H12
CXCL16
22-19-12
mouse IgG1, k


Plate 3
A01
Blank


Plate 3
A02
DLL1
MHD1-314
mouse IgG1, k


Plate 3
A03
DLL4
MHD4-46
mouse IgG1, k


Plate 3
A04
DR3
JD3
mouse IgG1, k


Plate 3
A05
EGFR
AY13
mouse IgG1, k


Plate 3
A06
CD357
108-17
mouse IgG1, k


Plate 3
A07
GPR19
K152D10
mouse IgG1, k


Plate 3
A08
GPR56
CG4
mouse IgG1, k


Plate 3
A09
HLA-E
3D12
mouse IgG1, k


Plate 3
A10
HVEM
122
mouse IgG1, k


Plate 3
A11
Ig light chain κ
MHK-49
mouse IgG1, k


Plate 3
A12
IgM
MHM-88
mouse IgG1, k


Plate 3
B01
CD360
2G1-K12
mouse IgG1, k


Plate 3
B02
Integrin α9β1
Y9A2
mouse IgG1, k


Plate 3
B03
Jagged 2
MHJ2-523
mouse IgG1, k


Plate 3
B04
Ksp37
TDA3
mouse IgG1, k


Plate 3
B05
LAP
TW4-2F8
mouse IgG1, k


Plate 3
B06
LY6G6D
13.8
mouse IgG1, k


Plate 3
B07
MERTK
590H11G1E3
mouse IgG1, k


Plate 3
B08
MSC
W7C6
mouse IgG1, k


Plate 3
B09
MSC, NPC
W4A5
mouse IgG1, k


Plate 3
B10
TNAP
W8B2
mouse IgG1, k


Plate 3
B11
MUC-13
TCC16
mouse IgG1, k


Plate 3
B12
NKp80
5D12
mouse IgG1, k


Plate 3
C01
Notch 1
MHN1-519
mouse IgG1, k


Plate 3
C02
Notch3
MHN3-21
mouse IgG1, k


Plate 3
C03
Notch 4
MHN4-2
mouse IgG1, k


Plate 3
C04
NPC
57D2
mouse IgG1, k


Plate 3
C05
CD352
NT-7
mouse IgG1, k


Plate 3
C06
PSMA
LNI-17
mouse IgG1, k


Plate 3
C07
ROR1
2A2
mouse IgG1, k


Plate 3
C08
Siglec-10
5G6
mouse IgG1, k


Plate 3
C09
CD328
S7.7
mouse IgG1, k


Plate 3
C10
Siglec-8
7C9
mouse IgG1, k


Plate 3
C11
Siglec-9
K8
mouse IgG1, k


Plate 3
C12
SSEA-5
8E11
mouse IgG1, k


Plate 3
D01
SUSD2
W5C5
mouse IgG1, k


Plate 3
D02
TCR α/β
IP26
mouse IgG1, k


Plate 3
D03
TCR γ/δ
B1
mouse IgG1, k


Plate 3
D04
Tim-4
9F4
mouse IgG1, k


Plate 3
D05
TLT-2
MIH61
mouse IgG1, k


Plate 3
D06
TM4SF20
C9
mouse IgG1, k


Plate 3
D07
TRA-2-49
TRA-2- 49/6E
mouse IgG1, k


Plate 3
D08
TRA-2-54
TRA-2-54/2J
mouse IgG1, k


Plate 3
D09
TSLPR
1B4
mouse IgG1, k


Plate 3
D10
VEGFR-3
9D9F9
mouse IgG1, k


Plate 3
D11
IgG2a, κ Isotype Ctrl
MOPC-173
mouse IgG2a, k


Plate 3
D12
APCDD1
7.13
mouse IgG2a, k


Plate 3
E01
CD272
MIH26
mouse IgG2a, k


Plate 3
E02
CD198
L263G8
mouse IgG2a, k


Plate 3
E03
CCRL2
K097F7
mouse IgG2a, k


Plate 3
E04
CD102
CBR-IC2/2
mouse IgG2a, k


Plate 3
E05
CD104
58XB4
mouse IgG2a, k


Plate 3
E06
CD124
G077F6
mouse IgG2a, k


Plate 3
E07
CD130
2E1B02
mouse IgG2a, k


Plate 3
E08
CD144
BV9
mouse IgG2a, k


Plate 3
E09
CD152 (CTLA-4)
BNI3
mouse IgG2a, k


Plate 3
E10
CD155
TX24
mouse IgG2a, k


Plate 3
E11
CD158b
DX27
mouse IgG2a, k


Plate 3
E12
CD184
12G5
mouse IgG2a, k


Plate 3
F01
CD186
K041E5
mouse IgG2a, k


Plate 3
F02
CD192
K036C2
mouse IgG2a, k


Plate 3
F03
CD197
G043H7
mouse IgG2a, k


Plate 3
F04
CD199
L053E8
mouse IgG2a, k


Plate 3
F05
CD209
9E9A8
mouse IgG2a, k


Plate 3
F06
CD217
W15177A
mouse IgG2a, k


Plate 3
F07
CD230 (Prion)
3F4
mouse IgG2a, k


Plate 3
F08
CD24
ML5
mouse IgG2a, k


Plate 3
F09
CD243
UIC2
mouse IgG2a, k


Plate 3
F10
CD26
BA5b
mouse IgG2a, k


Plate 3
F11
CD269
19F2
mouse IgG2a, k


Plate 3
F12
CD282
TL2.1
mouse IgG2a, k


Plate 3
G01
CD284
HTA125
mouse IgG2a, k


Plate 3
G02
CD301
H037G3
mouse IgG2a, k


Plate 3
G03
CD303
201A
mouse IgG2a, k


Plate 3
G04
CD304
12C2
mouse IgG2a, k


Plate 3
G05
CD307e
509f6
mouse IgG2a, k


Plate 3
G06
CD323
SHM33
mouse IgG2a, k


Plate 3
G07
CD357
108-17
mouse IgG2a, k


Plate 3
G08
CD36
5-271
mouse IgG2a, k


Plate 3
G09
CD369
15E2
mouse IgG2a, k


Plate 3
G10
CD370
8F9
mouse IgG2a, k


Plate 3
G11
CD371
50C1
mouse IgG2a, k


Plate 3
G12
CD45RO
UCHL1
mouse IgG2a, k


Plate 3
H01
CD51
NKI-M9
mouse IgG2a, k


Plate 3
H02
CD59
p282 (H19)
mouse IgG2a, k


Plate 3
H03
CD7
CD7-6B7
mouse IgG2a, k


Plate 3
H04
CD71
CY1G4
mouse IgG2a, k


Plate 3
H05
CD84
CD84.1.21
mouse IgG2a, k


Plate 3
H06
CD88
S5/1
mouse IgG2a, k


Plate 3
H07
CD355
Cr24.1
mouse IgG2a, k


Plate 3
H08
erbB3
1B4C3
mouse IgG2a, k


Plate 3
H09
FPR3
K102B9
mouse IgG2a, k


Plate 3
H10
Ganglioside GD2
14G2a
mouse IgG2a, k


Plate 3
H11
GPR83
K07JP05
mouse IgG2a, k


Plate 3
H12
HLA-A, B, C
W6/32
mouse IgG2a, k


Plate 4
A01
Blank


Plate 4
A02
HLA-DR
L243
mouse IgG2a, k


Plate 4
A03
Ig light chain λ
MHL-38
mouse IgG2a, k


Plate 4
A04
IgD
IA6-2
mouse IgG2a, k


Plate 4
A05
IL-28RA
MHLICR2a
mouse IgG2a, k


Plate 4
A06
integrin β5
AST-3T
mouse IgG2a, k


Plate 4
A07
KLRG1
SA231A2
mouse IgG2a, k


Plate 4
A08
LOX-1
15C4
mouse IgG2a, k


Plate 4
A09
MICA/MICB
6D4
mouse IgG2a, k


Plate 4
A10
SUSD2
W3D5
mouse IgG2a, k


Plate 4
A11
Notch 2
MHN2-25
mouse IgG2a, k


Plate 4
A12
TACSTD2
NY18
mouse IgG2a, k


Plate 4
B01
TIGIT (VSTM3)
A15153G
mouse IgG2a, k


Plate 4
B02
IgG2b, κ Isotype Ctrl
MPC-11
mouse IgG2b, k


Plate 4
B03
C3aR
hC3aRZ8
mouse IgG2b, k


Plate 4
B04
CCX-CKR (CCRL1)
13E11
mouse IgG2b, k


Plate 4
B05
CD110
S-HCL-3
mouse IgG2b, k


Plate 4
B06
CD129
AH9R7
mouse IgG2b, k


Plate 4
B07
CD158
HP-MA4
mouse IgG2b, k


Plate 4
B08
CD181
8F1/CXCR1
mouse IgG2b, k


Plate 4
B09
CD193
5E8
mouse IgG2b, k


Plate 4
B10
CD196
G034E3
mouse IgG2b, k


Plate 4
B11
CD1d
51.1
mouse IgG2b, k


Plate 4
B12
CD20
2H7
mouse IgG2b, k


Plate 4
C01
CD22
S-HCL-1
mouse IgG2b, k


Plate 4
C02
CD220
B6.220
mouse IgG2b, k


Plate 4
C03
CD235ab
HIR2
mouse IgG2b, k


Plate 4
C04
CD258
T5-39
mouse IgG2b, k


Plate 4
C05
CD274
29E.2A3
mouse IgG2b, k


Plate 4
C06
CD319
162.1
mouse IgG2b, k


Plate 4
C07
CD32
FUN-2
mouse IgG2b, k


Plate 4
C08
CD326
9C4
mouse IgG2b, k


Plate 4
C09
CD338
5D3
mouse IgG2b, k


Plate 4
C10
CD368
9B9
mouse IgG2b, k


Plate 4
C11
CD45RA
HI100
mouse IgG2b, k


Plate 4
C12
CD45RB
MEM-55
mouse IgG2b, k


Plate 4
D01
CD49e
NKI-SAM-1
mouse IgG2b, k


Plate 4
D02
CD52
HI186
mouse IgG2b, k


Plate 4
D03
CD66a/c/e
ASL-32
mouse IgG2b, k


Plate 4
D04
CD85h
24
mouse IgG2b, k


Plate 4
D05
CD85
GHI/75
mouse IgG2b, k


Plate 4
D06
CD86
IT2.2
mouse IgG2b, k


Plate 4
D07
CD92
VIM15b
mouse IgG2b, k


Plate 4
D08
CXCR7
8F11-M16
mouse IgG2b, k


Plate 4
D09
Delta Opioid Receptor
DOR7D2A4
mouse IgG2b, k


Plate 4
D10
Dopamine Receptor D1
L205G1
mouse IgG2b, k


Plate 4
D11
EphA2
SHM16
mouse IgG2b, k


Plate 4
D12
FcεRlα
AER-37 (CRA-1)
mouse IgG2b, k


Plate 4
E01
GARP
7B11
mouse IgG2b, k


Plate 4
E02
CD215
JM7A4
mouse IgG2b, k


Plate 4
E03
Lymphotoxin β Receptor
31G4D8
mouse IgG2b, k


Plate 4
E04
MRGX2
K125H4
mouse IgG2b, k


Plate 4
E05
TMEM8A
SA065C3
mouse IgG2b, k


Plate 4
E06
CD254
MIH24
mouse IgG2b, k


Plate 4
E07
CD318
CUB1
mouse IgG2b, k


Plate 4
E08
IgG3, k Isotype Ctrl
MG3-35
mouse IgG3, k


Plate 4
E09
CD255
CARL-1
Mouse IgG3, k


Plate 4
E10
SSEA-4
MC-813-70
Mouse IgG3, k


Plate 4
E11
IgM, κ Isotype Ctrl
MM-30
Mouse IgM, k


Plate 4
E12
Sialyl Lewis X (dimeric)
FH6
Mouse IgM, k


Plate 4
F01
TRA-1-81
TRA-1-81
Mouse IgM, k


Plate 4
F02
CD160
BY55
Mouse IgM, k


Plate 4
F03
CD57
HNK-1
Mouse IgM, k


Plate 4
F04
CD66b
G10F5
Mouse IgM, k


Plate 4
F05
TRA-1-60-R
TRA-1-60-R
Mouse IgM, k


Plate 4
F06
IgG1, κ Isotype Ctrl
RTK2071
Rat IgG1, k


Plate 4
F07
CD115
9-4D2-1E4
Rat IgG1, k


Plate 4
F08
CD201
RCR-401
Rat IgG1, k


Plate 4
F09
IgG2a, κ Isotype Ctrl
RTK2758
Rat IgG2a, k


Plate 4
F10
CD120b
3G7A02
Rat IgG2a, k


Plate 4
F11
CD210
3F9
Rat IgG2a, k


Plate 4
F12
CD267
1A1
Rat IgG2a, k


Plate 4
G01
CD294
BM16
Rat IgG2a, k


Plate 4
G02
CD49f
GoH3
Rat IgG2a, k


Plate 4
G03
CD85
MKT5.1
Rat IgG2a, k


Plate 4
G04
CD85d
42D1
Rat IgG2a, k


Plate 4
G05
IgG Fc
M1310G05
Rat IgG2a, k


Plate 4
G06
Integrin β7
FIB504
Rat IgG2a, k


Plate 4
G07
XCR1
S15046E
Rat IgG2a, k


Plate 4
G08
Podoplanin
NC-08
Rat IgG2a, l


Plate 4
G09
IgG2b, κ Isotype Ctrl
RTK4530
Rat IgG2b, k


Plate 4
G10
CD132
TUGh4
Rat IgG2b, k


Plate 4
G11
CD195
J418F1
Rat IgG2b, k


Plate 4
G12
CX3CR1
2A9-1
Rat IgG2b, k


Plate 4
H01
IgM, κ Isotype Ctrl
RTK2118
Rat IgM, k


Plate 4
H02
SSEA-3
MC-631
Rat IgM, k


Plate 4
H03
CD177
MEM-166
mouse IgG1, k


Plate 4
H04
IL1RAP
89412
mouse IgG1, k
















TABLE 3







Antibodies and Reagents

















Catalog


Name of Product
Target
Fluorochrome
Company
Isotype
#





LEGENDScreena
SCREEN
PE
BioLegend
10 different
700001


Human PE Kit


LEGENDScreena
SCREEN
PE
BioLegend
10 different
700007


Human PE Kit


PE/Cy7 anti-human CD3
CD3
PE-Cy7
BioLegend
mouse
300316


Antibody



IgG2a, k


APC/Cy7 anti-human
CD19
APC-Cy7
BioLegend
mouse
302218


CD19 Antibody



IgG1, k


Alexa Fluor{umlaut over ( )} 488 anti-
CD34
AF488
BioLegend
mouse
343518


human CD34 Antibody



IgG1, k


APC/Cy7 anti-human
CD34
APC-Cy7
BioLegend
mouse
343614


CD34 Antibody



IgG2a, k


BV421 Mouse Anti-
CD34
BV421
BD
mouse
562577


Human CD34



IgG1, k


BV711 Mouse Anti-
CD38
BV711
BD
mouse
563965


Human CD38 Clone



IgG1, k


HIT2 (RUO)


PE-Cy7 Mouse Anti-
CD45RA
PE-Cy7
BD
mouse
560675


Human CD45RA



IgG2b, k


Alexa Fluor 488 anti-
CD123
AF488
BioLegend
mouse
306036


human CD123 Antibody



IgG1, k


PerCP/Cyanine5.5 anti-
CD90
PerCP-Cy5.5
BioLegend
mouse
328117


human CD90 (Thy1)



IgG1, k


Antibody


APC anti-human CD45
CD45
APC
BioLegend
mouse
982304


Antibody



IgG1, k


PE anti-human CD177
CD177
PE
BioLegend
mouse
315806


Antibody



IgG1, k


Human IL-1 RAcP/IL-1
IL1RAP
PE
RnD
mouse
FAB676P


R3 PE-conjugated


Systems
IgG1, k


Antibody


PE anti-human CD62E
CD62E
PE
BioLegend
mouse
336008


Antibody



IgG1, k


PE anti-human CD54
CD54
PE
BioLegend
mouse
353105


Antibody



IgG1, k


PE anti-human CD244
CD244
PE
BioLegend
mouse
393507


(2B4) Antibody



IgG1, k


PE anti-human Jagged 2
Jagged
PE
BioLegend
mouse
346904


Antibody
2


IgG1, k


PE anti-human CD323
CD323
PE
BioLegend
mouse
356703


(JAM3) Antibody



IgG2a, k


PE anti-CD105
CD105
PE
BioLegend
mouse
800503


(Endoglin) Antibody



IgG1, k


PE anti-human CD369
CD369
PE
BioLegend
mouse
355403


(Dectin-1/CLEC7A)



IgG2a, k


Antibody


PE anti-human CD352
SLAMF6
PE
BioLegend
mouse
317207


(NTB-A) Antibody



IgG1, k


Anti-SLAMF6 antibody
SLAMF6

Abcam
mono IgG
ab238421


[EPR22170]



rabbit


PE Mouse IgG1, κ
ISO
PE
BioLegend
mouse
400112


Isotype Ctrl Antibody



IgG1, k


PE Mouse IgG2a, κ
ISO
PE
BioLegend
mouse
400214


Isotype Ctrl (FC)



IgG2a


Antibody


Rabbit IgG, monoclonal
ISO

Abcam
mono IgG
ab199376


[EPR25A]



rabbit


7-AAD Viability Staining
7AAD

BioLegend

420404


Solution


4′,6-Diamidine-2′-
DAPI

Sigma-

D9542


phenylindole


Aldrich


dihydrochloride
















TABLE 4







Top Cell Surface Marker Candidates



























Rank







NBM#1
NBM#2
NBM#3
AML#48
AML#83
AML#80
AML −
Rank
Mean


Rank
Specificity
Gene Symbol
MFI
MFI
MFI
MFI
MFI
MFI
NBM
AML/NBM
Rank





















1
CD123
IL3RA
86.7
1000
1059
2893
4057
1121
3
12
7.5


2
CD369
CLEC7A

1106
88.7

2650
1920
15
1
8


3
CD352
SLAMF6, NTB-A
153
689
627
1754
1495
3253
14
6
10


4
CD54
ICAM1
−427
1302
1134
633
4308
8953
2
18
10


5
CD105
ENG
320
1764
1154
1199
3678
6067
6
14
10


6
CD370
CLEC9A
33.3
653
1197
241
2025
4185
18
4
11


7
CD323
JAM3

1384
1067

3292
3345
7
24
15.5


8
CD93
CD93
140
1381
1906
1890
1784
5837
9
22
15.5


9
CD11c
ITGAX
−1528
988
550
−249
1206
3379
19
15
17


10
TRA-2-54
ALPL

692
465

4091
1686
26
8
17


11
IL1RAP
IL1RAP
−53.3
564
305
440
1818
1655
22
20
21


12
CD252
TNFSF4
−300

194
42.7
4159
1931
35
7
21


13
CD261
TNFRSF10A
−414
1130
434
−849
2609
2359
16
33
24.5


14
CD56
NCAM1
−80
587
302
19.4
3138
1165
49
13
31


15
CD49d
ITGA4
1295
2558
3163
2574
2495
7646
20
43
31.5


16
TLT-2
TRML2
−883
646
420
7.77
1621
1567
44
21
32.5


17
CD11b
ITGAM
−541
758
358
34.9
1784
1931
36
31
33.5


18
CD314
KLRK1
−501
1251
2231
−793
2911
4049
13
55
34


19
CD49c
ITGA3
−681
1244
1774
−2513
2442
3525
30
44
37


20
CD266
TNFRSF12A
−1309
641
64.2
73.8
694
602
77
5
41


21
CD165
ADAMTSL1
60
1448
1902
303
4281
2578
62
23
42.5


22
CD344
FZD4
−675
2283
1392
−745
3040
4116
56
41
48.5


23
Jagged 2
JAG2
−7099
2676
1561
637
4057
3785
5
93
49


24
CD7
CD7
−387
661
676
6668
1667
1183
41
58
49.5


25
CD156c
ADAM10
856
3189
4176
3403
4903
4352
12
88
50


26
CD354
TREM1
40
1280
567
562
1392
2168
81
27
54


27
CD94
KLRD1
−2968
383
516
−42.7
851
1136
71
38
54.5


28
CD263
TNFRSF10C
−340
1523
903
−233
2505
2262
43
69
56


29
CD124
IL4R
127
1310
1152
507
3145
1658
83
29
56


30
CD1d
CD1D
−795
1327
136
−813
2231
2609
47
67
57


31
CD144
CDH5
133
1106
1138
−350
1892
3641
54
60
57


32
Siglec-8
SIGLEC8
−2353
1478
59.1
−315
2099
1788
10
105
57.5


33
PSMA
FOLH1
−133
817
705
50.5
1532
1376
65
53
59


34
CD134
TNFSRF4
40
982
943
284
1529
1856
74
45
59.5


35
CD258
TNFSRF14
−73.3
1874
53.9
−483
2177
1518
119
3
61


36
TCR α/β
TRAC
−3013
1227
939
159
1670
1798
51
72
61.5


37
CD275
ICOSLG
80
1278
722
491
1822
1735
76
48
62


38
CD182
CXCR2
180
483
438
745
1002
−594
82
42
62


39
SSEA-5

−1099
1418
152
117
2095
2414
27
99
63


40
CD45RB
PTPRC
153
395
151
503
1957
500
109
17
63


41
Siglec-10
SIGLEC10
−748
1424
1290
−257
2266
3234
52
79
65.5


42
MICA/MICB
MICA/MICB
−534
1076
504
−3.88
1199
1337
79
52
65.5


43
IgD
IGHD
−2154
978
661
−11.6
1358

21
113
67


44
TCR γ/δ

−3825
1964
1332
−307
2975
2283
40
94
67


45
CD36L1
SCARB1

1072
974

1600
2128
80
54
67


46
TRA-2-49
ALPL

1405
884

1950
1758
75
64
69.5


47
CD269
TNFRSF17

614
406

958
1360
112
28
70


48
CD304
NRP1
−80
283
275
54.4
862
593
117
25
71


49
CD244
SLAMF4, 2B4
1817
4008
3077
4077
2461
5676
4
139
71.5


50
CD51
ITGAV
−86.7
457
370
−1099
1888
605
135
10
72.5









Flow Cytometry Target Validation


Protein expression of the top candidates from the screen was confirmed using separate flow cytometry analyses of mononuclear cells from AML and NBM bone marrow samples. Analyses were performed on an LSR Fortessa (BD Bioscience, USA), corresponding isotype controls were used to determine positive cells. All antibodies and reagents used are listed in Table 3.


Antibody Dependent Cellular Cytotoxicity


Antibody dependent cellular cytotoxicity (ADCC) assays were performed as previously described in Landberg et al., 201824. Target cells were labeled with the membrane dye PKH26 (Sigma-Aldrich, USA) and subsequently incubated with antibodies of varying concentrations for 30 minutes. Freshly isolated or frozen NK cells from healthy donors were then added in a 10:1 ratio compared to target cells. Corresponding isotype antibodies and wells with only NK and target cells were used as controls. The ADCC effect was assessed by flow cytometry after 12-18 hours using an LSR Fortessa (BD Bioscience, USA), with the viability dye DAPI (Sigma-Aldrich, USA) and CountBright Absolute Counting Beads (Thermo Fisher Inc, USA) added to each well. Specific ADCC-induced cell death was calculated with the formula: percentage viable cellsantibody/percentage viable cellsno antibody, and percentage viable cellisotype/percentage viable cellsno antibody respectively.


AML Xenografts


Viably frozen mononuclear cells from bone marrow of AML patients were thawed and T cell depleted using CD3 microbead separation (Miltenyi Biotec) or OKT3 anti-CD3 antibody (BioXCell). For primary and secondary transplantations, ≥5 million cells were transplanted by tail vein injection to sublethally irradiated (250 cGy) NOD.Cg-PrkdcscidII2rgtm1WjI/SzJ-SGM3 (NSGS) mice, a variant of the NSG mouse overexpressing hGM-CSF, hIL-3 and hSCF (Jackson laboratory). Mice were euthanized upon signs of serious illness. In vivo experiments were approved by the regional Animal Ethics Committee of Malmö/Lund.


Statistical Analyses


Statistical tests were performed using Prism 6 (GraphPad Software, USA). Students T-test or Mann-Whitney U test was used when comparing two groups. Spearman's rank test was used to determine correlations between biological replicates when conducting the antibody screen.


Results


Antibody Based Screen Identifies Multiple Candidate Cell Surface Markers


To identify new cell surface markers specifically expressed on immature AML cells, an arrayed antibody screening system was used to evaluate 362 different cell surface markers within the immature 7AADCD3CD19CD34+CD38 fraction of TP53 mutated AML bone marrow and NBM controls (FIGS. 1A and B). High consistency in marker expression was seen between the different biological replicates analyzed with the same iteration of the screen although the correlation co-efficient was higher for NBM samples (NBM #2 compared to NBM #3 r=0.6811, p<0.0001; FIG. 1C) than for AML samples (AML #80 compared to AML #83 r=0.647, p<0.0001; FIG. 1D). Two different ranking systems were combined to obtain a top list of the most promising cell surface markers (FIG. 1E). Markers that appeared to be specifically expressed in the CD3CD19 CD34+CD38 compartment of TP53 mutated AML compared to NBM included the known markers CD123/IL3RA,25 IL1RAP,23 CD93,26 CD7,27 CD244,28 CD56/NCAM1,29 Jagged 2/JAG2,30 and CD105/ENG,31 as well as multiple new candidate markers not previously described in AML.


Flow Cytometric Validation Show Overexpression of SLAMF6 in TP53 Mutated AML Samples


Based on the antibody screen, eight novel markers were chosen for further validation (FIG. 2). SLAMF6 was confirmed to be significantly overexpressed on immature CD34+CD38 cells from primary TP53 mutated AML bone marrow compared to corresponding cells from normal bone marrow (p=0.006 comparing percent positive cells). CD54/ICAM1 also showed a trend towards higher MFI in AML compared to NBM (p=0.12 comparing median MFI). CD244, CD105/Endoglin, CD323/JAM3, and Jagged 2/JAG2 were expressed in AML but also showed a similar expression in the immature compartment of normal bone marrow. CD62E/SELE and CD369/CLEC7A expression could not be confirmed in either of the three tested AML samples.


SLAMF6 is Expressed on Immature TP53 AML Cells


To further delineate SLAMF6 expression in TP53 mutated AML, different cellular compartments were examined in AML samples with retained CD34/CD38 phenotypic hierarchies. All three TP53 mutated AML samples showed high SLAMF6 expression in the immature CD34+CD38 compartment as compared to the more mature CD34 compartments (FIG. 3A). The opposite was seen in normal bone marrow where SLAMF6 was absent in the CD34+CD38 HSC compartment but expressed in more mature cells, presumably B, T and NK cells (FIG. 3B). The gene expression level of SLAMF6 was also investigated in FACS sorted normal hematopoietic progenitor populations and compared to the expression levels to bulk MNC from TP53 mutated AMLs (n=18), showed a striking overexpression in the AML samples (p=0.0007 for Mann-Whitney test comparing AML to hematopoietic stem cells (HSC, n=6), FIG. 3C). All tested AML samples (n=14, Table 1) showed SLAMF6 expression on CD3+ T-cells and CD19+ B-cells cells independent of genetic subtype (FIG. 3D).


SLAMF6 is Expressed in AMLs of Diverse Genetic Subtypes


Given that all tested TP53 mutated AML samples showed SLAMF6 expression, the gene expression levels of SLAMF6 in MNC were investigated in the publicly available TCGA data set.32 A higher mean expression in TP53 mutated (n=14) compared to wild type AML (n=151) was observed (p=0.001, FIG. 4A). The SLAMF6 protein expression was next examined by flow cytometry. A TP53 mutated AML sample with a CD34 negative phenotype showed high SLAMF6 expression (FIG. 4B). Three samples; AML 33, AML 34 and AML 144, showed SLAMF6 expression in their leukemic compartment (FIG. 4C). AML 33 (CD34 negative, mutations in DNMT3A, NPM1, FLT3-ITD, IDH1) showed expression in the myeloid compartment, AML 34 (CD34 positive, mutations in DNMT3A and IDH1) showed SLAMF6 expression in the myeloid as well as the more immature CD34+CD38 compartment, and a sample with myelodysplastic syndrome MDS144 (mostly CD34 negative, mutations in RUNX1, ASXL1, TET2, and BCOR) showed expression only in a subset of the myeloid cells. Three additional samples; AML 32, MDS35 and AML 66 showed a low expression of SLAMF6 in their myeloid compartment (FIG. 4D). Four samples; AML 21, AML 28, MDS70 and AML 94 completely lacked SLAMF6 expression in their myeloid compartment (FIG. 4E). SLAMF6 was thus highly expressed in all evaluated AML samples carrying a TP53 mutation as well as in three out of ten AML samples of other genetic subtypes.


SLAMF6 Antibody Induces ADCC Mediated Killing of KG1 Cells


To evaluate SLAMF6 as a target for antibody-based therapies, a series of AML cell lines were first investigated for expression of SLAMF6. KG1 and K562 cells both expressed high levels of SLAMF6, while OCI-AML 3 showed no expression (FIG. 5A). To evaluate if SLAMF6 antibodies were able to elicit cell killing by recruiting human NK effector cells, ADCC experiments were performed using KG1 cells. As shown in FIG. 5B, a specific killing of SLAMF6 expressing KG1 cells was observed.


SLAMF6 Expression is Retained after Serial Xenografting


To establish a disease model allowing in vivo studies for antibody-based targeting of SLAMF6, primary AML samples were transplanted to sublethally irradiated NSGS mice. Three TP53 mutated AML samples were serially transplanted and two of these (AML 48 and AML 80) showed high leukemic engraftment in secondary mice. SLAMF6 expression was highly retained in both of these samples, showing the feasibility of studying SLAMF6 in vivo (FIG. 5C).


Discussion


To improve the survival of patients with neoplastic hematologic disorders (including AML), a better understanding of the disease- and relapse-causing leukemic stem cells and possibilities to specifically target such cells are needed. By identifying cell surface markers specifically expressed on AML stem cells, their prospective isolation for functional interrogation becomes feasible. Such cell surface markers may also provide attractive targets for directed treatments as shown for several markers including CD33, CD123 and IL1RAP.9, 14, 33 Because AML is a heterogeneous disease both in terms of underlying molecular cause and response to therapy, searching for cell surface markers in specific genetic subtypes of AML might increase the chance of identifying such markers. This in turn could provide specific biological insights into AML subtypes with treatment implications in parity with ATRA treatment for t(15;17) acute promyelocytic leukemia.34 In this study, TP53 mutated AML was a focus, which is one of the subtypes recognized by European Leukemia Net as having the worst prognosis of all AML subtypes.35 Using an arrayed antibody screen of 362 cell surface markers, specifically upregulated markers were screened for on CD3CD19CD34+CD38 AML cells compared to corresponding cells from normal bone marrow. Using this approach, several previously described markers were identified including CD123, IL1RAP and CD93, thus validating our screening approach. Importantly, SLAMF6 was identified as a new marker being upregulated on immature TP53 mutated AML cells and SLAMF6 antibodies were showed that can recruit human NK cells to elicit cell killing of AML cells.


SLAMF6 is one of nine members of the SLAM family of paralogue genes located on chromosome band 1q23, most of which play a role in immune regulation and some that have been suggested as therapeutic targets in different malignancies.36 Elotuzumab is a naked antibody targeting SLAMF7 that has been shown to both mark myeloma cells for effector cell mediated killing and induce an immune response against the myeloma cells through the antibody's activating effect upon binding to normal NK cells.37, 38 This dual mode of action immune therapy is a promising, novel treatment concept. Elotuzumab has shown promising effects in clinical trials for treatment of myeloma.39 SLAMF6 is known to be expressed on human B, T and NK cells. Upon homophilic self-ligation of SLAMF6, internal signaling through tyrosine phosphorylation of SLAMF6 cytoplasmic tail, recruitment of SAP or EAT-2 is involved in NK cell and T cell activation.4041 SLAMF6 also plays a role in T cell exhaustion and an anti-SLAMF6 antibody was shown to reactivate exhausted CD8+ T cells, another potential antineoplastic effect that targeting SLAMF6 with an antibody could elicit.42 However, SLAMF6 can also inhibit cellular functions through recruitment of SHP-1/2 in the absence of SAP, making the exact effects of SLAMF6 signaling or binding context- and cell-dependent.43 In the present study, SLAMF6 was found to be upregulated in the immature CD34+CD38 subpopulation of AML cells, which in most subtypes of AML has been shown to contain the highest AML stem cell activity as measured by long-term engraftment in immunodeficient mice.15 Notably, SLAMF6 was not expressed on immature normal CD34+CD38 bone marrow cells, suggesting that directed therapies against SLAMF6 would spare normal hematopoietic stem cells. SLAMF6 expression was however retained in CD3+ T cells and CD19+ B-cells in all AML samples analyzed, independent of genetic alterations in the AML sample. KG1 cells were also shown to express high levels of SLAMF6 and these cells were specifically killed in ADCC experiments using an anti-SLAMF6 antibody. Importantly, SLAMF6 was retained on AML cells after serial transplantation to NSGS mice.


In conclusion, SLAMF6 was identified as a cell surface marker upregulated on immature AML cells, for example those carrying a TP53 mutation. SLAMF6 was further demonstrated to provide a new target for antibody-based therapies in AML, thus opening up new avenues for the development of antibody-based therapeutic strategies for AML, including those subtypes with poor prognosis (such as TP53 mutated AML). Further, although the cell death mechanism of action demonstrated in this experiment is ADCC, the induction of cell death would be achievable with other mechanisms of action based on this finding that SLAMF6 is present on the immature AML cells. For example, SLAMF6 could be targeted with an antibody that comprises a radiolabel or cytotoxic moiety.


EXAMPLE 2

SLAMF6 Antibody Induces ADCC of AML Patient Cells


Summary


SLAMF6 antibodies can induce ADCC to kill AML patient cells.


Introduction


Killing of cancer cells through ADCC with a SLAMF6 antibody has never before been demonstrated. Here, it is demonstrated that SLAMF6-expressing primary AML patient samples and xenografted AML patient samples enriched for leukemia stem cells can be killed ex vivo by ADCC using SLAMF6 antibodies.


Materials and Methods


AML Patient Samples


Bone marrow and peripheral blood samples from AML patients were collected at the Department of Clinical Genetics, Skene University Hospital after written informed consent. Mononuclear cells were prepared by lymphoprep separation (GE Healthcare) and viably frozen. Protein expression of SLAMF6 on the leukemia cells was determined by flow cytometry with a SLAMF6 antibody (Biolegend).


Patient-Derived Xenografts


To generate patient-derived xenografts, primary AML patient cells were thawed, and T cells depleted by either CD3 microbead separation (Miltenyi Biotec) or treatment with the OKT3 anti-CD3 antibody (BioXCell). A total of ≥5 million cells were then transplanted by tail vein injection to sublethally irradiated NOD.Cg-PrkdcscidII2rgtm1WjI/SzJ-SGM3 (NSG-S) mice (250 cGy), a substrain of the NSG mouse overexpressing hGM-CSF, hIL-3 and hSCF (Jackson laboratory). Mice were euthanized upon signs of serious illness.


Antibody Dependent Cellular Cytotoxicity


ADCC assays were performed as described in Example 1. Target cells were labeled with the membrane dye PKH26 (Sigma-Aldrich, USA) and subsequently incubated with rabbit monoclonal SLAMF6 antibody or an isotype control (Biolegend) for 30 minutes. Freshly isolated NK cells from healthy donors were then added in a 10:1 ratio compared to target cells. Corresponding isotype antibodies and wells with only NK and target cells were used as controls. The ADCC effect was assessed by flow cytometry after 12-18 hours using an LSR Fortessa (BD Bioscience), with the viability dye DAPI (Sigma-Aldrich) and CountBright Absolute Counting Beads (Thermo Fisher Inc) added to each well.


Results


Treatment with a SLAMF6 antibody induced cell death in AML patient cells ex vivo by recruitment of NK effector cells and killing through ADCC (FIG. 6A). To enrich for leukemia stem cells by serial transplantation, patient cells from AML-83 were xenografted and passaged for two generations in vivo. ADCC was effective against both the primary AML patient cells and the leukemia stem cell-enriched patient cells. Protein expression of SLAMF6 on the surface of the patient samples was determined by FACS (FIG. 6B).


Discussion


These data show that an antibody against SLAMF6 can elicit killing of AML patient cells and of leukemia stem cell-enriched AML samples ex vivo, by binding to the target cell and recruiting effector cells to induce ADCC. This demonstrates that SLAMF6 antibodies have therapeutic activity against AML patient cells.


EXAMPLE 3

SLAMF6 expression on AML stem cells and AML cell lines


Summary


SLAMF6 is expressed on leukemic stem cells in a majority of AML patients in AML of diverse genetic background.


Introduction


To determine the relevance of SLAMF6 as a target for therapy in AML, SLAMF6 protein expression was investigated in a cohort of 42 primary AML patient samples and nine AML cell lines. The expression was further studied in the leukemic stem cell containing compartment with a CD3CD19CD34+CD38low immunophenotype.


Materials and Methods


Bone marrow and peripheral blood samples were collected after written informed consent in accordance to the Declaration of Helsinki. Samples were collected from patients with AML and myelodysplastic syndrome (MDS). Mononuclear cells (MNC) were isolated using Lymphoprep (GE Healthcare Bio-Sciences AB, Sweden) and subsequently viably frozen. Patients included in the study and their clinical characteristics are shown in Table 5. The study was approved by a regional ethics committee in Lund (Dnr 2011/289). SLAMF6 expression was determined by flowcytometry on an LSR Fortessa (BD Bioscience, USA) with commercially available antibodies targeting CD3, CD19, CD34, CD38 and SLAMF6 as well as a viability marker.


Results


A cohort of 42 primary AML samples was evaluated for SLAMF6 expression. For CD34 expressing AML samples, the CD34+ and the CD34+CD38low cells within the CD3−CD19− compartment known to be enriched for leukemic stem cells were specifically evaluated. Samples were classified as high (“SLAMF6high”) when >50% of cells expressed SLAMF6, intermediate (“SLAMF6int”) when 10-50% of cells expressed SLAMF6 and negative (“SLAMF6neg”) when <10% of cells expressed SLAMF6. Within the CD3−CD19− myeloid compartment, 26% of AML samples were classified as SLAMF6high and 33% as SLAMF6int, within the CD34+ compartment 45% were classified to be SLAMF6high and 21% SLAMF6int and within the CD34+CD38low compartment 41% were SLAMF6high and 24% were SLAMF6int (FIG. 7). Further, SLAMF6 expression is not limited to one specific genetic background but is instead highly expressed in patients with a variety of different genetic alterations (FIG. 8). Furthermore, SLAMF6 is expressed in multiple AML cell lines (FIG. 9).


Discussion


SLAMF6 is shown to be aberrantly expressed on leukemic stem cells from primary AML samples carrying a large variety of genetic alterations.









TABLE 5







AML Patient Characteristics



























CD34+


Pat#
Gender
Disease
FAB
ELN
Karyotype
Mutations
CD34%
Myeloid
CD34+
CD38low




















7
F
tAML
M4
tAML

IDH2, FLT3, KRAS,
1
Int






diagnosis



NPM1, WT1, DNMT3A


9
F
AML
M5
LR
46, XX
DNMT3A, TET2, NPM1
0
Neg






diagnosis


10
M
sAML
M4
sAML
46, XY, inv(9)(p11q12)
RUNX1, ASXL1, IDH2,
99
Neg
Neg
Neg




diagnosis



JAK2, SRSF2,


21
M
AML
M2
IR
46, XY
IDH2, STAG2, BCOR
94
Neg
Neg
Neg




diagnosis


23
M
AML
M4
LR
46, XY, der(16)(q22)
NRAS
46
Int
High
High




diagnosis


24
M
AML
M2
LR
46, XY
IDH2, NPM1, FLT3-ITD
0
Neg






diagnosis


25
M
AML
M2
LR
45, X, −Y
NPM1, TET2
5
Neg






diagnosis


28
M
sAML
M2
sAML
46, XY
NPM1, FLT3-ITD, TET2
1
Neg






diagnosis


32
M
AML
M2
HR
47, XY, +13
RUNX1, ASXL1
73
Int
Int
Int




diagnosis


33
F
AML
M4
sAML
46, XX
DNMT3A, NPM1, IDH1,
3
High






relapse



CEBPA


34
F
AML
M5
IR
46, XX
DNMT3A, IDH1, NRAS,
96
High
High
High




diagnosis



CEBPA


35
F
MDS
NA
HR
46, XX
RUNX1, ASXL1, DNMT3A,
59
Int
Int
Int




RAEB-2



IDH2


37
M
AML
M5
HR
Complex1
TP53, FLT3
2
Int






diagnosis


48
F
tAML
M2
tAML
Complex2
TP53
63
High
High
High




dignosis


55
M
tAML
M2
tAML
45, XY, −7/45, X, −Y
SRSF2, ASXL1, EZH2,
42
High
High
High




diagnosis



PTPN11


61
F
AML
M2
LR
Complex3
RUNX1, ASXL1, RB1
52
High
High
High




diagnosis


62
F
AML
M4
HR
Complex4
TP53, DNMT3A,
15
Neg
Neg
Neg




diagnosis


66
M
sAML
M2
sAML
46, XY
IDH1, NPM1, JAK2,
4
Neg






diagnosis



CEBPA


70
M
MPN/MDS
NA
NA
46, XY
RUNX1, ASXL1, TET2
10
Neg
Neg
Neg


80
M
AML
M2
HR
48, XXYc, +i(12)(p10),
TP53
35
Int
High
High




diagnosis


der(17)t(13;17)(q1?4;p1?3)


83
M
sAML
M2
sAML
Complex5
TP53, TET2, NF1
15
Int
High
High




diagnosis


85D
F
AML
M2
HR
Complex6
TP53, DNMT3A
60
Neg
Neg
Neg




diagnosis


85R
F
AML
M2
HR
N/A
TP53, DNMT3A
91
Int
Int
Int




relapse


94
M
AML
M0
HR
47, XY, +10
DNMT3A, IDH1, FLT3-
98
Neg
Neg
Neg




diagnosis



ITD,








RUNX1, BCOR, STAG2


97
F
AML
M4
LR
46, XX
NPM1, FLT3-ITD, IDH1,
39
Int
Int
Int




diagnosis



DNMT3A


104
M
tAML
M0
tAML
46, XY
NPM1, FLT3-ITD, IDH2,
0
Neg






diagnosis



DNMT3A


105
F
AML
M4
IR
46, XX
NPM1, FLT3-ITD,
10
High
High
High




diagnosis



DNMT3A,


110
M
AML
M2
IR
46, XY
IDH2, SRSF2, STAG2,
7
Neg






diagosis


111
M
sAML
M2
sAML
47, XY, +8/47, XY, +mar
RUNX1, BCOR
4
Neg






diagnosis


117
M
AML
M2
LR
46, XY, t(8;21)(q22;q22)
KIT, NRAS
57
High
High
Int




diagnosis


123
F
AML
M4
LR
46, XX, t(16;16)(p13;q22)

70
High
High
High




diagnosis


124
M
AML
M4
LR
46, XY, inv(16)(p13q22)
KIT
91
High
High
High




diagnosis


126
F
AML
M1
HR
46, XX
IDH2, CBFC, CEBPA,
60
Neg
Neg
Neg




diagnosis



RAD21, MEIS1


136
F
AML
M4
LR
46, XX
NPM1, IDH2
4
High






diagnosis


138
M
AML
M4
HR
48, XY, +13, +19
RUNX1, TET2, ZRSR2
98
Neg
Neg
Neg




diagnosis


144
F
MPN/MDS
NA
NA
47, XX, +8
RUNX1, ASXL1, TET2,
15
Int
Neg
Neg








BCOR


151
M
sAML
M1
sAML
46, XY, ?del(Y)(q11)
RUNX1, ASXL1, TET2,
96
Neg
Neg
Neg




diagnosis



IDH1, SRSF2, PHF6


154
M
tAML
M0
tAML
46, XY, inv(2)(p21q31),
RUNX1, SF3B1, NF1,
20
Int
High
High




diagnosis


t(3;12)(q26;p13)
PTPN11, ASXL2, IKZF1


155
M
AML
M5
HR
Complex7
TP53, FLT3
2
High






diagnosis


157
M
sAML
M4
sAML
46, XY
SRSF2, NRAS
49
Int
Int
Int




diagnosis


161
F
AML
M2
LR
46, XX, t(8;21)(q22;q22)
FLT3-ITD, KIT, KRAS,
64
Int
Int
Int




diagnosis



RUNX1


172
F
AML
M4
HR
46, XX, t(3;12)(q26;p13)
FLT3-ITD, ASXL1, RUNX1
26
Int
High
High




diagnosis





Abbreviations;


sAML: secondary AML,


tAML: therapy-related AML,


ELN: European Leukemia net risk classification 2017,


LR: low risk,


IR: intermediate risk,


HR: high risk


Complex1: 47, XY, +6, +i(8)(q10), −18, −22, +mar/45-46, XY, −3, +add(6)(p21), +i(8)(q10), der(16)t(3;16)(p12;q11), −22


Complex2: 43-47, XX, del(5)(q13q33), der(8)t(8;12)(p22;p13), add(11)(p15), −12, add(13)(p11), −18, del(20)(q11), add(22)(q13), +mar


Complex3: 45, X, −X, der(8)t(8;21)(q22;q22), del(9)(q13), del(13)(q21q21), del(17)(p?13), der(21)t(8;21)ins(21;?)(q22;?)


Complex4: 46, XX, der(3)t(1;3)(p13;q27), −5, add(7)(q22), der(16)t(?5;16)(q?;p11), der(17)t(5;17)(q?;q21), der(21)t(11;21)(q13;q22), +mar


Complex5: 45, XY, der(13;14)(q10;q10)/42-44, idem, −5, −7, −9, −10, −11 , ?hsr(11)(q23), −14, −16, −20 , +4mar


Complex6: 43-49, XX, −3, add(6)(p21), −13, add(13)(q34), add(17)(p11), +2-5r/46, XX, del(5)(q13q33)


Complex7: 47, XY, +8, t(9;11)(p21;q23)/46-47, idem, der(9)del(9)(p12)del(9)(q12), der(17)t(9;17)(q31;p13)/46-47, idem, der(7;10)(q10;q10), +add(8)(p11)






EXAMPLE 4

Activation of SLAMF6 on T Cells Promotes Killing of AML Cells


Summary


Stimulation of T cells with an activating SLAMF6 antibody induces T cell-mediated killing of AML cells.


Introduction


SLAMF6 is a self-ligand, binding to other SLAMF6 molecules on the surface of interacting cells. Since SLAMF6 is expressed both on leukemia stem cells and on certain normal immune cells (e.g. T, B and NK cells), modulating these interactions could have therapeutic potential. Therefore, the effect on T cell-mediated killing of AML cells by T cell stimulation was determined with an activating SLAMF6 antibody.


Materials and Methods


T cells were isolated by CD3 microbead separation (Miltenyi Biotec) of leukocyte concentrate collected from healthy donors and viably frozen. T cell-mediated killing was assessed by incubating 80,000 T cells and 20,000 HNT-34 target cells with a SLAMF6 antibody or an isotype control (Biolegend) for 72 hours before quantification on an LSR Fortessa (BD Biosciences) with CountBright Absolute Counting Beads (Thermo Fisher) and antibodies against CD3 and CD33 (Biolegend).


Results


T cell stimulation with an activating antibody against SLAMF6 markedly increased T cell-mediated killing of AML target cells at all tested concentrations (FIG. 10).


Discussion


These data show that targeting SLAMF6 on immune cells modulates their response to leukemia and induces T cell-mediated killing of AML cells. A therapeutic agent could thus act either on AML cells, on interacting immune cells or on both cell types in combination, to elicit cell killing of leukemic cells.


EXAMPLE 5

Knockout of SLAMF6 from AML Cells Promotes T Cell-Mediated Killing


Summary


SLAMF6 protects AML cells from T cell-mediated killing. Knocking out SLAMF6 in AML cells promotes T cell expansion, activation and killing of the AML cells.


Introduction


The functional importance of SLAMF6 on AML cells was determined by knocking out SLAMF6 in AML cells by CRISPR-Cas9 and analyzing the effect on T cell-mediated killing.


Materials and Methods


SLAMF6 knockout cell lines were generated by introduction of Cas9 protein (PNA Bio) and one of two different SLAMF6 gRNA constructs, or a negative control gRNA against luciferase, by electroporation with an ECM 830 Electroporation System (Harvard Apparatus), followed by sorting of successfully transfected cells after 24 h with a FACS Aria (BD Biosciences). Knockout was verified by FACS with a SLAMF6 antibody (Biolegend) before initiation of experiments. T cells were isolated by CD3 microbead separation (Miltenyi Biotec) of leukocyte concentrate collected from healthy donors. T cell activation and T cell-mediated killing were assessed by incubation of 80,000 T cells and 20,000 target cells for 72 hours before quantification on an LSR Fortessa (BD Biosciences) with CountBright Absolute Counting Beads (Thermo Fisher) and antibodies against CD3 and CD33 (Biolegend).


Results


Removal of SLAMF6 from KG-1 AML cells by CRISPR-Cas9 (FIG. 11D) resulted in expansion and activation of interacting T cells, and increased T cell-mediated killing of the AML cells (FIG. 11A-C).


Discussion


This finding demonstrates that SLAMF6 protects leukemia cells against T cell-mediated killing and that targeting of SLAMF6 on AML cells or modulation of the SLAMF6-SLAMF6 interaction between leukemia cells and immune cells promotes an anti-leukemia immune response.


REFERENCES



  • 1. Dombret H, Gardin C. An update of current treatments for adult acute myeloid leukemia. Blood. 2016 Jan. 7; 127(1):53-61.

  • 2. Stein E M, Tallman M S. Emerging therapeutic drugs for AML. Blood. 2016 Jan. 7; 127(1):71-8.

  • 3. Estey E H. Acute myeloid leukemia: 2019 update on risk-stratification and management. Am J Hematol. 2018 Oct. 16; 93(10):1267-91.

  • 4. Rucker F G, Schlenk R F, Bullinger L, Kayser S, Teleanu V, Kett H, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood. 2012 Feb. 23; 119:2114-21.

  • 5. Stengel A, Kern W, Haferlach T, Meggendorfer M, Fasan A, Haferlach C. The impact of TP53 mutations and TP53 deletions on survival varies between AML, ALL, MDS and CLL: an analysis of 3307 cases. Leukemia. 2017 March; 31(3):705-11.

  • 6. Middeke J M, Herold S, Rücker-Braun E, Berdel W E, Stelljes M, Kaufmann M, et al. TP53 mutation in patients with high-risk acute myeloid leukaemia treated with allogeneic haematopoietic stem cell transplantation. Brit J Haematol. 2016 Jan. 13; 172(6):914-22.

  • 7. Wong T N, Ramsingh G, Young A L, Miller C A, Touma W, Welch J S, et al. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia. Nature. 2015 Feb. 16; 518(7540):552-5.

  • 8. Jin L, Lee E M, Ramshaw H S, Busfield S J, Peoppl A G, Wilkinson L, et al. Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Cell Stem Cell. 2009 Jul. 2; 5(1):31-42.

  • 9. Ågerstam H, Karlsson C, Hansen N, Sandén C, Askmyr M, Palffy von S, et al. Antibodies targeting human IL1RAP (IL1R3) show therapeutic effects in xenograft models of acute myeloid leukemia. Proc Natl Acad Sci USA. 2015 Aug. 25; 112(34):10786-91.

  • 10. Majeti R, Chao M P, Alizadeh A A, Pang W W, Jaiswal S, Gibbs K D Jr, et al. CD47 Is an Adverse Prognostic Factor and Therapeutic Antibody Target on Human Acute Myeloid Leukemia Stem Cells. Cell. 2009 Jul. 23; 138(2):286-99.

  • 11. Kikushige Y, Shima T, Takayanagi S-I, Urata S, Miyamoto T, Iwasaki H, et al. TIM-3 Is a Promising Target to Selectively Kill Acute Myeloid Leukemia Stem Cells. Cell Stem Cell. 2010 Dec. 3; 7(6):708-17.

  • 12. van Rhenen A, van Dongen GAMS, Kelder A, Rombouts E J, Feller N, Moshaver B, et al. The novel AML stem cell associated antigen CLL-1 aids in discrimination between normal and leukemic stem cells. Blood. 2007 Oct. 1; 110(7):2659-66.

  • 13. Cowan A J, Laszlo G S, Estey E H, Walter R B. Antibody-based therapy of acute myeloid leukemia with gemtuzumab ozogamicin. Front Biosci (Landmark Ed). 2013 Jun. 1; 18:1311-34.

  • 14. Appelbaum F R, Bernstein I D. Gemtuzumab ozogamicin for acute myeloid leukemia. Blood. 2017 Nov. 30; 130(22):2373-6.

  • 15. Thomas D, Majeti R. Biology and relevance of human acute myeloid leukemia stem cells. Blood. 2017 Mar. 23; 129(12):1577-85.

  • 16. Radhakrishnan S V, Bhardwaj N, Luetkens T, Atanackovic D. Novel anti-myeloma immunotherapies targeting the SLAM family of receptors. OncoImmunology. 2017; 6(5):e1308618.

  • 17. Yigit B, Wang N, Herzog R W, Terhorst C. SLAMF6 in health and disease: Implications for therapeutic targeting. Clin Immunol. 2018 Oct. 23.

  • 18. Munitz A, Bachelet I, Fraenkel S, Katz G, Mandelboim O, Simon H-U, et al. 2B4 (CD244) Is Expressed and Functional on Human Eosinophils. J Immunol. 2004 Dec. 20; 174(1):110-8.

  • 19. Lewis T, Olson D J, Gordon K A, Sandall S L, Miyamoto J, Westendorf L, et al. Abstract 1195: SGN-CD352A: A novel humanized anti-CD352 antibody-drug conjugate for the treatment of multiple myeloma. Cancer Res. 2016 Jul. 15; 76(14 Supplement): 1195.

  • 20. Korver W, Singh S, Liu S, Zhao X, Yonkovich S, Sweeney A, et al. The lymphoid cell surface receptor NTB-A: a novel monoclonal antibody target for leukaemia and lymphoma therapeutics. Brit J Haematol. 2007 May; 137(4):307-18.

  • 21. Yigit B, Halibozek P J, Chen S-S, O'Keeffe M S, Arnason J, Avigan D, et al. A combination of an anti-SLAMF6 antibody and ibrutinib efficiently abrogates expansion of chronic lymphocytic leukemia cells. Oncotarget. 2016 May 3; 7(18):26346-60.

  • 22. Askmyr M, Agerstam H, Hansen N, Gordon S, Arvanitakis A, Rissler M, et al. Selective killing of candidate AML stem cells by antibody targeting of IL1RAP. Blood. 2013 May 2; 121(18):3709-13.

  • 23. Barreyro L, Will B, Bartholdy B, Zhou L, Todorova T I, Stanley R F, et al. Overexpression of IL-1 receptor accessory protein in stem and progenitor cells and outcome correlation in AML and MDS. Blood. 2012 Aug. 9; 120(6):1290-8.

  • 24. Landberg N, Palffy von S, Askmyr M, Lilljebjörn H, Sandén C, Rissler M, et al. CD36 defines primitive chronic myeloid leukemia cells less responsive to imatinib but vulnerable to antibody-based therapeutic targeting. Haematologica. 2018 March; 103(3):447-55.

  • 25. Jordan C T, Upchurch D, Szilvassy S J, Guzman M L, Howard D S, Pettigrew A L, et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia. 2000 October; 14(10):1777-84.

  • 26. Iwasaki M, Liedtke M, Gentles A J, Cleary M L. CD93 Marks a Non-Quiescent Human Leukemia Stem Cell Population and Is Required for Development of MLL-Rearranged Acute Myeloid Leukemia. Cell Stem Cell. 2015 Oct. 1; 17(4):412-21.

  • 27. Saxena A, Sheridan D P, Card R T, McPeek A M, Mewdell C C, Skinnider L F. Biologic and clinical significance of CD7 expression in acute myeloid leukemia. Am J Hematol. 1998 August; 58(4):278-84.

  • 28. Quek L, Otto G W, Garnett C, Lhermitte L, Karamitros D, Stoilova B, et al. Genetically distinct leukemic stem cells in human CD34− acute myeloid leukemia are arrested at a hemopoietic precursor-like stage. J Exp Med. 2016 Jul. 25; 213(8):1513-35.

  • 29. Sasca D, Szybinski J, Schüler A, Shah V, Heidelberger J, Haehnel P S, et al. NCAM1 (CD56) promotes leukemogenesis and confers drug resistance in AML. Blood. 2019 May 23; 133(21):2305-19.

  • 30. Gal H, Amariglio N, Trakhtenbrot L, Jacob-Hirsh J, Margalit O, Avigdor A, et al. Gene expression profiles of AML derived stem cells; similarity to hematopoietic stem cells. Leukemia. 2006 December; 20(12):2147-54.

  • 31. Dourado K M C, Baik J, Oliveira V K P, Beltrame M, Yamamoto A, Theuer C P, et al. Endoglin: a novel target for therapeutic intervention in acute leukemias revealed in xenograft mouse models. Blood. 2017 May 4; 129(18):2526-36.

  • 32. The Cancer Genome Atlas Research Network. Genomic and Epigenomic Landscapes of Adult De Novo Acute Myeloid Leukemia. N Engl J Med. 2013 May 30; 368(22):2059-74.

  • 33. Testa, Pelosi, Castelli. CD123 as a Therapeutic Target in the Treatment of Hematological Malignancies. Cancers. 2019 September; 11(9):1358-31.

  • 34. Lo-Coco F, Avvisati G, Vignetti M, Thiede C, Orlando S M, Iacobelli S, et al. Retinoic Acid and Arsenic Trioxide for Acute Promyelocytic Leukemia. N Engl J Med. 2013 Jul. 11; 369(2):111-21.

  • 35. Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum F R, Büchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017 Jan. 26; 129(4):424-47.

  • 36. Calpe S, Wang N, Romero X, Berger S B, Lanyi A, Engel P, et al. The SLAM and SAP Gene Families Control Innate and Adaptive Immune Responses. Adv Immunol. 2008. Vol. 97: 177-250.

  • 37. Tai Y-T, Dillon M, Song W, Leiba M, Li X-F, Burger P, et al. Anti-CS1 humanized monoclonal antibody HuLuc63 inhibits myeloma cell adhesion and induces antibody-dependent cellular cytotoxicity in the bone marrow milieu. Blood. 2008 Aug. 15; 112(4):1329-37.

  • 38. Collins S M, Bakan C E, Swartzel G D, Hofmeister C C, Efebera Y A, Kwon H, et al. Elotuzumab directly enhances NK cell cytotoxicity against myeloma via CS1 ligation: evidence for augmented NK cell function complementing ADCC. Cancer Immunol Immun. 2013 Oct. 26; 62(12):1841-9.

  • 39. Dimopoulos M A, Dytfeld D, Grosicki S, Moreau P, Takezako N, Hori M, et al. Elotuzumab plus Pomalidomide and Dexamethasone for Multiple Myeloma. N Engl J Med. 2018 Nov. 8; 379(19):1811-22.

  • 40. Flaig R M, Stark S, Watzl C. Cutting Edge: NTB-A Activates NK Cells via Homophilic Interaction. J Immunol. 2004 May 20; 172(11):6524-7.

  • 41. Eissmann P, Watzl C. Molecular Analysis of NTB-A Signaling: A Role for EAT-2 in NTB-A-Mediated Activation of Human NK Cells. J Immunol. 2006 Aug. 18; 177(5):3170-7.

  • 42. Yigit B, Wang N, Hacken ten E, Chen S-S, Bhan A K, Suarez-Fueyo A, et al. SLAMF6 as a Regulator of Exhausted CD8+ T Cells in Cancer. Cancer Immunol Res. 2019 Jul. 17:1-33.

  • 43. Kageyama R, Cannons J L, Zhao F, Yusuf I, Lao C, Locci M, et al. The Receptor Ly108 Functions as a SAP Adaptor-Dependent On-Off Switch for T Cell Help to B Cells and NKT Cell Development. Immunity. 2012 Jun. 29; 36(6):986-1002.

  • 44. Guo et al., 2008, Nature 453(7194):529-33

  • 45. Plant et al., 1995, Analyt Biochem 226(2), 342-348

  • 46. Fields & Song, Nature 340:245-246 (1989)

  • 47. Orlandi et al, 1989. Proc. Natl. Acad. Sci. U.S.A. 86:3833-3837

  • 48. Winter et al., 1991, Nature 349:293-299

  • 49. Kohler et al., 1975. Nature 256:4950497

  • 50. Kozbor et al., 1985. J. Immunol. Methods 81:31-42

  • 51. Cote et al., 1983. Proc. Natl. Acad. Sci. USA 80:2026-2030

  • 52. Cole et al., 1984. Mol. Cell. Biol. 62:109-120

  • 53. Jones et al., 1986. Nature 321:522-525

  • 54. Riechmann et al., 1988, Nature 332:323-329

  • 55. Presta, 1992, Curr. Op. Struct. Biol. 2:593-596

  • 56. H Zola (CRC Press, 1988) “Monoclonal Antibodies: A manual of techniques”

  • 57. J G R Hurrell (CRC Press, 1982) “Monoclonal Hybridoma Antibodies: Techniques and Applications”

  • 58. Harlow & Lane, 1988, “Antibodies: A Laboratory Manual”, Cold Spring Harbor Laboratory, New York)

  • 59. Hoogenboom & Winter, 1991, J. Mol. Biol. 227:381

  • 60. Marks et al., 1991, J. Mol. Biol. 222:581

  • 61. Cole et al., 1985, In: Monoclonal antibodies and Cancer Therapy, Alan R. Liss, pp. 77

  • 62. Boerner et al., 1991. J. Immunol. 147:86-95

  • 63. Skerra, Curr Opin Biotechnol. 2007 August; 18(4):295-304

  • 64. Hoppe-Seyler & Butz, 2000, J Mol Med. 78 (8): 426-30

  • 65. Bunka D H & Stockley P G, 2006, Nat Rev Microbiol. 4 (8): 588-96

  • 66. Drabovich et al., 2006, Anal Chem. 78 (9): 3171-8

  • 67. Remington's Pharmaceutical Sciences, 18th edition, A. R Gennaro, Ed., Mack Publishing Company (1990)

  • 68. Handbook of Pharmaceutical Excipients, 3rd edition, A. Kibbe, Ed., Pharmaceutical Press (2000)

  • 69. U.S. Pat. No. 4,235,871

  • 70. U.S. Pat. No. 5,851,451

  • 71. EP 0 213 303

  • 72. Yigit et al., 2019, Cancer Immunology Research

  • 73. Wu et al., 2016, Nature Immunology



EMBODIMENT PARAGRAPHS

Accordingly, the present application also provides aspects according to the following numbered paragraphs:


1. An agent comprising or consisting of a binding moiety with specificity for Signaling Lymphocytic Activating Molecule Family Member 6 (SLAMF6) for use in inducing cell death and/or inhibiting the growth and/or proliferation of pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder, wherein the cells express SLAMF6.


2. An agent comprising or consisting of a binding moiety with specificity for Signaling Lymphocytic Activating Molecule Family Member 6 (SLAMF6) for use in detecting pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder, wherein the cells express SLAMF6.


3. An agent according to paragraph 1 or paragraph 2 wherein the neoplastic hematologic disorder is a leukemia, optionally wherein

    • (a) the pathological stem cells are leukemic stem cells; and/or
    • (b) the progenitor cells are leukemic progenitor cells.


4. An agent according to any one of the preceding paragraphs wherein the cells expressing SLAMF6 also express CD34+CD38.


5. An agent according to any one of the preceding paragraphs wherein the neoplastic hematologic disorder is associated with cells comprising a TP53 mutation.


6. An agent according to paragraph 5 wherein the cells expressing SLAMF6 also express CD34+CD38 and wherein the cells comprise a TP53 mutation.


7. An agent according to any one of the preceding paragraphs wherein the neoplastic hematologic disorder is selected from the group consisting of chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML).


8. An agent according to any one of the preceding paragraphs wherein the neoplastic hematologic disorder is acute myeloid leukemia (AML).


9. An agent according to any one of the preceding paragraphs wherein the binding moiety has specificity for human SLAMF6.


10. An agent according to any one of the preceding paragraphs wherein SLAMF6 is localised on the surface of a cell.


11. An agent according to any one of the preceding paragraphs wherein the agent is capable of modulating an interaction between an immune cell and leukemic stem cells and/or an immune cell and leukemic cells.


12. An agent according to paragraph 11 wherein the immune cells are selected from the group consisting of: B cells, T cells and/or NK cells; preferably wherein the immune cells express SLAMF6.


13. An agent according to any one of the preceding paragraphs wherein the agent is capable of killing the pathological stem cells and/or progenitor cells.


14. An agent according to paragraph 13 wherein the agent is capable of inducing apoptosis of the stem cells and/or progenitor cells.


15. An agent according to paragraph 13 or 14 wherein killing of the cells is induced by antibody-dependent cell-mediated cytotoxicity (ADCC) and/or by a T cell mediated mechanism.


16. An agent according to any one of the preceding paragraphs wherein the agent comprises or consists of a polypeptide.


17. An agent according to paragraph 16 wherein the agent comprises or consists of an antibody or an antigen-binding fragment thereof with binding specificity for SLAMF6, or a variant, fusion or derivative of said antibody or antigen-binding fragment, or a fusion of a said variant or derivative thereof, which retains the binding specificity for SLAMF6.


18. An agent according to paragraph 17 wherein the agent comprises or consists of an antibody or antigen-binding fragment thereof with binding specificity for SLAMF6.


19. An agent according to paragraph 18 wherein the agent comprises or consists of an intact antibody.


20. An agent according to paragraph 18 wherein the agent comprises or consists of an antigen-binding fragment of an antibody.


21. An agent according to paragraph 20 wherein antigen-binding fragment is selected from the group consisting of Fv fragments (e.g. single chain Fv, disulphide-bonded Fv and domain antibodies) and Fab-like fragments (e.g. Fab fragments, Fab′ fragments and F(ab)2 fragments).


22. An agent according to any one of paragraphs 17 to 21 wherein the antibody is a recombinant antibody.


23. An agent according to any one of paragraphs 17 to 21 wherein the antibody is a monoclonal antibody.


24. An agent according to any one of paragraphs 17 to 21 wherein the antibody is a polyclonal antibody.


25. An agent according to any one of paragraphs 17 to 24 wherein the antibody or antigen-binding fragment thereof is human or humanised.


26. An agent according to any one of paragraphs 1 to 16 wherein the agent comprises or consists of a non-immunoglobulin binding moiety.


27. An agent according to any one of paragraphs 1 to 16 wherein the agent comprises or consists of an aptamer.


28. An agent according to paragraph 27 wherein the agent comprises or consists of a peptide aptamer.


29. An agent according to paragraph 27 wherein the agent comprises or consists of a nucleic acid aptamer.


30. An agent according to any one of paragraphs 1 to 15 wherein the agent comprises or consists of a small chemical entity.


31. An agent according to any one of the preceding paragraphs further comprising a moiety for increasing the in vivo half-life of the agent.


32. An agent according to paragraph 31 wherein the moiety for increasing the in vivo half-life is selected from the group consisting of polyethylene glycol (PEG), human serum albumin, glycosylation groups, fatty acids and dextran.


33. An agent according to paragraph 31 or 32 wherein the agent is PEGylated.


34. An agent according to any one of the preceding paragraphs further comprising a cytotoxic moiety.


35. An agent according to paragraph 34 wherein the cytotoxic moiety comprises or consists of a radioisotope.


36. An agent according to paragraph 35 wherein the radioisotope is selected from the group consisting of astatine-211, bismuth-212, bismuth-213, iodine-131, yttrium-90, lutetium-177, samarium-153 and palladium-109.


37. An agent according to paragraph 34 wherein the cytotoxic moiety comprises or consists of a toxin (such as saporin or calicheamicin).


38. An agent according to paragraph 34 wherein the cytotoxic moiety comprises or consists of a chemotherapeutic agent (such as an antimetabolite).


39. An agent according to any one of the preceding paragraphs further comprising a detectable moiety.


40. An agent according to paragraph 39 wherein the detectable moiety comprises or consists of a radioisotope.


41. An agent according to paragraph 40 wherein the radioisotope is selected from the group consisting of: technetium-99m; indium-111; gallium-67; gallium-68; arsenic-72; zirconium-89; iodine-12; thallium-201.


42. An agent according to paragraph 39 wherein the detectable moiety comprises or consists of a paramagnetic isotope.


43. An agent according to paragraph 42 wherein the paramagnetic isotope is selected from the group consisting of: gadolinium-157; manganese-55, dysprosium-162, chromium-52; iron-56.


44. A pharmaceutical composition comprising an effective amount of an agent as defined in any one of the preceding paragraphs and a pharmaceutically-acceptable diluent, carrier or excipient.


45. A pharmaceutical composition according to paragraph 44 adapted for parenteral delivery.


46. A pharmaceutical composition according to paragraph 44 adapted for intravenous delivery.


47. A kit comprising an agent as defined in any one of paragraphs 1 to 43 or a pharmaceutical composition as defined in any one of paragraphs 44 to 46.


48. Use of an agent as defined in any one of paragraphs 1 to 43 in the preparation of a medicament for inducing cell death and/or inhibiting the growth and/or proliferation of pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder, wherein the cells express SLAMF6.


49. Use of an agent as defined in any one of paragraphs 1 to 43 in the preparation of a diagnostic agent for detecting pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder, wherein the cells express SLAMF6.


50. Use of an agent as defined in any one of paragraphs 1 to 43 for detecting pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder, wherein the cells express SLAMF6.


51. The use according to paragraph 48, 49 or 50 wherein the neoplastic hematologic disorder is a leukemia.


52. A use according to any one of paragraphs 48 to 51 wherein the neoplastic hematologic disorder is selected from the group consisting of chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML).


53. The use according to paragraph 52 wherein the neoplastic hematologic disorder is acute myeloid leukemia (AML).


54. A method for inducing cell death and/or inhibiting the growth and/or proliferation of pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder in an individual, comprising the step of administering to the individual an effective amount of an agent as defined in any one of paragraphs 1 to 43, or a pharmaceutical composition as defined in paragraph 44 to 46, wherein the cells express SLAMF6.


55. A method according to paragraph 54 wherein the neoplastic hematologic disorder is a leukemia.


56. A method for detecting pathological stem cells and/or progenitor cells associated with neoplastic hematologic disorder in an individual, comprising the step of administering to the individual an effective amount of an agent as defined in any one of paragraphs 1 to 43, or a pharmaceutical composition as defined in paragraph 44 to 46 wherein the cells express SLAMF6.


57. An in vitro method for diagnosing or prognosing a neoplastic hematologic disorder, the method comprising:

    • (a) providing a bone marrow or peripheral blood sample of haematopoietic cells from an individual to be tested;
    • (b) isolating a subpopulation of CD34+, CD38 cells from the haematopoietic cells; and
    • (c) determining whether stem cells, contained within the CD34+, CD38 cells, express the cell surface marker SLAMF6;
    • wherein stem cells that exhibit the cell surface marker profile CD34+, CD38 and SLAMF6+ are indicative of the individual having or developing leukemia
    • optionally wherein the method also includes a step comprising quantification of levels of immune cells (such as B cells, T cells and/or NK cells), preferably wherein the immune cells express SLAMF6.


58. A method according to any one of paragraphs 54 to 57 wherein the neoplastic hematologic disorder is a leukemia.


59. A method according to any one of paragraphs 54 to 58 wherein the neoplastic hematologic disorder is selected from the group consisting of chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML).


60. The method according to paragraph 59 wherein the neoplastic hematologic disorder is acute myeloid leukemia (AML).


61. An agent for use in medicine substantially as described herein with reference to the description.


62. A pharmaceutical composition substantially as described herein with reference to the description.


63. Use of an agent substantially as described herein with reference to the description.


64. A method of treatment or diagnosis as described herein with reference to the description.


65. A kit substantially as defined herein with reference to the description.

Claims
  • 1. A method for: (i) inducing cell death of pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder; and/or(ii) inhibiting the growth and/or proliferation of pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder; or(iii) detecting pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder,
  • 2. (canceled)
  • 3. A method according to claim 1, wherein the neoplastic hematologic disorder: (a) is a leukemia;(b) is associated with cells comprising a TP53 mutation; and/or(c) is selected from the group consisting of chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML), preferably acute myeloid leukemia (AML).
  • 4. A method according to claim 1, wherein the cells expressing SLAMF6 also express CD34+CD38−, optionally wherein the cells comprise a TP53 mutation.
  • 5. A method according to claim 1, wherein the binding moiety has specificity for human SLAMF6, and/or wherein SLAMF6 is localised on the surface of a cell.
  • 6. A method according to claim 1, wherein the agent is capable of modulating an interaction between an immune cell and leukemic cells and/or an immune cell and leukemic stem cells, optionally wherein the immune cells are selected from the group consisting of: B cells, T cells and/or NK cells; preferably wherein the immune cells express SLAMF6.
  • 7. A method according to claim 1, wherein the agent: (a) is capable of killing the pathological stem cells and/or progenitor cells, optionally by antibody-dependent cell-mediated cytotoxicity (ADCC) and/or by a T cell mediated mechanism;(b) is capable of inducing apoptosis of the stem cells and/or progenitor cells, optionally by antibody-dependent cell-mediated cytotoxicity (ADCC) and/or by a T cell mediated mechanism.
  • 8. A method according to claim 1, wherein the agent comprises or consists of: (a) a polypeptide; and/or(b) an antibody or an antigen-binding fragment thereof with binding specificity for SLAMF6, or a variant, fusion or derivative of said antibody or antigen-binding fragment, or a fusion of a said variant or derivative thereof, which retains the binding specificity for SLAMF6; and/or(c) an antibody or antigen-binding fragment thereof with binding specificity for SLAMF6, optionally wherein the agent comprises or consists of an intact antibody or an antigen-binding fragment of an antibody, such as an antigen-binding fragment selected from the group consisting of Fv fragments (e.g. single chain Fv, disulphide-bonded Fv and domain antibodies) and Fab-like fragments (e.g. Fab fragments, Fab′ fragments and F(ab)2 fragments); and/or(d) a recombinant antibody; or(e) a monoclonal antibody; or(f) a polyclonal antibody;optionally wherein the antibody or antigen-binding fragment thereof is human or humanised; or(g) a non-immunoglobulin binding moiety; and/or(h) an aptamer, such as a peptide aptamer or a nucleic acid aptamer; or(i) a small chemical entity.
  • 9. A method according to claim 1, wherein the agent further comprises: (a) a moiety for increasing the in vivo half-life of the agent, optionally wherein the moiety for increasing the in vivo half-life is selected from the group consisting of polyethylene glycol (PEG), human serum albumin, glycosylation groups, fatty acids and dextran; and/orwherein the agent is PEGylated; and/or(b) a cytotoxic moiety, optionally wherein the cytotoxic moiety comprises or consists of a radioisotope, such as a radioisotope selected from the group consisting of astatine-211, bismuth-212, bismuth-213, iodine-131, yttrium-90, lutetium-177, samarium-153 and palladium-109; orwherein the cytotoxic moiety comprises or consists of a toxin (such as saporin or calicheamicin) or a chemotherapeutic agent (such as an antimetabolite); and/or(c) a detectable moiety, optionally wherein the detectable moiety comprises or consists of a radioisotope, such as a radioisotope selected from the group consisting of: technetium-99m; indium-111; gallium-67; gallium-68; arsenic-72; zirconium-89; iodine-12; thallium-201; orwherein the detectable moiety comprises or consists of a paramagnetic isotope, such as a paramagnetic isotope selected from the group consisting of: gadolinium-157; manganese-55, dysprosium-162, chromium-52; iron-56.
  • 10. A pharmaceutical composition comprising an effective amount of an agent comprising or consisting of a binding moiety with specificity for SLAMF6 and a pharmaceutically-acceptable diluent, carrier or excipient, optionally adapted for parenteral delivery or intravenous delivery.
  • 11-12. (canceled)
  • 13. A method according to claim 1, wherein the method is for inducing cell death and/or inhibiting the growth and/or proliferation of pathological stem cells and/or progenitor cells associated with a neoplastic hematologic disorder in an individual, comprising the step of administering to the individual an effective amount of an agent comprising or consisting of a binding moiety with specificity for SLAMF6, wherein the cells express SLAMF6.
  • 14. A method according to claim 1, wherein the method is for detecting pathological stem cells and/or progenitor cells associated with neoplastic hematologic disorder in an individual and comprises the step of administering to the individual an effective amount of an agent comprising or consisting of a binding moiety with specificity for SLAMF6, wherein the cells express SLAMF6.
  • 15. An in vitro method for diagnosing or prognosing a neoplastic hematologic disorder, the method comprising: (a) providing a bone marrow or peripheral blood sample of haematopoietic cells from an individual to be tested;(b) isolating a subpopulation of CD34+, CD38− cells from the haematopoietic cells; and(c) determining whether stem cells, contained within the CD34+, CD38− cells, express the cell surface marker SLAMF6;wherein stem cells that exhibit the cell surface marker profile CD34+, CD38− and SLAMF6+ are indicative of the individual having or developing leukemia;
  • 16. A method according to claim 1, wherein the neoplastic hematologic disorder is a leukemia, such as a neoplastic hematologic disorder selected from the group consisting of chronic myeloid leukemia (CML), myeloproliferative disorders (MPD), myelodysplastic syndrome (MDS), acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML), preferably acute myeloid leukemia (AML).
  • 17. (canceled)
Priority Claims (1)
Number Date Country Kind
1951413-2 Dec 2019 SE national
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2020/084893 12/7/2020 WO