Biomarker signature for predicting tumor response to anti-CD200 therapy

Information

  • Patent Grant
  • 11761963
  • Patent Number
    11,761,963
  • Date Filed
    Wednesday, September 26, 2018
    5 years ago
  • Date Issued
    Tuesday, September 19, 2023
    7 months ago
Abstract
Provided herein are methods for treating cancer in a patient who has been determined to have positive expression of CD200 receptor (CD200R1) and one or more biomarkers (i.e., ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and/or CD14) by administering to the patient a CD200 inhibitor. Also provided are methods for monitoring responsiveness of a patient having cancer to treatment with a CD200 inhibitor, the method comprising: determining expression levels of CD200R1 and one or more biomarkers (i.e., ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and/or CD14) in a biological sample from the patient, wherein increased expression levels of CD200R1 and the one or more biomarkers, as compared to expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Mar. 18, 2020, is named AXJ-244US_Sequence_Listing txt and is 130,540 bytes in size.


BACKGROUND

The human immune system employs a variety of immune surveillance mechanisms, which can identify malignant cells within a host organism and kill the cells before a cancer develops (see, e.g., Geertsen et al. (1999) Int J Mol Med 3(1):49-57; Kerebijn et al. (1999) Crit. Rev Oncol Hematol 31(1):31-53; and Pardoll (2003) Annu Rev Immunol 21:807-39). However, cancer cells are known to evade detection by the immune system. One potential mechanism by which cancer cells escape immunosurveillance is expression or overexpression of CD200 (OX-2) protein and/or expression or overexpression of CD200 receptor.


CD200 is an immune checkpoint protein expressed by a number of immune cells, including B, T cells and macrophages, as well as non-immune cells, including endothelial cells and neurons. CD200 binds to its receptor (CD200R1), expressed on antigen-presenting cells (APCs) and T cells, and is believed to play an important role in normal immune homeostasis. However, CD200 protein has also been shown to be expressed or overexpressed on a variety of human cancer cells including, e.g., B cell chronic lymphocytic leukemia cells, prostate cancer cells, breast cancer cells, colon cancer cells, and brain cancer cells (see, e.g., Kawasaki et al. (2007) Biochem Biophys Res Commun 364(4):778-782; Kretz-Rommel et al. (2007); and Siva et al. (2008) Cancer Immunol Immunother 57(7):987-96). Overexpression of CD200 by tumor cells implicates CD200 in tumor-mediated immunosuppression and regulation of anti-tumor activity and is associated with worse outcomes.


The variable response rates of patients to monoclonal antibody therapies and chemotherapies means that methods are needed for accurately predicting which patients are likely to respond to therapeutic treatment, so that the treatment can be administered to only those patients who are likely to receive benefits that outweigh the financial costs and potential deleterious effects of treatment (including possible damage to the patient due to tumor growth over time during the administration of ineffective treatments). Particular biomarkers or sets of biomarkers (e.g., gene products such as proteins or RNAs) in tumors may be found for which a particular concentration range or expression level for each biomarker (e.g., in the set) correlates with tumor responsiveness to a particular therapy.


Accordingly, the following disclosure provides novel biomarker criteria that allow for optimization of tumor therapy using CD200 inhibitors, improved methods for treating cancer patients, and methods for monitoring the progression and abatement of cancer.


SUMMARY

Provided herein are methods for treating cancer in a patient who has been determined to have positive expression of CD200 receptor (CD200R1) and one or more biomarkers (i.e., Inducible T-cell COStimulator (ICOS), T Cell Immunoreceptor with Ig and ITIM Domains (TIGIT), Tumor Necrosis Factor Receptor Superfamily Member 9 (TNFRSF9), Hepatitis A Virus Cellular Receptor 2 (HAVCR2), and Programmed Cell Death 1 (PDCD1), Fc Fragment Of IgG Receptor IIa (FCGR2A), Fc Fragment Of IgG Receptor Ia (FCGR1A), Cluster of Differentiation 163 (CD163), and/or CD14), by administering to the patient a CD200 inhibitor.


In one embodiment, a method for treating a patient having cancer who has been determined to have positive expression of CD200 receptor (CD200R1) and one or more biomarkers (e.g., two or more, three or more, four or more, five or more, 6 or more, 7 or more, 8 or more, or 9) in a biological sample from the patient is provided, the method comprising administering to the patient a CD200 inhibitor in an amount and with a frequency sufficient to reduce the cancer burden in the patient (e.g., by about 30, 40, 50, 60, 70, 80, 90, or 100%). In one embodiment, the biomarker is ICOS. In another embodiment, the biomarker is TIGIT. In another embodiment, the biomarker is TNFRSF9. In another embodiment, the biomarker is HAVCR2. In another embodiment, the biomarker is PDCD1. In another embodiment, the biomarker is FCGR2A. In another embodiment, the biomarker is FCGR1A. In another embodiment, the biomarker is CD163. In another embodiment, the biomarker is CD14.


In one embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of two biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of three biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of four biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of five biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have an positive expression of CD200R1 and positive expression of six biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of seven biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of eight biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1, ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. The patient can have positive expression of any possible combination of the biomarkers disclosed herein.


In one embodiment, the method includes determining if a patient having cancer has positive expression of CD200 receptor (CD200R1) and one or more biomarkers (i.e., Inducible T-cell COStimulator (ICOS), T Cell Immunoreceptor with Ig and ITIM Domains (TIGIT), Tumor Necrosis Factor Receptor Superfamily Member 9 (TNFRSF9), Hepatitis A Virus Cellular Receptor 2 (HAVCR2), and Programmed Cell Death 1 (PDCD1), Fc Fragment Of IgG Receptor IIa (FCGR2A), Fc Fragment Of IgG Receptor Ia (FCGR1A), Cluster of Differentiation 163 (CD163), and/or CD14) and administering to the patient a CD200 inhibitor if the patient has positive expression of CD200R1 and one or more biomarkers.


In one embodiment, positive expression of CD200R1 in the biological sample is equal to or greater than expression of CD200R1 in a normal biological sample of the same type. In another embodiment, positive expression of the one more biomarkers in the biological sample is equal to or greater than expression of the one or more biomarkers in a normal biological sample of the same type.


Also, provided are methods for monitoring responsiveness of a subject having cancer to treatment with a CD200 inhibitor, the method comprising: determining expression levels of CD200R1 and one or more (e.g., two or more, three or more, four or more, five or more, 6 or more, 7 or more, 8 or more, or 9) biomarkers (i.e., ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and/or CD14) in a biological sample from the patient, wherein increased expression levels of CD200R1 and the one or more biomarkers, as compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor.


In one embodiment, increased expression levels of CD200R1 and one biomarker selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, expression levels of CD200R1 and two biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels ncentrations in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and three biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and four biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and five biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to expression levels in a biological f the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and six biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and seven biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to expression levels in a sample of biological the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and eight biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1, ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to expression levels in a sample of biological the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. The patient can have elevated expression levels of any possible combination of the biomarkers disclosed herein.


Any suitable CD200 inhibitor can be used in the methods described herein. In one embodiment, the CD200 inhibitor is a small molecule. In another embodiment, the CD200 inhibitor is a polypeptide. In another embodiment, the CD200 inhibitor is a polypeptide analog. In another embodiment, the CD200 inhibitor is a peptidomimetic. In another embodiment, the CD200 inhibitor is an aptamer.


In another embodiment, the CD200 inhibitor is an antibody, or an antigen-binding fragment thereof. For example, the antibody, or antigen-binding fragment thereof, can be a humanized antibody, a recombinant antibody, a diabody, a chimerized or chimeric antibody, a monoclonal antibody, a deimmunized antibody, a fully human antibody, a single chain antibody, an Fv fragment, an Fd fragment, an Fab fragment, an Fab′ fragment, or an F(ab′)2 fragment.


An exemplary anti-CD200 antibody is samalizumab (also known as “ALXN6000”).


In one embodiment, the anti-CD200 antibody antigen-binding fragment thereof, comprises a heavy chain variable region CDR1 having the sequence set forth in SEQ ID NO: 7, a heavy chain variable region CDR2 having the sequence set forth in SEQ ID NO: 8, a heavy chain variable region CDR3 having the sequence set forth in SEQ ID NO: 9, a light chain variable region CDR1 having the sequence set forth in SEQ ID NO: 4, a light chain variable region CDR2 having the sequence set forth in SEQ ID NO: 5, and a light chain variable region CDR3 having the sequence set forth in SEQ ID NO: 6.


In another embodiment, the anti-CD200 antibody comprises heavy and light chain variable regions having the sequences set forth in SEQ ID NOs: 13 and 12, respectively.


In another embodiment, the anti-CD200 antibody comprises heavy and light chains having the sequences as set forth in SEQ ID NOs: 11 and 10, respectively.


In another embodiment, the anti-CD200 antibody, or antigen binding fragment thereof, comprises the CDR1, CDR2 and CDR3 domains of a heavy chain variable region having the sequence set forth in SEQ ID NO: 13, and the CDR1, CDR2 and CDR3 domains of a light chain variable region having the sequence set forth in SEQ ID NO: 12.


In another embodiment, the anti-CD200 antibody, or antigen binding fragment thereof, comprising the CDR1, CDR2, and CDR3 domains of a heavy chain region having the sequence set forth in SEQ ID NO: 11, and the CDR1, CDR2, and CDR3 domains of a light chain region having the sequence set forth in SEQ ID NO: 10.


In another embodiment, the anti-CD200 antibody is a human antibody. In another embodiment, a composition of anti-CD200 antibodies, or fragments thereof, is use in the methods described herein, wherein the composition (e.g., a sterile composition) comprises a pharmaceutically acceptable carrier.


In another embodiment, the anti-CD200 antibody or antigen-binding fragment thereof inhibits the interaction between CD200 and CD200R1. In another embodiment, the method of treatment results in a CD200 saturation of at least 60%, 65%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.


In one embodiment, the anti-CD200 antibody is administered at a dose of about 300 mg/m2 to about 600 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 300 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 400 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 500 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 600 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 700 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 300 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 800 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 900 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 1000 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 1100 mg/m2.


In another embodiment, the anti-CD200 antibody is administered at a dose of about 5 mg/kg to about 50 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 10 mg/kg to about 30 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 15 mg/kg to about 25 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 10 mg/kg to about 20 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 10 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 15 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 20 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 25 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 40 mg/kg.


Expression levels CD200R1 and the one or more biomarkers can be measured by quantitation of protein and/or RNA levels in a biological sample from the patient (e.g., tumor tissue, tumor cells, blood, or a blood fraction) using any suitable technique. In one embodiment, expression levels are measured by quantitation of protein and/or RNA levels, using at least one of an immunoassay, immunochemistry assay, immunohistochemistry assay, nucleoprobe assay, in situ hybridization, fluorescent RNA probes, RT-PCR, microarray transcription assay, and/or RNA transcription assay. In another embodiment, expression levels are measured using an immunoassay (e.g., an enzyme-linked immunosorbent assay (ELISA) or a radioimmunoassay (RIA)).


In one embodiment, expression levels of CD200R1 and one or more biomarkers are measured in two or more types of biological samples. In another embodiment, expression levels of CD200R1 and one or more biomarkers are measured in one type of biological sample and levels of a second biomarker are measured in a second type of biological sample.


In one embodiment, positive expression of CD200R1 in the biological sample is equal to or greater than expression of CD200R1 in a normal biological sample of the same type. In another embodiment, positive expression of the one more biomarkers in the biological sample is equal to or greater than expression of the one or more biomarkers in a normal biological sample of the same type.


In one embodiment, the method further comprises measuring CD200 expression in the biological sample and identifying patients with tumors having elevated expression of CD200, wherein the elevated expression of CD200 in the biological sample is greater than median expression levels of CD200 in normal tissue.


In one embodiment, the patient is an adult and the cancer is selected from the group consisting of diffuse large B cell lymphoma (DLBL), lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), glioblastoma (GBM), low grade glioma (LGG), clear cell RCC (KIRC), chromophobe (KICH), papillary cell RCC (KIRP), melanoma (SKCM), ovarian cancer (OV), colon cancer (COAD), rectum cancer (READ), uterine endometrial cancer (UCEC).


In another embodiment, the patient is a pediatric patient, and the cancer is selected from atypical teratoid rhabdoid tumor (AT/RT), ependymoma, osteosarcoma, rhabdomyosarcoma, Ewing sarcoma, pilocytic astrocytoma, neuroblastoma, and retinoblastoma.


In one embodiment, the treatment produces at least one therapeutic effect, for example, morphologic complete remission, cytogenetic complete remission, morphologic CR with incomplete blood count recovery, partial remission, and/or stable disease.


Other features and advantages of the methods of treatment will be apparent from the following description, the examples, and from the claims. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 depicts the results of a gene expression analysis, wherein publically available human tumor gene expression data (TCGA) was mined and analyzed for the expression of CD200 by tumor type.



FIG. 2 is a flow chart that outlines the steps of the genomic analysis of Example 2.



FIG. 3 depicts the Pearson correlation results of the TCGA dataset analysis for ovarian cancer, and shows that CD200R1, but not CD200, significantly correlates with the expression of immune cell markers, across all tumor types examined.



FIG. 4 depicts normalized CD200 expression in pediatric tumors.



FIGS. 5A and 5B depict CD4 medium-centered expression and correlation with CD200 in pediatric tumors.



FIGS. 6A and 6B depict CD68 medium-centered expression and correlation with CD200 in pediatric tumors.



FIG. 7 shows the tissue microarrays and slides that were used for Example 3.



FIG. 8 shows the tissue samples that were analyzed for Example 3.



FIGS. 9A-9D depict the results of the IHC assay for control tissues (FIG. 9A), rhabdomyosarcoma (FIG. 9B), nephroblastoma (FIG. 9C), and neuroblastoma (FIG. 9D) (Example 3).



FIG. 10 depicts tumor CD200 expression as assessed by IHC (Example 3).



FIG. 11 depicts tumor infiltrates in all samples as shown by IHC (Example 3).



FIG. 12 depicts levels of immune infiltrates in brain neoplasia as shown by IHC (Example 3).



FIG. 13 depicts levels of immune infiltrates neuroblastomas as shown by IHC (Example 3).



FIGS. 14A-14D depict levels of immune infiltrates nephroblastomas as shown by IHC (Example 3).



FIGS. 15A-15D depict levels of immune infiltrates osteosarcomas as shown by IHC (Example 3).



FIG. 16 depicts samalizumab competent signature GSVA scores in pediatric tumors (Example 2).



FIG. 17 depicts samalizumab competent signature GSVA scores in adult tumors (Example 1).





DETAILED DESCRIPTION

As described herein and exemplified in the working Examples, the inventors have developed a “samalizumab competent” gene signature from genes which co-correlate with CD200R1 expression across tumor types, which includes biomarkers of T cells and macrophages.


This signature was used to identify tumor types which harbor CD200R1-expressing immune infiltrates that are responsive to CD200 inhibitors, including but not limited to samalizumab. Accordingly, analysis of expression and/or activity levels of these signature biomarkers can be employed to evaluate and/or treat patients having cancer and/or monitor treatment response to a CD200 inhibitor, such as samalizumab.


I. Definitions

In order that the present description may be more readily understood, certain terms are first defined. Additional definitions are set forth throughout the detailed description. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art, and conventional methods of immunology, protein chemistry, biochemistry, recombinant DNA techniques, and pharmacology are employed.


As used herein, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. The use of “or” or “and” means “and/or” unless stated otherwise. Furthermore, use of the term “including” as well as other forms, such as “include”, “includes”, and “included,” is not limiting.


The term “about” as used herein when referring to a measurable value such as an amount, a temporal duration and the like, is encompasses variations of up to ±10% from the specified value. Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, etc., used herein are to be understood as being modified by the term “about”.


As used herein, “cancer” refers a broad group of diseases characterized by the uncontrolled growth of abnormal cells in the body. Unregulated cell division may result in the formation of malignant tumors or cells that invade neighboring tissues and may metastasize to distant parts of the body through the lymphatic system or bloodstream. As used herein, the term includes pre-malignant, as well as malignant cancers.


An “immune response” refers to a biological response within a vertebrate against foreign agents, which response protects the organism against these agents and diseases caused by them. An immune response is mediated by the action of a cell of the immune system (for example, a T lymphocyte, B lymphocyte, natural killer (NK) cell, macrophage, eosinophil, mast cell, dendritic cell or neutrophil) and soluble macromolecules produced by any of these cells or the liver (including antibodies, cytokines, and complement) that results in selective targeting, binding to, damage to, destruction of, and/or elimination from the vertebrate's body of invading pathogens, cells or tissues infected with pathogens, cancerous or other abnormal cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues. An immune response or reaction includes, e.g., activation or inhibition of a T cell, e.g., an effector T cell or a Th cell, such as a CD4+ or CD8+ T cell, or the inhibition of a Treg cell.


“Immunotherapy” refers to the treatment of a subject afflicted with, or at risk of contracting or suffering a recurrence of, a disease by a method comprising inducing, enhancing, suppressing or otherwise modifying an immune response. “Immunostimulating therapy” or “immunostimulatory therapy” refers to a therapy that results in increasing (inducing or enhancing) an immune response in a subject for, e.g., treating cancer. “Potentiating an endogenous immune response” means increasing the effectiveness or potency of an existing immune response in a subject. This increase in effectiveness and potency may be achieved, for example, by overcoming mechanisms that suppress the endogenous host immune response or by stimulating mechanisms that enhance the endogenous host immune response.


As used herein, the terms “inhibits” or “blocks” (e.g., referring to inhibition/blocking of activity, function and/or the expression of CD200 and/or its receptor) are used interchangeably and encompass both partial and complete inhibition/blocking.


As used herein, the term “normal,” when used to modify the term “individual” or “subject” refers to an individual or group of individuals who does/do not have a particular disease or condition (e.g., cancer) and is also not suspected of having or being at risk for developing the disease or condition. The term “normal” is also used herein to qualify a biological specimen or sample (e.g., a biological fluid) isolated from a normal or healthy individual or subject (or group of such subjects), for example, a “normal control sample” or “normal control biological fluid”.


As used herein, the term “positive expression” refers to an expression level of a biomarker in a biological sample that is approximately equal to, or greater than, to the expression of the same biomarker in a normal biological sample of the same type. In this instance, biomarker may refer to the expression of a gene (i.e., RNA) and/or the expression of a peptide.


“Polypeptide,” “peptide,” and “protein” are used interchangeably and mean any peptide-linked chain of amino acids, regardless of length or post-translational modification. The proteins described herein can contain or be wild-type proteins or can be variants that have not more than 50 (e.g., not more than one, two, three, four, five, six, seven, eight, nine, ten, 12, 15, 20, 25, 30, 35, 40, or 50) conservative amino acid substitutions. Conservative substitutions typically include substitutions within the following groups: glycine and alanine; valine, isoleucine, and leucine; aspartic acid and glutamic acid; asparagine, glutamine, serine and threonine; lysine, histidine and arginine; and phenylalanine and tyrosine.


As used herein, percent (%) amino acid sequence identity is defined as the percentage of amino acids in a candidate sequence that are identical to the amino acids in a reference sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST software. Appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared can be determined by known methods.


The term “antibody” as used herein refers to polypeptides comprising at least one antibody derived antigen binding site (e.g., VH/VL region or Fv, or CDR), and includes whole antibodies and any antigen binding fragments (i.e., “antigen-binding portions”) or single chains thereof. Antibodies include known forms of antibodies. For example, the antibody can be a human antibody, a humanized antibody, a bispecific antibody, or a chimeric antibody. A whole “antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, in which each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region; and each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.


The exact boundaries of CDRs can be defined differently according to different methods. In some embodiments, the positions of the CDRs or framework regions within a light or heavy chain variable domain can be as defined by Kabat et al. [(1991) “Sequences of Proteins of Immunological Interest.” NIH Publication No. 91-3242, U.S. Department of Health and Human Services, Bethesda, Md.]. In such cases, the CDRs can be referred to as “Kabat CDRs” (e.g., “Kabat LCDR2” or “Kabat HCDR1”). In other embodiments, the positions of the CDRs of a light or heavy chain variable region can be as defined by Chothia et al. (1989) Nature 342:877-883. Accordingly, these regions can be referred to as “Chothia CDRs” (e.g., “Chothia LCDR2” or “Chothia HCDR3”). In other embodiments, the positions of the CDRs of the light and heavy chain variable regions can be as defined by a Kabat-Chothia combined definition. In such embodiments, these regions can be referred to as “combined Kabat-Chothia CDRs”. Thomas et al. [(1996) Mol Immunol 33(17/18):1389-1401] exemplifies the identification of CDR boundaries according to Kabat and Chothia definitions. In other embodiments, the positions of the CDRs or framework regions within a light or heavy chain variable domain can be as defined by the international ImMunoGeneTics database (IMGT) standard. Marie-Paule Lefranc et al. [(2003) Developmental & Comparative Immunology 27(1):55-77] exemplifies the identification of and CDR boundaries according to IMGT standard. Accordingly, these regions can be referred to as “IMGT CDRs” (e.g., “IMGT-LCDR2” or “IMGT-HCDR3”).


The antibody also can be of any of the following isotypes: IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgAsec, IgD, and IgE. The antibody may be a naturally occurring antibody or may be an antibody that has been altered by a protein engineering technique (e.g., by mutation, deletion, substitution, and/or conjugation to a non-antibody moiety). For example, an antibody may include one or more variant amino acids (compared to a naturally occurring antibody) which change a property (e.g., a functional property) of the antibody. For example, numerous such alterations are known in the art which affect, e.g., half-life, effector function, and/or immune responses to the antibody in a patient. The term antibody also includes artificial or engineered polypeptide constructs which comprise at least one antibody-derived antigen binding site.


The term “human antibody,” as used herein, is intended to include antibodies having variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences as described, for example, by Kabat et al. (See Kabat, et al. (1991) Sequences of proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242).


The term “antigen-binding portion” of an antibody (or simply “antibody portion”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., CD200), e.g., a Fab, Fab′2, ScFv, SMIP, AFFIBODY® antibody mimetic (Affibody AB AKTIEBOLAG, Sweden), nanobody, or a domain antibody. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. Other forms of single chain antibodies, such as diabodies are also encompassed. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see, e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123). In one embodiment, the composition contains an antigen-binding portions described in U.S. Pat. Nos. 6,090,382 and 6,258,562, each incorporated by reference herein.


The term “monoclonal antibody,” as used herein, includes an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Monoclonal antibodies are advantageous in that they may be synthesized by a hybridoma culture, essentially uncontaminated by other immunoglobulins. The modifier “monoclonal” indicates the character of the antibody as being amongst a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. The monoclonal antibodies to be used in accordance with the formulations disclosed herein may be made by the hybridoma method first described by Kohler, et al., (1975) Nature 256: 495 or other methods known in the art. A “polyclonal antibody” is an antibody which was produced among or in the presence of one or more other, non-identical antibodies. In general, polyclonal antibodies are produced from a B-lymphocyte in the presence of several other B-lymphocytes which produced non-identical antibodies. Usually, polyclonal antibodies are obtained directly from an immunized animal.


An “isolated” antibody or antigen binding fragment is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with research, diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes. In some embodiments, an antibody is purified to greater than 95% by weight of antibody, and in some embodiments, to greater than 99% by weight of antibody.


The term “pharmaceutical formulation” or “pharmaceutical composition” refers to preparations which are in such form as to permit the biological activity of the active ingredients to be unequivocally effective, and which contain no additional components which are significantly toxic to the subjects to which the formulation would be administered.


As used herein, an “aqueous” pharmaceutical composition is a composition suitable for pharmaceutical use, wherein the aqueous carrier comprises water. A composition suitable for pharmaceutical use may be sterile, homogeneous, and/or isotonic. Aqueous pharmaceutical compositions may be prepared directly in an aqueous form and/or may be reconstituted from a lyophilisate and/or powder form.


A “sterile” composition is aseptic or free or essentially free from all living microorganisms and their spores.


As used herein, the terms “specific binding,” “selective binding,” “selectively binds,” and “specifically binds,” refer to antibody binding to an epitope on a predetermined antigen but not to other antigens. Typically, the antibody (i) binds with an equilibrium dissociation constant (KD) of approximately less than 10−7 M, such as approximately less than 10−8 M, 10−9 M or 10−10 M or even lower when determined by, e.g., surface plasmon resonance (SPR) technology in a BIACORE® 2000 surface plasmon resonance instrument using the predetermined antigen, e.g., recombinant human CD200, as the analyte and the antibody as the ligand, or Scatchard analysis of binding of the antibody to antigen positive cells, and (ii) binds to the predetermined antigen with an affinity that is at least two-fold greater than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely-related antigen. Accordingly, unless otherwise indicated, an antibody that “specifically binds to human CD200” refers to an antibody that binds to soluble or cell bound human CD200 with a KD of 10−7 M or less, such as approximately less than 10−8 M, 10−9 M, or 10−10 M or even lower.


An “epitope” refers to the site on a protein (e.g., a human CD200 protein) that is bound by an antibody. “Overlapping epitopes” include at least one (e.g., two, three, four, five, or six) common amino acid residue(s).


The term “surface plasmon resonance”, as used herein, refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Piscataway, N.J.). For further descriptions, see Jonsson, U., et al. (1993) Ann. Biol. Clin. 51:19-26; Jonsson, U., et al. (1991) Biotechniques 11:620-627; Johnsson, B., et al. (1995) J. Mol. Recognit. 8:125-131; and Johnnson, B., et al. (1991) Anal. Biochem. 198:268-277.


The term “Koff”, as used herein, is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex.


The term “Kd”, as used herein, is intended to refer to the dissociation constant of a particular antibody-antigen interaction.


Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure pertains. Preferred methods and materials are described below, although methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the presently disclosed methods and compositions. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.


Other features and advantages of the present disclosure, e.g., methods for treating cancer in a subject, will be apparent from the following description, the examples, and from the claims.


1. Biomarkers


Biomarkers provided herein can be used as an indicator to, e.g., evaluate whether a patient having cancer will be responsive to treatment with a CD200 inhibitor (including, but not limited to samalizumab) and/or to monitoring response to treatment with a CD200 inhibitor).


A. Signature Biomarkers


The GenBank (National Center for Biotechnology Information (NCBI)) reference numbers for the protein and gene sequences associated with each of the biomarkers described herein are listed below in Tables 1-2, the sequences of which are all expressly incorporated herein by reference. Additionally information regarding the individual biomarkers follows Tables 1-2.









TABLE 1





T Cell Markers















Inducible T-cell COStimulator


(also known as “ICOS”, “CD278”, “Activation-Inducible Lymphocyte Immunomediatory


Molecule”, “AILIM”, “Inducible T-Cell Co-Stimulator”, “Inducible Costimulator”, “CD278


Antigen”, “CD278”, and “CVID1”).


NCBI Reference Sequence: NP_036224.1 (SEQ ID NO: 14)


NCBI Reference Sequence: NM_012092.3 (SEQ ID NO: 15)


T Cell Immunoreceptor with Ig and ITIM Domains


(also known as “TIGIT,” “WUCAM,” “Vstm3,” “T-Cell Immunoreceptor With Ig And ITIM


Domains V-Set And Immunoglobulin Domain-Containing Protein,” “V-Set And


Transmembrane Domain-Containing Protein,” “V-Set And Immunoglobulin Domain


Containing 9”, “V-Set And Transmembrane Domain Containing 3,” “VSIG9,” “VSTM3,” “T


Cell Immunoreceptor With Ig And ITIM Domains,” “Washington University Cell Adhesion


Molecule,” and “WUCAM”)


NCBI Reference Sequence: NP_776160.2 (SEQ ID NO: 16)


NCBI Reference Sequence: NM_173799.3, mRNA (SEQ ID NO: 17)


Tumor Necrosis Factor Receptor Superfamily Member 9


(also known as “TNFRSF9”, “CD37”, “4-1BB”, “induced by lymphocyte activation”, “ILA”,


“TNF Receptor Superfamily Member 9,” “T-Cell Antigen 4-1BB Homolog,” “4-1BB Ligand


Receptor,” “CD137 Antigen,” “CDwl37,” “CD137,”, “Interleukin-Activated Receptor,


Homolog Of Mouse Ly63,” “Tumor Necrosis Factor Receptor Superfamily, Member 9,”


“Tumor Necrosis Factor Receptor Superfamily Member 9,” “Homolog Of Mouse 4-1BB,”


“Receptor Protein 4-1BB,” “T Cell Antigen ILA,” and “T-Cell Antigen ILA”)


NCBI Reference Sequence: NP_001552.2 (SEQ ID NO: 18)


NCBI Reference Sequence: NM_001561.5 (SEQ ID NO: 19)


Hepatitis A Virus Cellular Receptor 2


(also known as “HAVCR2,” “T-Cell Immunoglobulin And Mucin Domain-Containing


Protein,” “T-Cell Immunoglobulin Mucin Family Member,”


“T-Cell Immunoglobulin Mucin Receptor,” “T-Cell Membrane Protein”, “HAVcr-2,” “TIMD-


3”, “Tim-3”, “TIMD3”, “TIM3”, “T Cell Immunoglobulin Mucin”, “Kidney Injury Molecule-


3”, “CD366”, and “KIM-3”)


NCBI Reference Sequence: NP_116171.3 (SEQ ID NO: 20)


NCBI Reference Sequence: NM_032782.4 (SEQ ID NO: 21)


Programmed Cell Death 1


(also known as “PDCD1”, “Systemic Lupus Erythematosus Susceptibility”, “Protein PD-1”,


“HPD-1”, “PD1”, “Programmed Cell Death 1 Protein”, “Programmed Cell Death Protein 1”,


“CD279 Antigen”, “CD279”, “HPD-L”, “HSLE1”, “SLEB2”, and “PD-1”)


NCBI Reference Sequence: NP_005009.2 (SEQ ID NO: 22)


NCBI Reference Sequence: NM_005018.2 (SEQ ID NO: 23)
















TABLE 2





Macrophage Markers















Fc Fragment Of IgG Receptor IIa


(also known as “FCGR2A”, “Fc Fragment Of IgG, Low Affinity IIa”, “Receptor (CD32)”,


“Immunoglobulin G Fc Receptor II”, “IgG Fc Receptor II-A”, “Fc-Gamma-RIIa”, “FcRII-A”,


“FCGR2A1”, “CDw32”, “IGFR2”, “FCG2”,


“CD32”, “Fc Fragment Of IgG, Low Affinity IIa, Receptor For (CD32)”, “Low Affinity


Immunoglobulin Gamma Fc Region Receptor II-A”, “Fc Gamma Receptor IIa,” “Fc-Gamma


RII-A”, “CD32 Antigen”, “CD32A” “FCGR2”, and “FcGR 3”)


NCBI Reference Sequence: NP_001129691.1 (SEQ ID NO: 24) [Isoform 1]


NCBI Reference Sequence: NM_001136219.1 (SEQ ID NO: 25)


NCBI Reference Sequence: NP_067674.2 (SEQ ID NO: 26) [Isoform 2]


NCBI Reference Sequence: NM_021642.3 (SEQ ID NO: 27)


Fc Fragment Of IgG Receptor Ia


(also known as “FCGR1A”, “Fc Fragment Of IgG Receptor Ia”, “Fc Fragment Of IgG, High


Affinity Ia, Receptor For (CD64)”, “Fc Fragment Of IgG, High Affinity Ia, Receptor (CD64)”,


“Fc Gamma Receptor Ia”, “IgG Fc Receptor I”, “Fc-Gamma RIA”, “Fc-Gamma RI”,


“FcgammaRIa” and “IGFR1”)


NCBI Reference Sequence: NP_000557.1 (SEQ ID NO: 28)


NCBI Reference Sequence: NM_000566.3 (SEQ ID NO: 29)


Cluster of Differentiation 163


(also known as “CD163”, “CD163 Molecule”, “Hemoglobin Scavenger Receptor”, “CD163


Antigen”, “M130”, “Scavenger Receptor Cysteine-Rich Type 1 Protein M130”, “Macrophage-


Associated Antigen”, “SCARI1”, and “MM130”)


NCBI Reference Sequence: NP_004235.4 (SEQ ID NO: 30)


NCBI Reference Sequence: NM_004244.5 (SEQ ID NO: 31)


NCBI Reference Sequence: NP_981961.2 (SEQ ID NO: 32)


NCBI Reference Sequence: NM_203416.3 (SEQ ID NO: 33)


CD14


(also known as “CD14 Molecule”, “CD14 Antigen 2”, and “Myeloid Cell-Specific Leucine-


Rich Glycoprotein”).


NCBI Reference Sequence: NP_000582.1_(SEQ ID NO: 34)


NCBI Reference Sequence: NM_000591.3 (SEQ ID NO: 35)


NCBI Reference Sequence: NP_001035110.1 (SEQ ID NO: 36)


NCBI Reference Sequence: NM_001040021.2 (SEQ ID NO: 37)


NCBI Reference Sequence: NP_001167575.1 (SEQ ID NO: 38)


NCBI Reference Sequence: NM_001174104.1 (SEQ ID NO: 39)


NCBI Reference Sequence: NP_001167576.1 (SEQ ID NO: 40)


NCBI Reference Sequence: NM_001174105.1 (SEQ ID NO: 41)









ICOS is an immune checkpoint protein that in humans is encoded by the ICOS gene (see, e.g., Hutloff A, et al. (January 1999), Nature 397 (6716): 263-6; and Yoshinaga et al. (December 1999), Nature 402 (6763): 827-32). ICOS is a CD28-superfamily costimulatory molecule that is expressed on activated T cells. It is thought to be important for Th2 cells, in particular (see, e.g., Rudd C E, et al. (July 2003), Nature Reviews: Immunology 3 (7): 544-56; and Dong et al. (January 2001), Nature 409 (6816): 97-101). It forms homodimers and plays an important role in cell-cell signaling, immune responses and regulation of cell proliferation.


TIGIT is an immune receptor present on some T cells and Natural Killer Cells(NK) (see, e.g., Yu X, et al. (January 2009). Nat Immunol. 10 (1): 48-57). TIGIT Binds with high affinity to the poliovirus receptor (PVR), which causes increased secretion of IL10 and decreased secretion of IL12B and suppresses T-cell activation by promoting the generation of mature immunoregulatory dendritic cells.


TNFRSF9 is a member of the tumor necrosis factor (TNF) receptor family (see, e.g., Schwarz H, et al., (1993), Gene 134 (2): 295-8). TNFRSF9 can be expressed by activated T cells, but to a larger extent on CD8 than on CD4 T cells. In addition, TNFRSF9 expression is found on dendritic cells, B cells, follicular dendritic cells, natural killer cells, granulocytes, and cells of blood vessel walls at sites of inflammation.


HAVCR2 belongs to the immunoglobulin superfamily, and TIM family of proteins. CD4-positive T helper lymphocytes can be divided into types 1 (Th1) and 2 (Th2) on the basis of their cytokine secretion patterns. Th1 cells are involved in cell-mediated immunity to intracellular pathogens and delayed-type hypersensitivity reactions, whereas, Th2 cells are involved in the control of extracellular helminthic infections and the promotion of atopic and allergic diseases. This protein is a Th1-specific cell surface protein that regulates macrophage activation, and inhibits Th1-mediated auto- and alloimmune responses, and promotes immunological tolerance (see, e.g., Monney L, et al. (February 2002), Nature 415 (6871): 536-41).


PDCD1 is a cell surface membrane protein of the immunoglobulin superfamily that plays an important role in down-regulating the immune system and promoting self-tolerance by suppressing T cell inflammatory activity. PDCD1 is an immune checkpoint and guards against autoimmunity through a dual mechanism of promoting apoptosis (programmed cell death) in antigen specific T-cells in lymph nodes while simultaneously reducing apoptosis in regulatory T cells (anti-inflammatory, suppressive T cells) (see, e.g., Francisco L M, et al. (July 2010), Immunological Reviews 236: 219-42; and Fife B T, Pauken K E (January 201), Ann. NY Acad. of Sci. 1217: 45-59).


Receptors for the Fc portion of IgG, such as FCGR2A, play an essential role in the protection of the organism against foreign antigens by removing antigen-antibody complexes from the circulation (Hibbs, M. et al., Proc. Nat. Acad. Sci. 85: 2240-2244, 1988). Receptors are present on monocytes, macrophages, neutrophils, natural killer (NK) cells, and T and B lymphocytes, and they participate in diverse functions such as phagocytosis of immune complexes and modulation of antibody production by B cells. Alternative splicing results in multiple transcript variants.


Fc-gamma receptors (FCGRs), such as FCGR1A, are integral membrane glycoproteins that exhibit complex activation or inhibitory effects on cell functions after aggregation by complexed immunoglobulin G (IgG). FCGR1A is a 72-kD activating FCGR found exclusively on antigen-presenting cells (APCs) of macrophage and dendritic cell (DC) lineages and has a high affinity for monomeric IgG1 (Rodrigo, W., et al, J. Virol. 80: 10128-10138, 2006).


CD163 is a protein that in humans is encoded by the CD163 gene. CD163 is the high affinity scavenger receptor for the hemoglobin-haptoglobin complex and in the absence of haptoglobin—with lower affinity—for hemoglobin alone (see, e.g., Schaer D J, et al., Blood. 2006 Jan. 1; 107(1):373-80). It has also been shown to mark cells of monocyte/macrophage lineage (see, e.g., Lau S K, et al., Am. J. Clin. Path. 122 (5): 794-801). The receptor was discovered in 1987 (see, e.g., Onofre G, et al., Acta Medica (Hradec Kralove). 52 (2): 57-61). CD163 functions as an acute phase-regulated receptor involved in the clearance and endocytosis of hemoglobin/haptoglobin complexes by macrophages, and may thereby protect tissues from free hemoglobin-mediated oxidative damage. This protein may also function as an innate immune sensor for bacteria and inducer of local inflammation. Alternatively spliced transcript variants encoding different isoforms have been described for this gene.


CD14 was first identified on the surface of monocytes and macrophages (see Griffin J D, et al., J. Clin. Invest. 1981; 68: 932-41). At the first leucocyte typing workshop in Paris in 1982, several monoclonal antibodies binding to the same epitope on human monocytes were assigned to a provisional CD14 cluster, which was labelled as a leucocyte differentiation antigen. Later, the level of expression of CD14 on these cells was reported to be in the order of 30,000-45,000 copies (see Van Voorhis, et al., J. Exp. Med. 1983; 158: 126-45; and Vasselon T, et al., J. Immunol. 1997; 159: 4498-505). Another very thorough analysis using both reference beads and Scatchard analysis estimated the number to be greater: approximately 110,000 molecules per monocyte (Antal-Szalmas P, et al. J. Leukoc. Biol. 1997; 61: 721-8). Because of its abundance on these cells, CD14 is widely used as a monocyte/macrophage marker in immunohistochemistry as well as in flow cytometry. CD14 is a 55 kDa glycoprotein with multiple leucine-rich repeats (Setoguchi M, et al., Biochim. Biophys. Acta. 1989; 1008: 213-22; and Ferrero E, et al., J. Immunol. 1990; 145: 331-6). It is encoded on chromosome 5q23-31, together with IL-3, GM-CSF, epidermal growth factor (EGF) receptor, beta2 adrenergic receptor and platelet-derived growth factor (PDGF) (see Goyert S M, et al. Science 1988; 239: 497-500). CD14 is attached to the cell membrane via a glycosylphosphatidylinositol (GPI) anchor, which is encoded on the X chromosome (Haziot A, et al., J. Immunol. 1988; 141: 547-52; and Takeda J, et al., Cell 1993; 73: 703-11).


In some embodiments, CD200R1 expression levels are determined in combination with one or more biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In some embodiments, at least two, three, four, five, six, seven, eight, or nine, of the biomarker proteins set forth in Tables 1 and 2 can be used in combination as a panel, in addition to CD200R1. The expression levels and/or activity of one or more of the biomarkers in Tables 1 and 2 (or any of the subsets of biomarkers mentioned herein) can be measured in combination with the expression level and/or activity of CD200R1.


B. Biomarker Measurements


Measuring or determining protein expression levels in a biological sample may be performed by any suitable method (see, e.g., Harlow and Lane (1988) “Antibodies: A Laboratory Manual,” Cold Spring Harbor Laboratory: Cold Spring Harbor, N.Y.).


In general, protein levels are determined by contacting a biological sample obtained from a subject with binding agents for one or more of the biomarker proteins; detecting, in the biological sample the expression level (e.g., levels) of one or more of the biomarker proteins that bind to the binding agents; and comparing the levels of one or more of the biomarker proteins in the sample with the levels of the corresponding protein biomarkers in a control sample (e.g., a normal sample). In certain embodiments, a suitable binding agent is a ribosome, with or without a peptide component, an RNA molecule, or a polypeptide (e.g., a polypeptide that comprises a polypeptide sequence of a protein marker, a peptide variant thereof, or a non-peptide mimetic of such a sequence).


Suitable binding agents also include an antibody specific for a biomarker protein described herein (e.g., an antibody specific for any biomarker listed in Table 1 or Table 2). Suitable antibodies for use in the methods of the present invention include monoclonal and polyclonal antibodies and antigen-binding fragments (e.g., Fab fragments or scFvs) of antibodies. Antibodies, including monoclonal and polyclonal antibodies, fragments and chimeras, may be prepared using methods known in the art (see, for example, Kohler and Milstein (1975) Nature 256:495-497; Kozbor et al. (1985) J Immunol Methods 81:31-42; Cote et al. (1983) Proc Natl Acad Sci USA 80:2026-203; and Zhang et al. (2002) J Biol Chem 277:39379-39387). Antibodies to be used in the methods of the invention can be purified by methods well known in the art. Antibodies may also be obtained from commercial sources.


In certain embodiments, the binding agent is directly or indirectly labeled with a detectable moiety. The role of a detectable agent is to facilitate the detection step of the diagnostic method by allowing visualization of the complex formed by binding of the binding agent to the protein marker (or fragment thereof). The detectable agent can be selected such that it generates a signal that can be measured and whose intensity is related (preferably proportional) to the amount of protein marker present in the sample being analyzed. Methods for labeling biological molecules such as polypeptides and antibodies are well-known in the art. Any of a wide variety of detectable agents can be used in the practice of the present invention. Suitable detectable agents include, but are not limited to: various ligands, radionuclides, fluorescent dyes, chemiluminescent agents, microparticles (such as, for example, quantum dots, nanocrystals, phosphors and the like), enzymes (such as, e.g., those used in an ELISA, i.e., horseradish peroxidase, beta-galactosidase, luciferase, alkaline phosphatase), colorimetric labels, magnetic labels, and biotin, digoxigenin or other haptens and proteins for which antisera or monoclonal antibodies are available.


In certain embodiments, the binding agents (e.g., antibodies) may be immobilized on a carrier or support (e.g., a bead, a magnetic particle, a latex particle, a microtiter plate well, a cuvette, or other reaction vessel). Examples of suitable carrier or support materials include agarose (SEPHAROSE®, Pharmacia), cellulose, nitrocellulose, dextran, cross-linked dextran gel (SEPHADEX®, Pharmacia), liposomes, carboxymethyl cellulose, polyacrylamides, polystyrene, gabbros, filter paper, magnetite, ion-exchange resin, plastic film, plastic tube, glass, polyamine-methyl vinyl-ether-maleic acid copolymer, amino acid copolymer, ethylene-maleic acid copolymer, nylon, silk, and the like. Binding agents may be indirectly immobilized using second binding agents specific for the first binding agents (e.g., mouse antibodies specific for the protein markers may be immobilized using sheep anti-mouse IgG Fc fragment specific antibody coated on the carrier or support).


Protein expression levels in a biological sample may be determined using immunoassays. Examples of such assays are time resolved fluorescence immunoassays (TR-FIA), radioimmunoas says, enzyme immunoassays (e.g., ELISA), immunofluorescence immunoprecipitation, latex agglutination, hemagglutination, Western blot, and histochemical tests, which are conventional methods well-known in the art. Methods of detection and quantification of the signal generated by the complex formed by binding of the binding agent with the protein marker will depend on the nature of the assay and of the detectable moiety (e.g., fluorescent moiety).


In one example, the presence or amount of protein expression of a gene (e.g., a biomarker protein depicted in Table 1 or Table 2) can be determined using a Western blotting technique. For example, a lysate can be prepared from a biological sample, or the biological sample (e.g., biological fluid) itself, can be contacted with Laemmli buffer and subjected to sodium-dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). SDS-PAGE-resolved proteins, separated by size, can then be transferred to a filter membrane (e.g., nitrocellulose) and subjected to immunoblotting techniques using a detectably-labeled antibody specific to the protein of interest. The presence or amount of bound detectably-labeled antibody indicates the presence or amount of protein in the biological sample.


In another example, an immunoassay can be used for detecting and/or measuring the protein expression of a biomarker protein (e.g., one depicted in Table 1 or Table 2). As above, for the purposes of detection, an immunoassay can be performed with an antibody that bears a detection moiety (e.g., a fluorescent agent or enzyme). Proteins from a biological sample can be conjugated directly to a solid-phase matrix (e.g., a multi-well assay plate, nitrocellulose, agarose, encoded particles, or magnetic beads) or it can be conjugated to a first member of a specific binding pair (e.g., biotin or streptavidin) that attaches to a solid-phase matrix upon binding to a second member of the specific binding pair (e.g., streptavidin or biotin). Such attachment to a solid-phase matrix allows the proteins to be purified away from other interfering or irrelevant components of the biological sample prior to contact with the detection antibody and also allows for subsequent washing of unbound antibody. Here, as above, the presence or amount of bound detectably-labeled antibody indicates the presence or amount of protein in the biological sample.


Alternatively, the protein expression levels may be determined using mass spectrometry based methods or image-based methods known in the art for the detection of proteins. Other suitable methods include 2D-gel electrophoresis, proteomics-based methods such as the identification of individual proteins recovered from the gel (e.g., by mass spectrometry or N-terminal sequencing) and/or bioinformatics.


Methods for detecting or measuring protein expression can, optionally, be performed in formats that allow for rapid preparation, processing, and analysis of multiple samples. This can be, for example, in multi-well assay plates (e.g., 96 wells or 386 wells) or arrays (e.g., protein chips). Stock solutions for various reagents can be provided manually or robotically, and subsequent sample preparation, pipetting, diluting, mixing, distribution, washing, incubating (e.g., hybridization), sample readout, data collection (optical data) and/or analysis (computer aided image analysis) can be done robotically using commercially available analysis software, robotics, and detection instrumentation capable of detecting the signal generated from the assay. Examples of such detectors include, but are not limited to, spectrophotometers, luminometers, fluorimeters, and devices that measure radioisotope decay. Exemplary high-throughput cell-based assays (e.g., detecting the presence or level of a target protein in a cell) can utilize commercially available reader technologies, i.e., ARRAYSCAN™ VTI HCS Reader or KINTETICSCAN® HCS Reader technology (Thermo Fisher Scientific, Waltham, Mass.).


In some embodiments, the protein expression level (or activity) of at least two biomarker proteins (e.g., at least three proteins, at least four proteins, at least five proteins, at least six proteins, at least seven proteins, at least eight proteins, at least nine proteins, or at least 10 proteins) can be assessed and/or measured.


Expression of the biomarker can also be detected at the nucleic acid level (e.g., based on RNA levels). In one embodiment, RNA is detected using an RNA-ISH assay. Another method for determining the level of RNA in a sample involves the process of nucleic acid amplification from homogenized tissue, e.g., by RT-PCR (reverse transcribing the RNA and then, amplifying the resulting cDNA employing PCR or any other nucleic acid amplification method, followed by the detection of the amplified molecules. In another embodiment, RNA expression is assessed by quantitative fluorogenic RT-PCR (qPCR).


In one embodiment, expression levels of CD200R1 and one or more biomarkers are measured in two or more types of biological samples. In another embodiment, expression levels of CD200R1 and one or more biomarkers are measured in one type of biological sample and levels of a second biomarker are measured in a second type of biological sample.


In one embodiment, the methods described herein involve comparing the measured expression level or activity of a biomarker protein (as measured in a biological sample obtained from a subject) to a control sample. In some embodiments, control sample is obtained from the subject prior to administering to the subject the CD200 inhibitor (e.g., samalizumab). In some embodiments, the control sample can be (or can be based on), e.g., a collection of samples obtained from one or more (e.g., two, three, four, five, six, seven, eight, nine, 10, 15, 20, 25, 30, 35, or 40 or more) healthy individuals that have not been administered CD200 inhibitor. In some embodiments, the control sample can be (or can be based on), e.g., a pooled sample obtained from two or more (e.g., two, three, four, five, six, seven, eight, nine, 10, 15, 20, 25, 30, 35, or 40 or more) individuals. In some embodiments of any of the methods described herein, the pooled samples can be from healthy individuals, or at least, individuals who do not have or are not suspected of having cancer. In another embodiment, determining whether the expression level or activity of an biomarker has increased following treatment with a CD200 inhibitor can involve comparing the expression level or activity of the biomarker in a biological sample obtained from a subject prior to treatment to the expression level of the biomarker in a sample of the same biological type obtained from the patient after treatment with the inhibitor (e.g., one day, two days, three days, four days, five days, six days, 1 week, 2 weeks, 3 weeks, a month, 6 weeks, two months, or three months after treatment with the inhibitor).


In some embodiments, determining whether a CD200 inhibitor has produced a desired effect (e.g., a reduction in cancer burden (e.g., by about 30, 40, 50, 60, 70, 80, 90, or 100%)) in a human can be performed by querying whether the post-treatment expression level of the biomarker falls within a predetermined range indicative of responsiveness to a CD200 inhibitor by a human. In some embodiments, determining whether a CD200 inhibitor has produced a desired effect in a human can include querying if the post-treatment expression level or activity of one or more biomarkers falls above or below a predetermined cut-off value. A cut-off value is typically the expression level or activity of a given biomarker in a given biological sample above or below which is considered indicative of a certain phenotype—e.g., responsiveness to therapy with a CD200 inhibitor.


In some embodiments of any of the methods described herein, the same practitioner may administer the CD200 inhibitor to the subject prior to determining whether a change in the expression level or activity of one or more biomarkers has occurred, whereas in some embodiments, the practitioner who administers the inhibitor to the subject is different from the practitioner who determines whether a response has occurred in the subject. In some embodiments, the practitioner may obtain a biological sample from the subject prior to administration of the inhibitor. In some embodiments, the practitioner may obtain a biological sample from the subject following the administration of the inhibitor to the subject. In some embodiments, the post-treatment sample can be obtained from the subject less than 48 (e.g., less than 47, 46, 45, 44, 43, 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, nine, eight, seven, six, five, four, three, two, or even less than one) hour following administration of the inhibitor to the subject. In some embodiments, the post-treatment sample can be obtained from the subject less than 20 (e.g., less than 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, nine, eight, seven, six, five, four, three, two, or one) day(s) after administering to the subject the inhibitor. In some embodiments, the biological sample is obtained from the subject no more than 20 (e.g., no more than 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, nine, eight, seven, six, five, four, three, two, or one) day(s) after the inhibitor is administered to the subject.


In some embodiments, the expression level of CD200R1 and at least one (e.g., at least two, three, four, five, six, seven, eight, or nine) biomarker is increased by at least 5 (e.g., at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, or 70) % following administration of the inhibitor.


In some embodiments, the expression level of CD200R1 and at least one (e.g., at least two, three, four, five, six, seven, eight, or nine) biomarkers is increased to within 50 (e.g., 49, 48, 47, 46, 45, 44, 43, 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1) % of the normal expression level of the biomarker following administration of one or more doses of the inhibitor.


In some embodiments of any of the methods described herein, the expression level of ICOS is increased by at least about 40% (e.g., 40, 45, 50, 60, 665, 70, 80, 85, 90, 95, or up to 100%) following administration of a CD200 inhibitor (e.g., samalizumab). In some embodiments of any of the methods described herein, the expression level of TIGIT is increased by at least about 40% (e.g., 40, 45, 50, 60, 665, 70, 80, 85, 90, 95, or up to 100%) following administration of a CD200 inhibitor (e.g., samalizumab). In some embodiments of any of the methods described herein, the expression level of TNFRSF9 is increased by at least about 40% (e.g., 40, 45, 50, 60, 665, 70, 80, 85, 90, 95, or up to 100%) following administration of a CD200 inhibitor (e.g., samalizumab). In some embodiments of any of the methods described herein, the expression level of HAVCR2 is increased by at least about 40% (e.g., 40, 45, 50, 60, 665, 70, 80, 85, 90, 95, or up to 100%) following administration of a CD200 inhibitor (e.g., samalizumab). In some embodiments of any of the methods described herein, the expression level of PDCD1 is increased by at least about 40% (e.g., 40, 45, 50, 60, 665, 70, 80, 85, 90, 95, or up to 100%) following administration of a CD200 inhibitor (e.g., samalizumab). In some embodiments of any of the methods described herein, the expression level of FCGR2A is increased by at least about 40% (e.g., 40, 45, 50, 60, 665, 70, 80, 85, 90, 95, or up to 100%) following administration of a CD200 inhibitor (e.g., samalizumab). In some embodiments of any of the methods described herein, the expression level of FCGR1A is increased by at least about 40% (e.g., 40, 45, 50, 60, 665, 70, 80, 85, 90, 95, or up to 100%) following administration of a CD200 inhibitor (e.g., samalizumab). In some embodiments of any of the methods described herein, the expression level of CD163 is increased by at least about 40% (e.g., 40, 45, 50, 60, 665, 70, 80, 85, 90, 95, or up to 100%) following administration of a CD200 inhibitor (e.g., samalizumab). In some embodiments of any of the methods described herein, the expression level of CD14 is increased by at least about 40% (e.g., 40, 45, 50, 60, 665, 70, 80, 85, 90, 95, or up to 100%) following administration of a CD200 inhibitor (e.g., samalizumab).


In some embodiments of any of the methods described herein, the expression level of ICOS is increased by at least about 1.5-fold, 2-fold, 2.5-fold, or 3-fold following administration of a CD200 inhibitor (e.g., samalizumab) compared to the expression level of ICOS in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor. In another embodiment of any of the methods described herein, the expression level of TIGIT is increased by at least about 1.5-fold, 2-fold, 2.5-fold, or 3-fold following administration of a CD200 inhibitor (e.g., samalizumab) compared to the expression level of TIGIT in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor. In another embodiment of any of the methods described herein, the expression level of TNFRSF9 is increased by at least about 1.5-fold, 2-fold, 2.5-fold, or 3-fold following administration of a CD200 inhibitor (e.g., samalizumab) compared to the expression level of TNFRSF9 in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor. In another embodiment of any of the methods described herein, the expression level of HAVCR2 is increased by at least about 1.5-fold, 2-fold, 2.5-fold, or 3-fold following administration of a CD200 inhibitor (e.g., samalizumab) compared to the expression level of HAVCR2 in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor. In another embodiment of any of the methods described herein, the expression level of PDCD1 is increased by at least about 1.5-fold, 2-fold, 2.5-fold, or 3-fold following administration of a CD200 inhibitor (e.g., samalizumab) compared to the expression level of TIGIT in a biological sample of the same type obtained from the subject prior to treatment with the PDCD1 inhibitor. In another embodiment of any of the methods described herein, the expression level of FCGR2A is increased by at least about 1.5-fold, 2-fold, 2.5-fold, or 3-fold following administration of a CD200 inhibitor (e.g., samalizumab) compared to the expression level of FCGR2A in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor. In another embodiment of any of the methods described herein, the expression level of FCGR1A is increased by at least about 1.5-fold, 2-fold, 2.5-fold, or 3-fold following administration of a CD200 inhibitor (e.g., samalizumab) compared to the expression level of FCGR1A in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor. In another embodiment of any of the methods described herein, the expression level of CD163 is increased by at least about 1.5-fold, 2-fold, 2.5-fold, or 3-fold following administration of a CD200 inhibitor (e.g., samalizumab) compared to the expression level of CD163 in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor. In another embodiment of any of the methods described herein, the expression level of CD14 is increased by at least about 1.5-fold, 2-fold, 2.5-fold, or 3-fold following administration of a CD200 inhibitor (e.g., samalizumab) compared to the expression level of CD14 in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor.


In another embodiment of any of the methods described herein, the expression level of CD200R1 is increased by at least about 1.5-fold, 2-fold, 2.5-fold, or 3-fold following administration of a CD200 inhibitor (e.g., samalizumab) compared to the expression level of CD200R1 in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor.


2. CD200 Inhibitors


The terms “CD200”, “OX-2” and “OX-2/CD200” are used interchangeably herein and refers to the highly conserved type I transmembrane glycoprotein including multiple transcript variants: CD200 isoform a (SEQ ID NO: 1; NCBI Reference Sequence: NP_005935.4), CD200 isoform b (SEQ ID NO:2; NCBI Reference Sequence: NP_001004196.2), CD200 isoform c (SEQ ID NO:3; NCBI Reference Sequence: NP_001305755.1 or NP_001305759.1), CD200 isoform d (SEQ ID NO: 46; NCBI Reference Sequence: NP_001305757.1).


CD200 interacts with the CD200 receptor (also known as “CD200R,” “CD200R1,” “Cell Surface Glycoprotein OX2 Receptor,” “CD200 Cell Surface Glycoprotein Receptor,” “MOX2 Receptor,” “MOX2R,” “OX2R,” “Cell Surface Glycoprotein CD200 Receptor,” “Cell Surface Glycoprotein Receptor CD200,” “HCRTR2,” and “CRTR2”), which induces immune suppression by skewing the immune response from a Th1-cytokine producing response, to a response characterized by an increased frequency of immunosuppressive regulatory T cells and suppression of memory T cell function. CD200R1 is restricted to the surfaces of myeloid lineage cells. Alternative splicing of CD200R1 gene results in multiple transcript variants, including CD200R1 isoform a (SEQ ID NO: 42; NCBI Reference Sequence: NP_620161.1), CD200R1 isoform b (SEQ ID NO: 43; NCBI Reference Sequence: NP_620385.1), CD200R1 isoform c (SEQ ID NO: 44; NCBI Reference Sequence: NP_620386.1), and CD200R1 isoform d (SEQ ID NO: 45; NCBI Reference Sequence: NP_740750.1).


The terms “CD200 antagonist” and “CD200 inhibitor” as used herein include any agent that is capable of inhibiting the activity, function and/or the expression of CD200 or its receptor. In certain embodiments, the antagonist disrupts the interaction of CD200 and CD200R1. In other embodiments, the CD200 antagonist is capable of decreasing the immunosuppressive effects of CD200 or are capable of targeting CD200-expressing cells for depletion or elimination.


Any compound which binds to and inhibits, or otherwise inhibits the activity, function and/or the expression of CD200 or its receptor may be utilized in accordance with the present disclosure. For example, an inhibitor of CD200 can be, e.g., a small molecule, a nucleic acid or nucleic acid analog, a peptidomimetic, or a macromolecule that is not a nucleic acid or a protein. These agents include, but are not limited to, small organic molecules, RNA aptamers, L-RNA aptamers, Spiegelmers, antisense compounds, double stranded RNA, small interfering RNA, locked nucleic acid inhibitors, and peptide nucleic acid inhibitors. In some embodiments, a CD200 inhibitor may be a protein or protein fragment.


Other compounds which may be utilized include, but are not limited to, proteins, protein fragments, peptides, small molecules, RNA aptamers, L-RNA aptamers, spiegelmers, antisense compounds, serine protease inhibitors, molecules which may be utilized in RNA interference (RNAi) such as double stranded RNA including small interfering RNA (siRNA), locked nucleic acid (LNA) inhibitors, peptide nucleic acid (PNA) inhibitors, etc.


An inhibitor of CD200 can be, e.g., a small molecule, a polypeptide, a polypeptide analog, a nucleic acid, or a nucleic acid analog.


“Small molecule” as used herein, is meant to refer to an agent, which has a molecular weight preferably of less than about 6 kDa and most preferably less than about 2.5 kDa. Many pharmaceutical companies have extensive libraries of chemical and/or biological mixtures comprising arrays of small molecules, often fungal, bacterial, or algal extracts, which can be screened with any of the assays of the application. This application contemplates using, among other things, small chemical libraries, peptide libraries, or collections of natural products. Tan et al. described a library with over two million synthetic compounds that is compatible with miniaturized cell-based assays (J Am Chem Soc (1998) 120:8565-8566). It is within the scope of this application that such a library may be used to screen for agents that bind to a target antigen of interest (e.g., CD200). There are numerous commercially available compound libraries, such as the Chembridge DIVERSet™ diverse screening library. Libraries are also available from academic investigators, such as the Diversity set from the NCI developmental therapeutics program. Rational drug design may also be employed. Rational drug design can also be achieved based on known compounds, e.g., a known inhibitor of CD200 (e.g., an antibody, or antigen-binding fragment thereof, that binds to CD200).


Peptidomimetics can be compounds in which at least a portion of a subject polypeptide is modified, and the three dimensional structure of the peptidomimetic remains substantially the same as that of the subject polypeptide. Peptidomimetics may be analogues of a subject polypeptide of the disclosure that are, themselves, polypeptides containing one or more substitutions or other modifications within the subject polypeptide sequence. Alternatively, at least a portion of the subject polypeptide sequence may be replaced with a non-peptide structure, such that the three-dimensional structure of the subject polypeptide is substantially retained. In other words, one, two or three amino acid residues within the subject polypeptide sequence may be replaced by a non-peptide structure. In addition, other peptide portions of the subject polypeptide may, but need not, be replaced with a non-peptide structure. Peptidomimetics (both peptide and non-peptidyl analogues) may have improved properties (e.g., decreased proteolysis, increased retention or increased bioavailability). Peptidomimetics generally have improved oral availability, which makes them especially suited to treatment of disorders in a human or animal. It should be noted that peptidomimetics may or may not have similar two-dimensional chemical structures, but share common three-dimensional structural features and geometry. Each peptidomimetic may further have one or more unique additional binding elements.


Nucleic acid inhibitors can be used to bind to and inhibit a target antigen of interest. The nucleic acid antagonist can be, e.g., an aptamer. Aptamers are short oligonucleotide sequences that can be used to recognize and specifically bind almost any molecule, including cell surface proteins. The systematic evolution of ligands by exponential enrichment (SELEX) process is powerful and can be used to readily identify such aptamers. Aptamers can be made for a wide range of proteins of importance for therapy and diagnostics, such as growth factors and cell surface antigens. These oligonucleotides bind their targets with similar affinities and specificities as antibodies do (see, e.g., Ulrich (2006) Handb Exp Pharmacol. 173:305-326).


In some embodiments, the CD200 inhibitor is a non-antibody scaffold protein. These proteins are, generally, obtained through combinatorial chemistry-based adaptation of pre-existing antigen-binding proteins. For example, the binding site of human transferrin for human transferrin receptor can be modified using combinatorial chemistry to create a diverse library of transferrin variants, some of which have acquired affinity for different antigens. Ali et al. (1999) J Biol Chem 274:24066-24073. The portion of human transferrin not involved with binding the receptor remains unchanged and serves as a scaffold, like framework regions of antibodies, to present the variant binding sites. The libraries are then screened, as an antibody library is, against a target antigen of interest to identify those variants having optimal selectivity and affinity for the target antigen. Non-antibody scaffold proteins, while similar in function to antibodies, are touted as having a number of advantages as compared to antibodies, which advantages include, among other things, enhanced solubility and tissue penetration, less costly manufacture, and ease of conjugation to other molecules of interest. Hey et al. (2005) TRENDS Biotechnol 23(10):514-522.


One of skill in the art would appreciate that the scaffold portion of the non-antibody scaffold protein can include, e.g., all or part of: the Z domain of S. aureus protein A, human transferrin, human tenth fibronectin type III domain, Kunitz domain of a human trypsin inhibitor, human CTLA-4, an ankyrin repeat protein, a human lipocalin, human crystallin, human ubiquitin, or a trypsin inhibitor from E. elaterium. Hey et al., (2005).


In some embodiments, the CD200 inhibitor is an antibody, or antigen-binding fragment thereof, which binds to CD200 (e.g., an “anti-CD200 antibody.”)


In some embodiments, an anti-CD200 antibody described herein binds to an epitope within the extracellular portion of 65 a CD200 protein. For example, in some embodiments, the anti-CD200 antibody can bind to CD200 protein at an epitope within or overlapping with: (i) amino acids 1 to 233 of the amino acid sequence depicted in SEQ ID NO: 1; (ii) amino acids 1 to 258 of the amino acid sequence depicted in SEQ ID NO:2; or amino acids 1 to 229 of the amino acid sequence depicted in SEQ ID NO:3.


In some embodiments, the anti-CD200 antibody binds to an epitope in the human CD200 protein lacking the leader sequence. For example, an anti-CD200 antibody described herein can bind to a CD200 protein at an epitope within or overlapping with amino acids 31 to 233 of the amino acid sequence depicted in SEQ ID NO: 1, which corresponds to the extracellular portion of the mature form of human CD200 isoform A less the amino terminal leader sequence. In some embodiments, an anti-CD200 antibody described herein can bind to a CD200 protein at an epitope within or overlapping with amino acids 56 to 258 of the amino acid sequence depicted in SEQ ID NO: 2, which corresponds to the extracellular portion of the mature form of human CD200 isoform B less the amino terminal leader sequence. In some embodiments, an anti-CD200 antibody described herein can bind to a CD200 protein at an epitope within or overlapping with amino acids 27 to 229 of the amino acid sequence depicted in SEQ ID NO: 3, which corresponds to the extracellular portion of the mature form of human CD200 less the amino terminal leader sequence.


In some embodiments, the anti-CD200 antibody specifically binds to a human CD200 protein (e.g., the human CD200 protein having the amino acid sequence depicted in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO:46 or the extracellular domains of the mature forms of the CD200 proteins). Methods for identifying the epitope to which a particular antibody binds are also know in the art.


Anti-CD200 antibodies for use in the methods provided herein are CD200 antagonists and include whole antibodies, or antibody fragments capable of binding to CD200, particularly anti-CD200 antibodies which disrupt the interaction between CD200 and CD200R1. Exemplary anti-CD200 antibodies, or antigen binding fragments thereof, which can be used in the methods described herein include, but are not limited to, those disclosed in U.S. Pat. Nos. 7,408,041; 8,075,884; and WO 2012/106634 (the contents of each of which are herein incorporated by reference in their entirety).


In one embodiment, the anti-CD200 antibody, or antigen binding fragment thereof, comprises the CDR1, CDR2, and CDR3 domains of a heavy chain variable region having the sequence set forth in SEQ ID NO: 13, and the CDR1, CDR2, and CDR3 domains of a light chain variable region having the sequence set forth in SEQ ID NO: 12.


In another embodiment, the anti-CD200 antibody, or antigen binding fragment thereof, comprising the CDR1, CDR2 and CDR3 domains of a heavy chain region having the sequence set forth in SEQ ID NO: 11, and the CDR1, CDR2 and CDR3 domains of a light chain region having the sequence set forth in SEQ ID NO: 10.


In another embodiment, the anti-CD200 antibody, or antigen binding fragment thereof, comprises: (a) a light chain variable domain that comprises (i) a light chain CDR1 comprising the sequence set forth in SEQ ID NO: 4, (ii) a light chain CDR2 comprising the sequence set forth in SEQ ID NO: 5, and (iii) a light chain CDR3 comprising the sequence set forth in SEQ ID NO: 6; and (b) a heavy chain variable domain comprising (i) a heavy chain CDR1 comprising the sequence set forth in SEQ ID NO: 7, (ii) a heavy chain CDR2 comprising the sequence set forth in SEQ ID NO: 8 and (iii) a heavy chain CDR3 comprising the sequence set forth in SEQ ID NO: 9.


In another embodiment, the antibody comprises a light chain region sequence as set forth in SEQ ID NO: 10 and/or a heavy chain variable region sequence as set forth in SEQ ID NO: 11. In one embodiment, the antibody comprises a light chain sequence as set forth in SEQ ID NO: 12 and/or a heavy chain sequence as set forth in SEQ ID NO: 13. In one embodiment, the anti-CD200 antibody is samalizumab (also known as ALXN6000; Alexion Pharmaceuticals, Inc.).


Antibodies and antigen binding fragments thereof may be obtained according to established hybridoma and recombinant procedures. Suitable methods for producing an antibody (e.g., an anti-CD200 antibody) or antigen-binding fragments thereof may be obtained according to established hybridoma and recombinant procedures as previously disclosed (see, e.g., U.S. Pat. Nos. 7,427,665; 7,435,412; and 7,408,041). For example, a process for the production of an antibody disclosed herein includes culturing a host (e.g., E. coli or a mammalian cell), which has been transformed with a hybrid vector. The vector includes one or more expression cassettes containing a promoter operably linked to a first DNA sequence encoding a signal peptide linked in the proper reading frame to a second DNA sequence encoding the antibody protein. The antibody protein is then collected and isolated. Optionally, the expression cassette may include a promoter operably linked to polycistronic, for example bicistronic, DNA sequences encoding antibody proteins each individually operably linked to a signal peptide in the proper reading frame.


Multiplication of hybridoma cells or mammalian host cells in vitro is carried out in suitable culture media, which include the customary standard culture media (such as, for example Dulbecco's Modified Eagle Medium (DMEM) or RPMI 1640 medium), optionally replenished by a mammalian serum (e.g. fetal calf serum), or trace elements and growth sustaining supplements (e.g. feeder cells such as normal mouse peritoneal exudate cells, spleen cells, bone marrow macrophages, 2-aminoethanol, insulin, transferrin, low density lipoprotein, oleic acid, or the like). Multiplication of host cells which are bacterial cells or yeast cells is likewise carried out in suitable culture media known in the art. For example, for bacteria, suitable culture media include, but are not limited to, medium LE, NZCYM, NZYM, NZM, Terrific Broth, SOB, SOC, 2xYT, and/or M9 Minimal Medium. For yeast, suitable culture media include, but are not limited to, medium YPD, YEPD, Minimal Medium, and/or Complete Minimal Dropout Medium.


In vitro production provides relatively pure antibody preparations and allows scale-up to give large amounts of the desired antibodies. Techniques for bacterial cell, yeast, plant, or mammalian cell cultivation are known in the art and include homogeneous suspension culture (e.g., in an airlift reactor or in a continuous stirrer reactor), and immobilized or entrapped cell culture (e.g., in hollow fibers, microcapsules, on agarose microbeads or ceramic cartridges).


Large quantities of the desired antibodies can also be obtained by multiplying mammalian cells in vivo. For this purpose, cells producing the desired antibodies are injected into histocompatible mammals to cause growth of antibody-producing tumors. Optionally, the animals are primed with a hydrocarbon, especially mineral oils such as pristane (tetramethyl-pentadecane), prior to the injection. After one to three weeks, the antibodies are isolated from the body fluids of those mammals. For example, hybridoma cells obtained by fusion of suitable myeloma cells with antibody-producing spleen cells from Balb/c mice, or transfected cells derived from hybridoma cell line Sp2/0 that produce the desired antibodies are injected intraperitoneally into Balb/c mice optionally pre-treated with pristine. After one to two weeks, ascitic fluid is taken from the animals.


The antibody which is formulated is preferably essentially pure and desirably essentially homogeneous (e.g., free from contaminating proteins, etc.). “Essentially pure” antibody means a composition comprising at least about 90% by weight of the antibody, based on total weight of the composition, preferably at least about 95% by weight of the antibody. “Essentially homogeneous” antibody means a composition comprising at least about 99% by weight of antibody, based on total weight of the composition.


Techniques for purification of therapeutic antibodies to pharmaceutical grade are well known in the art. For example, the immunoglobulins in the culture supernatants or in the ascitic fluid may be concentrated, e.g., by precipitation with ammonium sulfate, dialysis against hygroscopic material such as polyethylene glycol, filtration through selective membranes, or the like. If necessary and/or desired, the antibodies are purified by the customary chromatography methods, for example gel filtration, ion-exchange chromatography, chromatography over DEAE-cellulose and/or (immuno-) affinity chromatography, e.g., affinity chromatography with a one or more surface polypeptides derived from a CLL cell line according to this disclosure, or with Protein-A or G.


The foregoing, and other, techniques are discussed in, for example, Kohler and Milstein, (1975) Nature 256:495-497; U.S. Pat. No. 4,376,110; Harlow and Lane, Antibodies: a Laboratory Manual, (1988) Cold Spring Harbor, the disclosures of which are all incorporated herein by reference. Techniques for the preparation of recombinant antibody molecules is described in the above references and also in, for example WO 97/08320; U.S. Pat. Nos. 5,427,908 and 5,508,717; Smith, 1985, Science, Vol. 225, pp 1315-1317; Parmley and Smith 1988, Gene 73, pp 305-318; De La Cruz et al, 1988, J. Biol. Chem., 263 pp 4318-4322; U.S. Pat. Nos. 5,403,484; 5,223,409; WO88/06630; WO 92/15679; U.S. Pat. Nos. 5,780,279; 5,571,698; and 6,040,136; Davis et al., Cancer Metastasis Rev., 1999; 18(4):421-5; Taylor, et al., Nucleic Acids Research 20 (1992): 6287-6295; and Tomizuka et al., Proc. Nat. Academy of Sciences USA 97(2) (2000): 722-727 (the contents of each are incorporated herein by reference).


3. Biological Samples and Sample Collection


Suitable biological samples for use in the methods described herein include whole blood (or a fraction thereof), tumor tissue, or tumor cells. A biological sample can be further fractionated, if desired, to a fraction containing particular analytes (e.g., proteins) of interest. For example, a whole blood sample can be fractionated into serum or into fractions containing particular types of proteins.


Biological samples suitable for the invention may be fresh or frozen samples collected from a subject, or archival samples with known diagnosis, treatment and/or outcome history. The biological samples can be obtained from a subject, e.g., a subject having, suspected of having, or at risk of developing, cancer. Any suitable methods for obtaining the biological samples can be employed, although exemplary methods include, e.g., phlebotomy, swab (e.g., buccal or other swab), lavage, or fine needle aspirate biopsy procedure. Biological samples can also be obtained from bone marrow.


In some embodiments, a protein extract may be prepared from a biological sample. In some embodiments, a protein extract contains the total protein content. Methods of protein extraction are well known in the art. See, e.g., Roe (2001) Protein Purification Techniques: A Practical Approach, 2nd Edition, Oxford University Press. Numerous different and versatile kits can be used to extract proteins from bodily fluids and tissues, and are commercially available from, for example, BioRad Laboratories (Hercules, Calif.), BD Biosciences Clontech (Mountain View, Calif.), Chemicon International, Inc. (Temecula, Calif.), Calbiochem (San Diego, Calif.), Pierce Biotechnology (Rockford, Ill.), and Invitrogen Corp. (Carlsbad, Calif.).


Methods for obtaining and/or storing samples that preserve the activity or integrity of cells in the biological sample are well known to those skilled in the art. For example, a biological sample can be further contacted with one or more additional agents such as appropriate buffers and/or inhibitors, including protease inhibitors, the agents meant to preserve or minimize changes (e.g., changes in osmolarity or pH) in protein structure. Such inhibitors include, for example, chelators such as ethylenediamine tetraacetic acid (EDTA), ethylene glycol tetraacetic acid (EGTA), protease inhibitors such as phenylmethylsulfonyl fluoride (PMSF), aprotinin, and leupeptin. Appropriate buffers and conditions for storing or otherwise manipulating whole cells are described in, e.g., Pollard and Walker (1997), “Basic Cell Culture Protocols,” volume 75 of Methods in Molecular Biology, Humana Press; Masters (2000) “Animal cell culture: a practical approach,” Volume 232 of Practical Approach Series, Oxford University Press; and Jones (1996) “Human cell culture protocols,” volume 2 of Methods in Molecular Medicine, Humana Press.


A sample also can be processed to eliminate or minimize the presence of interfering substances. For example, a biological sample can be fractionated or purified to remove one or more materials (e.g., cells) that are not of interest. Methods of fractionating or purifying a biological sample include, but are not limited to, flow cytometry, fluorescence activated cell sorting, and sedimentation.


4. Methods for Treatment


Also provided herein are methods for treating cancer in a subject (e.g., a human). In one embodiment, the patient is an adult and the cancer is selected from the group consisting of diffuse large B cell lymphoma (DLBL), lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), glioblastoma (GBM), low grade glioma (LGG), clear cell RCC (KIRC), chromophobe (KICH), papillary cell RCC (KIRP), melanoma (SKCM), ovarian cancer (OV), colon cancer (COAD), rectum cancer (READ), and uterine endometrial cancer (UCEC).


In another embodiment, the patient is a pediatric patient, and the cancer is selected from atypical teratoid rhabdoid tumor (AT/RT), ependymoma, osteosarcoma, rhabdomyosarcoma, Ewing sarcoma, pilocytic astrocytoma, neuroblastoma, and retinoblastoma.


The terms “treat,” “treating,” and “treatment,” as used herein, refer to therapeutic measures described herein. The methods of treatment employ administration to a subject (such as a human) the combination disclosed herein in order to cure, delay, reduce the severity of, or ameliorate one or more symptoms of the disease or disorder or recurring disease or disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment.


The terms “effective amount” or “therapeutically effective amount” are used interchangeably and refer to an amount of formulation or antibody effective to alleviate or ameliorate one or more symptom(s) of cancer or to prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. Therapeutically effective dosages may be determined by using in vitro and in vivo methods.


In one embodiment, a method for treating a patient having cancer who has been determined to have positive expression of CD200 receptor (CD200R1) and one or more biomarkers (e.g., two or more, three or more, four or more, five or more, 6 or more, 7 or more, 8 or more, or 9) in a biological sample from the patient is provided, the method comprising administering to the patient a CD200 inhibitor in an amount and with a frequency sufficient to reduce the cancer burden in the patient (e.g., by about 30, 40, 50, 60, 70, 80, 90, or 100%). In one embodiment, the biomarker is ICOS. In another embodiment, the biomarker is TIGIT. In another embodiment, the biomarker is TNFRSF9. In another embodiment, the biomarker is HAVCR2. In another embodiment, the biomarker is PDCD1. In another embodiment, the biomarker is FCGR2A. In another embodiment, the biomarker is FCGR1A. In another embodiment, the biomarker is CD163. In another embodiment, the biomarker is CD14.


In one embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of two biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of three biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of four biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of five biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of six biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of seven biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1 and positive expression of eight biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. In another embodiment, the patient has been determined to have positive expression of CD200R1, ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14. The patient can have positive expression of any possible combination of the biomarkers disclosed herein.


In one embodiment, the method includes determining if a patient having cancer has positive expression of CD200 receptor (CD200R1) and one or more biomarkers (i.e., Inducible T-cell COStimulator (ICOS), T Cell Immunoreceptor with Ig and ITIM Domains (TIGIT), Tumor Necrosis Factor Receptor Superfamily Member 9 (TNFRSF9), Hepatitis A Virus Cellular Receptor 2 (HAVCR2), and Programmed Cell Death 1 (PDCD1), Fc Fragment Of IgG Receptor IIa (FCGR2A), Fc Fragment Of IgG Receptor Ia (FCGR1A), Cluster of Differentiation 163 (CD163), and/or CD14) and administering to the patient a CD200 inhibitor if the patient has an positive expression of CD200R1 and one or more biomarkers.


The CD200 inhibitor can be administered to a subject, e.g., a human subject, using a variety of methods that depend, in part, on the route of administration. The route can be, e.g., intravenous injection or infusion (IV), subcutaneous injection (SC), intraperitoneal (IP) injection, or intramuscular injection.


Administration can be achieved by, e.g., local infusion, injection, or by means of an implant. The implant can be of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. The implant can be configured for sustained or periodic release of the composition to the subject. See, e.g., U.S. patent publication no. 20080241223; U.S. Pat. Nos. 5,501,856; 4,863,457; and 3,710,795; and European patent nos. EP488401 and EP430539, the disclosures of each of which are incorporated herein by reference in their entirety. The composition can be delivered to the subject by way of an implantable device based on, e.g., diffusive, erodible or convective systems, e.g., osmotic pumps, biodegradable implants, electrodiffusion systems, electroosmosis systems, vapor pressure pumps, electrolytic pumps, effervescent pumps, piezoelectric pumps, erosion-based systems, or electromechanical systems.


A suitable dose of a CD200 inhibitor (e.g., an anti-CD200 antibody or fragment thereof), which dose is capable of treating cancer in a subject, can depend on a variety of factors including, e.g., the age, sex, and weight of a subject to be treated and the particular inhibitor compound used. For example, a different dose of an siRNA specific for human CD200 may be required to treat a subject with cancer as compared to the dose of an anti-CD200 antibody required to treat the same patient. Other factors affecting the dose administered to the subject include, e.g., the type or severity of the cancer. Other factors can include, e.g., other medical disorders concurrently or previously affecting the subject, the general health of the subject, the genetic disposition of the subject, diet, time of administration, rate of excretion, drug combination, and any other additional therapeutics that are administered to the subject. It should also be understood that a specific dosage and treatment regimen for any particular subject will depend upon the judgment of the treating medical practitioner (e.g., doctor or nurse).


The inhibitor can be administered as a fixed dose, or in a milligram per kilogram “mg/kg” dose. In one embodiment, the CD200 inhibitor is an anti-CD200 antibody administered at a dose of about 300 mg/m2 to about 600 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 300 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 400 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 500 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 600 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 700 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 300 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 800 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 900 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 1000 mg/m2. In another embodiment, the anti-CD200 antibody is administered at a dose of about 1100 mg/m2.


In another embodiment, the anti-CD200 antibody is administered at a dose of about 5 mg/kg to about 50 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 10 mg/kg to about 30 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 15 mg/kg to about 25 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 10 mg/kg to about 20 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 10 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 15 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 20 mg/kg. In another embodiment, the anti-CD200 antibody is administered at a dose of about 25 mg/kg.


A pharmaceutical composition can include a therapeutically effective amount of an inhibitor of CD200 (e.g., an anti-CD200 antibody or antigen-binding fragment thereof). Such effective amounts can be readily determined by one of ordinary skill in the art based, in part, on the effect of the administered inhibitor, or the combinatorial effect of the antibody and one or more additional active agents, if more than one agent is used. A therapeutically effective amount of an inhibitor of a CD200 inhibitor (e.g., such as samalizumab) can also vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody (and one or more additional active agents) to elicit a desired response in the individual, e.g., amelioration of at least one condition parameter, e.g., amelioration of at least one symptom of aHUS. For example, a therapeutically effective amount of a CD200 inhibitor can inhibit (lessen the severity of or eliminate the occurrence of) of any one of the symptoms of cancer. A therapeutically effective amount is also one in which any toxic or detrimental effects of the composition are outweighed by the therapeutically beneficial effects.


Toxicity and therapeutic efficacy of CD200 inhibitors can be determined by known pharmaceutical procedures in cell cultures or experimental animals. These procedures can be used, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compositions, or inhibitors (e.g., anti-CD200 antibodies) of the compositions, that exhibit high therapeutic indices are preferred. While compositions that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue and to minimize potential damage to normal cells and, thereby, reduce side effects.


In some embodiments, the CD200 inhibitor can be administered to a subject as a monotherapy. Alternatively, as described above, the inhibitor can be administered to a subject as a combination therapy with another treatment, e.g., another treatment for cancer. For example, the combination therapy can include administering to the subject (e.g., a human patient) one or more additional that provide a therapeutic benefit to the subject who has cancer. In one embodiment, the inhibitor is administered first in time and the one or more additional active agents are administered second in time. In some embodiments, the one or more additional active agents are administered first in time and the inhibitor is administered second in time.


5. Methods of Monitoring Responsiveness


Also, provided are methods for monitoring responsiveness of a subject having cancer to treatment with a CD200 inhibitor, the method comprising: determining the expression level of CD200R1 and one or more (e.g., two or more, three or more, four or more, five or more, 6 or more, 7 or more, 8 or more, or 9) biomarkers (i.e., ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and/or CD14) in a biological sample from the patient, wherein increased expression levels of CD200R1 and the one or more biomarkers, as compared to expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor.


In one embodiment, increased expression levels of CD200R1 and one biomarker selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and two biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and three biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and four biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and five biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and six biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and seven biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1 and eight biomarkers selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. In another embodiment, increased expression levels of CD200R1, ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and CD14, compared to the expression levels in a biological sample of the same type obtained from the subject prior to treatment with the CD200 inhibitor, indicates that the subject is responsive to treatment with the CD200 inhibitor. The patient can have elevated expression levels) of any possible combination of the biomarkers disclosed herein.


6. Additional Agents/Therapies


The anti-CD200 inhibitors described herein (e.g., samalizumab) can also be used in conjunction with other well-known therapies that are selected for their particular usefulness against the cancer that is being treated. Combinations of the present disclosure may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when inappropriate.


For example, the CD200 inhibitors described herein can further be used in combination (e.g., simultaneously or separately) with an additional treatment, such as irradiation, chemotherapy (e.g., using cytarabine, daunorubicin, camptothecin (CPT-11), 5-fluorouracil (5-FU), cisplatin, doxorubicin, irinotecan, paclitaxel, gemcitabine, cisplatin, paclitaxel, doxorubicin, 5-fu, or camptothecin+apo21/TRAIL (a 6× combo)).


The CD200 inhibitors described herein can further be used in combination with one or more anti-proliferative cytotoxic agents. Classes of compounds that may be used as anti-proliferative cytotoxic agents include, but are not limited to, the following:


Alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes): Uracil mustard, Chlormethine, Cyclophosphamide (CYTOXAN®), fosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylenemelamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Streptozocin, Dacarbazine, and Temozolomide.


Antimetabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors): methotrexate, 5-Fluorouracil, floxuridine, cytarabine, 6-Mercaptopurine, 6-Thioguanine, fludarabine phosphate, pentostatine, and gemcitabine.


Suitable anti-proliferative agents for use in the methods of disclosed herein, include, without limitation, taxanes, paclitaxel (paclitaxel is commercially available as TAXOL®)(tamoxifen), docetaxel, discodermolide (DDM), dictyostatin (DCT), Peloruside A, epothilones, epothilone A, epothilone B, epothilone C, epothilone D, epothilone E, epothilone F, furanoepothilone D, desoxyepothilone B1, [17]-dehydrodesoxyepothilone B, [18]dehydrodesoxyepothilones B, C12,13-cyclopropyl-epothilone A, C6-C8 bridged epothilone A, trans-9,10-dehydroepothilone D, cis-9,10-dehydroepothilone D, 16-desmethylepothilone B, epothilone B10, discoderomolide, patupilone (EPO-906), KOS-862, KOS-1584, ZK-EPO, ABJ-789, XAA296A (Discodermolide), TZT-1027 (soblidotin), ILX-651 (tasidotin hydrochloride), Halichondrin B, Eribulin mesylate (E-7389), Hemiasterlin (HTI-286), E-7974, Cyrptophycins, LY-355703, Maytansinoid immunoconjugates (DM-1), MKC-1, ABT-751, T1-38067, T-900607, SB-715992 (ispinesib), SB-743921, MK-0731, STA-5312, eleutherobin, 17beta-acetoxy-2-ethoxy-6-oxo-B-homo-estra-1,3,5(10)-trien-3-ol, cyclostreptin, isolaulimalide, laulimalide, 4-epi-7-dehydroxy-14,16-didemethyl-(+)-discodermolides, and cryptothilone 1, in addition to other microtubuline stabilizing agents known in the art.


In cases where it is desirable to render aberrantly proliferative cells quiescent in conjunction with or prior to treatment as described herein, hormones and steroids (including synthetic analogs), such as 17a-Ethinylestradiol, Diethylstilbestrol, Testosterone, Prednisone, Fluoxymesterone, Dromostanolone propionate, Testolactone, Megestrolacetate, Methylprednisolone, Methyl-testosterone, Prednisolone, Triamcinolone, Chlorotrianisene, Hydroxyprogesterone, Aminoglutethimide, Estramustine, Medroxyprogesteroneacetate, Leuprolide, Flutamide, Toremifene, ZOLADEX® (goserelin acetate), can also be administered to the patient. When employing the methods or compositions of the present disclosure, other agents used in the modulation of tumor growth or metastasis in a clinical setting, such as antimimetics, can also be administered as desired.


Methods for the safe and effective administration of chemotherapeutic agents are known to those skilled in the art. In addition, their administration is described in the standard literature. For example, the administration of many of the chemotherapeutic agents is described in the Physicians' Desk Reference (PDR), e.g., 1996 edition (Medical Economics Company, Montvale, N.J. 07645-1742, USA); the disclosure of which is incorporated herein by reference thereto.


The chemotherapeutic agent(s) and/or radiation therapy can be administered according to therapeutic protocols well known in the art. It will be apparent to those skilled in the art that the administration of the chemotherapeutic agent(s) and/or radiation therapy can be varied depending on the disease being treated and the known effects of the chemotherapeutic agent(s) and/or radiation therapy on that disease. Also, in accordance with the knowledge of the skilled clinician, the therapeutic protocols (e.g., dosage amounts and times of administration) can be varied in view of the observed effects of the administered therapeutic agents on the patient, and in view of the observed responses of the disease to the administered therapeutic agents.


7. Outcomes


Patients treated according to the methods disclosed herein preferably experience improvement in at least one sign of cancer. In one embodiment, improvement is measured by a reduction in the quantity and/or size of measurable tumor lesions. In another embodiment, lesions can be measured on chest x-rays or CT or MRI films. In another embodiment, cytology or histology can be used to evaluate responsiveness to a therapy. In another embodiment, the treatment produces at least one therapeutic effect, for example, morphologic complete remission, cytogenetic complete remission, morphologic CR with incomplete blood count recovery, partial remission, and/or stable disease.


In another embodiment, the patient treated exhibits a complete response, partial response, and/or stable disease. In another embodiment, the patient treated experiences tumor shrinkage and/or decrease in growth rate, i.e., suppression of tumor growth (e.g., by about 30, 40, 50, 60, 70, 80, 90, or 100%). In another embodiment, unwanted cell proliferation is reduced or inhibited (e.g., by about 30, 40, 50, 60, 70, 80, 90, or 100%). In yet another embodiment, one or more of the following can occur: the number of cancer cells can be reduced; tumor size can be reduced; cancer cell infiltration into peripheral organs can be inhibited, retarded, slowed, or stopped; tumor metastasis can be slowed or inhibited; tumor growth can be inhibited; recurrence of tumor can be prevented or delayed; one or more of the symptoms associated with cancer can be relieved to some extent.


In another embodiment, the methods of treatment produce a clinical benefit (e.g., Morphologic Complete Remission (Morphologic CR), cytogenetic complete remission (CRc), morphologic CR with incomplete blood count recovery (CRi), or partial remission (PR)).


Morphologic CR requires <5% blasts in bone marrow aspirate, neutrophils ≥1,000/μL, platelets ≥100,000/μL, no extramedullary disease, no blasts with Auer rods detected, and No circulating blasts (rare may be permitted)/No evidence of pre-treatment blast phenotype by flow cytometry (i.e. CD34, CD7 co-expression).


CRc requires <5% blasts in bone marrow aspirate, neutrophils ≥1,000/μL, platelets ≥100,000/μL, no extramedullary disease, no blasts with Auer rods detected, no circulating blasts (rare may be permitted)/no evidence of pre-treatment blast phenotype by flow cytometry (i.e. CD34, CD7 co-expression), and reversion to a normal karyotype.


CRi requires <5% blasts in bone marrow aspirate, neutrophils <1,000/μL or Platelets <100,000/μL, no extramedullary disease, no blasts with Auer rods detected, and no circulating blasts (rare may be permitted)/no evidence of pre-treatment blast phenotype by flow cytometry (i.e. CD34, CD7 co-expression).


PR requires all criteria for CR except for bone marrow blasts, must have greater than 50% decrease in blasts in bone marrow aspirate to a range of 5-25%, neutrophils ≥1,000/μL, platelets ≥100,000/μL, no extramedullary disease, and if Auer rods are detected the blast count in the bone marrow must be ≤5%.


In another aspect, the treatment produces a desired immunomodulatory effect in a human (e.g., a cancer patient). The immunomodulatory effect can be characterized by a change (e.g., an increase) in at least one biomarker, i.e., CD200R1 and one or more of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGR1A, CD163, and/or CD14. It is understood that any of the methods described herein can involve determining whether there has been a change (e.g., an increase) in one or more (e.g., three, four, five, six, seven, eight, or nine) of the biomarkers described herein. Where interrogation of more than one of the biomarkers is practiced, any combination of two or more (e.g., three, four, five, six, seven, eight, nine, or 10 or more) of the biomarkers can be analyzed.


Methods for determining immune response following treatment with an ani-CD200 antibody, or antigen-binding fragment thereof, are elaborated on in, e.g., U.S. Pat. No. 9,180,186.


8. Kits


Also provided are kits comprising various reagents and materials useful for carrying out the methods described herein. The procedures for measuring, diagnosing, evaluating, and/or assessing described herein may be performed by diagnostic laboratories, experimental laboratories, or individual practitioners. The invention provides kits which can be used in any or all of these settings. In some embodiments, the kits described herein comprise materials and reagents for, among other things, characterizing or processing biological samples (e.g., biological samples), measuring biomarker levels (e.g., protein or nucleic acid levels), monitoring treatment response in a subject according to the methods provided herein. In certain embodiments, an inventive kit comprises at least one or more reagents that specifically detect protein levels of one or more biomarker proteins described herein (e.g., those Tables 1 and 2) and, optionally, instructions for using the kit. The kit can include, e.g., any of the arrays described herein.


In some embodiments, the kits may include suitable control samples (e.g., biological samples from normal healthy individuals or a solution comprising a known, control amount of a particular analyte of interest). In some embodiments, kits of the invention may include instructions for using the kit according to one or more methods described herein and may comprise instructions for processing the biological sample obtained from the subject and/or for performing the test or instructions for interpreting the results.


While the present disclosure has been described with reference to the specific embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the disclosure. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, or process step or steps, to the objective, spirit and scope of the present disclosure. All such modifications are intended to be within the scope of the disclosure.


The following examples are intended to illustrate, not limit, the invention.


EXAMPLES
Example 1: Immune Effector Cells Signature Development

CD200 (OX-2) is an immune checkpoint protein expressed by a number of immune cells, including B, T cells and macrophages, as well as non-immune cells, including endothelial cells and neurons. CD200 binds to its receptor (CD200R1), expressed on antigen-presenting cells (APCs) and T cells and is believed to play an important role in normal immune homeostasis. However, overexpression of CD200 by tumor cells implicates the CD200 pathway in tumor-mediated immunosuppression and regulation of anti-tumor activity.


Recent evidence suggests that the presence of immune effector cells within tumors is critical for clinical response to immune checkpoint therapy. Similarly, the expression of the immune modulatory target within the tumor may correlate with response to therapy. The purpose of this study was to identify tumor types which may be most sensitive to samalizumab, based on CD200 expression and the presence of immune effector cell populations within the tumor. To this end, tumor gene expression data was mined to evaluate CD200 pathway expression in multiple tumor types, to develop a gene signature which may correlate with response to samalizumab, as described below. Gene expression values for CD200, CD200R1, and other immune cell marker genes were described as the percentage of samples with expression higher than the median for the set of all tumors. The data and methodology is discussed further below.


1. Data Sources and Description


In the field of cancer research, The Cancer Genome Atlas (TCGA) data portal is the largest and most commonly used public resource, providing somatic mutation, gene expression, gene methylation and copy number variation (CNV) data sets, amongst others, for several thousands of tumor samples from adult cancer patients.


RNA-Seq (RNA sequencing), also called whole transcriptome shotgun sequencing (WTSS), uses next-generation sequencing (NGS) to reveal the presence and quantity of RNA in a biological sample at a given moment in time (see, e.g., Ryan D. Morin, et al., BioTechniques. 45 (1): 81-94 (2008); Chu Y, Corey D R (August 2012), Nucleic Acid Ther. 22 (4): 271-4; and Wang, Zhong, et al., Nature Reviews Genetics. 10 (1): 57-63). RNA-Seq is used to analyze the continually changing cellular transcriptome. Specifically, RNA-Seq facilitates the ability to look at alternative gene spliced transcripts, post-transcriptional modifications, gene fusion, mutations/SNPs and changes in gene expression over time, or differences in gene expression in different groups or treatments (see, e.g., Maher C A, et al. (March 2009), Nature. 458 (7234): 97-101). In addition to mRNA transcripts, RNA-Seq can look at different populations of RNA to include total RNA, small RNA, such as miRNA, tRNA, and ribosomal profiling (see, e.g., Ingolia N T, et al. (August 2012), Nat Protoc. 7 (8): 1534-50). RNA-Seq can also be used to determine exon/intron boundaries and verify or amend previously annotated 5′ and 3′ gene boundaries.


Publically available human tumor gene expression data (TCGA) from adult patients was mined and analyzed for the expression of CD200 by tumor type (see FIG. 1). RNAseq expression data was retrieved from the TCGA raw data FTP site. Specifically, expression data was retrieved from tcgadata.nci.nih.gov/tcgafiles/ftp_auth/distro_ftpusers/anonymous/tumor/<disease>/cgcc/unc.edu/illuminahiseq_rnasegv2/rnasegv2/. Clinical data was retrieved from tcga-data.nci.nih.gov/tcgafiles/ftp_auth/distro_ftpusers/anonymous/tumor/disease>/bcr/biotab/clin/.


The retrieved files contained sets of genes, raw read counts (RPKM), scaled estimates (“TPM” or “Transcripts Per Million”, see www.ncbi.nlm.nih.gov/pmc/articles/PMC4702907/), and clinical measures for each sample/patient. The data was then reformatted and merged, and saved as a simple raw input data file in TSV format for further processing by R scripts. TPM values were considered continuous data, and were compared using standard R statistical tools.


Tumor types included in the analysis were: diffuse large B cell lymphoma (DLBL), lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), glioblastoma (GBM), low grade glioma (LGG), clear cell RCC (KIRC), chromophobe (KICH), papillary cell RCC (KIRP), melanoma (SKCM), ovarian cancer (OV), colon cancer (COAD), rectum cancer (READ), uterine endometrial cancer (UCEC). The cutoff date of data download was March 2016.


2. Correlation of Gene Expression in Tumors


Pair-wise correlation of gene expression was evaluated for a list of genes including CD200, CD200R1, and a number of immune cell markers and targets: NT5E, IDO1, LRRC32, CTLA4, TNFRSF9, CD27, CD40, TNFRSF25, TNFRSF18, ICOS, TNFRSF4, PDCD1, CD274, PDCD1LG2, TIGIT, HAVCR2, LAG3, KIR2DL1, KIR2DL2, KIR2DL3, KIR2DS1, CXCR4, FOXP3, CD8B, CD3E, CD4, CD19, NCR1, CD68, CD14, CD163, MRC1, FCGR2A, FCGR1A, FCGR1B, TBX21, GATA3, PTPRC. Pearson correlation coefficients were calculated per tumor type, and per gene pair.


3. Generation of a Signature Indicative of Responsiveness to Anti-CD200 Therapy


Recent literature suggests that the presence of immune effector cells within tumors is critical for clinical response to immunotherapy. To find a tumor indication that may respond to anti-CD200 therapy, it was postulated that tumors with infiltrating effector cells expressing CD200 and/or CD200R1 might be responsive to anti-CD200 therapy. To this end the correlation between immune markers and CD200/CD200R1 were examined.


This assessment revealed that CD200R1, but not CD200, is significantly correlated with the expression of immune cell markers, across all tumor types examined (see FIG. 3). CD200 expression is not restricted to immune cells; both tumor cells and infiltrating immune cells contribute to overall expression of CD200 in tumor tissues. However, CD200R1 expression is restricted to immune effector cells (macrophages, monocytes, etc.). This suggested that CD200R1 was a better marker of immune cell infiltration in tumors. In particular, immune markers that exhibited the most significant correlation are markers of T cell and macrophage, two effector cell types known to express CD200R1.


Accordingly, a “samalizumab competent” gene signature was developed from genes which co-correlate with CD200R1 expression across tumor types, and includes markers of T cells and macrophages. This gene expression signature is composed of 10 genes: CD200R1, T cell markers such as ICOS, TIGIT, TNFRSF9, HAVCR2, and PDCD1, and macrophage markers such as FCGR2A, FCGR1A, CD163, and CD14. These T cell and macrophage markers exhibit the most significant correlation with CD200R1. This signature was used to identify tumor types which harbor CD200R1-expressing immune infiltrates that may mediate sensitivity to samalizumab.


The Gene Set Variation Analysis (GSVA) was utilized to calculate sample-wise gene set enrichment scores as a function of genes inside and outside the gene set (i.e. signature), analogously to a competitive gene set test (see, e.g., Hänzelmann, S., et al., BMC Bioinformatics 14, 7 (2013). The median signature score across all samples was then calculated. For every tumor type, the percentage of samples with signature scores above the overall median was calculated. Tumor types were then ranked based on the percentage values. As shown in FIG. 17, high samalizumab competent signature scores were found in large B cell lymphoma (79%), lung adenocarcinoma (77%), and glioblastoma (74%). Low signature scores were found in chromophobe (13%), uterine endometrial cancer (17%) and rectum cancer (19%). Immunohistochemistry (see Example 3) and the gene expression data were highly concordant.


The data suggest that tumors having the “samalizumab competent” gene signature will be more likely to respond to treatment with samalizumab or other anti-CD200 therapy than tumors that lack this gene signature.


Example 2: Pediatric Tumor Application

The resulting gene signature from Example 1 was then applied to a pediatric tumor database and the expression levels were evaluated and normalized in pediatric tumors.


1. Data source and processing Possible pediatric tumor expression projects were identified through key words searches in the ArrayExpress and Gene Expression Omnibus (GEO) databases using terms “pediatric tumor” and “pediatric cancer”. In order to be able to normalize expression across projects, projects utilizing UniGene Build #133 based arrays (e.g. HG-U133A, HG-U133 plus 2.0, and HG-U133+PM) were retained for further analysis. The raw data (.CEL files) of the selected projects were downloaded and processed uniformly using the R package oligo::rma with default settings (with quantile normalization and background). Biobase::exprs was used to extract the normalized expression data as a matrix for further processing.


Sample annotations were also extracted from the respective databases and used to eliminate non-pediatric samples. Samples were identified as pediatric based on explicit identification as such, an exclusively pediatric tumor type, or patient age under 18. Non-primary human tumor samples such as tumor cell line, xenograft, etc. were excluded entirely. The data was processed and normalized according to the analysis set forth in FIG. 2.


2. Mapping from Probe Set to Gene


A single probe set was selected for each gene. Only probe sets that uniquely identified a gene and were in use across all array types were considered. For each of the housekeeping genes (ACTB, OAZ1, UBC, RPLPO) used for normalization, the probe set with the highest average Spearman correlation with other housekeeping genes across all sample was chosen. For all other genes, the probe set with the highest mean expression across the most projects was chosen. After the mapping, each sample was translated into a list of analyzable genes with expression values for the sample.


3. Data Normalization Across Projects


An intensity multiplier was calculated for each sample based on the expression of four housekeeping genes: actin beta (ACTB), ornithine decarboxylase antizyme 1 (OAZ1), ubiquitin C (UBC), and ribosomal protein lateral stalk subunit P0 (RPLP0). First, for each housekeeping gene, the mean expression across all samples was calculated. Second, for each sample and for each housekeeping gene, a scale factor was calculated by dividing the sample expression value by the mean expression value. Third, for each sample, its intensity multiplier was calculated as the geometric mean of the scale factors of the four housekeeping genes.


For all samples, the gene expression values were normalized by multiplying with the sample intensity multiplier value.


4. Expression and Signature Analyses


Because of array coverages, six (ICOS, TNFRSF9, FCGR2A, PDCD1, CD163, CD14) out of the ten genes were used in this signature analysis. The Gene Set Variation Analysis (GSVA) was utilized to calculate sample-wise gene set enrichment scores as a function of genes inside and outside the gene set (i.e. signature), analogously to a competitive gene set test (see, e.g., Hänzelmann, S., et al., BMC Bioinformatics 14, 7 (2013). The median signature score across all samples was then calculated. For every tumor type, the percentage of samples with signature scores above the overall median was calculated. Tumor types were then ranked based on the percentage values. As shown in FIG. 16, high samalizumab scores were found in osteosarcoma (98%), neuroblastoma (74%), and rhabdomyosarcoma (73%). Low signature scores were found in retinoblastoma (8%), Wilms tumor (24%), and ALL B relapse (36%). Immunohistochemistry (see Example 3) and the gene expression data were highly concordant.


For each individual gene of interest, the median value of expression for that gene across all samples was first calculated. For every tumor type, the percentage of samples with the individual gene of interest with expression above the overall median was calculated.


Additionally, CD200 expression levels were measured. As shown in FIG. 4, high CD200 expression levels were found in Pilocytic Astrocytoma (96%), Neuroblastoma (88%), and Retinoblastoma (71%). Low expression levels were found in Osteosarcoma (2%), Rhabdomyosarcoma (4%), and ALL Relapse (5%). CD4 (T cell marker) medium-centered expression and its correlation with CD200 (see FIGS. 5A and 5B), as well as CD68 (macrophage marker) medium centered expression and its correlation with CD200 (see FIGS. 6A and 6B), were also measured. Cancer responsiveness may correlate with CD200 expression in addition to the gene signature.


Example 3: Immunohistochemistry Analysis of Adult and Pediatric Tumors

To confirm the gene expression data, a series of adult and pediatric tumor sections were analyzed by immunohistochemistry for expression of CD200 and infiltration of immune cells. A CD200 Immunohistochemistry (IHC) assay was developed for the analysis of patient biopsies in samalizumab clinical trials. Immune marker IHC assays were commercially available (Table 3). Commercially available tissue microarrays and slides encompassing both adult and pediatric tumor tissues were analyzed for tumor CD200 expression and the presence of the following select immune cell infiltrates by immunohistochemistry:












TABLE 3







Tissue Marker
Population









CD200
Tumor/immune cells



CD3
T-cells



CD8
Cytoxic T-cells



CD68
Monocytes/macrophages



FoxP3
Regulatory T cells










The tissue microarrays and slides utilized are shown in FIG. 7 and the analyzed tissue samples are shown in FIG. 8.


The results of the IHC assay for control tissues (FIG. 9A), Rhabdomyosarcoma (FIG. 9B), Nephroblastoma (FIG. 9C), and Neuroblastoma (FIG. 9D) are shown in FIGS. 9A-9D.


Tumor CD200 expression as assessed by IHC is set forth in FIG. 10. FIG. 11 depicts tumor infiltrates in all samples. Levels of immune infiltrates in brain neoplasia (FIG. 12), neuroblastomas (FIG. 13), nephroblastomas (FIGS. 14A-14D), and osteosarcomas (FIGS. 15A-15D) are also provided. The presence of immune effector cells in these CD200+ human tumors is supportive of targeting CD200 in these populations.












SEQUENCE LISTING SUMMARY








DESIGNATION
SEQUENCE





SEQ ID NO: 1
MERLVIRMPF SHLSTYSLVW VMAAVVLCTA QVQVVTQDER EQLYTPA


CD200 isoform A
SLK CSLQNAQEAL IVTWQKKKAV SPENMVTFSE NHGVVIQPAY


NCBI Reference
KDKINITQLG LQNSTITFWN ITLEDEGCYM CLFNTFGFGK ISGTACLTV


Sequence:
Y VQPIVSLHYK FSEDHLNITC SATARPAPMV FWKVPRSGIE NSTVTL


NP_005935.4
SHPN GTTSVTSILH IKDPKNQVGK EVICQVLHLG TVTDFKQTVN



KGYWFSVPLLLSIVSLVILL VLISILLYWK RHRNQDREP





SEQ ID NO: 2
MERLTLTRTI GGPLLTATLL GKTTINDYQV IRMPFSHLST YSLVW


CD200 isoform B
VMAAV VLCTAQVQVVTQDEREQLYT PASLKCSLQN AQEALIVTW


NCBI Reference
Q KKKAVSPENM VTFSENHGVV IQPAYKDKINITQLGLQNST ITFW


Sequence:
NITLED EGCYMCLFNT FGFGKISGTA CLTVYVQPIV SLHYKFSEDH


NP_001004196.2
LNITCSATAR PAPMVFWKVP RSGIENSTVT LSHPNGTTSV TSILHIK



DPK NQVGKEVICQVLHLGTVTDF KQTVNKGYWF SVPLLLSIVS



LVILLVLISI LLYWKRHRNQ DREP





SEQ ID NO: 3
MKGVTCVSSI PLVLGRSQER PASPSMPIVS LHYKFSEDHL NITCSA


CD200 isoform c
TARP APMVFWKVPRSGIENSTVTL SHPNGTTSVT SILHIKDPKN


NCBI Reference
QVGKEVICQV LHLGTVTDFK QTVNKGYWFSVPLLLSIVSL VILLV


Sequence:
LISIL LYWKRHRNQD REP


NP_001305755.1/



NP_001305759.1






SEQ ID NO: 4
KASQDINSYLS


Samalizumab



Light Chain



CDR1






SEQ ID NO: 5
RANRLVD


Samalizumab



Light Chain



CDR2






SEQ ID NO: 6
LQYDEFPYT


Samalizumab



Light Chain



CDR3






SEQ ID NO: 7
GYSFTDYIIL


Samalizumab



Heavy Chain



CDR1






SEQ ID NO: 8
HIDPYYGSSNYNLKFKG


Samalizumab



Heavy Chain



CDR2






SEQ ID NO: 9
SKRDYFDY


Samalizumab



Heavy Chain



CDR3






SEQ ID NO: 10
DIQMTQSPSS LSASIGDRVT ITCKASQDIN SYLSWFQQKP GKAPKLLIYR


Samalizumab
ANRLVDGVPS RFSGSGSGTD YTLTISSLQP EDFAVYYCLQ


Light Chain
YDEFPYTFGG GTKLEIKRTV AAPSVFIFPP SDEQLKSGTA



SVVCLLNNFY PREAKVQWKV DNALQSGNSQ ESVTEQDSKD



STYSLSSTLTLSKADYEKHK VYACEVTHQ GLSSPVTKSF NRGEC





SEQ ID NO: 11
QVQLQQSGSE LKKPGASVKI SCKASGYSFT DYIILWVRQN


Samalizumab
PGKGLEWIGH IDPYYGSSNY NLKFKGRVTI TADQSTTTAY


Heavy Chain
MELSSLRSED TAVYYCGRSK RDYFDYWGQG TTLTVSSAST



KGPSVFPLAP CSRSTSESTA ALGCLVKDYF PEPVTVSWNS



GALTSGVHTF PAVLQSSGLYSLSSVVTVPS SNFGTQTYTC



NVDHKPSNTK VDKTVERKCC VECPPCPAPP VAGPSVFLFP



PKPKDTLMIS RTPEVTCVVV DVSQEDPEVQ FNWYVDGVEV



HNAKTKPREE QFNSTYRVVSVLTVLHQDWL NGKEYKCKVS



NKGLPSSIEK TISKAKGQPR EPQVYTLPPS QEEMTKNQVS



LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF



FLYSRLTVDK SRWQEGNVFS CSVMHEALHN HYTQKSLSLS LGK





SEQ ID NO: 12
DIQMTQSP SSLSASIGDR VTITCKASQD INSYLSWFQQ KPGKAPKLLI


Samalizumab
YRANRLVDGV PSRFSGSGSG TDYTLTISSL QPEDFAVYYC


Light Chain
LQYDEFPYTF GGGTKLEIKR


Variable Region






SEQ ID NO: 13
QVQLQQSGS ELKKPGASVK ISCKASGYSF TDYIILWVRQ


Samalizumab
NPGKGLEWIG HIDPYYGSSN YNLKFKGRVT ITADQSTTTA


Heavy Chain
YMELSSLRSE DTAVYYCGRS KRDYFDYWGQ GTTLTVSS





Variable Region



SEQ ID NO: 14
MKSGLWYFFL FCLRIKVLTG EINGSANYEM FIFHNGGVQI


ICOS precursor;
LCKYPDIVQQ FKMQLLKGGQ ILCDLTKTKG SGNTVSIKSL


NP_036224.1
KFCHSQLSNN SVSFFLYNLD HSHANYYFCN LSIFDPPPFK


sapiens
VTLTGGYLHI YESQLCCQLK FWLPIGCAAF VVVCILGCIL



ICWLTKKKYS SSVHDPNGEY MFMRAVNTAK KSRLTDVTL





SEQ ID NO: 15
cgagagcctg aattcactgt cagctttgaa cactgaacgc gaggactgtt aactgtttct



Homosapiens

ggcaaacatg aagtcaggcc tctggtattt ctttctcttc tgcttgcgca ttaaagtttt


inducible T-cell
aacaggagaa atcaatggtt ctgccaatta tgagatgttt atatttcaca acggaggtgt


costimulator
acaaatttta tgcaaatatc ctgacattgt ccagcaattt aaaatgcagt tgctgaaagg


(ICOS)
ggggcaaata ctctgcgatc tcactaagac aaaaggaagt ggaaacacag tgtccattaa


NM_012092.3,
gagtctgaaa ttctgccatt ctcagttatc caacaacagt gtctcttttt ttctatacaa


mRNA
cttggaccat tctcatgcca actattactt ctgcaaccta tcaatttttg atcctcctcc



ttttaaagta actcttacag gaggatattt gcatatttat gaatcacaac tttgttgcca



gctgaagttc tggttaccca taggatgtgc agcctttgtt gtagtctgca ttttgggatg



catacttatt tgttggctta caaaaaagaa gtattcatcc agtgtgcacg accctaacgg



tgaatacatg ttcatgagag cagtgaacac agccaaaaaa tctagactca cagatgtgac



cctataatat ggaactctgg cacccaggca tgaagcacgt tggccagttt tcctcaactt



gaagtgcaag attctcttat ttccgggacc acggagagtc tgacttaact acatacatct



tctgctggtg ttttgttcaa tctggaagaa tgactgtatc agtcaatggg gattttaaca



gactgccttg gtactgccga gtcctctcaa aacaaacacc ctcttgcaac cagctttgga



gaaagcccag ctcctgtgtg ctcactggga gtggaatccc tgtctccaca tctgctccta



gcagtgcatc agccagtaaa acaaacacat ttacaagaaa aatgttttaa agatgccagg



ggtactgaat ctgcaaagca aatgagcagc caaggaccag catctgtccg catttcacta



tcatactacc tcttctttct gtagggatga gaattcctct tttaatcagt caagggagat



gcttcaaagc tggagctatt ttatttctga gatgttgatg tgaactgtac attagtacat



actcagtact ctccttcaat tgctgaaccc cagttgacca ttttaccaag actttagatg



ctttcttgtg ccctcaattt tctttttaaa aatacttcta catgactgct tgacagccca



acagccactc tcaatagaga gctatgtctt acattctttc ctctgctgct caatagtttt



atatatctat gcatacatat atacacacat atgtatataa aattcataat gaatatattt



gcctatattc tccctacaag aatatttttg ctccagaaag acatgttctt ttctcaaatt



cagttaaaat ggtttacttt gttcaagtta gtggtaggaa acattgcccg gaattgaaag



caaatttatt ttattatcct attttctacc attatctatg ttttcatggt gctattaatt



acaagtttag ttctttttgt agatcatatt aaaattgcaa acaaaatcat ctttaatggg



ccagcattct catggggtag agcagaatat tcatttagcc tgaaagctgc agttactata



ggttgctgtc agactatacc catggtgcct ctgggcttga caggtcaaaa tggtccccat



cagcctggag cagccctcca gacctgggtg gaattccagg gttgagagac tcccctgagc



cagaggccac taggtattct tgctcccaga ggctgaagtc accctgggaa tcacagtggt



ctacctgcat tcataattcc aggatctgtg aagagcacat atgtgtcagg gcacaattcc



ctctcataaa aaccacacag cctggaaatt ggccctggcc cttcaagata gccttcttta



gaatatgatt tggctagaaa gattcttaaa tatgtggaat atgattattc ttagctggaa



tattttctct acttcctgtc tgcatgccca aggcttctga agcagccaat gtcgatgcaa



caacatttgt aactttaggt aaactgggat tatgttgtag tttaacattt tgtaactgtg



tgcttatagt ttacaagtga gacccgatat gtcattatgc atacttatat tatcttaagc



atgtgtaatg ctggatgtgt acagtacagt actgaacttg taatttgaat ctagtatggt



gttctgtttt cagctgactt ggacaacctg actggctttg cacaggtgtt ccctgagttg



tttgcaggtt tctgtgtgtg gggtggggta tggggaggag aaccttcatg gtggcccacc



tggcctggtt gtccaagctg tgcctcgaca catcctcatc cccagcatgg gacacctcaa



gatgaataat aattcacaaa atttctgtga aatcaaatcc agttttaaga ggagccactt



atcaaagaga ttttaacagt agtaagaagg caaagaataa acatttgata ttcagcaact



gaaaaaaaaa aa





SEQ ID NO: 16
MRWCLLLIWA QGLRQAPLAS GMMTGTIETT GNISAEKGGS


T-cell
IILQCHLSST TAQVTQVNWEQQDQLLAICN ADLGWHISPS


immunoreceptor
FKDRVAPGPG LGLTLQSLTV NDTGEYFCIY HTYPDGTYTG


with Ig and ITIM
RIFLEVLESS VAEHGARFQI PLLGAMAATL VVICTAVIVV


domain precursor
VALTRKKKAL RIHSVEGDLRRKSAGQEEWS PSAPSPPGSC


[Homosapiens]
VQAEAAPAGL CGEQRGEDCA ELHDYFNVLS YRSLGNCSFF


NP_776160.2
TETG





SEQ ID NO: 17
cgtcctatct gcagtcggct actttcagtg gcagaagagg ccacatctgc ttcctgtaggccctctgggc



Homosapiens T-

agaagcatgc gctggtgtct cctcctgatc tgggcccagg ggctgaggca ggctcccctc gcctcaggaa


cell
tgatgacagg cacaatagaa acaacgggga acatttctgcagagaaaggt ggctctatca tcttacaatg


immunoreceptor
tcacctctcc tccaccacgg cacaagtgac ccaggtcaac tgggagcagc aggaccagct tctggccatt


with Ig and ITIM
tgtaatgctg acttggggtggcacatctcc ccatccttca aggatcgagt ggccccaggt cccggcctgg


domains (TIGIT),
gcctcaccct ccagtcgctg accgtgaacg atacagggga gtacttctgc atctatcaca


mRNA,
cctaccctgatgggacgtac actgggagaa tcttcctgga ggtcctagaa agctcagtgg ctgagcacgg


NM_173799.3
tgccaggttc cagattccat tgcttggagc catggccgcg acgctggtgg tcatctgcacagcagtcatc



gtggtggtcg cgttgactag aaagaagaaa gccctcagaa tccattctgt ggaaggtgac ctcaggagaa



aatcagctgg acaggaggaa tggagcccca gtgctccctcacccccagga agctgtgtcc aggcagaagc



tgcacctgct gggctctgtg gagagcagcg gggagaggac tgtgccgagc tgcatgacta cttcaatgtc



ctgagttaca gaagcctgggtaactgcagc ttcttcacag agactggtta gcaaccagag gcatcttctg



gaagatacac ttttgtcttt gctattatag atgaatatat aagcagctgt actctccatc



agtgctgcgtgtgtgtgtgt gtgtgtatgt gtgtgtgtgt tcagttgagt gaataaatgt catcctcttc



tccatcttca tttccttggc cttttcgttc tattccattt tgcattatgg caggcctagggtgagtaacg



tggatcttga tcataaatgc aaaattaaaa aatatcttga cctggtttta aatctggcag tttgagcaga



tcctatgtct ctgagagaca cattcctcat aatggccagcattttgggct acaaggtttt gtggttgatg



atgaggatgg catgactgca gagccatcct catctcattt tttcacgtca ttttcagtaa ctttcactca



ttcaaaggca ggttataagtaagtcctggt agcagcctct atggggagat ttgagagtga ctaaatcttg



gtatctgccc tcaagaactt acagttaaat ggggagacaa tgttgtcatg aaaaggtatt



atagtaaggagagaaggaga catacacagg ccttcaggaa gagacgacag tttggggtga ggtagttggc



ataggcttat ctgtgatgaa gtggcctggg agcaccaagg ggatgttgag gctagtctgggaggagcagg



agttttgtct agggaacttg taggaaattc ttggagctga aagtcccaca aagaaggccc tggcaccaag



ggagtcagca aacttcagat tttattctct gggcaggcatttcaagatc cttttgctgt gacatactca



tccattagac agcctgatac aggcctgtag cctcttccgg ccgtgtgtgc tggggaagcc ccaggaaacg



cacatgccca cacagggagccaagtcgtag catttgggcc ttgatctacc ttttctgcat caatacactc



ttgagccttt gaaaaaagaa cgtttcccac taaaaagaaa atgtggattt ttaaaatagg



gactcttcctaggggaaaaa ggggggctgg gagtgataga gggtttaaaa aataaacacc ttcaaactaa



cttcttcgaa cccttttatt cactccctga cgactttgtg ctggggttgg ggtaactgaaccgcttattt



ctgtttaatt gcattcaggc tggatcttag aagactttta tccttccacc atctctctca gaggaatgag



cggggaggtt ggatttactg gtgactgatt ttctttcatgggccaaggaa ctgaaagaga atgtgaagca



aggttgtgtc ttgcgcatgg ttaaaaataa agcattgtcc tgcttcctaa gacttagact ggggttgaca



attgttttag caacaagacaattcaactat ttctcctagg atttttatta ttattatttt ttcacttttc



taccaaatgg gttacatagg



aagaatgaac tgaaatctgt ccagagctcc aagtcctttg gaagaaagattagatgaacg



taaaaatgtt gttgtttgct gtggcagttt acagcatttt tcttgcaaaa ttagtgcaaa tctgttggaa



atagaacaca attcacaaat tggaagtgaa ctaaaatgtaatgacgaaaa gggagtagtg ttttgatttg



gaggaggtgt atattcggca gaggttggac tgagagttgg gtgttattta acataattat ggtaattggg



aaacatttat aaacactattgggatggtga taaaatacaa aagggcctat agatgttaga aatgggtcag



gttactgaaa tgggattcaa tttgaaaaaa atttttttaa atagaactca ctgaactaga



ttctcctctgagaaccagag aagaccattt catagttgga ttcctggaga catgcgctat ccaccacgta



gccactttcc acatgtggcc atcaaccact taagatgggg ttagtttaaa tcaagatgtgctgttataat



tggtataagc ataaaatcac actagattct ggagatttaa tatgaataat aagaatacta tttcagtagt



tttggtatat tgtgtgtcaa aaatgataat attttggatg tattgggtga aataaaatat taacattaaa



aaaaaaaa





SEQ ID NO: 18
MGNSCYNIVA TLLLVLNFER TRSLQDPCSN CPAGTFCDNN


tumor necrosis
RNQICSPCPP NSFSSAGGQRTCDICRQCKG VFRTRKECSS


factor receptor
TSNAECDCTP GFHCLGAGCS MCEQDCKQGQ ELTKKGCKDC


superfamily
CFGTFNDQKR GICRPWTNCS LDGKSVLVNG TKERDVVCGP


member 9
SPADLSPGAS SVTPPAPAREPGHSPQIISF FLALTSTALL FLLFFLTLRF


precursor [Homo
SVVKRGRKKL LYIFKQPFMR PVQTTQEEDGCSCRFPEEEE GGCEL



sapiens]




NP_001552.2






SEQ ID NO: 19
caaggaggga tcccacagat gtcacagggc tgtcacagag ctgtggtggg aatttcccatgagaccccgc



Homosapiens

ccctggctga gtcaccgcac tcctgtgttt gacctgaagt cctctcgagc tgcagaagcc tgaagaccaa


TNF receptor
ggagtggaaa gttctccggc agccctgaga tctcaagagtgacatttgtg agaccagcta atttgattaa


superfamily
aattctcttg gaatcagctt tgctagtatc atacctgtgc cagatttcat catgggaaac agctgttaca


member 9
acatagtagc cactctgttgctggtcctca actttgagag gacaagatca ttgcaggatc cttgtagtaa


(TNFRSF9),
ctgcccagct ggtacattct gtgataataa caggaatcag atttgcagtc cctgtcctcc


mRNA,
aaatagtttctccagcgcag gtggacaaag gacctgtgac atatgcaggc agtgtaaagg tgttttcagg


NM_001561.5
accaggaagg agtgttcctc caccagcaat gcagagtgtg actgcactcc agggtttcactgcctggggg



caggatgcag catgtgtgaa caggattgta aacaaggtca agaactgaca aaaaaaggtt gtaaagactg



ttgctttggg acatttaacg atcagaaacg tggcatctgtcgaccctgga caaactgttc tttggatgga



aagtctgtgc ttgtgaatgg gacgaaggag agggacgtgg tctgtggacc atctccagcc gacctctctc



cgggagcatc ctctgtgaccccgcctgccc ctgcgagaga gccaggacac tctccgcaga tcatctcctt



ctttcttgcg ctgacgtcga ctgcgttgct cttcctgctg ttcttcctca cgctccgttt



ctctgttgttaaacggggca gaaagaaact cctgtatata ttcaaacaac catttatgag accagtacaa



actactcaag aggaagatgg ctgtagctgc cgatttccag aagaagaaga aggaggatgtgaactgtgaa



atggaagtca atagggctgt tgggactttc ttgaaaagaa gcaaggaaat atgagtcatc cgctatcaca



gctttcaaaa gcaagaacac catcctacat aatacccaggattcccccaa cacacgttct tttctaaatg



ccaatgagtt ggcctttaaa aatgcaccac tttttttttt tttttgacag ggtctcactc tgtcacccag



gctggagtgc agtggcaccaccatggctct ctgcagcctt gacctctggg agctcaagtg atcctcctgc



ctcagtctcc tgagtagctg gaactacaag gaagggccac cacacctgac taactttttt



gttttttgtttggtaaagat ggcatttcac catgttgtac aggctggtct caaactccta ggttcacttt



ggcctcccaa agtgctggga ttacagacat gaactgccag gcccggccaa aataatgcaccacttttaac



agaacagaca gatgaggaca gagctggtga taaaaaaaaa aaaaaaaaag cattttctag ataccactta



acaggtttga gctagttttt ttgaaatcca aagaaaattatagtttaaat tcaattacat agtccagtgg



tccaactata attataatca aaatcaatgc aggtttgttt tttggtgcta atatgacata tgacaataag



ccacgaggtg cagtaagtacccgactaaag tttccgtggg ttctgtcatg taacacgaca tgctccaccg



tcagggggga gtatgagcag agtgcctgag tttagggtca aggacaaaaa acctcaggcc



tggaggaagttttggaaaga gttcaagtgt ctgtatatcc tatggtcttc tccatcctca caccttctgc



ctttgtcctg ctccctttta agccaggtta cattctaaaa attcttaact tttaacataatattttatac



caaagccaat aaatgaactg catatgatag



gtatgaagta cagtgagaaa attaacacct gtgagctcat tgtcctacca



cagcactaga gtgggggccg ccaaactcccatggccaaac ctggtgcacc atttgccttt gtttgtctgt



tggtttgctt gagacagtct tgctctgttg cccaggctgg aatggagtgg ctattcacag gcacaatcat



agcacactttagccttaaac tcctgggctc aagtgatcca cccgcctcag tctcccaagt agctgggatt



acaggtgcaa acctggcatg cctgccattg tttggcttat gatctaagga tagctttttaaattttattc



attttatttt tttttgagac agtgtctcac



tctgtctccc aggctggagt acagtggtac aatcttggat caccgcctcc



cagtttcaag tgatctccct gcctcagcctcctaagtagc tgggactaca ggtatgtgcc accacgcctg



gctaattttt atatttttag tagagacggg gtttcaccat gttgtccagg ctggtctcaa actcctgacc



tcaggtgatctgcccacctc tgcctcccaa agtgctggga ttacaggcat gagccaccat gcctggccat



ttcttacact tttgtatgac atgcctattg caagcttgcg tgcctctgtc ccatgttattttactctggg



atttaggtgg agggagcagc ttctatttgg aacattggcc atcgcatggc aaatgggtat ctgtcacttc



tgctcctatt tagttggttc tactataacc tttagagcaaatcctgcagc caagccaggc atcaataggg



cagaaaagta tattctgtaa ataggggtga ggagaagata tttctgaaca atagtctact gcagtaccaa



attgcttttc aaagtggctgttctaatgta ctcccgtcag tcatataagt gtcatgtaag tatcccattg



atccacatcc ttgctaccct ctggtactat caggtgccct taattttgcc aagccagtgg



gtatagaatgagatctcact gtggtcttag tttgcatttg cttggttact gatgagcacc ttgtcaaata



tttatatacc atttgtgttt atttttttaa ataaaatgct tgctcatgct tttttgcccatttgcaaaaa



aacttggggc cgggtgcagt ggctcatgcc



tgtagtccca gctctttggg aggccaaggt gggcagatcg cttgagccca



ggagttcgag accagccttg gcaacatggcgaaaccctgt ctttacaaaa aatacaaaaa ttagccgggt



gtggtggtgt gcacctgaag tcccagctac tcagtaggtt cgctttgagc ctgggaggca gaggttgcag



tgagctgggaccgcatcact acacttcagc ctgggcaaca gagaaaaacc ttttctcaga aacaaacaaa



cccaaatgtg gttgtttgtc ctgattccta aaaggtcttt atgtattcta gataataatctttggtcagt



tatatgtgtt aaaaaatatc ttctttgtgg



ccaggcacgg tagctcacac ctgtaatccc agcactttgc ggggctgagg



tgggtggatc atctgaggtc aagagttcaagatcagcctg gccaacacag tgaaacccca tctctactaa



acatgtacaa aacttagctg ggtatggtgg cgggtgcctg taaccccagc tgctccagag gctgtggcag



aagaatcgcttgaacccagg aggcagaggt tgcagcgagc caagattgtg ccattgcact ccagactggg



tgacaagagt gaaattctgc ctatctatct atctatctat ctatatctat atatatatatatatatatcc



tttgtaattt atttttccct ttttaaaatt



ttttataaaa ttctttttta tttttatttt tagcagaggt gaggtttctg aggtttcatt



atgttgccca ggctggtcttgaactcctga gctcaagtga tcctcccacc tcagccttcc aaagtgctgg



aattgcagac atgagccacc gcgcccctcc tgtttttctc taattaatgg tgtctttctt



tgtctttctggtaataagca aaaagttctt catttgattt ggttaaattt ataactgttt tctcatatgg



ttaacatttt ttcttgcctg gctaaagaaa tccttttctg cccaatacta taaagaggtttgcccacatt



ttattccaaa agttttaagt tttgtctttc atcttgaagt ctaatgtatc aggaactggc ttttgtgcct



gttgggaggt agtgatccaa ttccatgtct tgcatgtaggtaaccactgg tccctgcgcc atgtattcaa



tacgtcgtct ttctcctgcg ggtctgcaat ctcacctacc atccatcaag tttccatagg gccatgggtc



tgcttctggg ctccctgttctgttccattg tcaatttgtc tatcctgtgc cagtatcaca ctgtgtttat



tacaatagct ttgtaacagc tctcgatatc cggtaggaca tctccctcca ccttcttttt



ctacttcagaagtgtcttag ctaggtcagg cacggtggct cacgcctgta atcccagcac tttgggaggc



cgacgcggat ggatcacctg aggtcaggag ttttgagaca gcctggccaa catggtgaaaccccatctct



actaaaaaat acaaaaatta gtcaggcatg gtggcatgtg cctgtaatcc cagctatttg ggaggctgag



gccggagaat tgcttgaacc cggggggcgg aggttgcagtgagccgagat cgtaccattg cactccagcc



tgggtgacag agcgaaactc tgtctcagga aaaaaaagaa aagagatgtc ttggttattc ttggttcttt



attattcaat ataaattttagaagctgaat ttgaaaagat ttggattgga atttcattaa atctacaggt



caatttaggg agagttgata attttacaga attgagtcat ctggtgttcc aataagaata



agagaacaattattggctgt acaattcttg ccaaatagta ggcaaagcaa agcttaggaa gtatactggt



gccatttcag gaacaaagct aggtgcgaat atttttgtct ttctgaatca tgatgctgtaagttctaaag



tgatttctcc tcttggcttt ggacacatgg tgtttaatta cctactgctg actatccaca aacagaaaga



gactggtcat gccccacagg gttggggtat ccaagataatggagcgaggc tctcatgtgt cctaggttac



acaccgaaaa tccacagttt attctgtgaa



gaaaggaggc tatgtttatg atacagactg tgatattttt atcatagcct attctggtatcatgtgcaaa



agctataaat gaaaaacaca ggaacttggc atgtgagtca ttgctccccc taaatgacaa ttaataagga



aggaacattg agacagaata aaatgatccc cttctgggtttaatttagaa agttccataa ttaggtttaa



tagaaataaa tgtaaatttc tatgattaaa aataaattag cacatttagg gatacacaaa ttataaatca



ttttctaaat gctaaaaacaagctcaggtt tttttcagaa gaaagtttta attttttttc tttagtggaa



gatatcactc tgacggaaag ttttgatgtg



aggggcggat gactataaag tgggcatctt cccccacaggaagatgtttc



catctgtggg tgagaggtgc ccaccgcagc tagggcaggt tacatgtgcc ctgtgtgtgg taggacttgg



agagtgatct ttatcaacgt ttttatttaa aagactatctaataaaacac aaaactatga tgttcacagg



aaaaaaagaa taagaaaaaa agaaaaaaaaa





SEQ ID NO: 20
MFSHLPFDCV LLLLLLLLTR SSEVEYRAEV GQNAYLPCFY


hepatitis A virus
TPAAPGNLVP VCWGKGACPVFECGNVVLRT DERDVNYWTS


cellular 1
RYWLNGDFRK GDVSLTIENV TLADSGIYCC RIQIPGIMND


receptor (HAVCR2) 
EKFNLKLVIK PAKVTPAPTR QRDFTAAFPR MLTTRGHGPA


[Homosapiens]
ETQTLGSLPD INLTQISTLANELRDSRLAN DLRDSGATIR IGIYIGAGIC


NP_116171.3
AGLALALIFG ALIFKWYSHS KEKIQNLSLISLANLPPSGL ANAVAEGIRS



EENIYTIEEN VYEVEEPNEY YCYVSSRQQP SQPLGCRFAMP





SEQ ID NO: 21
agaacactta caggatgtgt gtagtgtggc atgacagaga actttggttt cctttaatgt


hepatitis A virus
gactgtagac ctggcagtgt tactataaga atcactggca atcagacacc cgggtgtgct


cellular 1
gagctagcac tcagtggggg cggctactgc tcatgtgatt gtggagtaga cagttggaag


receptor (HAVCR2) 
aagtacccag tccatttgga gagttaaaac tgtgcctaac agaggtgtcc tctgactttt


[Homosapiens]
cttctgcaag ctccatgttt tcacatcttc cctttgactg tgtcctgctg ctgctgctgc


NM_032782.4
tactacttac aaggtcctca gaagtggaat acagagcgga ggtcggtcag aatgcctatc



tgccctgctt ctacacccca gccgccccag ggaacctcgt gcccgtctgc tggggcaaag



gagcctgtcc tgtgtttgaa tgtggcaacg tggtgctcag gactgatgaa agggatgtga



attattggac atccagatac tggctaaatg gggatttccg caaaggagat gtgtccctga



ccatagagaa tgtgactcta gcagacagtg ggatctactg ctgccggatc caaatcccag



gcataatgaa tgatgaaaaa tttaacctga agttggtcat caaaccagcc aaggtcaccc



ctgcaccgac tcggcagaga gacttcactg cagcctttcc aaggatgctt accaccaggg



gacatggccc agcagagaca cagacactgg ggagcctccc tgatataaat ctaacacaaa



tatccacatt ggccaatgag ttacgggact ctagattggc caatgactta cgggactctg



gagcaaccat cagaataggc atctacatcg gagcagggat ctgtgctggg ctggctctgg



ctcttatctt cggcgcttta attttcaaat ggtattctca tagcaaagag aagatacaga



atttaagcct catctctttg gccaacctcc ctccctcagg attggcaaat gcagtagcag



agggaattcg ctcagaagaa aacatctata ccattgaaga gaacgtatat gaagtggagg



agcccaatga gtattattgc tatgtcagca gcaggcagca accctcacaa cctttgggtt



gtcgctttgc aatgccatag atccaaccac cttatttttg agcttggtgt tttgtctttt



tcagaaacta tgagctgtgt cacctgactg gttttggagg ttctgtccac tgctatggag



cagagttttc ccattttcag aagataatga ctcacatggg aattgaactg ggacctgcac



tgaacttaaa caggcatgtc attgcctctg tatttaagcc aacagagtta cccaacccag



agactgttaa tcatggatgt tagagctcaa acgggctttt atatacacta ggaattcttg



acgtggggtc tctggagctc caggaaattc gggcacatca tatgtccatg aaacttcaga



taaactaggg aaaactgggt gctgaggtga aagcataact tttttggcac agaaagtcta



aaggggccac tgattttcaa agagatctgt gatccctttt tgttttttgt ttttgagatg



gagtcttgct ctgttgccca ggctggagtg caatggcaca atctcggctc actgcaagct



ccgcctcctg ggttcaagcg attctcctgc ctcagcctcc tgagtggctg ggattacagg



catgcaccac catgcccagc taatttgttg tatttttagt agagacaggg tttcaccatg



ttggccagtg tggtctcaaa ctcctgacct catgatttgc ctgcctcggc ctcccaaagc



actgggatta caggcgtgag ccaccacatc cagccagtga tccttaaaag attaagagat



gactggacca ggtctacctt gatcttgaag attcccttgg aatgttgaga tttaggctta



tttgagcact gcctgcccaa ctgtcagtgc cagtgcatag cccttctttt gtctccctta



tgaagactgc cctgcagggc tgagatgtgg caggagctcc cagggaaaaa cgaagtgcat



ttgattggtg tgtattggcc aagttttgct tgttgtgtgc ttgaaagaaa atatctctga



ccaacttctg tattcgtgga ccaaactgaa gctatatttt tcacagaaga agaagcagtg



acggggacac aaattctgtt gcctggtgga aagaaggcaa aggccttcag caatctatat



taccagcgct ggatcctttg acagagagtg gtccctaaac ttaaatttca agacggtata



ggcttgatct gtcttgctta ttgttgcccc ctgcgcctag cacaattctg acacacaatt



ggaacttact aaaaattttt ttttactgtt aaaaaaaaaa aaaaaaaa





SEQ ID NO: 22
MQIPQAPWPV VWAVLQLGWR PGWFLDSPDR PWNPPTFSPA



Homosapiens

LLVVTEGDNA TFTCSFSNTSESFVLNWYRM SPSNQTDKLA


programmed cell
AFPEDRSQPG QDCRFRVTQL PNGRDFHMSV VRARRNDSGT


death protein 1
YLCGAISLAP KAQIKESLRA ELRVTERRAE VPTAHPSPSP


precursor
RPAGQFQTLV VGVVGGLLGSLVLLVWVLAV ICSRAARGTI


(PDCD1),
GARRTGQPLK EDPSAVPVFS VDYGELDFQW REKTPEPPVP


NP_005009.2[]
CVPEQTEYAT IVFPSGMGTS SPARRGSADG PRSAQPLRPE DGHCSWPL





SEQ ID NO: 23
agtttccctt ccgctcacct ccgcctgagc agtggagaag gcggcactct ggtggggctgctccaggcat



Homosapiens

gcagatccca caggcgccct ggccagtcgt ctgggcggtg ctacaactgg gctggcggcc aggatggttc


programmed cell
ttagactccc cagacaggcc ctggaacccc cccaccttctccccagccct gctcgtggtg accgaagggg


death 1 (PDCD1),
acaacgccac cttcacctgc agcttctcca acacatcgga gagcttcgtg ctaaactggt accgcatgag


mRNA
ccccagcaac cagacggacaagctggccgc cttccccgag gaccgcagcc agcccggcca


NM_005018.2
cacaactgcc caacgggcgt gacttccaca tgagcgtggt cagggcccgg cgcaatgaca



gcggcaccta cctctgtggg gccatctccc tggcccccaa ggcgcagatc aaagagagcctgcgggcaga



gctcagggtg acagagagaa gggcagaagt gcccacagcc caccccagcc cctcacccag



gccagccggc cagttccaaa ccctggtggt tggtgtcgtg ggcggcctgctgggcagcct ggtgctgcta



gtctgggtcc tggccgtcat ctgctcccgg gccgcacgag ggacaatagg agccaggcgc



accggccagc ccctgaagga ggacccctca gccgtgcctgtgttctctgt ggactatggg gagctggatt



tccagtggcg agagaagacc ccggagcccc ccgtgccctg tgtccctgag cagacggagt atgccaccat



tgtctttcct agcggaatgggcacctcatc ccccgcccgc aggggctcag ctgacggccc tcggagtgcc



cagccactga ggcctgagga tggacactgc tcttggcccc tctgaccggc ttccttggcc



accagtgttctgcagaccct ccaccatgag cccgggtcag cgcatttcct caggagaagc aggcagggtg



caggccattg caggccgtcc aggggctgag ctgcctgggg gcgaccgggg



ctccagcctgcacctgcacc aggcacagcc ccaccacagg actcatgtct caatgcccac agtgagccca



ggcagcaggt gtcaccgtcc cctacaggga gggccagatg cagtcactgc ttcaggtcctgccagcacag



agctgcctgc gtccagctcc ctgaatctct gctgctgctg ctgctgctgc tgctgctgcc tgcggcccgg



ggctgaaggc gccgtggccc tgcctgacgc cccggagcctcctgcctgaa cttgggggct ggttggagat



ggccttggag cagccaaggt gcccctggca gtggcatccc gaaacgccct ggacgcaggg



cccaagactg ggcacaggag tgggaggtacatggggctgg ggactcccca ggagttatct gctccctgca



ggcctagaga agtttcaggg aaggtcagaa gagctcctgg ctgtggtggg cagggcagga aacccctcca



cctttacacatgcccaggca gcacctcagg ccctttgtgg ggcagggaag ctgaggcagt aagcgggcag



gcagagctgg aggcctttca ggcccagcca gcactctggc ctcctgccgc cgcattccaccccagcccct



cacaccactc gggagaggga catcctacgg tcccaaggtc aggagggcag ggctggggtt



gactcaggcc cctcccagct gtggccacct gggtgttggg agggcagaagtgcaggcacc tagggccccc



catgtgccca ccctgggagc tctccttgga acccattcct gaaattattt aaaggggttg gccgggctcc



caccagggcc tgggtgggaa ggtacaggcgttcccccggg gcctagtacc cccgccgtgg cctatccact



cctcacatcc acacactgca cccccactcc tggggcaggg ccaccagcat ccaggcggcc



agcaggcacc tgagtggctgggacaaggga tcccccttcc ctgtggttct attatattat aattataatt



aaatatgaga gcatgctaag gaaaa





SEQ ID NO: 24
MTMETQMSQN VCPRNLWLLQ PLTVLLLLAS ADSQAAAPPK


low affinity
AVLKLEPPWI NVLQEDSVTLTCQGARSPES DSIQWFHNGN


immunoglobulin
LIPTHTQPSY RFKANNNDSG EYTCQTGQTS LSDPVHLTVL


gamma Fc region
SEWLVLQTPH LEFQEGETIM LRCHSWKDKP LVKVTFFQNG


receptor II-a
KSQKFSHLDP TFSIPQANHSHSGDYHCTGN IGYTLFSSKP


isoform 1
VTITVQVPSM GSSSPMGIIV AVVIATAVAA IVAAVVALIY


precursor [Homo
CRKKRISANS TDPVKAAQFE PPGRQMIAIR KRQLEETNND



sapiens]

YETADGGYMT LNPRAPTDDDKNIYLTLPPN DHVNSNN


NP_001129691.1






SEQ ID NO: 25
ctcttttcta agcttgtctc ttaaaaccca ctggacgttg gcacagtgct gggatgacta


low affinity
tggagaccca aatgtctcag aatgtatgtc ccagaaacct gtggctgctt caaccattga


immunoglobulin
cagttttgct gctgctggct tctgcagaca gtcaagctgc agctccccca aaggctgtgc


gamma Fc region
tgaaacttga gcccccgtgg atcaacgtgc tccaggagga ctctgtgact ctgacatgcc


receptor II-a
agggggctcg cagccctgag agcgactcca ttcagtggtt ccacaatggg aatctcattc


isoform 1
ccacccacac gcagcccagc tacaggttca aggccaacaa caatgacagc ggggagtaca


precursor
cgtgccagac tggccagacc agcctcagcg accctgtgca tctgactgtg ctttccgaat


(FCGR2A) [Homo
ggctggtgct ccagacccct cacctggagt tccaggaggg agaaaccatc atgctgaggt



sapiens] mRNA

gccacagctg gaaggacaag cctctggtca aggtcacatt cttccagaat ggaaaatccc


NM_001136219.1
agaaattctc ccatttggat cccaccttct ccatcccaca agcaaaccac agtcacagtg



gtgattacca ctgcacagga aacataggct acacgctgtt ctcatccaag cctgtgacca



tcactgtcca agtgcccagc atgggcagct cttcaccaat ggggatcatt gtggctgtgg



tcattgcgac tgctgtagca gccattgttg ctgctgtagt ggccttgatc tactgcagga



aaaagcggat ttcagccaat tccactgatc ctgtgaaggc tgcccaattt gagccacctg



gacgtcaaat gattgccatc agaaagagac aacttgaaga aaccaacaat gactatgaaa



cagctgacgg cggctacatg actctgaacc ccagggcacc tactgacgat gataaaaaca



tctacctgac tcttcctccc aacgaccatg tcaacagtaa taactaaaga gtaacgttat



gccatgtggt catactctca gcttgctgag tggatgacaa aaagagggga attgttaaag



gaaaatttaa atggagactg gaaaaatcct gagcaaacaa aaccacctgg cccttagaaa



tagctttaac tttgcttaaa ctacaaacac aagcaaaact tcacggggtc atactacata



caagcataag caaaacttaa cttggatcat ttctggtaaa tgcttatgtt agaaataaga



caaccccagc caatcacaag cagcctacta acatataatt aggtgactag ggactttcta



agaagatacc tacccccaaa aaacaattat gtaattgaaa accaaccgat tgcctttatt



ttgcttccac attttcccaa taaatacttg cctgtgacat tttgccactg gaacactaaa



cttcatgaat tgcgcctcag atttttcctt taacatcttt tttttttttg acagagtctc



aatctgttac ccaggctgga gtgcagtggt gctatcttgg ctcactgcaa acccgcctcc



caggtttaag cgattctcat gcctcagcct cccagtagct gggattagag gcatgtgcca



tcatacccag ctaatttttg tattttttat tttttttttt tagtagagac agggtttcgc



aatgttggcc aggccgatct cgaacttctg gcctctagcg atctgcccgc ctcggcctcc



caaagtgctg ggatgaccag catcagcccc aatgtccagc ctctttaaca tcttctttcc



tatgccctct ctgtggatcc ctactgctgg tttctgcctt ctccatgctg agaacaaaat



cacctattca ctgcttatgc agtcggaagc tccagaagaa caaagagccc aattaccaga



accacattaa gtctccattg ttttgccttg ggatttgaga agagaattag agaggtgagg



atctggtatt tcctggacta aattcccctt ggggaagacg aagggatgct gcagttccaa



aagagaagga ctcttccaga gtcatctacc tgagtcccaa agctccctgt cctgaaagcc



acagacaata tggtcccaaa tgactgactg caccttctgt gcctcagccg ttcttgacat



caagaatctt ctgttccaca tccacacagc caatacaatt agtcaaacca ctgttattaa



cagatgtagc aacatgagaa acgcttatgt tacaggttac atgagagcaa tcatgtaagt



ctatatgact tcagaaatgt taaaatagac taacctctaa caacaaatta aaagtgattg



tttcaaggtg atgcaattat tgatgaccta ttttattttt ctataatgat catatattac



ctttgtaata aaacattata accaaaaca





SEQ ID NO: 26
MTMETQMSQN VCPRNLWLLQ PLTVLLLLAS ADSQAAPPKA


low affinity
VLKLEPPWIN VLQEDSVTLTCQGARSPESD SIQWFHNGNL


immunoglobulin
IPTHTQPSYR FKANNNDSGE YTCQTGQTSL SDPVHLTVLS


gamma Fc region
EWLVLQTPHL EFQEGETIML RCHSWKDKPL VKVTFFQNGK


receptor II-a
SQKFSHLDPT FSIPQANHSHSGDYHCTGNI GYTLFSSKPV


isoform 2
TITVQVPSMG SSSPMGIIVA VVIATAVAAI VAAVVALIYC


precursor [Homo
RKKRISANST DPVKAAQFEP PGRQMIAIRK RQLEETNNDY



sapiens]

ETADGGYMTL NPRAPTDDDKNIYLTLPPND HVNSNN


NP_067674.2






SEQ ID NO: 27
ctcttttcta agcttgtctc ttaaaaccca ctggacgttg gcacagtgct gggatgacta


low affinity
tggagaccca aatgtctcag aatgtatgtc ccagaaacct gtggctgctt caaccattga


immunoglobulin
cagttttgct gctgctggct tctgcagaca gtcaagctgc tcccccaaag gctgtgctga


gamma Fc region
aacttgagcc cccgtggatc aacgtgctcc aggaggactc tgtgactctg acatgccagg


receptor II-a
gggctcgcag ccctgagagc gactccattc agtggttcca caatgggaat ctcattccca


isoform 2
cccacacgca gcccagctac aggttcaagg ccaacaacaa tgacagcggg gagtacacgt


precursor
gccagactgg ccagaccagc ctcagcgacc ctgtgcatct gactgtgctt tccgaatggc


(FCGR2A) [Homo
tggtgctcca gacccctcac ctggagttcc aggagggaga aaccatcatg ctgaggtgcc



sapiens] mRNA

acagctggaa ggacaagcct ctggtcaagg tcacattctt ccagaatgga aaatcccaga


NM_021642.3
aattctccca tttggatccc accttctcca tcccacaagc aaaccacagt cacagtggtg



attaccactg cacaggaaac ataggctaca cgctgttctc atccaagcct gtgaccatca



ctgtccaagt gcccagcatg ggcagctctt caccaatggg gatcattgtg gctgtggtca



ttgcgactgc tgtagcagcc attgttgctg ctgtagtggc cttgatctac tgcaggaaaa



agcggatttc agccaattcc actgatcctg tgaaggctgc ccaatttgag ccacctggac



gtcaaatgat tgccatcaga aagagacaac ttgaagaaac caacaatgac tatgaaacag



ctgacggcgg ctacatgact ctgaacccca gggcacctac tgacgatgat aaaaacatct



acctgactct tcctcccaac gaccatgtca acagtaataa ctaaagagta acgttatgcc



atgtggtcat actctcagct tgctgagtgg atgacaaaaa gaggggaatt gttaaaggaa



aatttaaatg gagactggaa aaatcctgag caaacaaaac cacctggccc ttagaaatag



ctttaacttt gcttaaacta caaacacaag caaaacttca cggggtcata ctacatacaa



gcataagcaa aacttaactt ggatcatttc tggtaaatgc ttatgttaga aataagacaa



ccccagccaa tcacaagcag cctactaaca tataattagg tgactaggga ctttctaaga



agatacctac ccccaaaaaa caattatgta attgaaaacc aaccgattgc ctttattttg



cttccacatt ttcccaataa atacttgcct gtgacatttt gccactggaa cactaaactt



catgaattgc gcctcagatt tttcctttaa catctttttt ttttttgaca gagtctcaat



ctgttaccca ggctggagtg cagtggtgct atcttggctc actgcaaacc cgcctcccag



gtttaagcga ttctcatgcc tcagcctccc agtagctggg attagaggca tgtgccatca



tacccagcta atttttgtat tttttatttt ttttttttag tagagacagg gtttcgcaat



gttggccagg ccgatctcga acttctggcc tctagcgatc tgcccgcctc ggcctcccaa



agtgctggga tgaccagcat cagccccaat gtccagcctc tttaacatct tctttcctat



gccctctctg tggatcccta ctgctggttt ctgccttctc catgctgaga acaaaatcac



ctattcactg cttatgcagt cggaagctcc agaagaacaa agagcccaat taccagaacc



acattaagtc tccattgttt tgccttggga tttgagaaga gaattagaga ggtgaggatc



tggtatttcc tggactaaat tccccttggg gaagacgaag ggatgctgca gttccaaaag



agaaggactc ttccagagtc atctacctga gtcccaaagc tccctgtcct gaaagccaca



gacaatatgg tcccaaatga ctgactgcac cttctgtgcc tcagccgttc ttgacatcaa



gaatcttctg ttccacatcc acacagccaa tacaattagt caaaccactg ttattaacag



atgtagcaac atgagaaacg cttatgttac aggttacatg agagcaatca tgtaagtcta



tatgacttca gaaatgttaa aatagactaa cctctaacaa caaattaaaa gtgattgttt



caaggtgatg caattattga tgacctattt tatttttcta taatgatcat atattacctt



tgtaataaaa cattataacc aaaaca





SEQ ID NO: 28
MWFLTTLLLW VPVDGQVDTT KAVITLQPPW VSVFQEETVT


high affinity
LHCEVLHLPG SSSTQWFLNGTATQTSTPSY RITSASVNDS


immunoglobulin
GEYRCQRGLS GRSDPIQLEI HRGWLLLQVS SRVFTEGEPL


gamma Fc
ALRCHAWKDK LVYNVLYYRN GKAFKFFHWN SNLTILKTNI


receptor I
SHNGTYHCSG MGKHRYTSAGISVTVKELFP APVLNASVTS


precursor [Homo
PLLEGNLVTL SCETKLLLQR PGLQLYFSFY MGSKTLRGRN



sapiens]

TSSEYQILTA RREDSGLYWC EAATEDGNVL KRSPELELQV


NP_000557.1
LGLQLPTPVW FHVLFYLAVGIMFLVNTVLW VTIRKELKRK



KKWDLEISLD SGHEKKVISS LQEDRHLEEE LKCQEQKEEQ



LQEGVHRKEP QGAT





SEQ ID NO: 29
aatatcttgc atgttacaga tttcactgct cccaccagct tggagacaac atgtggttct


high affinity
tgacaactct gctcctttgg gttccagttg atgggcaagt ggacaccaca aaggcagtga


immunoglobulin
tcactttgca gcctccatgg gtcagcgtgt tccaagagga aaccgtaacc ttgcactgtg


gamma Fc
aggtgctcca tctgcctggg agcagctcta cacagtggtt tctcaatggc acagccactc


receptor I
agacctcgac ccccagctac agaatcacct ctgccagtgt caatgacagt ggtgaataca


precursor
ggtgccagag aggtctctca gggcgaagtg accccataca gctggaaatc cacagaggct


(FCGR1A) [Homo
ggctactact gcaggtctcc agcagagtct tcacggaagg agaacctctg gccttgaggt



sapiens] mRNA

gtcatgcgtg gaaggataag ctggtgtaca atgtgcttta ctatcgaaat ggcaaagcct


NM_000566.3
ttaagttttt ccactggaat tctaacctca ccattctgaa aaccaacata agtcacaatg



gcacctacca ttgctcaggc atgggaaagc atcgctacac atcagcagga atatctgtca



ctgtgaaaga gctatttcca gctccagtgc tgaatgcatc tgtgacatcc ccactcctgg



aggggaatct ggtcaccctg agctgtgaaa caaagttgct cttgcagagg cctggtttgc



agctttactt ctccttctac atgggcagca agaccctgcg aggcaggaac acatcctctg



aataccaaat actaactgct agaagagaag actctgggtt atactggtgc gaggctgcca



cagaggatgg aaatgtcctt aagcgcagcc ctgagttgga gcttcaagtg cttggcctcc



agttaccaac tcctgtctgg tttcatgtcc ttttctatct ggcagtggga ataatgtttt



tagtgaacac tgttctctgg gtgacaatac gtaaagaact gaaaagaaag aaaaagtggg



atttagaaat ctctttggat tctggtcatg agaagaaggt aatttccagc cttcaagaag



acagacattt agaagaagag ctgaaatgtc aggaacaaaa agaagaacag ctgcaggaag



gggtgcaccg gaaggagccc cagggggcca cgtagcagcg gctcagtggg tggccatcga



tctggaccgt cccctgccca cttgctcccc gtgagcactg cgtacaaaca tccaaaagtt



caacaacacc agaactgtgt gtctcatggt atgtaactct taaagcaaat aaatgaactg



acttcaactg ggatacattt ggaaatgtgg tcatcaaaga tgacttgaaa tgaggcctac



tctaaagaat tcttgaaaaa cttacaagtc aagcctagcc tgataatcct attacatagt



ttgaaaaata gtattttatt tctcagaaca aggtaaaaag gtgagtgggt gcatatgtac



agaagattaa gacagagaaa cagacagaaa gagacacaca cacagccagg agtgggtaga



tttcagggag acaagaggga atagtataga caataaggaa ggaaatagta cttacaaatg



actcctaagg gactgtgaga ctgagagggc tcacgcctct gtgttcagga tacttagttc



atggcttttc tctttgactt tactaaaaga gaatgtctcc atacgcgttc taggcataca



agggggtaac tcatgatgag aaatggatgt gttattcttg ccctctcttt tgaggctctc



tcataacccc tctatttcta gagacaacaa aaatgctgcc agtcctaggc ccctgccctg



taggaaggca gaatgtaact gttctgtttg tttaacgatt aagtccaaat ctccaagtgc



ggcactgcaa agagacgctt caagtgggga gaagcggcga taccatagag tccagatctt



gcctccagag atttgcttta ccttcctgat tttctggtta ctaattagct tcaggatacg



ctgctctcat acttgggctg tagtttggag acaaaatatt ttcctgccac tgtgtaacat



agctgaggta aaaactgaac tatgtaaatg actctactaa aagtttaggg aaaaaaaaca



ggaggagtat gacacaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa



aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaa





SEQ ID NO: 30
MSKLRMVLLE DSGSADFRRH FVNLSPFTIT VVLLLSACFV


CD163 [Homo
TSSLGGTDKE LRLVDGENKCSGRVEVKVQE EWGTVCNNGW



sapiens]

SMEAVSVICN QLGCPTAIKA PGWANSSAGS GRIWMDHVSC


NP_004235.4
RGNESALWDC KHDGWGKHSN CTHQQDAGVT CSDGSNLEMR



LTRGGNMCSG RIEIKFQGRWGTVCDDNFNI DHASVICRQL



ECGSAVSFSG SSNFGEGSGP IWFDDLICNG NESALWNCKH



QGWGKHNCDH AEDAGVICSK GADLSLRLVD GVTECSGRLE



VRFQGEWGTI CDDGWDSYDAAVACKQLGCP TAVTAIGRVN



ASKGFGHIWL DSVSCQGHEP AIWQCKHHEW GKHYCNHNED



AGVTCSDGSD LELRLRGGGS RCAGTVEVEI QRLLGKVCDR



GWGLKEADVV CRQLGCGSALKTSYQVYSKI QATNTWLFLS



SCNGNETSLW DCKNWQWGGL TCDHYEEAKI TCSAHREPRL



VGGDIPCSGR VEVKHGDTWG SICDSDFSLE AASVLCRELQ



CGTVVSILGG AHFGEGNGQIWAEEFQCEGH ESHLSLCPVA



PRPEGTCSHS RDVGVVCSRY TEIRLVNGKT PCEGRVELKT



LGAWGSLCNS HWDIEDAHVL CQQLKCGVAL STPGGARFGK



GNGQIWRHMF HCTGTEQHMGDCPVTALGAS LCPSEQVASV



ICSGNQSQTL SSCNSSSLGP TRPTIPEESA VACIESGQLR



LVNGGGRCAG RVEIYHEGSW GTICDDSWDL SDAHVVCRQL



GCGEAINATG SAHFGEGTGPIWLDEMKCNG KESRIWQCHS



HGWGQQNCRH KEDAGVICSE FMSLRLTSEA SREACAGRLE



VFYNGAWGTV GKSSMSETTV GVVCRQLGCA DKGKINPASL



DKAMSIPMWV DNVQCPKGPDTLWQCPSSPW EKRLASPSEE



TWITCDNKIR LQEGPTSCSG RVEIWHGGSW GTVCDDSWDL



DDAQVVCQQL GCGPALKAFK EAEFGQGTGP IWLNEVKCKG



NESSLWDCPA RRWGHSECGHKEDAAVNCTD ISVQKTPQKA



TTGRSSRQSS FIAVGILGVV LLAIFVALFF LTKKRRQRQR



LAVSSRGENL VHQIQYREMN SCLNADDLDL MNSSENSHES



ADFSAAELIS VSKFLPISGMEKEAILSHTE KENGNL





SEQ ID NO: 31
atatgtagcc ttttcatttt catgaaagtg aagtgatttt tagaattctt agttgttttc



Homosapiens

tttagaagaa catttctagg gaataataca agaagattta ggaatcattg aagttataaa


CD163 mRNA,
tctttggaat gagcaaactc agaatggtgc tacttgaaga ctctggatct gctgacttca


complete cds
gaagacattt tgtcaacttg agtcccttca ccattactgt ggtcttactt ctcagtgcct


mRNA
gttttgtcac cagttctctt ggaggaacag acaaggagct gaggctagtg gatggtgaaa


NM_004244.5
acaagtgtag cgggagagtg gaagtgaaag tccaggagga gtggggaacg gtgtgtaata



atggctggag catggaagcg gtctctgtga tttgtaacca gctgggatgt ccaactgcta



tcaaagcccc tggatgggct aattccagtg caggttctgg acgcatttgg atggatcatg



tttcttgtcg tgggaatgag tcagctcttt gggattgcaa acatgatgga tggggaaagc



atagtaactg tactcaccaa caagatgctg gagtgacctg ctcagatgga tccaatttgg



aaatgaggct gacgcgtgga gggaatatgt gttctggaag aatagagatc aaattccaag



gacggtgggg aacagtgtgt gatgataact tcaacataga tcatgcatct gtcatttgta



gacaacttga atgtggaagt gctgtcagtt tctctggttc atctaatttt ggagaaggct



ctggaccaat ctggtttgat gatcttatat gcaacggaaa tgagtcagct ctctggaact



gcaaacatca aggatgggga aagcataact gtgatcatgc tgaggatgct ggagtgattt



gctcaaaggg agcagatctg agcctgagac tggtagatgg agtcactgaa tgttcaggaa



gattagaagt gagattccaa ggagaatggg ggacaatatg tgatgacggc tgggacagtt



acgatgctgc tgtggcatgc aagcaactgg gatgtccaac tgccgtcaca gccattggtc



gagttaacgc cagtaaggga tttggacaca tctggcttga cagcgtttct tgccagggac



atgaacctgc tatctggcaa tgtaaacacc atgaatgggg aaagcattat tgcaatcaca



atgaagatgc tggcgtgaca tgttctgatg gatcagatct ggagctaaga cttagaggtg



gaggcagccg ctgtgctggg acagttgagg tggagattca gagactgtta gggaaggtgt



gtgacagagg ctggggactg aaagaagctg atgtggtttg caggcagctg ggatgtggat



ctgcactcaa aacatcttat caagtgtact ccaaaatcca ggcaacaaac acatggctgt



ttctaagtag ctgtaacgga aatgaaactt ctctttggga ctgcaagaac tggcaatggg



gtggacttac ctgtgatcac tatgaagaag ccaaaattac ctgctcagcc cacagggaac



ccagactggt tggaggggac attccctgtt ctggacgtgt tgaagtgaag catggtgaca



cgtggggctc catctgtgat tcggacttct ctctggaagc tgccagcgtt ctatgcaggg



aattacagtg tggcacagtt gtctctatcc tggggggagc tcactttgga gagggaaatg



gacagatctg ggctgaagaa ttccagtgtg agggacatga gtcccatctt tcactctgcc



cagtagcacc ccgcccagaa ggaacttgta gccacagcag ggatgttgga gtagtctgct



caagatacac agaaattcgc ttggtgaatg gcaagacccc gtgtgagggc agagtggagc



tcaaaacgct tggtgcctgg ggatccctct gtaactctca ctgggacata gaagatgccc



atgttctttg ccagcagctt aaatgtggag ttgccctttc taccccagga ggagcacgtt



ttggaaaagg aaatggtcag atctggaggc atatgtttca ctgcactggg actgagcagc



acatgggaga ttgtcctgta actgctctag gtgcttcatt atgtccttca gagcaagtgg



cctctgtaat ctgctcagga aaccagtccc aaacactgtc ctcgtgcaat tcatcgtctt



tgggcccaac aaggcctacc attccagaag aaagtgctgt ggcctgcata gagagtggtc



aacttcgcct ggtaaatgga ggaggtcgct gtgctgggag agtagagatc tatcatgagg



gctcctgggg caccatctgt gatgacagct gggacctgag tgatgcccac gtggtttgca



gacagctggg ctgtggagag gccattaatg ccactggttc tgctcatttt ggggaaggaa



cagggcccat ctggctggat gagatgaaat gcaatggaaa agaatcccgc atttggcagt



gccattcaca cggctggggg cagcaaaatt gcaggcacaa ggaggatgcg ggagttatct



gctcagaatt catgtctctg agactgacca gtgaagccag cagagaggcc tgtgcagggc



gtctggaagt tttttacaat ggagcttggg gcactgttgg caagagtagc atgtctgaaa



ccactgtggg tgtggtgtgc aggcagctgg gctgtgcaga caaagggaaa atcaaccctg



catctttaga caaggccatg tccattccca tgtgggtgga caatgttcag tgtccaaaag



gacctgacac gctgtggcag tgcccatcat ctccatggga gaagagactg gccagcccct



cggaggagac ctggatcaca tgtgacaaca agataagact tcaggaagga cccacttcct



gttctggacg tgtggagatc tggcatggag gttcctgggg gacagtgtgt gatgactctt



gggacttgga cgatgctcag gtggtgtgtc aacaacttgg ctgtggtcca gctttgaaag



cattcaaaga agcagagttt ggtcagggga ctggaccgat atggctcaat gaagtgaagt



gcaaagggaa tgagtcttcc ttgtgggatt gtcctgccag acgctggggc catagtgagt



gtgggcacaa ggaagacgct gcagtgaatt gcacagatat ttcagtgcag aaaaccccac



aaaaagccac aacaggtcgc tcatcccgtc agtcatcctt tattgcagtc gggatccttg



gggttgttct gttggccatt ttcgtcgcat tattcttctt gactaaaaag cgaagacaga



gacagcggct tgcagtttcc tcaagaggag agaacttagt ccaccaaatt caataccggg



agatgaattc ttgcctgaat gcagatgatc tggacctaat gaattcctca gaaaattccc



atgagtcagc tgatttcagt gctgctgaac taatttctgt gtctaaattt cttcctattt



ctggaatgga aaaggaggcc attctgagcc acactgaaaa ggaaaatggg aatttataac



ccagtgagtt cagcctttaa gataccttga tgaagacctg gactattgaa tggagcagaa



attcacctct ctcactgact attacagttg catttttatg gagttcact tctcctagga



ttcctaagac tgctgctgaa tttataaaaa ttaagtttgt gaatgtgact acttagtggt



gtatatgaga ctttcaaggg aattaaataa ataaataaga atgttattga tttgagtttg



ctttaattac ttgtccttaa ttctattaat ttctaaatgg gcttcctaat tttttgtaga



gtttcctaga tgtattataa tgtgttttat ttgacagtgt ttcaatttgc atatacagta



ctgtatattt tttcttattt ggtttgaata attttcctat taccaaataa aaataaattt



atttttactt tagtttttct aagacaggaa aagttaatga tattgaaggg tctgtaaata



atatatggct aactttataa ggcatgactc acaacgattc tttaactgct ttttgttact



gtaattctgt tcactagaat aaaatgcaga gccacacctg gtgagggcac





SEQ ID NO: 32
MSKLRMVLLE DSGSADFRRH FVNLSPFTIT VVLLLSACFV


CD163
TSSLGGTDKE LRLVDGENKCSGRVEVKVQE EWGTVCNNGW


NCBI Reference
SMEAVSVICN QLGCPTAIKA PGWANSSAGS GRIWMDHVSC


Sequence:
RGNESALWDC KHDGWGKHSN CTHQQDAGVT CSDGSNLEMR


NP_981961.2:
LTRGGNMCSG RIEIKFQGRWGTVCDDNFNI DHASVICRQL



ECGSAVSFSG SSNFGEGSGP IWFDDLICNG NESALWNCKH



QGWGKHNCDH AEDAGVICSK GADLSLRLVD GVTECSGRLE



VRFQGEWGTI CDDGWDSYDAAVACKQLGCP TAVTAIGRVN



ASKGFGHIWL DSVSCQGHEP AIWQCKHHEW GKHYCNHNED



AGVTCSDGSD LELRLRGGGS RCAGTVEVEI QRLLGKVCDR



GWGLKEADVV CRQLGCGSALKTSYQVYSKI QATNTWLFLS



SCNGNETSLW DCKNWQWGGL TCDHYEEAKI TCSAHREPRL



VGGDIPCSGR VEVKHGDTWG SICDSDFSLE AASVLCRELQ



CGTVVSILGG AHFGEGNGQIWAEEFQCEGH ESHLSLCPVA



PRPEGTCSHS RDVGVVCSRY TEIRLVNGKT PCEGRVELKT



LGAWGSLCNS HWDIEDAHVL CQQLKCGVAL STPGGARFGK



GNGQIWRHMF HCTGTEQHMGDCPVTALGAS LCPSEQVASV



ICSGNQSQTL SSCNSSSLGP TRPTIPEESA VACIESGQLR



LVNGGGRCAG RVEIYHEGSW GTICDDSWDL SDAHVVCRQL



GCGEAINATG SAHFGEGTGPIWLDEMKCNG KESRIWQCHS



HGWGQQNCRH KEDAGVICSE FMSLRLTSEA SREACAGRLE



VFYNGAWGTV GKSSMSETTV GVVCRQLGCA DKGKINPASL



DKAMSIPMWV DNVQCPKGPDTLWQCPSSPW EKRLASPSEE



TWITCDNKIR LQEGPTSCSG RVEIWHGGSW GTVCDDSWDL



DDAQVVCQQL GCGPALKAFK EAEFGQGTGP IWLNEVKCKG



NESSLWDCPA RRWGHSECGHKEDAAVNCTD ISVQKTPQKA



TTGRSSRQSS FIAVGILGVV LLAIFVALFF LAVSSRGENL



VHQIQYREMN SCLNADDLDL MNSSGGHSEP H





SEQ ID NO: 33
atatgtagcc ttttcatttt catgaaagtg aagtgatttt tagaattctt agttgttttc


CD163
tttagaagaa catttctagg gaataataca agaagattta ggaatcattg aagttataaa


NCBI Reference
tctttggaat gagcaaactc agaatggtgc tacttgaaga ctctggatct gctgacttca


Sequence:
gaagacattt tgtcaacttg agtcccttca ccattactgt ggtcttactt ctcagtgcct


NM_203416.3:
gttttgtcac cagttctctt ggaggaacag acaaggagct gaggctagtg gatggtgaaa



acaagtgtag cgggagagtg gaagtgaaag tccaggagga gtggggaacg gtgtgtaata



atggctggag catggaagcg gtctctgtga tttgtaacca gctgggatgt ccaactgcta



tcaaagcccc tggatgggct aattccagtg caggttctgg acgcatttgg atggatcatg



tttcttgtcg tgggaatgag tcagctcttt gggattgcaa acatgatgga tggggaaagc



atagtaactg tactcaccaa caagatgctg gagtgacctg ctcagatgga tccaatttgg



aaatgaggct gacgcgtgga gggaatatgt gttctggaag aatagagatc aaattccaag



gacggtgggg aacagtgtgt gatgataact tcaacataga tcatgcatct gtcatttgta



gacaacttga atgtggaagt gctgtcagtt tctctggttc atctaatttt ggagaaggct



ctggaccaat ctggtttgat gatcttatat gcaacggaaa tgagtcagct ctctggaact



gcaaacatca aggatgggga aagcataact gtgatcatgc tgaggatgct ggagtgattt



gctcaaaggg agcagatctg agcctgagac tggtagatgg agtcactgaa tgttcaggaa



gattagaagt gagattccaa ggagaatggg ggacaatatg tgatgacggc tgggacagtt



acgatgctgc tgtggcatgc aagcaactgg gatgtccaac tgccgtcaca gccattggtc



gagttaacgc cagtaaggga tttggacaca tctggcttga cagcgtttct tgccagggac



atgaacctgc tatctggcaa tgtaaacacc atgaatgggg aaagcattat tgcaatcaca



atgaagatgc tggcgtgaca tgttctgatg gatcagatct ggagctaaga cttagaggtg



gaggcagccg ctgtgctggg acagttgagg tggagattca gagactgtta gggaaggtgt



gtgacagagg ctggggactg aaagaagctg atgtggtttg caggcagctg ggatgtggat



ctgcactcaa aacatcttat caagtgtact ccaaaatcca ggcaacaaac acatggctgt



ttctaagtag ctgtaacgga aatgaaactt ctctttggga ctgcaagaac tggcaatggg



gtggacttac ctgtgatcac tatgaagaag ccaaaattac ctgctcagcc cacagggaac



ccagactggt tggaggggac attccctgtt ctggacgtgt tgaagtgaag catggtgaca



cgtggggctc catctgtgat tcggacttct ctctggaagc tgccagcgtt ctatgcaggg



aattacagtg tggcacagtt gtctctatcc tggggggagc tcactttgga gagggaaatg



gacagatctg ggctgaagaa ttccagtgtg agggacatga gtcccatctt tcactctgcc



cagtagcacc ccgcccagaa ggaacttgta gccacagcag ggatgttgga gtagtctgct



caagatacac agaaattcgc ttggtgaatg gcaagacccc gtgtgagggc agagtggagc



tcaaaacgct tggtgcctgg ggatccctct gtaactctca ctgggacata gaagatgccc



atgttctttg ccagcagctt aaatgtggag ttgccctttc taccccagga ggagcacgtt



ttggaaaagg aaatggtcag atctggaggc atatgtttca ctgcactggg actgagcagc



acatgggaga ttgtcctgta actgctctag gtgcttcatt atgtccttca gagcaagtgg



cctctgtaat ctgctcagga aaccagtccc aaacactgtc ctcgtgcaat tcatcgtctt



tgggcccaac aaggcctacc attccagaag aaagtgctgt ggcctgcata gagagtggtc



aacttcgcct ggtaaatgga ggaggtcgct gtgctgggag agtagagatc tatcatgagg



gctcctgggg caccatctgt gatgacagct gggacctgag tgatgcccac gtggtttgca



gacagctggg ctgtggagag gccattaatg ccactggttc tgctcatttt ggggaaggaa



cagggcccat ctggctggat gagatgaaat gcaatggaaa agaatcccgc atttggcagt



gccattcaca cggctggggg cagcaaaatt gcaggcacaa ggaggatgcg ggagttatct



gctcagaatt catgtctctg agactgacca gtgaagccag cagagaggcc tgtgcagggc



gtctggaagt tttttacaat ggagcttggg gcactgttgg caagagtagc atgtctgaaa



ccactgtggg tgtggtgtgc aggcagctgg gctgtgcaga caaagggaaa atcaaccctg



catctttaga caaggccatg tccattccca tgtgggtgga caatgttcag tgtccaaaag



gacctgacac gctgtggcag tgcccatcat ctccatggga gaagagactg gccagcccct



cggaggagac ctggatcaca tgtgacaaca agataagact tcaggaagga cccacttcct



gttctggacg tgtggagatc tggcatggag gttcctgggg gacagtgtgt gatgactctt



gggacttgga cgatgctcag gtggtgtgtc aacaacttgg ctgtggtcca gctttgaaag



cattcaaaga agcagagttt ggtcagggga ctggaccgat atggctcaat gaagtgaagt



gcaaagggaa tgagtcttcc ttgtgggatt gtcctgccag acgctggggc catagtgagt



gtgggcacaa ggaagacgct gcagtgaatt gcacagatat ttcagtgcag aaaaccccac



aaaaagccac aacaggtcgc tcatcccgtc agtcatcctt tattgcagtc gggatccttg



gggttgttct gttggccatt ttcgtcgcat tattcttctt gactaaaaag cgaagacaga



gacagcggct tgcagtttcc tcaagaggag agaacttagt ccaccaaatt caataccggg



agatgaattc ttgcctgaat gcagatgatc tggacctaat gaattcctca ggaggccatt



ctgagccaca ctgaaaagga aaatgggaat ttataaccca gtgagttcag cctttaagat



accttgatga agacctggac tattgaatgg agcagaaatt cacctctctc actgactatt



acagttgcat ttttatggag ttcttcttct cctaggattc ctaagactgc tgctgaattt



ataaaaatta agtttgtgaa tgtgactact tagtggtgta tatgagactt tcaagggaat



taaataaata aataagaatg ttattgattt gagtttgctt taattacttg tccttaattc



tattaatttc taaatgggct tcctaatttt ttgtagagtt tcctagatgt attataatgt



gttttatttg acagtgtttc aatttgcata tacagtactg tatatttttt cttatttggt



ttgaataatt ttcctattac caaataaaaa taaatttatt tttactttag tttttctaag



acaggaaaag ttaatgatat tgaagggtct gtaaataata tatggctaac tttataaggc



atgactcaca acgattcttt aactgctttt tgttactgta attctgttca ctagaataaa



atgcagagcc acacctggtg agggcac





SEQ ID NO: 34
MERASCLLLL LLPLVHVSAT TPEPCELDDE DFRCVCNFSE


CD14
PQPDWSEAFQ CVSAVEVEIHAGGLNLEPFL KRVDADADPR


NCBI Reference
QYADTVKALR VRRLTVGAAQ VPAQLLVGAL RVLAYSRLKE


Sequence:
LTLEDLKITG TMPPLPLEAT GLALSSLRLR NVSWATGRSW


NP_000582.1
LAELQQWLKP GLKVLSIAQAHSPAFSCEQV RAFPALTSLD



LSDNPGLGER GLMAALCPHK FPAIQNLALR NTGMETPTGV



CAALAAAGVQ PHSLDLSHNS LRATVNPSAP RCMWSSALNS



LNLSFAGLEQ VPKGLPAKLRVLDLSCNRLN RAPQPDELPE



VDNLTLDGNP FLVPGTALPH EGSMNSGVVP ACARSTLSVG



VSGTLVLLQG ARGFA





SEQ ID NO: 35
cagagaaggc ttaggctccc gagtcaacag ggcattcacc gcctggggcg cctgagtcat


CD14
caggacactg ccaggagaca cagaacccta gatgccctgc agaatccttc ctgttacggt


NCBI Reference
ccccctccct gaaacatcct tcattgcaat atttccagga aaggaagggg gctggctcgg


Sequence:
aggaagagag gtggggaggt gatcagggtt cacagaggag ggaactgaat gacatcccag


NM_000591.3
gattacataa actgtcagag gcagccgaag agttcacaag tgtgaagcct ggaagccggc



gggtgccgct gtgtaggaaa gaagctaaag cacttccaga gcctgtccgg agctcagagg



ttcggaagac ttatcgacca tggagcgcgc gtcctgcttg ttgctgctgc tgctgccgct



ggtgcacgtc tctgcgacca cgccagaacc ttgtgagctg gacgatgaag atttccgctg



cgtctgcaac ttctccgaac ctcagcccga ctggtccgaa gccttccagt gtgtgtctgc



agtagaggtg gagatccatg ccggcggtct caacctagag ccgtttctaa agcgcgtcga



tgcggacgcc gacccgcggc agtatgctga cacggtcaag gctctccgcg tgcggcggct



cacagtggga gccgcacagg ttcctgctca gctactggta ggcgccctgc gtgtgctagc



gtactcccgc ctcaaggaac tgacgctcga ggacctaaag ataaccggca ccatgcctcc



gctgcctctg gaagccacag gacttgcact ttccagcttg cgcctacgca acgtgtcgtg



ggcgacaggg cgttcttggc tcgccgagct gcagcagtgg ctcaagccag gcctcaaggt



actgagcatt gcccaagcac actcgcctgc cttttcctgc gaacaggttc gcgccttccc



ggcccttacc agcctagacc tgtctgacaa tcctggactg ggcgaacgcg gactgatggc



ggctctctgt ccccacaagt tcccggccat ccagaatcta gcgctgcgca acacaggaat



ggagacgccc acaggcgtgt gcgccgcact ggcggcggca ggtgtgcagc cccacagcct



agacctcagc cacaactcgc tgcgcgccac cgtaaaccct agcgctccga gatgcatgtg



gtccagcgcc ctgaactccc tcaatctgtc gttcgctggg ctggaacagg tgcctaaagg



actgccagcc aagctcagag tgctcgatct cagctgcaac agactgaaca gggcgccgca



gcctgacgag ctgcccgagg tggataacct gacactggac gggaatccct tcctggtccc



tggaactgcc ctcccccacg agggctcaat gaactccggc gtggtcccag cctgtgcacg



ttcgaccctg tcggtggggg tgtcgggaac cctggtgctg ctccaagggg cccggggctt



tgcctaagat ccaagacaga ataatgaatg gactcaaact gccttggctt caggggagtc



ccgtcaggac gttgaggact tttcgaccaa ttcaaccctt tgccccacct ttattaaaat



cttaaacaac gggtcaaaaa aaaaaaaa





SEQ ID NO: 36
MERASCLLLL LLPLVHVSAT TPEPCELDDE DFRCVCNFSE


CD14
PQPDWSEAFQ CVSAVEVEIHAGGLNLEPFL KRVDADADPR


NCBI Reference
QYADTVKALR VRRLTVGAAQ VPAQLLVGAL RVLAYSRLKE


Sequence:
LTLEDLKITG TMPPLPLEAT GLALSSLRLR NVSWATGRSW


NP_001035110.1
LAELQQWLKP GLKVLSIAQAHSPAFSCEQV RAFPALTSLD



LSDNPGLGER GLMAALCPHK FPAIQNLALR NTGMETPTGV



CAALAAAGVQ PHSLDLSHNS LRATVNPSAP RCMWSSALNS



LNLSFAGLEQ VPKGLPAKLRVLDLSCNRLN RAPQPDELPE



VDNLTLDGNP FLVPGTALPH EGSMNSGVVP ACARSTLSVG



VSGTLVLLQG ARGFA





SEQ ID NO: 37
ttaaatatct gaggatattc agggacttgg atttggtggc aggagatcaa cataaaccaa


CD14
gacaaggaag aagtcaaaga aatgaatcaa gtagattctc tgggatataa gaggcagccg


NCBI Reference
aagagttcac aagtgtgaag cctggaagcc ggcgggtgcc gctgtgtagg aaagaagcta


Sequence:
aagcacttcc agagcctgtc cggagctcag aggttcggaa gacttatcga ccatggagcg


NM_001040021.2
cgcgtcctgc ttgttgctgc tgctgctgcc gctggtgcac gtctctgcga ccacgccaga



accttgtgag ctggacgatg aagatttccg ctgcgtctgc aacttctccg aacctcagcc



cgactggtcc gaagccttcc agtgtgtgtc tgcagtagag gtggagatcc atgccggcgg



tctcaaccta gagccgtttc taaagcgcgt cgatgcggac gccgacccgc ggcagtatgc



tgacacggtc aaggctctcc gcgtgcggcg gctcacagtg ggagccgcac aggttcctgc



tcagctactg gtaggcgccc tgcgtgtgct agcgtactcc cgcctcaagg aactgacgct



cgaggaccta aagataaccg gcaccatgcc tccgctgcct ctggaagcca caggacttgc



actttccagc ttgcgcctac gcaacgtgtc gtgggcgaca gggcgttctt ggctcgccga



gctgcagcag tggctcaagc caggcctcaa ggtactgagc attgcccaag cacactcgcc



tgccttttcc tgcgaacagg ttcgcgcctt cccggccctt accagcctag acctgtctga



caatcctgga ctgggcgaac gcggactgat ggcggctctc tgtccccaca agttcccggc



catccagaat ctagcgctgc gcaacacagg aatggagacg cccacaggcg tgtgcgccgc



actggcggcg gcaggtgtgc agccccacag cctagacctc agccacaact cgctgcgcgc



caccgtaaac cctagcgctc cgagatgcat gtggtccagc gccctgaact ccctcaatct



gtcgttcgct gggctggaac aggtgcctaa aggactgcca gccaagctca gagtgctcga



tctcagctgc aacagactga acagggcgcc gcagcctgac gagctgcccg aggtggataa



cctgacactg gacgggaatc ccttcctggt ccctggaact gccctccccc acgagggctc



aatgaactcc ggcgtggtcc cagcctgtgc acgttcgacc ctgtcggtgg gggtgtcggg



aaccctggtg ctgctccaag gggcccgggg ctttgcctaa gatccaagac agaataatga



atggactcaa actgccttgg cttcagggga gtcccgtcag gacgttgagg acttttcgac



caattcaacc ctttgcccca cctttattaa aatcttaaac aacgggtcaa aaaaaaaaaa



a





SEQ ID NO: 38
MERASCLLLL LLPLVHVSAT TPEPCELDDE DFRCVCNFSE


CD14
PQPDWSEAFQ CVSAVEVEIHAGGLNLEPFL KRVDADADPR


NCBI Reference
QYADTVKALR VRRLTVGAAQ VPAQLLVGAL RVLAYSRLKE


Sequence:
LTLEDLKITG TMPPLPLEAT GLALSSLRLR NVSWATGRSW


NP_001167575.1
LAELQQWLKP GLKVLSIAQAHSPAFSCEQV RAFPALTSLD



LSDNPGLGER GLMAALCPHK FPAIQNLALR NTGMETPTGV



CAALAAAGVQ PHSLDLSHNS LRATVNPSAP RCMWSSALNS



LNLSFAGLEQ VPKGLPAKLRVLDLSCNRLN RAPQPDELPE



VDNLTLDGNP FLVPGTALPH EGSMNSGVVP ACARSTLSVG



VSGTLVLLQG ARGFA





SEQ ID NO: 39
aattctaccc cccttggtgc caacagatga ggttcacaat ctcttccaca aaacatgcag


CD14
ttaaatatct gaggatattc agggacttgg atttggtggc aggagatcaa cataaaccaa


NCBI Reference
gacaaggaag aagtcaaaga aatgaatcaa gtagattctc tgggatataa ggaaaggaag


Sequence:
ggggctggct cggaggaaga gaggtgggga ggtgatcagg gttcacagag gagggaactg


NM_001174104.1
aatgacatcc caggattaca taaactgtca gaggcagccg aagagttcac aagtgtgaag



cctggaagcc ggcgggtgcc gctgtgtagg aaagaagcta aagcacttcc agagcctgtc



cggagctcag aggttcggaa gacttatcga ccatggagcg cgcgtcctgc ttgttgctgc



tgctgctgcc gctggtgcac gtctctgcga ccacgccaga accttgtgag ctggacgatg



aagatttccg ctgcgtctgc aacttctccg aacctcagcc cgactggtcc gaagccttcc



agtgtgtgtc tgcagtagag gtggagatcc atgccggcgg tctcaaccta gagccgtttc



taaagcgcgt cgatgcggac gccgacccgc ggcagtatgc tgacacggtc aaggctctcc



gcgtgcggcg gctcacagtg ggagccgcac aggttcctgc tcagctactg gtaggcgccc



tgcgtgtgct agcgtactcc cgcctcaagg aactgacgct cgaggaccta aagataaccg



gcaccatgcc tccgctgcct ctggaagcca caggacttgc actttccagc ttgcgcctac



gcaacgtgtc gtgggcgaca gggcgttctt ggctcgccga gctgcagcag tggctcaagc



caggcctcaa ggtactgagc attgcccaag cacactcgcc tgccttttcc tgcgaacagg



ttcgcgcctt cccggccctt accagcctag acctgtctga caatcctgga ctgggcgaac



gcggactgat ggcggctctc tgtccccaca agttcccggc catccagaat ctagcgctgc



gcaacacagg aatggagacg cccacaggcg tgtgcgccgc actggcggcg gcaggtgtgc



agccccacag cctagacctc agccacaact cgctgcgcgc caccgtaaac cctagcgctc



cgagatgcat gtggtccagc gccctgaact ccctcaatct gtcgttcgct gggctggaac



aggtgcctaa aggactgcca gccaagctca gagtgctcga tctcagctgc aacagactga



acagggcgcc gcagcctgac gagctgcccg aggtggataa cctgacactg gacgggaatc



ccttcctggt ccctggaact gccctccccc acgagggctc aatgaactcc ggcgtggtcc



cagcctgtgc acgttcgacc ctgtcggtgg gggtgtcggg aaccctggtg ctgctccaag



gggcccgggg ctttgcctaa gatccaagac agaataatga atggactcaa actgccttgg



cttcagggga gtcccgtcag gacgttgagg acttttcgac caattcaacc ctttgcccca



cctttattaa aatcttaaac aacgggtcaa aaaaaaaaaa a





SEQ ID NO: 40
MERASCLLLL LLPLVHVSAT TPEPCELDDE DFRCVCNFSE


CD14
PQPDWSEAFQ CVSAVEVEIHAGGLNLEPFL KRVDADADPR


NCBI Reference
QYADTVKALR VRRLTVGAAQ VPAQLLVGAL RVLAYSRLKE


Sequence:
LTLEDLKITG TMPPLPLEAT GLALSSLRLR NVSWATGRSW


NP_001167576.1
LAELQQWLKP GLKVLSIAQAHSPAFSCEQV RAFPALTSLD



LSDNPGLGER GLMAALCPHK FPAIQNLALR NTGMETPTGV



CAALAAAGVQ PHSLDLSHNS LRATVNPSAP RCMWSSALNS



LNLSFAGLEQ VPKGLPAKLRVLDLSCNRLN RAPQPDELPE



VDNLTLDGNP FLVPGTALPH EGSMNSGVVP ACARSTLSVG



VSGTLVLLQG ARGFA





SEQ ID NO: 41
aattctaccc cccttggtgc caacagatga ggttcacaat ctcttccaca aaacatgcag


CD14
ttaaatatct gaggatattc agggacttgg atttggtggc aggagatcaa cataaaccaa


NCBI Reference
gacaaggaag aagtcaaaga aatgaatcaa aggcagccga agagttcaca agtgtgaagc


Sequence:
ctggaagccg gcgggtgccg ctgtgtagga aagaagctaa agcacttcca gagcctgtcc


NM_001174105.1
ggagctcaga ggttcggaag acttatcgac catggagcgc gcgtcctgct tgttgctgct



gctgctgccg ctggtgcacg tctctgcgac cacgccagaa ccttgtgagc tggacgatga



agatttccgc tgcgtctgca acttctccga acctcagccc gactggtccg aagccttcca



gtgtgtgtct gcagtagagg tggagatcca tgccggcggt ctcaacctag agccgtttct



aaagcgcgtc gatgcggacg ccgacccgcg gcagtatgct gacacggtca aggctctccg



cgtgcggcgg ctcacagtgg gagccgcaca ggttcctgct cagctactgg taggcgccct



gcgtgtgcta gcgtactccc gcctcaagga actgacgctc gaggacctaa agataaccgg



caccatgcct ccgctgcctc tggaagccac aggacttgca ctttccagct tgcgcctacg



caacgtgtcg tgggcgacag ggcgttcttg gctcgccgag ctgcagcagt ggctcaagcc



aggcctcaag gtactgagca ttgcccaagc acactcgcct gccttttcct gcgaacaggt



tcgcgccttc ccggccctta ccagcctaga cctgtctgac aatcctggac tgggcgaacg



cggactgatg gcggctctct gtccccacaa gttcccggcc atccagaatc tagcgctgcg



caacacagga atggagacgc ccacaggcgt gtgcgccgca ctggcggcgg caggtgtgca



gccccacagc ctagacctca gccacaactc gctgcgcgcc accgtaaacc ctagcgctcc



gagatgcatg tggtccagcg ccctgaactc cctcaatctg tcgttcgctg ggctggaaca



ggtgcctaaa ggactgccag ccaagctcag agtgctcgat ctcagctgca acagactgaa



cagggcgccg cagcctgacg agctgcccga ggtggataac ctgacactgg acgggaatcc



cttcctggtc cctggaactg ccctccccca cgagggctca atgaactccg gcgtggtccc



agcctgtgca cgttcgaccc tgtcggtggg ggtgtcggga accctggtgc tgctccaagg



ggcccggggc tttgcctaag atccaagaca gaataatgaa tggactcaaa ctgccttggc



ttcaggggag tcccgtcagg acgttgagga cttttcgacc aattcaaccc tttgccccac



ctttattaaa atcttaaaca acgggtcaaa aaaaaaaaaa





SEQ ID NO: 42
MLCPWRTANL GLLLILTIFL VAEAEGAAQP NNSLMLQTSK


cell surface
ENHALASSSL CMDEKQITQNYSKVLAEVNT SWPVKMATNA


glycoprotein
VLCCPPIALR NLIIITWEII LRGQPSCTKA YKKETNETKE


CD200 receptor 1
TNCTDERITW VSRPDQNSDL QIRTVAITHD GYYRCIMVTP


isoform a
DGNFHRGYHL QVLVTPEVTLFQNRNRTAVC KAVAGKPAAH


precursor [Homo
ISWIPEGDCA TKQEYWSNGT VTVKSTCHWE VHNVSTVTCH



sapiens]

VSHLTGNKSL YIELLPVPGA KKSAKLYIPY IILTIIILTI VGFIWLLKVN


NCBI Reference
GCRKYKLNKTESTPVVEEDE MQPYASYTEK NNPLYDTTNK


Sequence:
VKASEALQSE VDTDLHTL


NP_620161.1






SEQ ID NO: 43
MLCPWRTANL GLLLILTIFL VAEAEGAAQP NNSLMLQTSK


cell surface
ENHALASSSL CMDEKQITQN YSKVLAEVNT SWPVKMATNA


glycoprotein
VLCCPPIALR NLIIITWEII LRGQPSCTKA YKKETNETKE


CD200 receptor 1
TNCTDERITW VSRPDQNSDL QIRTVAITHD GYYRCIMVTP


isoform b
DGNFHRGYHL QVLGKEHHILRYFTSPDL


precursor [Homo




sapiens]




NCBI Reference



Sequence:



NP_620385.1






SEQ ID NO: 44
MLCPWRTANL GLLLILTIFL VAASSSLCMD EKQITQNYSK


cell surface
VLAEVNTSWP VKMATNAVLC CPPIALRNLI IITWEIILRG


glycoprotein
QPSCTKAYKK ETNETKETNC TDERITWVSR PDQNSDLQIR


CD200 receptor 1
TVAITHDGYY RCIMVTPDGN FHRGYHLQVL GKEHHILRYF TSPDL


isoform c



precursor [Homo




sapiens]




NCBI Reference



Sequence:



NP_620386.1






SEQ ID NO: 45
MLCPWRTANL GLLLILTIFL VAASSSLCMD EKQITQNYSK


cell surface
VLAEVNTSWP VKMATNAVLCCPPIALRNLI IITWEIILRG


glycoprotein
QPSCTKAYKK ETNETKETNC TDERITWVSR PDQNSDLQIR


CD200 receptor 1
TVAITHDGYY RCIMVTPDGN FHRGYHLQVL VTPEVTLFQN


isoform d
RNRTAVCKAV AGKPAAHISWIPEGDCATKQ EYWSNGTVTV


precursor [Homo
KSTCHWEVHN VSTVTCHVSH LTGNKSLYIE LLPVPGAKKS



sapiens]

AKLYIPYIIL TIIILTIVGF IWLLKVNGCR KYKLNKTEST PVVE


NCBI Reference
EDEMQP YASYTEKNNPLYDTTNKVKA SEALQSEVDT DLHTL


Sequence:



NP_740750.1






SEQ ID NO: 46
MVTFSENHGV VIQPAYKDKI NITQLGLQNS TITFWNITLE DEGCYMC


CD200 isoform d
TFGFGKISGTACLTVYVQPI VSLHYKFSED HLNITCSATA RPAPMVFW


NCBI Reference
PRSGIENSTV TLSHPNGTTSVTSILHIKDP KNQVGKEVIC QVLHLGTVT


Sequence:
FKQTVNKGYW FSVPLLLSIV SLVILLVLISILLYWKRHRN QDREP


NP_001305757.1








Claims
  • 1. A method for treating a patient having cancer who has been determined to have positive expression of CD200 receptor (CD200R1) and one or more biomarkers in a biological sample from the patient, the method comprising administering to the patient a CD200 inhibitor in an amount and with a frequency sufficient to reduce the cancer burden in the patient, compared to the cancer burden in the patient prior to treatment with the CD200 inhibitor, wherein the one or more biomarkers are selected from the group consisting of Inducible T-cell COStimulator (ICOS), T Cell Immunoreceptor with Ig and ITIM Domains (TIGIT), Tumor Necrosis Factor Receptor Superfamily Member 9 (TNFRSP9), Hepatitis A Virus Cellular Receptor 2 (HAVCR2), Programmed Cell Death 1 (PDCD1), Fc Fragment Of IgG Receptor IIa (FCGR2A), Fc Fragment Of IgG Receptor Ia (FCGR1A), Cluster of Differentiation 163 (CD163), and Cluster of Differentiation 14 (CD14).
  • 2. The method of claim 1, wherein the CD200 inhibitor is selected from the group consisting of a small molecule, a polypeptide, a polypeptide analog, a peptidomimetic, and an aptamer.
  • 3. The method of claim 1, wherein the CD200 inhibitor is an antibody, or an antigen-binding fragment thereof.
  • 4. The method of claim 3, wherein the antibody, or antigen-binding fragment thereof, is selected from the group consisting of a humanized antibody, a recombinant antibody, a diabody, a chimerized or chimeric antibody, a monoclonal antibody, a deimmunized antibody, a fully human antibody, a single chain antibody, an Fv fragment, an Fd fragment, an Fab fragment, an Fab′ fragment, and an F(ab′)2 fragment.
  • 5. The method of claim 3, wherein the antibody comprises a heavy chain variable region CDR1 having the sequence set forth in SEQ ID NO: 7, a heavy chain variable region CDR2 having the sequence set forth in SEQ ID NO: 8, a heavy chain variable region CDR3 having the sequence set forth in SEQ ID NO: 9, a light chain variable region CDR1 having the sequence set forth in SEQ ID NO: 4, a light chain variable region CDR2 having the sequence set forth in SEQ ID NO: 5, and a light chain variable region CDR3 having the sequence set forth in SEQ ID NO: 6.
  • 6. The method of claim 3, wherein the antibody comprises a heavy chain variable region comprising SEQ ID NO: 13 and a light chain variable region comprising SEQ ID NO: 12.
  • 7. The method of claim 3, wherein the antibody is samalizumab or a variant of samalizumab.
  • 8. The method of claim 3, wherein the antibody or an antigen-binding fragment thereof, is administered at a dose of about 5 mg/kg to about 50 mg/kg.
  • 9. The method of claim 3, wherein the antibody is administered at a dose of about 1 mg/kg to about 20 mg/kg.
  • 10. The method of claim 3, wherein the antibody is administered at a dose of about 10 mg/kg, about 15 mg/kg, about 20 mg/kg, or about 40 mg/kg.
  • 11. The method of claim 1, wherein expression levels of CD200R1 and the one or more biomarkers are measured by quantitation of protein and/or RNA levels, using at least one of an immunoassay, immunochemistry assay, immunohistochemistry assay, nucleoprobe assay, in situ hybridization, fluorescent RNA probes, RT-PCR, microarray transcription assay, and RNA transcription assay.
  • 12. The method of claim 11, wherein the immunoassay is an enzyme-linked immunosorbent assay (ELISA) or a radioimmunoassay (MA).
  • 13. The method of claim 1, wherein the biological sample is at least one of tumor tissue, tumor cells, blood, and a blood fraction.
  • 14. The method of claim 1, wherein: (i) expression levels of CD200R1 and of one or more biomarkers are measured in two or more types of biological samples; or (ii) expression levels of CD200R1 and of one or more biomarkers are measured in one type of biological sample and levels of a second biomarker are measured in a second type of biological sample.
  • 15. The method of claim 1, wherein the positive expression of CD200R1 in the biological sample is equal to or greater than expression of CD200R1 in a normal biological sample of the same type.
  • 16. The method of claim 1, wherein the positive expression of the one more biomarkers in the biological sample is equal to or greater than expression of the one or more biomarkers in a normal biological sample of the same type.
  • 17. The method of claim 1, further comprising measuring CD200 expression in the biological sample and identifying patients with tumors having elevated expression of CD200, wherein the elevated expression of CD200 in the biological sample is greater than median expression levels of CD200 in normal tissue.
  • 18. The method of claim 1, wherein the patient has been determined to have elevated expression levels of CD200R1 and at least one biomarker selected from the group consisting of ICOS, TIGIT, TNFRSF9, HAVCR2, PDCD1, FCGR2A, FCGRIA, CD163, and CD14.
  • 19. The method of claim 1, wherein the patient is an adult and the cancer is selected from the group consisting of diffuse large B cell lymphoma (DLBL), lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), glioblastoma (GBM), low grade glioma (LGG), clear cell RCC (KIRC), chromophobe (KICH), papillary cell RCC (KIRP), melanoma (SKCM), ovarian cancer (OV), colon cancer (COAD), rectum cancer (READ), uterine endometrial cancer (UCEC).
  • 20. The method of claim 1, wherein the patient is a pediatric patient, and the cancer is selected from atypical teratoid rhabdoid tumor (AT/RT), ependymoma, osteosarcoma, rhabdomyosarcoma, Ewing sarcoma, pilocytic astrocytoma, neuroblastoma, and retinoblastoma.
  • 21. The method of claim 19, wherein the cancer is DLBL, LUAD, or GBM.
RELATED APPLICATIONS

This application is a 35 U.S.C. 371 national stage filing of International Application No. PCT/US2018/052792, filed on Sep. 26, 2018, which claims priority to U.S. Provisional Application No. 62/578,643, filed on Oct. 30, 2017, and U.S. Provisional Application No. 62/564,052, filed on Sep. 27, 2017. The entire contents of the aforementioned applications is incorporated herein by reference.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2018/052792 9/26/2018 WO
Publishing Document Publishing Date Country Kind
WO2019/067499 4/4/2019 WO A
US Referenced Citations (86)
Number Name Date Kind
3940475 Gross Feb 1976 A
4289747 Chu Sep 1981 A
4376110 David et al. Mar 1983 A
5223409 Ladner et al. Jun 1993 A
5225539 Winter Jul 1993 A
5403484 Ladner et al. Apr 1995 A
5427908 Dower et al. Jun 1995 A
5434131 Linsley et al. Jul 1995 A
5508717 Miller Apr 1996 A
5530101 Queen et al. Jun 1996 A
5571698 Ladner et al. Nov 1996 A
5580717 Dower et al. Dec 1996 A
5780279 Matthews et al. Jul 1998 A
5859205 Adair et al. Jan 1999 A
5902583 Buchsbaum et al. May 1999 A
5916560 Larsen et al. Jun 1999 A
6011138 Reff et al. Jan 2000 A
6040136 Garrard et al. Mar 2000 A
6632927 Adair et al. Oct 2003 B2
6652858 Gorczynski et al. Nov 2003 B2
6709832 Von Knebel Doeberitz et al. Mar 2004 B1
6737056 Presta May 2004 B1
6749854 Gorczynski et al. Jun 2004 B2
6955811 Gorczynski et al. Oct 2005 B2
6984625 Gorczynski Jan 2006 B2
7238352 Gorczynski et al. Jul 2007 B2
7368535 Gorczynski et al. May 2008 B2
7408041 Bowdish et al. Aug 2008 B2
7427665 Bowdish et al. Sep 2008 B2
7435412 Bowdish et al. Oct 2008 B2
7452536 Gorczynski et al. Nov 2008 B2
7598353 Bowdish et al. Oct 2009 B2
7714110 Bowdish et al. May 2010 B2
7887798 Gorczynski et al. Feb 2011 B2
7915000 Bowdish et al. Mar 2011 B2
8075884 Bowdish et al. Dec 2011 B2
8114403 Bowdish et al. Feb 2012 B2
8187877 Bowdish et al. May 2012 B2
8252285 Rother et al. Aug 2012 B2
8637014 Rother et al. Jan 2014 B2
8709415 Bowdish et al. Apr 2014 B2
8840885 Bowdish et al. Sep 2014 B2
8999328 Bowdish et al. Apr 2015 B2
9000133 Bowdish et al. Apr 2015 B2
9085623 Rother et al. Jul 2015 B2
9150661 Bowdish et al. Oct 2015 B2
9180186 Faas McKnight et al. Nov 2015 B2
9249229 Bowdish et al. Feb 2016 B2
9447187 Wang et al. Sep 2016 B2
RE46323 Rother et al. Feb 2017 E
9862767 Rother et al. Jan 2018 B2
20020031515 Caligiuri et al. Mar 2002 A1
20020168364 Gorczynski et al. Nov 2002 A1
20020192215 Hoek et al. Dec 2002 A1
20030017491 Shi et al. Jan 2003 A1
20040018972 Gorczynski et al. Jan 2004 A1
20040054145 Gorczynski Mar 2004 A1
20040175692 Bowdish et al. Sep 2004 A1
20040198661 Bowdish et al. Oct 2004 A1
20050074452 Bowdish et al. Apr 2005 A1
20050100957 Bowdish et al. May 2005 A1
20050107314 Gorczynski et al. May 2005 A1
20050129690 Bowdish et al. Jun 2005 A1
20050169870 Truitt et al. Aug 2005 A1
20060024231 Schnitzer et al. Feb 2006 A1
20070036786 Tuaillon et al. Feb 2007 A1
20070065438 Liversidge et al. Mar 2007 A1
20080131428 Young et al. Jun 2008 A1
20090017046 Bowdish et al. Jan 2009 A1
20090022745 Bowdish et al. Jan 2009 A1
20100239598 Bowdish et al. Sep 2010 A1
20100249382 Desjarlais et al. Sep 2010 A1
20100291085 Rother et al. Nov 2010 A1
20110129471 Bowdish et al. Jun 2011 A1
20110135633 Bowdish et al. Jun 2011 A1
20110236387 Bowdish et al. Sep 2011 A1
20120148579 Bowdish et al. Jun 2012 A1
20120321625 Rother et al. Dec 2012 A1
20130158236 Bowdish et al. Jun 2013 A1
20130172534 Bowdish et al. Jul 2013 A1
20130189258 Rother et al. Jul 2013 A1
20130202602 Faas McKnight et al. Aug 2013 A1
20140170143 Wang et al. Jun 2014 A1
20150368341 Bowdish et al. Dec 2015 A1
20160009803 Rother et al. Jan 2016 A1
20160033514 McKnight et al. Feb 2016 A1
Foreign Referenced Citations (39)
Number Date Country
2178561 Apr 2010 EP
2523976 Nov 2012 EP
2534178 Dec 2012 EP
2670487 Dec 2013 EP
8403508 Sep 1984 WO
8503508 Aug 1985 WO
8806630 Sep 1988 WO
9215679 Sep 1992 WO
9428027 Dec 1994 WO
9518825 Jul 1995 WO
9627011 Sep 1996 WO
9638557 Dec 1996 WO
9708320 Mar 1997 WO
9721450 Jun 1997 WO
9827230 Jun 1998 WO
9924565 May 1999 WO
0187336 Nov 2001 WO
0211762 Feb 2002 WO
0242332 May 2002 WO
0246227 Jun 2002 WO
02059280 Aug 2002 WO
02095030 Nov 2002 WO
03025202 Mar 2003 WO
2003074679 Sep 2003 WO
2004060295 Jul 2004 WO
2004078937 Sep 2004 WO
2004078938 Sep 2004 WO
2005007809 Jan 2005 WO
2006053301 May 2006 WO
2007084321 Jul 2007 WO
2008089022 Jul 2008 WO
2009014745 Jan 2009 WO
2011085343 Jul 2011 WO
2011100538 Aug 2011 WO
2012106634 Aug 2012 WO
2018075408 Apr 2018 WO
2018102594 Jun 2018 WO
2019126133 Jun 2019 WO
2019126536 Jun 2019 WO
Non-Patent Literature Citations (211)
Entry
Sun et al, Immunology Letter, Aug. 2016, vol. 178, pp. 105-113.
Kretz-Rommel et al, The Journal of Immunology, 2007, vol. 178, pp. 5595-5605.
Wong et al, Journal of Leukocyte Biology 2010, vol. 88, pp. 361-372.
Siva et al, (Cancer Immunology, Immunotherapy, 2008, vol. 57, pp. 987-996.
Mahadevan et al, Blood, 2010, vol. 21, pp. 2465.
Kotwica-Mojzych, (International Journal of Molecular, 2021, vol. 22, 1-21).
Xiong et al, (Clin Cancer Res; 2020; vol. 26:232).
Erin et al, (Oncotarget, 2018, vol. 9, No. 27; pp. 19147-19158).
Ebert et al., “Selective Immunosuppressive Action of a Factor Produced by Colon Cancer Cells,” Cancer Res. 60:6158-6161 (1990).
Elgert, K. D. “Immunology: Understanding the Immune System,” The Genetic Basis of Antibody Diversity, 123 (1996).
Ennishi, D. et al., “CD5 expression is potentially predictive of poor outcome among biomarkers in patients with diffuse large B-cell lymphoma receiving rituximab plus CHOP therapy,” Annals of Oncology, vol. 19:1921-1926 (2008).
Faguet et al., “Blood Kinectics, Tissue Distribution, and Radioimaging of Anti-Common Chronic Lymphatic Leukemia Antigen (cCLLa) Monoclonal Antibody CLL.sub.2 in Mice Transplanted With cCLLa-Bearing Human Leukemia Cells.” Blood, vol. 75, No. 9(1990) pp. 1853-1861.
Faisal et al., “Cell-surface Associated p43/Endothelial-monocyte-activating-polypeptide-II in Hepatocellular Carcinoma Cells Induces Apoptosis in T-lymphocytes,” Asian Journal of Surgery, 30(1):13-22 (2007).
Fallarino, F., et al., “Murine Plasmacytoid Dendritic Cells Initiate the Immunosupressive Pathway of Tryptophan Catabolism in Response to CD200 Receptor Engagement,” J. Immunol., 173:3748-3754 (2004).
Farber, U., et al., “Loss of heterozygosity on chromosome 3, bands q24→qter, in a diploid meningioma,” Cytogenet Cell Genet, 57:157-158 (1991).
Feurstein et al., “Induction of Autoimmunity in a Transgenic Model of B Cell Receptor Peripheral Tolerance: Changes in Coreceptors and B Cell Receptor-Induced Tyrosine-Phosphoproteins”, J. Immunol., vol. 163:5287-5297 (1999).
Friedberg, Jonathan W., “Unique Toxicities and Resistance Mechanisms Associated with Monoclonal Antibody Therapy,” Hematology, pp. 329-334 (2005).
Funakoshi et al., “Antitumor Effects of Nonconjugated Murine Lym-2 and Human-Mouse Chimeric CLL-1 Monoclonal Antibodies Against Various Human Lumphoma Cell Lines In Vitro and In Vivo.” Blood vol. 90, No. 8 (1997) pp. 3160-3166.
Ginaldi et al., “Levels of expression of CD52 in normal and leukemic B and T cells: correlation with in vivo therapeutic responses to Campath-1H,” Leukemia Res. 22(2):185-191 (1998).
Giusti et al., “Somatic diversification of S107 from an antiphosphocholine to an anti-DNA autoantibody is due to a single base change in its heavy chain variable region,” PNAS, 84(9):2926-2930 (1987).
Gorczynski and Marsden, “Modulation of CD200 receptors as a novel method of immunosuppression,” Expert Opin. Ther. Patents, 13(5):711-715(2003).
Gorczynski et al., “Anti-CD200R Ameliorates Collagen-Induced Arthritis in Mice,” Clinical Immunol., 104(3):256-264 (2002).
Gorczynski et al., “Does Successful Allopregnancy Mimic Transplantation Tolerance?”, Graft, 4(5):338-345(2001).
Gorczynski et al., “Evidence of a role for CD200 in regulation of immune rejection of leukaemic tumour cells in C57BL/6 mice,” Clin. Exp. Immunol., 126:220-229(2001).
Gorczynski, “CD200 and its receptors as targets for immunoregulation,” Current Opinion in Investigational Drugs, 6:483-488(2005).
Gorczynski, L., et al., “Evidence That an OX-2-Positive Cell Can Inhibit the Stimulation of Type 1 Cytokine Production by Bone Marrow-Derived B7-1 (and B7-2)-Positive Dendtritic Cells,” J. Immunol, 162:774-781 (1999).
Gorczynski, R., et al., “CD200 Is a Ligand for All Members of the CD200R Family of Immunoregulatory Molecules,” J. Immunol., 172:7744-7749 (2004).
Gorczynski, R., et al., “Dendritic Cells Expressing TGFBeta/IL-10, and CHO Cells With OX-2, Increase Graft Survival,” Transplantation Proceedings, 33:1565-1566 (2001).
Gorczynski, R.M., “Evidence for an Immunoregulatory Role of OX2 with its Counter Ligand (OX2L) in the Regulation of Transplant Rejection, Fetal Loss, Autoimmunity and Tumor Growth”, Archivum Immunologiae et Therapiae Experimentalis, Polish Academyof Sciences, vol. 49(4), pp. 303-309 (2001).
Gorczynski, R.M., “Role of Cytokines in Allograft Rejection,” Current Pharmaceutical Design, 7:1039-1057 (2001).
Gorczynski, R.M., “Synergy in Induction of Increased Renal Allograft Survival after Portal Vein Infusion of Dendtritic Cells Transduced to Express TGFbeta and IL-10, along with Administration of CHO Cells Expressing the Regulatory Molecule OX-2,”Clinical Immunology, 95(3):182-189 (2000).
Gorczynski, R.M., “Transplant tolerance modifying antibody to CD200 receptor, but not CD200, alters cytokine production profile from stimulated macrophages,” Eur. J. Immunol., 31:2331-2337 (2001).
Gorczynski, R.M., et al., “A CD200FC Immunoadhesin Prolongs Rat Islet Xenograft Survival in Mice,” Transplantation, 73(12):1948-1953 (2002).
Gorczynski, R.M., et al., “An Immunoadhesin Incorporating the Molecule OX-2 Is a Potent Immunosuppressant That Prolongs Allo- and Xenograft Survival,” J. Immunol., 163:1654-1660(1999).
Gorczynski, R.M., et al., “Anti-Rat OX-2 Blocks Increased Small Intestinal Transplant Survival After Portal Vein Immunization,” Transplantation Proceedings, 31:577-578 (1999).
Gorczynski, R.M., et al., “Augmented Induction of CD4+ CD25+ Treg using Monoclonal Antibodies to CD200R,” Transplantation, 79(4):488-491 (2005).
Gorczynski, R.M., et al., “Augmented Induction of CD4+ CD25+ Treg using Monoclonal Antibodies to CD200R,” Transplantation, 79(9), pp. 1180-1183 (2005).
Gorczynski, R.M., et al., “CD200 Immunoadhesin Supresses Collagen-Induced Arthritis in Mice,” Clinical Immunology, 101(3):328-334 (2001).
Gorczynski, R.M., et al., “Evidence for Persistent Expression of OX2 as a Necessary Component of Prolonged Renal Allograft Survival Following Portal Vein Immunization,” Clinical Immunol., 97(1):69-78 (2000).
Gorczynski, R.M., et al., “Induction of Tolerance-Inducing Antigen-Presenting Cells in Bone Marrow Cultures in Vitor Using Monoclonal Antibodies to CD200R,” Transplantation, 77(8):1138-1144 (2004).
Gorczynski, R.M., et al., “Interleukin-13, in Combination with Anti-Interleukin-12, Increases Graft Prolongation After Portal Venous Immunization with Cultured Allogeneic Bone Marrow-Derived Dentritic Cells,” Transplantation, 62(11):1592-1600(1996).
Gorczynski, R.M., et al., “Persistent expression of OX-2 is necessary for renal allograft survival,” FASEB Journal, 14(6):A1069 (2000).
Gorczynski, R.M., et al., “Receptor Engagement on Cells Expressing a Ligand for the Tolerance-Inducing Molecule OX2 Induces an Immunoregulatory Population That Inhibits Alloreactivity in Vitro and in Vivo,” J. Immunol., 165:4854-4860 (2000).
Gorczynski, R.M., et al., “Regulation of Gene Expression of Murine MD-1 Regulates Subsequent T Cell Activation and Cytokine Production,” J. of Immunology, 165:1925-1932 (2000).
Gorczynski, R.M., et al., “Structural and Functional Heterogeneity in the CD200R Family of Immunoregulatory Molecules and their Expression at the Fetomatemal Interface,” AJRI, 52:147-163 (2004).
Gorczynski, R.M., et al., “The Same Immunoregulatory Molecules Contribute to Successful Pregnancy and Transplantation,” AJRI, 48:18-26 (2002).
Greenwood, J.D. and Clark, M., “Protein Engineering of Antibody Molecules for Prophylactic and Therapeutic Applications in Man,” (ed. Clark, M.) Pub. Academic Titles, UK, pp. 4-5 (1993).
Gura, T. “Systems for Identifying New Drugs are Often Faulty”, Science, 278: 1041-1042 (1997).
Gussow and Seemann, “Humanization of Monoclonal Antibodies,” Meth. Enzymol. 203:99-121 (1991).
Hardy et al., “A lymphocyte-activating monoclonal antibody induces regression of human tumors in severe combined immunodeficient mice,” Proc. Natl. Acad. Sci. USA 94:5756-5760 (1997).
Hart, P.H., “Modulation of Monocyte Effector Functions by Lipopolysacc-haride and Interferon-Y,” Dept. of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville, Vic., 3050 (1987) (Abstract).
Hatherley, Deborah et al., “The CD200 and CD200 receptor cell surface proteins interact through their N-terminal immunoglobulin-like domains,” Eur. J. Immunol., vol. 34:1688-1694 (2004).
Heaney et al., “Severe asthma treatment: need for characterising patients,” Lancet, 365:974-976(2005).
Hegen et al., “Utility of animal models for identification of potential therapeutics for rheumatoid arthritis,” Ann. Rheum. Dis., vol. 67, pp. 1505-1515 (2008).
Hernandez-Ilizaliturri, F.J. et al., “Strategies to overcoming rituximab-chemotherapy resistance by targeting the autophagy pathway using bortezomib in combination with the Bcl-2 inhibitor obatoclax in non-Hodgkin's lymphomas (NHL),” Journal ofClinical Oncology, 2009 ASCO Annual Meeting Proceedings, vol. 27(15S), 1 page, Poster No. 8543 (2009).
Hoek, R.M., et al., “Down-Regulation of the Macrophage Lineage Through Interaction with OX2 (CD200),” Science, 290:1768-1771 (2000).
Hoek, R.M., et al., “Macrophage regulation by the B7.1/2 homologue OX2?”, FASB Journal, 14(6):A1232 (2000).
Holodick, Nichol E. et al., “Adult BM generates CD5+ B1 cells containing abundant N-region additions,” Eur. J. Immunol., vol. 39(9):2383-2394 (2009).
Petermann, Kimberly B. et al., “CD200 is induced by ERK and is a potential therapeutic target in melanoma,” The Journal of Clinical Investigation, vol. 117(12):3922-3929 (2007).
Presta, L., “Antibody engineering for therapeutics,” Current Opinion in Structural Biology, 13(4):519-525 (2003).
Preston et al., “The leukocyte/neuron cell surface antigen OX2 binds to a ligand on macrophages”, European J. of Immunol., 27(8):1911-1918(1997).
Ragheb et al., “Preparation and functional properties of monoclonal antibodies to human, mouse and rat OX-2”, Immunol. Letters, 68(2,3):311-315(1999).
Ragheb, R.F., “Exploration of OX-2 function in tolerance induction and graft acceptance using an anti-mouse OX-2 monoclonal antibody,” University of Toronto, Masters Abstracts International, 38(4):971-972 (2000).
Reddy, N.M. et al., “Rituximab resistance and its association with changes in the internal domain of CD20 antigen and down-regulation of pro-apoptotic protein Bax and Bak in both rituximab-resistant cell lines (RRCL) and diffuse large B-celllymphoma (DLBCL) patient (pt) samples,” Journal of Clinical Oncology, 2006 ASCO Annual Meeting Proceedings, vol. 24(18S), 1 page, Poster No. 17509 (2006).
Richards, S.J., et al., “Reported Sequence Homology Between Alzheimer Amyloid770 and the MCR OX-2 Antigen Does Not Predict Function,” Brain Research Bulletin, 38(3):305-306 (1995).
Rijkers, Eva S.K. et al., “The inhibitory CD200R is differentially expressed on human and mouse T and B lymphocytes,” Molecular Immunology, vol. 45:1126-1135 (2008).
Riley, “Melanoma and the Problem Malignancy,” J. Exp. Med., 204:1-9 (2004).
Rindfleisch et al., “Diagnosis and Management of Rheumatoid Arthritis,” American Family Physician, vol. 72(6), pp. 1037-1047(2005).
Rioux, P., Campath-1H (Cambridge Univeristy), IDrugs, vol. 2(2); pp. 153-167, Database Medline, abstract No. NLM16160950 (Abstract Only) (1999).
Romagnani, Sergio., “Short Analytical Review: TH1 and TH2 in Human Diseases,” Clin. Immunol. Immunopath, 80 (3):225-235(1996).
Rosenblum, M.D., et al., “CD200 is a novel p53-target gene involved in apoptosis-associated immune tolerance,” Blood, 103(7):2691-2698 (2004).
Rosenwald et al., “Relation of Gene Expression Phenotype to Immunoglobulin Mutation Genotype in B Cell Chronic Lymphocytic Leukemia,” J. of Exp. Medicine, 194(11):1639-1647(2001).
Rudicoff et al., “Single amino acid substitution altering antigen-binding specificity,” Proc. Natl. Acad. Sci. USA 79:1979-1983 (1982).
Sahin et al., “New monoclonal antibody specific for a 6.5 kDa glycoprotein which presents mainly on a B cell of chronic lymphocytic leukemia (CLL)” Immunology Letters, 2001, 76, 1-6.
Schlom, Jeffrey :Monoclonal Antiboides They're More and Less Than You Think, Molecular and Cellular Research for Future Diagnosis and Therapy, 95-134, 1991.
Schultes et al., “Immunotherapy of Human Ovarian Carcinoma With Ovarex.TM. Mab-B43.13 in a Human-PBL-SCID/BG Mouse Model,” Hybridoma, 18(1):47-55 (1999).
Sebestyen et al., Syndecan-1 (CD138) expression in human non-Hodgkin lymphomas. British Journal of Hematology. vol. 104, 1999, p. 412-419.
Sehgal, et al., “Generation of the Primary Antibody Repertoire in Rabbits: Expression of a Diverse Set of Igk-V Genes May Compensate for Limited Combinatorial Diversity at the Heavy Chain Locus,” Immunogenetics, vol. 50, pp. 31-42 (1999). cited byapplicant.
Shields, R. L., et al., “High Resolution Mapping of the Binding Site on Human IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and Design of IgG1 Variants with Improved Binding to the Fc gamma R,” Journal of Biological Chemistry,276(9):6591-6604 (2001).
Simelyte et al., “CD200-Fc, A Novel Antiarthritic Biologic Agent That Targets Proinflammatory Cytokine Expression in the Joints of Mice With Collagen-Induced Arthritis,” Arthritis & Rheumatism, vol. 58(4), pp. 1038-1043 (2008).
Smith-Gill, Sandra J., “Biology of Antibody-Mediated Responses,” Biologic Therapy of Cancer: Principles and Practice, Chapter 2, pp. 39-51 (1995).
Snyder et al., “Enhanced Targeting and Killing of Tumor Cells Expressing the CXC Chemokine Receptor 4 by Transducible Anticancer Peptides,” Cancer Res 65(23):10646-10650 (2005).
Srivastava, P.K., “Immunotherapy of human cancer: lessons from mice,” Nature Immunology, 1(5):363-366 (2000).
Steinman, Lawrence., “Assessment of Animal Models for MS and Demyelinating Disease in the Design of Rational Therapy,” Neuron, 24:511-514(1999).
Stuart et al., “Monkeying Around with Collagen Autoimmunity and Arthritis,” Lab. Invest., 54(1):1-3(1986).
Syme, R., et al., “Comparison of CD34 and Monocyte-Derived Dendritic Cells from Mobilized Peripheral Blood from Cancer Patients,” Stem Cells, 23:74-81 (2005).
Tanaka et al., “The Anti-Human Tumor Effect and Generation of Human Cytotoxic T Cells in SCID Mice Given Human Peripheral Blood Lymphocytes by the in Vivo transfer of the Interleukin-6 Gene Using Adenovirus Vector,” Cancer Research, 57:1335-1343(1997).
Tang et al., Pathogenesis of collagen-induced arthritis: modulation of disease by arthritogenic T-Cell epitope location, J. of Immunology, 113: 384-391 (2004).
Tangri and Raghupathy, “Expression of Cytokines in'Placentas of Mice Undergoing Immunologically Mediated Spontaneous Fetal Resorptions,” Biology of Reprod., 49:850-856(1993).
Tao et al., “Structural Features of Human Immunoglobulin G that Determine Isotype-specific Differences in Complement Activation,” J. Exp. Med., vol. 178, pp. 661-667 (1993).
Taylor, N., et al., “Enhanced Tolerance to Autoimmune Uveitis in CD200-Deficient Mice Correlates with a Pronounced Th2 Switch in Response to Antigen Challenge,” J. Immunol., 174:143-154 (2005).
Tedder, Thomas F et al., “Isolation and structure of a cDNA encoding the B1 (CD20) cell-surface antigen of human B lymphocytes,” Proc. Natl. Acad. Sci. USA, vol. 85:208-212 (1988).
Teeling, Jessica L. et al., “The Biological Activity of Human CD20 Monoclonal Antibodies Is Linked to Unique Epitopes on CD20,” The Journal of Immunology, vol. 177:362-371 (2006).
Thomsen et al., “Reconstitution of a human immune system in immunodeficient mice: models of human alloreaction in vivo,” Tissue Antigens, 66:73-82 (2005).
Toder et al., “Mouse Model for the Treatment of Immune Pregnancy Loss,” Am. J. of Reprod. Immunol., 26:42-46 (1991).
Webb, M., et al., “Localisation of the MRC OX-2 Glycoprotein on the Surfaces of Neurones,” J. Neurochemistry, 43:1061-1067 (1984).
Wilczynski, J.R., “Immunoligical Analogy Between Allograft Rejection, Recurrent Abortion and Pre-Eclampsia—the Same Basic Mechanism?,” Human Ummunology, 67:492-511 (2006).
Wright et al., “The unusual distribution of the neuronal/lymphoid cell surface CD200 (OX2) glycoprotein is conserved in himans,” Immunology 102:173-179 (2001).
Wright, G.J., et al., “Lymphoid/Neuronal Cell Surface OX2 Glycoprotein Recognizes a Novel Receptor on Macrophages Implicated in the Control of Their Function,” Immunity, 13:233-242 (2000).
Wright, G.J., et al., “The lymphoid/neuronal OX-2 glycoprotein interacts with a novel protein expressed by macrophages,” Tissue Antigens, 55(Supplement 1): 11 (2000).
Wright, G.J., et al., “Viral homologues of cell surface proteins OX2 and CD47 have potential to regulate macrophage function,” Annual Congress of the British Society for Immunology, vol. 101 (Supplement 1): 50; Dec. 5-8, 2000.
Yamaguchi et al., “Application of tumor marker for immunotargeting therapy of cancer,” Nihon Rinsho, vol. 54(6), pp. 1674-1679 (1996).
Yamaguchi et al., “Biological Response Modifier and Missile Cancer Chemotherapy,” Biotherapy, vol. 10(4), pp. 605-609 (1996).
Yamaguchi et al., “Immunomissile Therapy Using Tumor Markers: Application of tumor marker for immunotargeting therapy of cancer,” Nihon Rinsho, vol. 54(6), pp. 1674-1679 (1996).
Yang, C., et al., “Functional maturation and recent thymic emigrants in the periphery: development of alloreactivity correlates with the cyclic expression of CD45RC isoforms,” Eur. J. Immunol., 22:2261-2269 (1992).
Yu, X., et al., “The role of B7-CD28 co-stimulation in tumor rejection,” International Immunology, 10(6):791-797 (1998).
Zhang, S., et al., “Molecular Mechanisms of CD200 Inhibition of Mast Cell Activation,” J. Immunol., 173:6786-6793 (2004).
Zheng, P., et al., “B7-CTLA4 interaction enhances both production of antitumor cytotoxic T lymphocytes and resistance to tumor challenge,” Proc. Natl. Acad. Sci. USA, 95:6284-6289 (1998).
Zhu et al., “Radioimmunotherapy of Human B-Cell Chronic Lymphocytic Leukemia in Nude Mice.” Cancer Research. 54, 5111-5117 (1994).
Zips, D., et al., “New Anticancer Agents: In Vitro and In Vivo Evaluation,” in vivo, 19:1-7 (2005).
Zou et al. Human Glioma-Induced Immunosuppression Involves Soluble Factor(s) That Alters Monocyte Cytokine Profile and Surface Markers. Apr. 15, 1999. vol 162, pp. 4882-4892.
Alizadeh et al., “Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling,” Nature, 403:503-511(2000).
Almasri, Nidal M. et al., “Reduced Expression of CD20 Antigen as a Characteristic Marker for Chronic Lymphocytic Leukemia,” American Journal of Hematology, vol. 40:259-263 (1992).
Bach, “Immunosuppressive therapy of autoimmune diseases,” Immunology Today, 14(6)322-326(1993).
Banerjee, D., et al., “Blocking CD200-CD200 receptor axis augments Nos. 2 expression and aggravates experimental autoimmune uveoretinitis in Lewis rats,” Ocular Immunology and Inflammation, 12(2):115-125 (2004).
Barclay and Ward, “Purification and Chemical Characterisation of Membrane Glycoproteins From Rat Thymocytes and Brain, Recognised by Monoclonal Antibody MRC OX2,” European J. Biochemistry, 129:447-458(1982).
Barclay et al.,“CD200 and membrane protein interactions in the control of myeloid cells,” Trends in Immunology, 23(6): 285-290 (2002).
Barclay, A.N., “Different reticular elements in rat lymphoid tissue identified by localization of Ia, Thy-1 and MRC OX 2 antigens,” Immunology, 44:727-736 (1981).
Barclay, A.N., et al., “Neuronal/Lymphoid Membrane Glycoprotein MRC OX-2 is a Member of the Immunoglobulin Superfamily with a Light-Chain-Like Structure,” Biochem. Soc. Symp., 51:149-157 (1985).
Bauvois et al., Constitutive expression of CD26/dipeptidylpepidase IV on peripheral blood B lymphocytes of patients with B chronic lymphocytic leukaemia. British Journal of Cancer 1999., vol. 79. p. 1042-1048.
Bello, Celeste et al., “Monoclonal Antibodies for B-Cell Lymphomas: Rituximab and Beyond,” Hematology, pp. 233-242 (2007).
Blazer, B.R., et al., “CD28/B7 Interactions Are Required for Sustaining the Graft-Versus_Leukemia Effect of Delayed Post-Bone Marrow Transplantation Splenocyte Infusion in Murine Recipients of Myeloid or Lymphoid Leukemia Cells,” J. Immunol., 159:3460-3473 (1997).
Bodey et al. “Human Cancer Detection and Immunotherapy with Conjugated and Non-Conjugated Monoclonal Antibodies” Anticancer Research 16: 661-674 (1996).
Bohen, S.P., “Variation in gene expression patterns in follicular lymphoma and the response to rituximab,” PNAS, 100(4):1926-1930(2003).
Boon, Thierry., “Toward a Genetic Analysis of Tumor Rejection Antigens,” Advances in Cancer Res., 58:177-210 (1992).
Borriello et al., “MRC OX-2 Defines a Novel T Cell Costimulatory Pathway,” J. Immunol., 158:4549-4554(1997).
Borriello, F., et al., “Characterization and localization of Mox2, the gene encoding the murine homolog of the rat MRC OX-2 membrane glycoprotein,” Mammalian Genome, 9(2):114-118 (1998).
Broderick et al., “Constitutive Retinal CD200 Expression Regulates Resident Microglia and Activin State of Inflammatory Cells During Experimental Autoimmune Uveoretinitis,” Am. J of Pathology, 161(5):1669-1677(2002).
Bruggemann et al., “A Matched Set of Rat/Mouse Chimeric Antibodies: Identification and Biological Properties of Rat H Chain Constant Regions .mu., .gamma.I, .gamma.2a, .gamma.2b, .gamma.2c, .epsilon., and .alpha.1,” The Journal of Immunology, vol. 142(9), pp. 3145-3150 (1989).
Bukovsky, A., et al., “Association of lymphoid cell markers with rat ascitic malignant cells,” IRCS Med. Sci., 11:866-867 (1983).
Bukovsky, A., et al., “Association of some cell surface antigens of lymphoid cells and cell surface differentiation antigens with early rat pregnancy,” Immunology, 52:631-640 (1984).
Bukovsky, A., et al., “lhe localization of Thy-1.1, MRC OX 2 and la antigens in the rat ovary andfollopian tube,” Immunology, 48:587-596 (1983).
Bukovsky, A., et al., “The ovarian follicle as a model for the cell-mediated control of tissue growth,” Cell Tissue Res., 236:717-724 (1984).
Burge, Daniel J. et al., “Pharmacokinetic and Pharmacodynamic Properties of TRU-015, a CD20-Directed Small Modular Immunopharmaceutical Protein Therapeutic, in Patients with Rheumatoid Arthritis: A Phase I, Open-Label, Dose-Escalation ClinicalStudy,” Clinical Therapeutics, vol. 30(10):1806-1816 (2008).
Caldas et al., “Humanization of the anti-CD18 antibody 6.7: an unexpected effect of a framework residue in binding to antigen,” Molecular Immunology, 39(15):941-952 (2003).
Chaouat and Clark, FAS/FAS Ligand Interaction at the Placental Interface is not Required for the Success of Allogeneic Pregnancy in Anti-Paternal MHC Preimmunized Mice, Presented at the 6th Congress of the Adria-Alps Soc. of Immunol. of Reprod.,(2000) / Amer. J. of Reprod. Immunol., 45:108-115(2001).
Chaudhary et al., “A recombinant immunotoxin consisting of two antibody variable domains fused to Pseudomonas exotoxin,” Nature, vol. 339, 394-397 (1989).
Chen, D., et al., “Discrete Monoclonal Antibodies Define Functionally Important Epitopes in the CD200 Molecule Responsible for Immunosupression Function,” Transplantation, 79:282-288 (2005).
Chen, D., et al., “Synthetic peptides from the N-terminal regions of CD200 and CD200R1 modulate immunosupressive and anti-inflammatory effects of CD200-CD200R1 interaction,” International Immunology, 17(3):289-296 (2005).
Chen, Z., et al., “Cloning and characterization of the murine homologue of the rat/human MRC OX-2 gene,” Database Medline, Biochemica et Biophysica Acta, 1362(1):6-10 (1997).
Cherwinski, H.M., et al., “The CD200 Receptor Is a Novel and Potent Regulator of Murine and Human Mast Cell Function,” J. Immunol., 174:1348-1356 (2005).
Chien et al., “Significant structural and functional change of an antigen-binding site by a distant amino acid substitution: proposal of a structural mechanism,” Proc. Natl. Acad. Sci. USA 86:5532-5536 (1989).
Chitnis et al., “The Role of CD200 in Immune-Modulation and Neural Protection in EAE,” Abstract 12th International Congress of Immunology and 4th Annual Conference of FOCIS, Montreal, Jul. 21, 2004. Abstract Only.
Chitnis, T., et al., “Elevated Neuronal Expression of CD200 Protects Wld.sup.s Mice from Inflammation-Mediated Neurodegeneration,” American Journal of Pathology, 170(5):1695-1712 (2007).
Clark et al., “Fg12 prothrombinase expression in mouse trophoblast and decidua triggers abortion but may be countered by OX-2,” Mol. Human Reprod., 7:185-194(2001).
Clark et al., “Labile CD200 tolerance signal important in transfusion-related immunomodulation (TRIM) prevention of recurrent miscarriages,” Amer. J. Reprod. Immunol., 45:361(2001). Abstract Only.
Clark et al., “Procoagulants in fetus rejection: the role of the OX-2 (CD200) tolerance signal,” Seminars in Immunol., 13 (4)255-263(2001).
Clark et al., “The OX-2 Tolerance Signal Molecule at the Fetomatemal Interface Determines Pregnancy Outcome,” Amer. Journal of Reprod Immunol., 43:326(2000). Abstract Only.
Clark et al., Amer. Soc. for Reprod. Medicine, 55th Annual Meeting (1999). Abstract Only.
Clark, D.A., “Intralipid as Treatment for Recurrent Unexplained Abortion?”, Am. J. of Reprod. Immunol., 32:290-293 (1994).
Clark, M.J., et al., “MRC OX-2 antigen: a lymphoid/neuronal membrane glycoprotein with a structure like a single immunoglobulin light chain,” EMBO Journal, 4(1): 113-118 (1985).
Clarke, M.J., “MRC OX-2 lymphoid brain glycoprotein: S1 mapping suggests higher levels of abnormal RNA in the thymus than in the brain,” Biochemical Society Transactions, 14:80-81 (1986).
Cochlovius et al., “Cure of Burkitt's Lymphome in Severe Combined Immunodeficiency Mice by T Cells, Tetravalent CD3 .times. CD19 Tandem Diabody, and CD28 Costimulation,” Cancer Res. 60:4336-4341 (2000).
Cohen, P.L., “Systemic Autoimmunity,” in Fundamental Immunology, Fourth edition, W.E. Paul, Editor, Lippincott-Raven Publishers, Philadelphia, Ch. 33, p. 1067-1088(1999).
Coles, S.J. et al., “The immunosuppressive ligands PD-L1 and CD200 are linked in AML T-cell immunosuppression: identification of a new immunotherapeutic synapse,” Leukemia, vol. 29(9): 1952-1954 (2015).
Cui, Weiguo et al., “CD200 and its receptor, CD200R, modulate bone mass via the differentiation of osteoclasts,” PNAS, vol. 104(36):14436-14441 (2007).
Davidson, A., et al. “Autoimmune Diseases,” (editors Mackay and Rosen) New England Journal of Medicine, 345 (5):340-350 (2001).
DeNardo et al., “Increased Survival Associated with Radiolabeled Lym-1 Therapy for Non-Hodgkin's Lymphoma and Chronic Lymphocyctic Leukemia.” Cancer Supplement (1997) vol. 80, No. 12. pp 2706-2711.
Dennis, C., “Off by a whisker,” Nature, 442,:739-741 (2006).
Dick et al., “Control of Myeloid Activity During Retinal Inflammation,” J. of Leukocyte Bio., 74:161-166(2003).
Dorai et al., “Aglycosylated Chimeric Mouse/Human IgG1 Antibody Retains Some Effector Function,” Hybridoma, vol. 10(2): 211-217 (1991).
Huang, “Structural chemistry and therapeutic intervention of protein-protein interactions in immune response, human immunodeficiency virus entry, and apoptosis,” Pharmacol. Therapeutics, 86:201-215(2000).
Hutchings, N.J., et al., “Interactions of Cytoplasmic Region of OX2R Are Consistent with an Inhibitory Function,” Annual Congress of the British Society for Immunology, 101 (Supplement 1): 24, Abstract #10.6 (2000).
International Preliminary Report on Patentability, PCT/US2018/052792, dated Mar. 31, 2020, 6 pages.
International Search Report and Written Opinion, PCT/US2018/052792, dated Dec. 7, 2018, 8 pages.
Iwanuma et al., “Antitumor Immune Response of Human Peripheral Blood Lymphocytes Coengrafted with Tumor into Severe Combined Immunodeficient Mice,” Cancer Research, 57:2937-2942(1997).
Jain, “The next frontier of molecular medicine: Delivery of therapeutics,” Nature Medicine, 4(6):655-657(1998).
Jansky, L., et al., “Dynamics of Cytokine Production in Human Peripheral Blood Mononuclear Cells Stimulated by LPS or Infected by Borrelia,” Physiol. Res., 52:593-598 (2003).
Jeurissen, S.H.M., et al., “Characteristics and functional aspects of nonlymphoid cells in rat germinal centers, recognized by two monoclonal antibodies ED5 and ED6,” Eur, J. Immunol., 16:562-568 (1986).
Kausar, Fariha et al., “Ocrelizumab: a step forward in the evolution of B-cell therapy,” Expert Opin. Biol. Ther., vol. 9 (7):889-895 (2009).
Keil et al., “The Tolerance-Promoting Molecule OX-2 is Expressed in Fetal Trophoblast Cells that Cocoon the ‘Fetal Allograft’ and May Prevent Pregnancy Loss Caused by Cytokine-Activation of FGL2 Prothrombinase,” Amer. J. Reprod. Immunol.,45:343(2001) (abstract).
Kim et al., “Divergent Effects of 4-1BB Antibodies on Antitumor Immunity and on Tumor-reactive T-Cell Generation,” Cancer Res., 61:2031-2037(2001).
Kjaergaard et al., “Therapeutic Efficacy of OX-40 Receptor antibody Depends on Tumor Immunogenicity and Anatomic Site of Tumor Growth,” Cancer Res. 60:5514-5521(2000).
Kneitz, C., et al., “Inhibition of Tcell/B cell interaction by B-CLL cells,” Leukemia, vol. 13, pp. 98-104 (1999).
Kretz-Rommel, “CD200 Expression on Tumor Cells Suppresses Antitumor Immunity: New Approaches to Cancer Immunotherapy,” J. Immunol. 178:5595-5605 (2007).
Kretz-Rommel, A., et al., “CD200 Expression on Tumor Cells Suppresses Anti-Tumor Immunity: New Approaches to Cancer Immunotherapy,” J. Immunother., 29(6):666 (2006).
Kretz-Rommel, A., et al., “Immune Evasion by CD200: New Approaches to Targeted Therapies for Chronic Lymphocytic Leukemia,” J. Immunother., 28(6):650 (2005).
Kretz-Rommel, A., et al., “The Immuno-Regulatory Protein CD200 Is Overexpressed in a Subset of B-Cell Chronic Lymphocytic Leukemias and Plays a Role in Down-Regulatin the TH1 Immune Response,” J. Immunother., 27(6):S46 (2004).
Kretz-Rommel, Anke et al., “Blockade of CD200 in the Presence or Absence of Antibody Effector Function: Implications for Anti-CD200 Therapy,” The Journal of Immunology, vol. 180:699-705 (2008).
Kroese, F.G.M., et al., “Germinal centre formation and follicular antigen trapping in the spleen of lethally X-irradiated and reconstituted rats,” Immunology, 57:99-104 (1986).
Kroese, F.G.M., et al., “The ontogeny of germinal centre forming capacity of neonatal rat spleen,” Immunology, 60:597-602 (1987).
Levene, Adam P. et al., “Therapeutic monoclonal antibodies in oncology,” J.R. Soc. Med., vol. 98:146-152 (2005).
Liu et al., “Effect of combined T- and B-cell depletion of allogenic HLA-mismatched bone marrow graft on the magnitude and kinetics of Epstein-Barr virus load in the peripheral blood of bone marrow transplant recipients,” Clin. Transplant.18:518-524 (2004).
Mariuzza et al., “The Structural Basis of Antigen-Antibody Recognition,” Ann. Rev. Biophys. Biophys. Chem. 16:139-159 (1987).
Marsh, M.N., “Functional and Structural Aspects of the Epithelial Lymphocyte, with Implications for Coeliac Disease and Tropical Sprue,” Scandinavian Journal of Gastroenterology 114: 55-75 (1985).
Marti, G.E. et al., “CD20 and CD5 Expression in B-Chronic Lymphocytic Leukemia,” Annals of the New York Academy of Sciences, vol. 651:480-483 (1992).
Matutes et al. Morphological and Immuniphenotypic Features of Chronic Lymphocytic Leukemia. Rev. Clin. Exp. Hematol. vol. 4.1, Mar. 2000 p. 22-47.
McCaughan et al., “Characterization of the Human Homolog of the Rat MRC OX-2 Membrane Glycoprotein,” Immunogenetics, 25:329-335(1987).
McCaughan, G.M., et al., “Identification of the human homologue of the rat lymphoid/brain antigen MRC OX-2,” Australian and New Zealand Journal of Medicine 17: 142 (Abstract) (1987).
McCaughan, G.W., et al., “The Gene for MRC OX-2 Membrane Glycoprotein Is Localized on Human Chromosome 3,” Immunogenetics, 25:133-135 (1987).
McMaster, W.R., et al., “Identification of la glycoproteins in rat thymus and purification from rat spleen,” Eur. J. Immunol., 9:426-433 (1979).
McWhirter et al., Supplemental Materials, “Antibodies selected from combinatorial libraries block a tumor antigen that plays a key role in immunomodulation,” PNAS, vol. 103, pp. 1041-1046 (2006).
McWhirter, J.R, et al., “Antibodies selected from combinatorial libraries block a tumor antigen that plays a key role in immunomodulation,” PNAS, 103(4): 1041-1046 (2006).
Milani, Cannon et al., “Veltuzumab, an anti-CD20 mAb for the treatment of non-Hodgkin's lymphoma, chronic lymphocytic leukemia and immune thrombocytopenic purpura,” Current Opinion in Molecular Therapeutics, vol. 11 (2):200-207 (2009). cited byapplicant.
Mjaaland et al., “Modulation of immune responses with monoclonal antibodies. I. Effects on regional lymph node morphology and on anti-hapten responses to haptenized monoclonal antibodies”, Eur. J. Immunol., 20:1457-1461 (1990).
Mjaaland, S., et al., “The Localization of Antigen in Lymph Node Follicles of Congenitally Athymic Nude Rats,” Scand. J. Immunol., 26:141-147 (1987).
Mohammad, R.M., et al., “Establishment of a human B-CLL xenograft model: utility as a preclinical therapeutic model,” Leukemia, 10:130-137 (1996).
Mori et al., “Establishment of a new anti-cancer drugs-resistant cell line derived from B-chronic lymphocyctic leukemia,” Proceedings, Fifty-Ninth Annual Meeting of the Japanese Cancer Association, p. 583, #3788 (Sep. 1, 2000).
Morris, R.J., et al., “Sequential Expression of OX2 and Thy-1 Glycoproteins on the Neuronal Surface during Development,” Dev. Neurosci., 9:33-44 (1987).
Morschhauser, Franck et al., “Humanized Anti-CD20 Antibody, Veltuzumab, in Refractory/Recurrent Non-Hodgkin's Lymphoma: Phase I/II Results,” Journal of Clinical Oncology, vol. 27(20):3346-3353 (2009).
Mueller et al., “Aglycosylated chimeric mouse/human IgG1 antibody retains some effector function,” Hybridoma, vol. 10 (2), pp. 211-217 (1991).
Mueller et al., “Humanized Porcine VCAM-Specific Monoclonal Antibodies With Chimeric** IgG2/G4 Constant Regions Block Human Leukocyte Binding to Porcine Endothelial Cells,” Molecular Immunology, vol. 34(6), pp. 441-452 (1997).
Myers et al., “Characterization of a Peptide Analog of a Determinant of Type II Collagen that Suppresses Collagen-Induced Arthritis,” J. of Immunology, 3589-3595(1998).
Nagelkerken L., et al., “Accessory Cell Function of Thoracic Duct Nonlymphoid Cells, Dentritic Cells, and Splenic Adherent Cells in the Brown-Norway Rat,” Cellular Immunology, 93:520-531 (1985).
Nathan and Muller, “Putting the Brakes on innate immunity: a regulatory role for CD200?”, Nat Immunol., 2(1):17-19 (2001).
Ni et al., “An immunoadhesin incorporating the molecule OX-2 is a potent immunosuppressant which prolongs allograft survival”, FASEB Journal 13(5):A983(1999). Abstract Only.
Ohno et al., “Antigen-binding specificities of antibodies are primarily determined by seven residues of VH,” Proc. Natl. Acad. Sci., vol. 82, pp. 2945-2949 (1985).
Pakula and Sauer, “Genetic analysis of protein stability and function,” Annu. Rev. Genet., vol. 23, pp. 289-310 (1989).
Panka et al., “Variable region framework differences result in decreased or increased affinity of variant anti-digoxin antibodies,” Proc, Natl. Acad. Sci. USA, vol. 85:3080-3084 (1988).
Pardoll, Drew., “Therapeutic Vaccination for Cancer,” Clin. Immunol., 95(1):S44-S62(2000).
Paterson, D.J., et al., “Antigens of Activated Rat T Lymphocytes Including a Molecule of 50,000 Mr Detected Only on CD4 Positive T Blasts,” Molecular Immunology, 24(12):1281-1290 (1987).
Related Publications (1)
Number Date Country
20200232990 A1 Jul 2020 US
Provisional Applications (2)
Number Date Country
62578643 Oct 2017 US
62564052 Sep 2017 US