METHODS OF TREATING RED BLOOD CELL DISORDERS

Information

  • Patent Application
  • 20240083995
  • Publication Number
    20240083995
  • Date Filed
    March 02, 2022
    2 years ago
  • Date Published
    March 14, 2024
    a month ago
Abstract
The present invention relates, in part, to methods for treating red blood cell disorders, such as an MDS and/or an anemia, by down-regulating IL-22 signaling.
Description
BACKGROUND OF THE INVENTION

Myelodysplastic syndromes (MDS) are heterogeneous hematopoietic stem and progenitor cell neoplasms characterized clinically by bone marrow (BM) failure and resultant cytopenias (Komrokji et al. (2010) Hematol. Oncol. Clin. North Am. 24:443-457). MDS are the most commonly diagnosed myeloid neoplasms in the United States (Bejar and Steensma (2014) Blood 124(18):2793-2803), with a 3-year survival rate of only 35-45% (Ma (2012) Am. J. Med. 125:S2-S5; Rollison et al. (2008) Blood 112:45-52). According to the most recent MDS risk assessment tool (Revised International Prognostic Scoring System, IPSS-R), the median time to 25% AML transformation ranged from 10.8 years (low-risk MDS group, LR-MDS) to 0.7 years (high-risk MDS group, HR-MDS) (Greenberg et al. (1997) Blood 89:2079-2088; Greenberg et al. (2012) Blood 120:2454-2465; Malcovati et al. (2007) J. Clin. Oncol. 25:3503-3510). Because the majority of patients at diagnosis are ≥60 years of age (Ma (2012) Am. J. Med. 125:S2-S5), most patients are ineligible for BM transplantation due to older age-related comorbidities. Lenalidomide (List et al. (2006) N. Engl. J. Med. 355:1456-1465; Fenaux et al. (2011) Blood 118:3765-3776; Sekeres et al. (2012) Blood 120:4945-4951) and azanucleosides (azacitidine (Silverman et al. (2002) J. Clin. Oncol. 20:2429-2440), decitabine (Lubbert et al. (2011) J. Clin. Oncol. 29:1987-1996; Steensma et al. (2009) J. Clin. Oncol. 27:3842-3848)) remain the only currently approved therapies for treating MDS patients. However, lenalidomide extends survival in LR-MDS patients by only 14-17 months and in the HR-MDS group by 4-6 months. The mean duration of response is ˜10-14 months for treatment with azanucleosides (Fenaux et al. (2009) Lancet Oncol. 10:223-232; Prebet et al. (2014) J. Clin. Oncol. 32:1242-1248). Currently, there are no approved therapies for patients with refractory disease particularly after azanucleoside therapy (Montalban-Bravo and Garcia-Manero (2018) Am. J. Hematol. 93:129-147). No new FDA-approved drugs for MDS have emerged in the past decade (DeZern (2015) Hematol. Am. Soc. Hematol. Educ. Program 2015:308-316) highlighting the critical need to identify new therapeutic targets that will improve the outlook for MDS patients.


Deletion of chromosome 5q (del(5q)), either isolated or accompanied by additional cytogenetic abnormalities, is the most commonly detected chromosomal abnormality in MDS, reported in 10%-30% of patients (Giagounidis et al. (2006) Clin. Cancer Res. 12:5-10; Haase et al. (2007) Blood 110:4385-4395; Hofmann et al. (2004) Hematol J. 5:1-8; Sole et al. (2000) Br. J. Haematol. 108:346-356; Bejar et al. (2011) J. Clin. Oncol. 29:504-515). Anemia is the most common hematologic manifestation of MDS, particularly in patients with del(5q) MDS, along with peripheral blood pancytopenia (Komrokji et al. (2013) Best Pract. Res. Clin. Haematol. 26:365-375). Previous studies using haploinsufficient 5q gene deletions revealed diminished erythroid progenitors (Kumar et al. (2011) Blood 118:4666-4673; Ribezzo et al. (2019) Leukemia 33:1759-1772; Schneider et al. (2014) Cancer Cell 26:509-520; Schneider et al. (2016) Nat. Med. 22:288-297), but the molecular mechanisms underlying this defect remain unclear. The severe anemia in del(5q) MDS patients has been linked to haploinsufficiency of ribosomal proteins such as RPS14 and RPS19 (Ebert et al. (2008) Nature 451:335-339; Dutt et al. (2011) Blood 117:2567-2576). Some ribosomal protein genes lie outside of 5q33, the most commonly deleted 5q region in MDS. One such gene, right open reading frame kinase 2 (RIOK2), lies at the q15 band on human chromosome 5 (5q15) within the genomic breakpoints observed in del(5q) syndromes (Royer-Pokora et al. (2006) Cancer Genet. Cytogenet. 167:66-69; Tang et al. (2015) Am. J. Clin. Pathol. 144:78-86). RIOK2, an atypical serine-threonine protein kinase, functions in the synthesis of the 40S ribosomal subunit (Zemp et al. (2009) J. Cell Biol. 185:1167-1180).


In addition to the need to improve the outlook for MDS patients in general, there is also a need to improve the diagnosis and treatment of anemia, since various types of anemia, such as those caused or worsened by an inability to produce sufficient red blood cells as in MDS, do not have adequate treatments.


SUMMARY OF THE INVENTION

The present invention is based, at least in part, on the discovery that mice having Riok2 haploinsufficiency exhibit anemia and high T-cell-derived IL-22 production. This anemia phenotype can be ameliorated by downregulating IL-22 signaling, such as by deleting one or both copies of the IL-22 gene or the IL-22 receptor (IL-22RA) gene, and/or by treatment with an anti-IL-22 signaling agent (such as a down-regulator of anti-IL-22, down-regulator of IL22RA, and the like). In accordance with disclosures provided herein, various red blood cell disorders (e.g., anemia, myelodysplastic syndromes, anemia caused by myelodysplastic syndromes, anemia caused by insufficiency of serine/threonine-protein kinase RIOK2, anemia caused by one or more mutations and/or deletions on human chromosome 5 or in an ortholog thereof, macrocytic anemia, Diamond Blackfan anemia, Schwachmann-Diamond syndrome, anemia caused by drugs, such as phenylhydrazine or other stressors of erythroid differentiation, and the like) of a subject can be treated by administering to the subject a down-regulator of IL-22 signaling. Such an administration can treat the red blood cell disorders by promoting differentiation from an erythroid progenitor cell toward a mature red blood cell in the subject. Moreover, it was unexpectedly determined herein that erythroid progenitors express the IL-22 receptor A protein.


Immunobiology of hematologic diseases, such as anemia, is an area of study that is largely under-explored. Thus, therapies targeting immune mediators have not been tested in this realm. Use of anti-IL-22-signaling agents to treat anemia, either as a single agent or in a combinatorial approach with currently existing or experimental therapies, according to the present invention are believed to lead to a prolonged beneficial effect, and can also address the problem of developing resistance to single agent therapies.


Accordingly, in one aspect, a method of treating one or more red blood cell disorders in a subject, the method comprising administering to the subject an effective amount of a down-regulator of interleukin-22 (IL-22) signaling, is provided.


Numerous embodiments are further provided that can be applied to any aspect of the present invention and/or combined with any other embodiment described herein. For example, in one embodiment, the one or more red blood disorders comprise anemia. In another embodiment, the one or more red blood disorders comprise one or more myelodysplastic syndromes (MDS), optionally wherein the one or more MDS are mediated by one or more mutations and/or deletions in the long arm of human chromosome 5, or in an orthologous region of an orthologous chromosome thereof. In still another embodiment, the one or more red blood disorders comprise an insufficiency of serine/threonine-protein kinase RIOK2. In yet another embodiment, the one or more red blood disorders comprise an increase in levels of one or more biomarkers listed in Table 1, optionally wherein the one or more biomarkers is IL-22. In another embodiment, the down-regulator comprises an anti-IL-22 antibody or antigen-binding fragment thereof, an anti-IL-22RA1 antibody or antigen-binding fragment thereof, an anti-IL-10Rbeta antibody or antigen-binding fragment thereof, an agent that inhibits the copy number, amount, and/or activity of at least one biomarker listed in Table 1, or a combination thereof. In still another embodiment, the anti-IL-22 antibody or antigen-binding fragment thereof comprises IL22JOP™ monoclonal antibody, such as fezakinumab. In yet another embodiment, the down-regulator comprises an anti-IL-22RA1 antibody or antigen-binding fragment thereof. In another embodiment, the down-regulator comprises an anti-IL-22RA1/IL-10R2-heterodimer antibody or antigen-binding fragment thereof. In still another embodiment, the down-regulator comprises IL-22 binding protein or a fragment thereof. In yet another embodiment, the down-regulator comprises an antagonist of aryl hydrocarbon receptor, such as stemregenin 1, CH-223191, or 6,2′,4′-trimethoxyflavone. In another embodiment, the method further comprises administering to the subject an effective amount of lenalidomide, azacitidine, decitabine, or a combination thereof. In still another embodiment, the method further comprises administering to the subject an effective amount of an erythropoiesis-stimulating agent, such as erythropoietin, epoetin alfa, epoetin beta, epoetin omega, epoetin zeta, IL-9, or darbepoetin alfa.


In another aspect, a method of promoting differentiation of an erythroid progenitor cell toward a mature red blood cell in a subject, the method comprising administering to the subject an effective amount of a down-regulator of interleukin-22 (IL-22) signaling, is provided.


As described above, numerous embodiments are further provided that can be applied to any aspect of the present invention and/or combined with any other embodiment described herein. For example, in one embodiment, the down-regulator comprises an anti-IL-22 antibody or antigen-binding fragment thereof, an anti-IL-22RA1 antibody or antigen-binding fragment thereof, an anti-IL-10Rbeta antibody or antigen-binding fragment thereof, an agent that inhibits the copy number, amount, and/or activity of at least one biomarker listed in Table 1, or a combination thereof. In another embodiment, the anti-IL-22 antibody or antigen-binding fragment thereof comprises IL22JOP™ monoclonal antibody, such as fezakinumab. In still another embodiment, the down-regulator comprises an anti-IL-22RA1 antibody or antigen-binding fragment thereof. In yet another embodiment, the down-regulator comprises an anti-IL-22RA1/IL-10R2-heterodimer antibody or antigen-binding fragment thereof. In another embodiment, the down-regulator comprises IL-22 binding protein or a fragment thereof. In still another embodiment, the down-regulator comprises an antagonist of aryl hydrocarbon receptor, such as stemregenin 1, CH-223191, or 6,2′,4′-trimethoxyflavone. In yet another embodiment, the erythroid progenitor is selected from the group consisting of erythroid progenitors of stage RI, RII, RIII, and RIV.


In still another aspect, a method of determining whether a subject afflicted with or at risk for developing an MDS and/or an anemia would benefit from therapy with a down-regulator of IL-22 signaling, the method comprising a) obtaining a biological sample from the subject; b) determining the copy number, amount, and/or activity of at least one biomarker listed in Table 1; c) determining the copy number, amount, and/or activity of the at least one biomarker in a control; and d) comparing the copy number, amount, and/or activity of the at least one biomarker detected in steps b) and c), wherein the presence of, or a significant increase in, the copy number, amount, and/or activity of at least one biomarker listed in Table 1 in the subject sample relative to the control copy number, amount, and/or activity of the at least one biomarker indicates that the subject afflicted with or at risk for developing the MDS and/or the anemia would benefit from therapy with the down-regulator of IL-22 signaling, is provided.


As described above, numerous embodiments are further provided that can be applied to any aspect of the present invention and/or combined with any other embodiment described herein. For example, in one embodiment, the method further comprises recommending, prescribing, or administering the down-regulator of IL-22 signaling if the subject is determined to benefit from the agent. In another embodiment, the method further comprises recommending, prescribing, or administering at least one additional MDS and/or anemia therapy that is administered before, after, or concurrently with the down-regulator of IL-22 signaling. In still another embodiment, the method further comprises recommending, prescribing, or administering cancer therapy other than a down-regulator of IL-22 signaling if the subject is determined not to benefit from the down-regulator of IL-22 signaling. In yet another embodiment, the down-regulator is selected from the group consisting of an anti-IL-22RA1 antibody or antigen-binding fragment thereof, an anti-IL-10Rbeta antibody or antigen-binding fragment thereof, an agent that inhibits the copy number, amount, and/or activity of at least one biomarker listed in Table 1, and combinations thereof. In another embodiment, the control sample comprises cells.


In yet another aspect, A method for predicting the clinical outcome of a subject afflicted with an MDS and/or an anemia to treatment with a down-regulator of IL-22 signaling, the method comprising a) determining the copy number, amount, and/or activity of at least one biomarker listed in Table 1 in a subject sample; b) determining the copy number, amount, and/or activity of the at least one biomarker in a control having a good clinical outcome; and c) comparing the copy number, amount, and/or activity of the at least one biomarker in the subject sample and in the control, wherein the presence of, or a significant increase in, the copy number, amount, and/or activity of at least one biomarker listed in Table 1 in the subject sample as compared to the copy number, amount and/or activity in the control, is an indication that the subject has a favorable clinical outcome, is provided.


In another aspect, a method for monitoring the efficacy of a down-regulator of IL-22 signaling in treating an MDS and/or an anemia in a subject, wherein the subject is administered a therapeutically effective amount of the down-regulator of IL-22 signaling, the method comprising a) detecting in a subject sample at a first point in time the copy number, amount, and/or activity of at least one biomarker listed in Table 1; b) repeating step a) at a subsequent point in time; and c) comparing the amount or activity of at least one biomarker listed in Table 1 detected in steps a) and b) to monitor the progression of the cancer in the subject, wherein the absence of, or a significant decrease in, the copy number, amount, and/or activity of, the at least one biomarker listed in Table 1 in the subject sample as compared to the copy number, amount and/or activity in the control, is an indication that the down-regulator of IL-22 signaling effectively treats the MDS and/or the anemia in the subject.


In still another aspect, a method of assessing the efficacy of an agent that inhibits the copy number, amount, and/or activity of at least one biomarker listed in Table 1 for treating an MDS and/or an anemia in a subject, comprising a) detecting in a sample at a first point in time the copy number, amount, and/or or activity of at least one biomarker listed in Table 1; b) repeating step a) during at least one subsequent point in time after contacting the sample with the agent; and c) comparing the copy number, amount, and/or activity detected in steps a) and b), wherein the absence of, or a significant decrease in, the copy number, amount, and/or activity of, the at least one biomarker listed in Table 1, in the subsequent sample as compared to the copy number, amount, and/or activity in the sample at the first point in time, indicates that the agent effectively treats the MDS and/or the anemia.


As described above, numerous embodiments are further provided that can be applied to any aspect of the present invention and/or combined with any other embodiment described herein. For example, in one embodiment, the subject has undergone treatment, completed treatment, and/or is in remission for the MDS and/or the anemia between the first point in time and the subsequent point in time. In another embodiment, the first and/or at least one subsequent sample is selected from the group consisting of in vitro samples, optionally wherein the in vitro sample comprising cells. In still another embodiment, the first and/or at least one subsequent sample is selected from the group consisting of ex vivo and in vivo samples. In yet another embodiment, the first and/or at least one subsequent sample is a portion of a single sample or pooled samples obtained from the subject. In another embodiment, the sample comprises blood, bone marrow fluid, or Th22 T lymphocytes. In still another embodiment, biomarker mRNA and/or protein are detected. In yet another embodiment, the MDS and/or the anemia is selected from the group consisting of macrocytic anemia, anemia associated with chronic kidney disease (CKD), anemia caused by insufficiency of serine/threonine-protein kinase RIOK2, anemia caused by one or more mutations and/or deletions in human chromosome 5 or in an ortholog thereof, stress-induced anemia, Diamond Blackfan anemia, and Schwachman-Diamond syndrome. In another embodiment, wherein the subject is a mammal, optionally wherein the mammal is a human, a mouse, and/or an animal model of an MDS and/or an anemia.





BRIEF DESCRIPTION OF THE DRAWINGS

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the office upon request and payment of the necessary fee.



FIG. 1A-FIG. 1E show localization and expression of Riok2. FIG. 1A shows the location of RIOK2 on human chromosome 5. FIG. 1B shows expression of Riok2 in mouse BM cells. FIG. 1C shows Riok2 mRNA expression by qPCR in BM cells from Riok2 haploinsufficient mice and Vav1-cre controls. FIG. 1D shows frequency of genotypes indicated on the X-axis among 4 litters from 4 different breeding crosses of the genotypes mentioned. FIG. 1E shows in vivo protein synthesis rates in the indicates cell types from Riok2 haploinsufficient mice and Vav1-cre controls. * p<0.05, **** p<0.0001.



FIG. 2A-FIG. 2G show that Riok2 haploinsufficient (Riok2f/+Vav1cre) mice display anemia and myeloproliferation. FIG. 2A shows peripheral blood (PB) RBC numbers, hemoglobin (Hb), and hematocrit (HCT) in Riok2f/+Vav1cre mice in comparison to Riok2+/+Vav1cre controls (n=5/group). FIG. 2B shows frequency of erythroid progenitor populations among viable bone marrow cells in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls (n=5/group). FIG. 2C shows frequency of apoptotic erythroid progenitors among viable bone marrow cells in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls (n=5/group). FIG. 2D shows PB RBC numbers, Hb, and HCT in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls undergoing phenylhydrazine (PhZ)-induced stress erythropoiesis (n=7/group). FIG. 2E shows percentage of monocytes and neutrophils in the PB of Riok2f/+Vav1cre mice in comparison to Riok2+/+Vav1cre controls (n=5-6/group). FIG. 2F shows percentage of GMPs in the BM of Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls (n=3-4/group). FIG. 2G shows percentage CD11b+ cells obtained from Lin-Sca-1+c-kit+ cells from Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls cultured in MethoCult™ for 7 days (n=5/group). Unpaired two-tailed t-test (FIGS. 2A-2C and 2E-2G) and 1-way ANOVA with Tukey's correction for multiple comparison (FIG. 2D) used to calculate statistical significance. * p<0.05, ** p<0.01. Data are shown as mean±s.e.m and are representative of two (FIGS. 2D and 2F) or three (FIGS. 2A-2C and 2G) independent experiments.



FIG. 3A-FIG. 3E further show that Riok2 haploinsufficient mice display anemia. FIG. 3A shows a gating strategy used for the identification of erythroid progenitors in the BM. FIG. 3B shows results of a cell cycle analysis of erythroid progenitors from Riok2 haploinsufficient mice in comparison to Vav1-cre controls. FIG. 3C shows cdkn1a mRNA expression by qPCR in erythroid progenitors from Riok2 haploinsufficient mice and Vav1-cre controls. FIG. 3D shows a Kaplan-Meier survival curve for Riok2 haploinsufficient mice and Vav1-cre controls subjected to lethal dose of PhZ. FIG. 3E shows PB RBC numbers, Hb, and HCT in mice transplanted with either Riok2 haploinsufficient mice or Vav1-cre BM cells. * p<0.05, ** p<0.01.



FIG. 4A-FIG. 4D show immune activation signatures resulting from quantitative proteomics of Riok2 haploinsufficient progenitors. FIG. 4A shows proteomic analysis of changes in protein expression in erythroid progenitors from Riok2 haploinsufficient mice and Vav1-cre controls. FIG. 4B shows a comparison of upregulated proteins with their respective p-values and log fold-change values in erythroid progenitors from Riok2-haploinsufficient mice and Rps14-haploinsufficient mice with their respective controls. FIG. 4C shows overlap of all the upregulated proteins in each of the dataset with respect to their respective controls. FIG. 4D shows secreted IL-22 levels from in vitro-polarized Th22 cells from Rps14 haploinsufficient mice and Vav1-cre controls. * p<0.05.



FIG. 5A-FIG. 5G show that expression of lineage-associated T cell factors is comparable between Riok2 haploinsufficient and sufficient cells. Concentration of IL-2 (FIG. 5A), IFN-gamma (FIG. 5B), IL-4 (FIG. 5C), IL-5 (FIG. 5D), IL-13 (FIG. 5E), IL-17A (FIG. 5F) and % Foxp3+ cells (FIG. 5G) from in vitro polarized T cells of the indicated genotypes are shown.



FIG. 6A-FIG. 6G show that Riok2 haploinsufficient T cells secrete increased IL-22 and that IL-22 neutralization alleviates anemia. FIGS. 6A and 6B show secreted IL-22 (FIG. 6A) and percentage of IL-22+CD4+ T cells (FIG. 6B) from in vitro-polarized Th22 cells from Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls (n=5/group). FIG. 6C shows IL-22 levels in the serum (left panel) and bone marrow supernatant (right panel) in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls (n=5/group). FIG. 6D shows PB RBC numbers, Hb, and HCT in the indicated strains undergoing PhZ-induced stress erythropoiesis (n=4-5/group). FIG. 6E shows frequency of erythroid progenitor populations among viable bone marrow cells in the indicated strains undergoing PhZ-induced stress erythropoiesis (n=4-5/group). FIG. 6F shows PB RBC numbers, Hb, and HCT in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls undergoing PhZ-induced stress erythropoiesis treated with either an isotype control or anti-IL-22 antibody (n=4-5/group). FIG. 6G shows frequency of apoptotic erythroid progenitors among viable bone marrow cells in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls undergoing PhZ-induced stress erythropoiesis treated with either an isotype control or anti-IL-22 antibody (n=4-5/group). Unpaired two-tailed t-test (FIG. 6A-FIG. 6C) and 1-way ANOVA with Tukey's correction for multiple comparison (FIG. 6D-FIG. 6G) used to calculate statistical significance. * p<0.05, ** p<0.01. Data are shown as mean±s.e.m and are representative of two (FIG. 6C and FIG. 6F) or three (FIGS. 6A, 6B, 6D, and 6E) independent experiments.



FIG. 7A-FIG. 7E show that recombinant IL-22 exacerbates PhZ-induced anemia in wt mice. FIG. 7A shows PB RBC numbers, Hb, and HCT in wt C57BL/6J mice administered PBS or rIL-22 and subsequently treated with PhZ. FIG. 7B shows PB reticulocytes in mice treated as in FIG. 7A. FIG. 7C shows percentage of RII-RIV erythroid progenitors in the BM of PBS- or rIL-22-treated C57BL/6J mice 7 days after PhZ administration. FIG. 7D shows percentage of apoptotic RII erythroid progenitors in mice treated as in FIG. 7C. FIG. 7E shows an effect of recombinant IL-22 (500 ng/mL) in an in vitro erythropoiesis assay (left panel) and dose-dependent effect of recombinant IL-22 (right panel). * p<0.05, ** p<0.01, **** p<0.0001



FIG. 8A-FIG. 8B show that IL-22 neutralization alleviates anemia in wt mice undergoing PhZ-induced stress erythropoiesis. FIG. 8A shows PB RBC numbers, Hb, and HCT in naïve wt C57BL/6J mice treated with either an isotype control or anti-IL-22 antibody. FIG. 8B shows PB RBC numbers, Hb, and HCT in wt C57BL/6J mice undergoing PhZ-induced stress erythropoiesis treated with either an isotype control or anti-IL-22 antibody. *** p<0.001.



FIG. 9A-FIG. 9E show that genetic deletion of IL-22RA1 alleviates anemia in Riok2 haploinsufficient mice. FIG. 9A shows IL-22RA1 expression on erythroid progenitors in wild-type (wt) mice as assessed by flow cytometry using antibody from Novus Biologicals targeting the extracellular domain of IL-22RA1. FIG. 9B shows PB RBC numbers, Hb, and HCT in the indicated strains undergoing PhZ-induced stress erythropoiesis (n=5-6/group). FIG. 9C shows frequency of erythroid progenitor populations among viable bone marrow cells in the indicated strains undergoing PhZ-induced stress erythropoiesis (n=5/group). FIG. 9D shows PB RBC numbers, Hb, and HCT in the indicated strains undergoing PhZ-induced stress erythropoiesis (n=6/group). FIG. 9E shows frequency of erythroid progenitor populations among viable bone marrow cells in the indicated strains undergoing PhZ-induced stress erythropoiesis (n=5/group). Unpaired two-tailed t-test (FIGS. 9D and 9E and 1-way ANOVA with Tukey's correction for multiple comparison (FIGS. 9B and 9C) used to calculate statistical significance. * p<0.05. Data are shown as mean±s.e.m and are representative of two (FIGS. 9D and 9E) or three (FIG. 9A-9C) independent experiments. The presence of the IL-22RA1 receptor on erythroid progenitors has not been previously discovered and is an important component of the use of IL-22 signaling blockade.



FIG. 10A-FIG. 10C show that erythroid progenitors express IL-22RA1. FIG. 10A shows a gating strategy employed for assessing IL-22RA1 expression by erythroid progenitors. FIG. 10B shows a gating strategy to show that majority of IL-22RA1+ cells in the mouse BM are erythroid progenitors. FIG. 10C shows IL-22RA1 expression on erythroid progenitors assessed using flow cytometry and a second antibody targeting a different epitope.



FIG. 11A-FIG. 11F shows that MDS patients exhibit increased IL-22 levels and IL-22 associated signature. FIG. 11A shows IL-22 concentration in the BM fluid of healthy controls and del(5q) and nondel(5q) MDS patients. FIG. 11B shows correlation between RIOK2 mRNA and IL-22 concentration in the del(5q) cohort shown in FIG. 11A. FIG. 11C shows frequency of IL-22 producing CD4 T cells in the PB of healthy controls and MDS patients. FIG. 11D shows expression of IL-22 signature genes in CD34+ cells from healthy controls and del(5q) and nondel(5q) MDS patients. FIG. 11E shows plasma IL-22 concentration in healthy subjects and CKD patients with or without secondary anemia. FIG. 11F shows correlation between IL-22 concentration and hemoglobin levels in CKD patients shown in FIG. 11E. r denotes Pearson correlation coefficient. Unpaired two-tailed t-test (FIG. 11C) and 1-way ANOVA with Tukey's correction for multiple comparison (FIGS. 11A and 11D) used to calculate statistical significance. * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001.



FIG. 12 shows increased frequency of IL-22+CD4+ cells in MDS subjects. The figure shows frequency of CD4+IL-22+ cells among total PBMCs in the peripheral blood of MDS patients and healthy subjects.



FIG. 13 is a chart showing some of the functions of IL-22 in progenitor and immune cells.



FIG. 14A-FIG. 14I show that Riok2 haploinsufficient (Riok2f/+Vav1cre) mice display anemia and myeloproliferation. FIG. 14A shows peripheral blood (PB) RBC numbers, hemoglobin (Hb), and hematocrit (HCT) in Riok2f/+Vav1cre mice in comparison to Riok2+/+Vav1cre controls (n=5/group). FIG. 14B shows frequency of erythroid progenitor/precursor populations among viable bone marrow (BM) cells in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls (n=5/group). FIG. 14C shows frequency of apoptotic erythroid precursors among viable BM cells in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls (n=5/group). FIG. 14D shows PB RBC numbers, Hb, and HCT in Riok2f/+Vav mice and Riok2+/+Vav1cre controls undergoing phenylhydrazine (PhZ)-induced stress erythropoiesis (n=7/group). FIG. 14E shows frequency of RIII and RIV erythroid precursor populations among viable BM cells in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls day 6 after PhZ treatment (n=4/group). FIG. 14F shows number of CFU-e colonies in Epo-containing MethoCult assay using Linc-kit+CD71+ cells from Riok2f/+Vav1cre mice (n=4) in comparison to Riok2+/+Vav1cre controls (n=6). FIG. 14G shows percentage of monocytes (CD11b+Ly6GLy6Chi) and neutrophils (CD11b+Ly6G+) in the PB of Riok2f/+Vav1cre mice (n=6) in comparison to Riok2+/+Vav1cre controls (n=5). FIG. 14H shows Percentage of Ki-67+ granulocyte-macrophage progenitors (GMPs) in the BM of Riok2f/+Vav1cre mice (n=3) and Riok2+/+Vav1cre controls (n=4). FIG. 14I shows Percentage of CD11b+ cells obtained from LinSca-1+c-kit+BM cells from Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls cultured in MethoCult for 7 days (n=5/group). Unpaired two-tailed t-test (FIG. 14A to C, E to I) and 1-way ANOVA with Tukey's correction for multiple comparisons (FIG. 14D) used to calculate statistical significance. * p<0.05, ** p<0.01, *** p<0.001. Data are shown as mean±s.e.m and are representative of two (FIG. 14C, D, F-H) or three (FIG. 14A, B, E, I) independent experiments.



FIG. 15A-FIG. 15D show quantitative proteomics of Riok2 haploinsufficient erythroid precursors reveals immune activation signatures. FIG. 15A to C show GSEA performed on proteomics data shown in FIG. 4A to reveal similarity with Rps14 haploinsufficient data (FIG. 15A), activation of immune response (FIG. 15B) and enrichment of IL-22 signature genes (FIG. 15C). NES=Normalized enrichment score, FDR=False discovery rate. FIG. 15D shows MetaCore analysis of the Riok2 proteomics dataset shown in FIG. 4A. Two sample moderated t-test with multiple hypothesis corrections used to calculate the statistical significance in FIG. 4B.



FIG. 16A-FIG. 16N show Riok2 haploinsufficiency-driven p53 upregulation drives increased IL-22. FIG. 16A shows secreted IL-22 and FIG. 16B shows percentage of IL-22+CD4+ T cells from in vitro polarized TH22 cells from Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls (n=5/group). FIG. 16C shows IL-22 levels in the serum (left) and bone marrow fluid (BMF) (right) in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls (n=5/group). FIG. 16D shows number of IL-22+CD4+cells in the spleens of Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls (n=5/group). FIG. 16E shows Volcano plot showing transcriptomic changes in purified IL-22+ cells from Riok2f/+Vav1cre mice in comparison to Riok2+/+Vav1cre controls. n=5/group. FIG. 16F shows GSEA analysis showing activation of the p53 pathway in IL-22+ cells from Riok2f/+Vav1cre mice in comparison to Riok2+/+Vav1cre controls. FIG. 16G shows Snapshot of differentially expressed genes in the p53 pathway shown in (FIG. 16F) in Riok2f/+Vav1cre and Riok2+/+Vav1cre mice. FIG. 16H shows flow cytometry plot showing p53 expression in in vitro polarized TH22 cells from Riok2f/+Vav1cre and Riok2+/+Vav1cre controls. FIG. 16I shows graphical representation of data shown in (FIG. 16H). n=5/group. FIG. 16J shows predicted p53 binding site in the Il22 promoter region. SEQ ID NO: 7. AGTTAAGTTTGGAAATATCG. FIG. 16K shows chromatin immunoprecipitation showing p53 occupancy at the Il22 promoter in T cells. n=2 independent experiments. FIG. 16I shows secreted IL-22 from wt TH22 cells cultured in the presence or absence of p53 inhibitor, pifithrin-α, p-nitro (1 μM). n=5 mice/group. FIG. 16M shows secreted IL-22 from WT TH22 cells cultured in the presence or absence of p53 activator, Nutlin-3 (100 nM). n=4 mice/group. FIG. 16N shows secreted IL-22 from in vitro polarized TH22 cells from the indicated strains. n=5/group. Unpaired two-tailed t-test (FIG. 16A to D, I, K-M) and 1-way ANOVA with Tukey's correction for multiple comparison (n) used to calculate statistical significance. * p<0.05, ** p<0.01, *** p<0.001. Data are shown as mean±s.e.m and are representative of two (FIG. 16C-D, H, I, K-N) or three (FIG. 16A, B) independent experiments. Data in (FIG. 16K) is represented as mean±s.d. and is pooled from two independent experiments.



FIG. 17A-FIG. 17D show IL-22 neutralization alleviates stress-induced anemia in Riok2 sufficient and haploinsufficient mice. FIG. 17A shows PB RBC numbers, Hb, and HCT in the indicated strains undergoing PhZ-induced stress erythropoiesis. n=6,5,5, and 5 mice for Riok2+/+Il22+/+Vav1cre, Riok2+/+Il22+/+Vav1cre, Riok2f/+Il22+/+Vav1cre, Riok2f/+Il22+/−Vav1cre respectively. FIG. 17B shows frequency of erythroid progenitor/precursor populations among viable BM cells in the indicated strains undergoing PhZ-induced stress erythropoiesis (n=4-5/group). FIG. 17C shows PB RBC numbers, Hb, and HCT in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls undergoing PhZ-induced stress erythropoiesis treated with either an isotype control or anti-IL-22 antibody (n=4-5/group). FIG. 17D shows frequency of apoptotic erythroid precursors among viable BM cells in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls undergoing PhZ-induced stress erythropoiesis treated with either an isotype control or anti-IL-22 antibody. n=4,5,4, and 5 mice for isotype-treated Riok2+/+Vav1cre, anti-IL-22-treated Riok2+/+Vav1cre, isotype-treated Riok2f/+Vav1cre, and anti-IL-22-treated Riok2f/+Vav1cre mice, respectively. 1-way ANOVA with Tukey's correction for multiple comparison (FIG. 17A to D) used to calculate statistical significance. * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001. Data are shown as mean±s.e.m and are representative of two (FIG. 17C, D) or three (FIG. 17A, B) independent experiments.



FIG. 18A-FIG. 18G show recombinant IL-22 exacerbates PhZ-induced anemia in wt mice. FIG. 18A shows PB RBC numbers, Hb, and HCT in wt C57BL/6J mice administered PBS (n=5) or rIL-22 (n=4) and subsequently treated with PhZ. n=4-5 mice/group. FIG. 18B shows PB reticulocytes in mice treated as in (FIG. 18A). n=4 mice/group. FIG. 18C shows percentage of RII-RIV erythroid precursors in the BM of PBS- or rIL-22-treated C57BL/6J mice 7 days after PhZ administration. n=4 mice/group. FIG. 18D shows percentage of apoptotic RH erythroid precursors in mice treated as in (FIG. 18C). n=4 mice/group. FIG. 18D shows effect of recombinant IL-22 (500 ng/mL) on the frequency (left) and cell number (right) in an in vitro erythropoiesis assay and FIG. 18F shows dose dependent effect of recombinant IL-22. n=5 and 4 for PBS and IL-22 groups, respectively. FIG. 18G shows p53 expression in in in vitro erythropoiesis culture treated with rIL-22 or PBS. n=5 and 4 mice for PBS and IL-22 groups, respectively. Data are shown as mean±s.e.m and are representative of three (FIG. 18A, B) or two (FIG. 18C to G) independent experiments. Unpaired two-tailed t-test (FIG. 18A to D, G), multiple unpaired two-tailed t-tests with Holm-Sidak method (FIG. 18E) and 1-way ANOVA with Tukey's correction for multiple comparisons (FIG. 18F) used to calculate statistical significance. * p <0.05, ** p<0.01, *** p<0.001, **** p<0.0001.



FIG. 19A-FIG. 19G show genetic deletion of Il22ra1 alleviates anemia in Riok2 haploinsufficient mice. FIG. 19A shows IL-22RA1 expression on BM erythroid precursors in wild-type (WT) mice assessed by flow cytometry using antibody from Novus Biologicals targeting the extracellular domain of IL-22RA1. FIG. 19B shows PB RBC numbers, Hb, and HCT in the indicated strains undergoing PhZ-induced stress erythropoiesis. n=6,5,6, and 4 mice for Riok2+/+Il22r1a+/+Vav1cre, Riok2+/+Il22ra1f/fVav1cre, Riok2f/+Il22ra1+/+Vav1cre, Riok2f/+Il22ra1f/fVav1cre, respectively. FIG. 19C shows frequency of erythroid progenitor/precursor populations among viable BM cells in the indicated strains undergoing PhZ-induced stress erythropoiesis (n=5/group). FIG. 19D shows flow cytometry plots showing p53 expression in IL-22RA1+ and IL-22RA1 erythroid precursors in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls. n=5/group. FIG. 19E shows graphical representation of data shown in (FIG. 19D). FIG. 19F shows gene expression of Trp53 (p53) and listed p53 target genes in IL-22RA1+ and IL-22RA1 erythroid precursors from Riok2f/+Vav1cre and Riok2+/+Vav1cre mice assessed by qRT-PCR. n=3/group. FIG. 19G shows frequency of apoptotic cells (assessed by flow cytometry) with (n=4) or without (n=5) p53 inhibitor, pifithrin-α, p-nitro (1 μM), in an in vitro erythropoiesis assay using Lin BM cells from Riok2f/+Vav1cre and Riok2+/+Vav1cre mice. 2-way ANOVA with Tukey's correction for multiple comparisons (FIGS. 19E, G) and 1-way ANOVA with Tukey's correction for multiple comparison (FIGS. 19B, C) used to calculate statistical significance. * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001. Data are shown as mean±s.e.m and are representative of two (FIGS. 19D-G) or three (FIGS. 19A to C) independent experiments.



FIG. 20A-FIG. 20C show erythroid-specific deletion of IL-22RA1 alleviates stress-induced anemia. FIG. 20A shows PB RBC numbers, Hb, and HCT in the indicated strains undergoing PhZ-induced stress erythropoiesis (n=6/group). FIG. 20B shows frequency of erythroid progenitor/precursor populations among viable BM cells in the indicated strains undergoing PhZ-induced stress erythropoiesis (n=5/group). FIG. 20C shows PB RBC numbers, Hb, and HCT in Il22ra1+/+Eporcre (n=5) and Il22ra1f/fEporcre (n=4) mice administered rIL-22 and subsequently treated with PhZ. n=4-5 mice/group. Unpaired two-tailed t-test (FIG. 20A-C) used to calculate statistical significance. * p<0.05, *** p<0.001. Data are shown as mean±s.e.m and are representative of two (FIG. 20A-C) independent experiments.



FIG. 21A-FIG. 21G show MDS patients exhibit increased IL-22 levels and IL-22 associated signature. FIG. 21A shows IL-22 concentration in the BM fluid of healthy controls (n=12), del(5q) MDS (n=11), and non-del(5q) MDS (n=22) patients. FIG. 21B shows correlation between RIOK2 mRNA and BM IL-22 concentration in the del(5q) cohort shown in (a), n=10. FIG. 21C shows S100A8 concentration in the samples shown in (a). n=11, 10, and 19 for healthy, del(5q) MDS, and non-del(5q) MDS, respectively. FIG. 21D shows correlation between IL-22 concentration and S100A8 concentration in BM fluid of del(5q) (left, n=10) and non-del(5q) (right, n=19) samples. FIG. 21E shows frequency of IL-22 producing CD4+ T cells in the PB of healthy controls (n=11), del(5q) (n=3) and non-del(5q) (n=24) MDS patients. FIG. 21F shows plasma IL-22 concentration in healthy subjects (n=10) and chronic kidney disease (CKD) patients with (n=13) or without (n=13) secondary anemia. FIG. 21G shows correlation between plasma IL-22 concentration and hemoglobin (HGB) in CKD patients with (n=13) or without (n=13) anemia. Kruskal-Wallis test with Dunn's correction for multiple comparisons (FIG. 21A to C, E), 1-way ANOVA with Tukey's correction for multiple comparisons (FIG. 21F) used to calculate statistical significance. Pearson correlation co-efficient (FIG. 21B, D, G), used to calculate statistical significance and correlation coefficient. ** p<0.01, *** p<0.001, **** p<0.0001. Data are shown as mean±s.e.m (FIG. 21C). Solid lines represent median and dashed lines represent quartiles (FIG. 21A, E, F).



FIG. 22A-FIG. 22E show localization and expression of Riok2. FIG. 22A shows schematic representation of the Riok2tmla(KOMP)Wtsi allele and generation of Riok2 foxed mice. FIG. 22B shows agarose gel showing genotyping of Riok2 foxed mice. Riok2Δ indicates deletion of Riok2. No band expected in the Riok2wt lane. FIG. 22C shows Riok2 mRNA expression by qRT-PCR in BM cells from Riok2 haploinsufficient mice and Vav1cre controls. n=5 mice/group. FIG. 22D shows frequency of the genotypes indicated on the X-axis among 4 litters from 4 different breeding crosses of the genotypes mentioned. FIG. 22E shows in vivo protein synthesis rates in the indicated cell types from Riok2 haploinsufficient mice (n=2) and Vav1cre controls (n=8). Unpaired two-tailed t-test (FIG. 22C), multiple unpaired two-tailed t-tests with Holm-Sidak method (FIG. 22E) used to calculate statistical significance. Data are shown as mean±s.e.m (FIG. 22C, D) or mean±s.d. (FIG. 22E) and are representative of two (FIG. 22C, E) or four (FIG. 22B, D) independent experiments. ** p<0.01, *** p<0.001, **** p<0.0001.



FIG. 23A-FIG. 23K further show that Riok2 haploinsufficient mice display anemia and myeloproliferation. FIG. 23A shows gating strategy used for the identification of erythroid progenitor/precursor cells in the BM. FIG. 23B shows Number of erythroid progenitor populations among viable BM cells in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls. n=5/group. FIG. 23C shows cell cycle analysis of erythroid progenitor/precursor cells from Riok2 haploinsufficient mice in comparison to Vav1cre controls. n=5 mice/group. FIG. 23D shows Cdkn1a mRNA expression by qRT-PCR in erythroid progenitors from Riok2 haploinsufficient mice and Vav1cre controls. n=3 mice/group. FIG. 23E shows Kaplan-Meier survival curve for Riok2 haploinsufficient mice and Vav1cre controls subjected to lethal dose of PhZ. FIG. 23F shows number of RIII and RIV erythroid precursor populations among viable BM cells in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls day 6 after PhZ treatment. n=4/group. FIG. 23G shows PB RBC numbers, Hb, and HCT in mice transplanted with either Riok2 haploinsufficient mice or Vav1cre BM cells. n=5 mice/group. FIG. 23H shows PB RBC numbers, Hb, and HCT in mice with tamoxifen-inducible deletion of Riok2. Tamoxifen administered on days 3-7. n=8 and 7 for Riok2+/+Ert2cre and Riok2f/+Ert2cre mice, respectively. FIG. 23I shows representative flow cytometry plots showing frequency of monocytes (CD11b+Ly6GLc6Chi) and neutrophils (CD11b+Ly6G+) in the PB of Riok2f/+Vav1cre and Riok2+/+Vav1cre mice. FIG. 23J shows representative flow cytometry plots showing Ki-67+ GMPs in the BM of Riok2f/+Vav1cre and Riok2+/+Vav1cre mice. FIG. 23K shows number of CFU-GM colonies in MethoCult from LinSca-1+c-kit+ BM cells from Riok2f/+Vav1cre mice (n=5) and Riok2+/+Vav1cre controls (n=4) after a 7-day culture period. n=4-5/group. Multiple unpaired two-tailed t-tests with Holm-Sidak method (FIG. 23B, C), unpaired two-tailed t-test (FIG. 23D, F, G, K), log-rank test (FIG. 23E), and 2-way ANOVA with Sidak's correction for multiple comparisons (FIG. 23H) used to calculate statistical significance. Data are shown as mean±s.e.m and are representative of two (FIG. 23B to K) independent experiments. * p<0.05, ** p<0.01, *** p<0.001.



FIG. 24A-FIG. 24C show that Riok2 haploinsufficiency alters early hematopoietic progenitors in an age-dependent fashion. FIG. 24A shows frequency and number of indicated cell types in the bone marrow of Riok2f/+Vav1cre and Riok2+/+Vav1cre mice. n=4/group. LT-HSC=long term hematopoietic stem cells, ST-HSC=short term hematopoietic stem cells, MPP=multipotent progenitors, CLP=common lymphoid progenitors. FIG. 24B shows % CD45.2 (donor) chimerism in PB from competitive BM transplant with CD45.1 recipient cells. Time point ‘−1’ reflects first bleeding 4 weeks after transplantation and one day before tamoxifen induced deletion of Riok2. Donor (CD45.2) chimerism of the HSC compartment in the BM of competitive transplantation experiments. n=5/group. FIG. 24C shows frequency of donor (CD45.2+) early hematopoietic progenitors 24 weeks after tamoxifen treatment in a competitive transplantation assay as described in (FIG. 24B). n=5 and 4 for Riok2+/+Ert2cre and Riok2f/+Ert2cre mice, respectively. Unpaired two-tailed t-test (FIGS. 24A, C) and 2-way ANOVA with Sidak's multiple comparison test (FIG. 24B) used to calculate statistical significance. Data are shown as mean±s.e.m and are representative of two (FIGS. 24A-C) independent experiments. * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001.



FIG. 25A-FIG. 25G show that Riok2 haploinsufficient erythroid precursors express increased S100 proteins. FIG. 25A shows expression of ribosomal proteins quantified by proteomics in Riok2f/+Vav1cre and Riok2+/+Vav1cre erythroid precursors. S100A8 (FIG. 25B) and S100A9 (FIG. 25C) expression assessed by flow cytometry in BM erythroid precursors from Riok2f/+Vav1cre and Riok2+/+Vav1cre mice. n=4 mice/group. FIGS. 25D and E show S100a8 and S100a9 mRNA expression in erythroid precursors isolated from Riok2f/+Vav1cre and Riok2+/+Vav1cre mice. n=4 mice/group. FIG. 25F shows p53 expression assessed by flow cytometry in BM erythroid precursors from Riok2f/+Vav1cre and Riok2+/+Vav1cre mice. FIG. 25G shows graphical representation of data shown in (FIG. 25E). n=5 mice/group. Data are shown as mean±s.e.m and are representative of two (FIGS. 25B to G) independent experiments. Unpaired two-tailed t-test (FIGS. 25B to G) used to calculate statistical significance. ** p<0.01, *** p<0.001, **** p<0.0001.



FIG. 26A-FIG. 26N show that expression of lineage-associated T cell cytokines is comparable between Riok2 haploinsufficient and sufficient T cells. FIG. 26A to G show concentration of IL-2 (FIG. 26A), IFN-γ (FIG. 26B), IL-4 (FIG. 26C), IL-5 (FIG. 26D), IL-13 (FIG. 26E), IL-17A (FIG. 26F) and frequency of Foxp3+cells (FIG. 26G) from in vitro polarized T cells of the indicated genotypes. n=3 mice/group. FIGS. 26H to I show number of IL-22+ NKT cells (FIG. 26H) and ILCs (FIG. 26I) in the spleens of Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls (n=4/group). FIG. 26J shows frequency of IL_23p19+ DCs in Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls. n=4 mice/group. FIG. 26K shows PB RBC numbers, Hb, and HCT in Riok2 f Cd4cre mice (n=3) in comparison to Riok2+/+Cd4cre controls (n=5). FIG. 26L shows secreted IL-22 from in vitro polarized TH22 cells from Rps14 haploinsufficient mice and Vav1cre controls. n=4 mice/group. FIG. 26M shows secreted IL-22 from in vitro polarized TH22 cells from ApcMin mice and littermate controls. n=4 mice/group. FIG. 26N shows viable cells (expressed as percentage of total cells in culture) for the indicated treatments assessed by flow cytometry. n=5 mice/group. Data are shown as mean±s.e.m and are representative of two (FIGS. 26A to N) independent experiments. Unpaired two-tailed t-test FIG. 26A to N) used to calculate statistical significance. * p<0.05, ** p<0.01.



FIG. 27A-FIG. 27D show that neutralization of IL-22 signaling increases number of erythroid precursors. FIG. 27A to D show number of RI-RIV erythroid populations among viable BM cells in the indicated strains undergoing PhZ-induced stress erythropoiesis. For (FIG. 27A), n=5,5,4, and 4 for Riok2+/+Il22+/+Vav1cre, Riok2+/+Il22+/+Vav1cre, Riok2f/+Il22+/+Vav1cre, Riok2f/+Il22f/−Vav1cre respectively. For (FIG. 27D), n=5/group. Data are shown as mean±s.e.m and are representative of three (FIGS. 27A, C) or two (FIGS. 27B, D) independent experiments. 1-way ANOVA with Tukey's correction (FIGS. 27A, C) or unpaired two-tailed t-test (FIGS. 27B, D) used to calculate statistical significance. * p<0.05, ** p<0.01.



FIG. 28A-FIG. 28C show that IL-22 neutralization alleviates anemia in wt mice undergoing PhZ-induced stress erythropoiesis. FIG. 28A shows PB RBC numbers, Hb, and HCT in naïve wt C57BL/6J mice treated with isotype control (Rat IgG2aκ, 50 mg/mouse) or anti-IL-22 antibody (50 mg/mouse). n=5 mice/group. FIG. 28B shows PB RBC numbers, Hb, and HCT in wt C57BL/6J mice undergoing PhZ-induced stress erythropoiesis treated with isotype control or anti-IL-22 antibody. n=5 mice/group. FIG. 28C shows Percentage of RI-RIV erythroid precursors in the BM of mice treated as in (FIG. 28B). n=4 and 5 for Il22ra1+/+Eporcre and Il22ra1f/fEporcre mice, respectively. Data are shown as mean±s.e.m and are representative of three (FIGS. 28A, B) or two (FIG. 28C) independent experiments. Unpaired two-tailed t-test (FIGS. 28A to C) used to calculate statistical significance. * p<0.05, *** p<0.001.



FIG. 29A-FIG. 29D show that erythroid precursors express IL-22RA1. FIG. 29A shows gating strategy employed for assessing IL-22RA1 expression on erythroid precursors. FIG. 29B shows gating strategy to show that majority of IL-22RA1+cells in the mouse BM are erythroid precursors. FIG. 29C shows IL-22RA1 expression on erythroid precursors assessed using flow cytometry and a second antibody targeting a different epitope of IL-22RA1. FIG. 29D shows Il22ra1 mRNA expression in the indicated cell types assessed by qRT-PCR. T cells and liver represent negative and positive controls, respectively. n=4 mice/group. Data are shown as mean±s.e.m (FIG. 29D) and are representative of three (FIGS. 29A to C) or two (FIG. 29D) independent experiments.



FIG. 30A-FIG. 30B show increased IL-22 and its signature genes in del(5q) MDS subjects IL-22. FIG. 30A shows representative flow cytometry plots showing frequency of CD4+IL-22+cells among total PBMCs in the peripheral blood of MDS patients and healthy subjects. Pre-gated on viable CD3e+CD4+cells. Cumulative data shown in FIG. 25E. FIG. 30B shows expression of indicated IL-22 signature genes in CD34+cells from healthy controls and del(5q) and non-del(5q) MDS patients. n=17, 47, and 136 for healthy, del(5q) MDS, and non-del(5q) MDS, respectively. Kruskal-Wallis test with Dunn's correction for multiple comparisons (FIG. 30B) used to calculate statistical significance * p <0.05, ** p<0.01, *** p<0.001, **** p<0.0001. Solid lines represent median and dashed lines represent quartiles (FIG. 30B).



FIG. 31A-FIG. 31C show that Riok2 haploinsufficiency recapitulates del(5q) MDS transcriptional changes. FIG. 31A to B show GSEA enrichment plots comparing proteins up-regulated (FIG. 31A) and down-regulated (FIG. 31B) upon Riok2 haploinsufficiency to the transcriptional changes seen in del(5q) MDS. FIG. 31C shows schematic of mechanism underlying Riok2 haploinsufficiency-induced, IL-22—induced anemia.



FIG. 32A-FIG. 32B show that anti-IL-22 inhibits recombinant IL-22-induced IL-10 production. FIG. 32A and FIG. 32B show COLO-205 cells stimulated with recombinant mouse IL-22 (FIG. 32A) and recombinant human IL-22 (FIG. 32B) in the presence of anti-IL-22 or matching isotype. After 24 hours, cell free-supernatant was collected and subjected to IL-10 quantification by ELISA.



FIG. 33 shows that IL-22 neutralization with anti-IL-22 antibody alleviates anemia in wild type (wt) mice undergoing PhZ-induced stress erythropoiesis. FIG. 33 shows PB RBC numbers, Hb, and HCT in wt C57BL/6J mice undergoing PhZ-induced stress erythropoiesis treated with either an isotype control or anti-IL-22. * p<0.05, ** p<0.01, *** p<0.001.


For any figure showing a bar histogram, curve, or other data associated with a legend, the bars, curve, or other data presented from left to right for each indication correspond directly and in order to the boxes from top to bottom of the legend.





DETAILED DESCRIPTION OF THE INVENTION

The present invention is based, at least in part, on the discovery that down-regulators of IL-22 signaling can treat red blood cell disorders, such as anemia. In particular, it is described herein that hematopoietic cell-specific haploinsufficient deletion of Riok2 (Riok2f/+Vav1cre) leads to reduced erythroid progenitor frequency due to increased apoptosis and cell cycle arrest leading to anemia. Quantitative proteomics of Riok2f/+ Vav1cre erythroid progenitors identified elevated expression of multiple antimicrobial and alarmin proteins, an indication of immune system activation. An exclusive increase in IL-22 secretion from Riok2f/+Vav1cre CD4+ T cells after polarization towards different T cell lineages was observed. In addition and unexpectedly, it was discovered that the IL-22 receptor, IL-22RA1, is present on erythroid progenitor cells. Genetic blockade of IL-22 signaling by deletion of Il22 or Il22ra1 alleviated anemia in Riok2f/+Vav1cre mice. Additionally, erythroid cell-specific deletion of Il22ra1 in Riok2 sufficient phenylhydrazine-treated mice led to increased erythroid progenitor frequency and peripheral blood RBCs confirming a function of IL-22 signaling in stress-induced erythropoiesis. Further, treatment with a neutralizing monoclonal IL-22 antibody alleviated phenylhydrazine-driven anemia not only in Riok2f/+Vav1cre mice but also in wild-type mice indicating a broader role for targeting IL-22 in disorders with defective erythropoiesis. Levels of IL-22 and its downstream signaling effectors were increased in two independent cohorts of MDS patients and in a published large-scale sequencing study (Pellagatti et al. (2010) Leukemia 24:756-764) of CD34+cells from MDS patients. Moreover, patients with anemia secondary to chronic kidney disease (CKD) were also demonstrated herein to have elevated levels of IL-22 as compared to CKD patients with normal hematocrits. The results described herein demonstrate an unexpected role for IL-22 signaling in erythroid differentiation and provides therapeutic opportunities for reversing anemias, including stress-induced anemias, and MDS disorders. Down-regulators of IL-22 not only act against an increase of hepcidin from hepatocytes, but can promote differentiation of erythroid progenitors to red blood cells. These effects of down-regulators of IL-22 are useful for diagnosing, prognosing, and treating a variety of red blood cell disorders, such as anemia, since the effects include increasing the number of red blood cells in a subject.


Accordingly, the present invention provides methods of treating one or more red blood cell disorders (e.g., anemia) in a subject by administering to the subject an effective amount of a down-regulator of IL-22 signaling. The present invention also provides methods of promoting differentiation from an erythroid progenitor cell toward a mature red blood cell in a subject by administering to the subject an effective amount of a down-regulator of IL-22 signaling.


I. Definitions

The articles “a” and “an” are used herein to refer to one or to more than one (i.e. to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.


The term “altered amount” or “altered level” refers to increased or decreased copy number (e.g., germline and/or somatic) of a biomarker nucleic acid, e.g., increased or decreased expression level in a sample, as compared to the expression level or copy number of the biomarker nucleic acid in a control sample. The term “altered amount” of a biomarker also includes an increased or decreased protein level of a biomarker protein in a sample, as compared to the corresponding protein level in a normal, control sample. Furthermore, an altered amount of a biomarker protein may be determined by detecting posttranslational modification such as methylation status of the marker, which may affect the expression or activity of the biomarker protein.


The amount of a biomarker in a subject is “significantly” higher or lower than the normal amount of the biomarker, if the amount of the biomarker is greater or less, respectively, than the normal level by an amount greater than the standard error of the assay employed to assess amount, and preferably at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 300%, 350%, 400%, 500%, 600%, 700%, 800%, 900%, 1000% or than that amount. Alternately, the amount of the biomarker in the subject can be considered “significantly” higher or lower than the normal amount if the amount is at least about two, and preferably at least about three, four, or five times, higher or lower, respectively, than the normal amount of the biomarker. Such “significance” can also be applied to any other measured parameter described herein, such as for expression, inhibition, cytotoxicity, cell growth, and the like.


The term “altered activity” of a biomarker refers to an activity of the biomarker which is increased or decreased in a disease state, e.g., in a sample from a subject having MDS and/or an anemia, as compared to the activity of the biomarker in a normal, control sample. Altered activity of the biomarker may be the result of, for example, altered expression of the biomarker, altered protein level of the biomarker, altered structure of the biomarker, or, e.g., an altered interaction with other proteins involved in the same or different pathway as the biomarker or altered interaction with transcriptional activators or inhibitors.


The term “altered structure” of a biomarker refers to the presence of mutations or allelic variants within a biomarker nucleic acid or protein, e.g., mutations which affect expression or activity of the biomarker nucleic acid or protein, as compared to the normal or wild-type gene or protein. For example, mutations include, but are not limited to substitutions, deletions, or addition mutations. Mutations may be present in the coding or non-coding region of the biomarker nucleic acid.


The term “administering” is intended to include modes and routes of administration which allow an agent to perform its intended function. Examples of routes of administration for treatment of a body which can be used include injection (subcutaneous, intravenous, parenterally, intraperitoneally, intrathecal, etc.), oral, inhalation, and transdermal routes. The injection can be bolus injections or can be continuous infusion. Depending on the route of administration, the agent can be coated with or disposed in a selected material to protect it from natural conditions which may detrimentally affect its ability to perform its intended function. The agent may be administered alone, or in conjunction with a pharmaceutically acceptable carrier. The agent also may be administered as a prodrug, which is converted to its active form in vivo.


Unless otherwise specified herein, the terms “antibody” and “antibodies” broadly encompass naturally-occurring forms of antibodies (e.g., IgG, IgA, IgM, IgE) and recombinant antibodies such as single-chain antibodies, murine, chimeric, humanized, and human antibodies and multi-specific antibodies, as well as fragments and derivatives of all of the foregoing, which fragments and derivatives have at least an antigenic binding site. Antibody derivatives may comprise a protein or chemical moiety conjugated to an antibody.


In addition, intrabodies are well-known antigen-binding molecules having the characteristic of antibodies, but that are capable of being expressed within cells in order to bind and/or inhibit intracellular targets of interest (Chen et al. (1994) Human Gene Ther. 5:595-601). Methods are well-known in the art for adapting antibodies to target (e.g., inhibit) intracellular moieties, such as the use of single-chain antibodies (scFvs), modification of immunoglobulin VL domains for hyperstability, modification of antibodies to resist the reducing intracellular environment, generating fusion proteins that increase intracellular stability and/or modulate intracellular localization, and the like. Intracellular antibodies can also be introduced and expressed in one or more cells, tissues or organs of a multicellular organism, for example for prophylactic and/or therapeutic purposes (e.g., as a gene therapy) (see, at least PCT Publs. WO 08/020079, WO 94/02610, WO 95/22618, and WO 03/014960; U.S. Pat. No. 7,004,940; Cattaneo and Biocca (1997) Intracellular Antibodies: Development and Applications (Landes and Springer-Verlag publs.); Kontermann (2004) Methods 34:163-170; Cohen et al. (1998) Oncogene 17:2445-2456; Auf der Maur et al. (2001) FEBS Lett. 508:407-412; Shaki-Loewenstein et al. (2005) J. Immunol. Meth. 303:19-39).


The term “antibody” as used herein also includes an “antigen-binding portion” of an antibody (or simply “antibody portion”). The term “antigen-binding portion”, as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. 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 polypeptides (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; and Osbourn et al. 1998, Nature Biotechnology 16: 778). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. Any VH and VL sequences of specific scFv can be linked to human immunoglobulin constant region cDNA or genomic sequences, in order to generate expression vectors encoding complete IgG polypeptides or other isotypes. VH and VL can also be used in the generation of Fab, Fv or other fragments of immunoglobulins using either protein chemistry or recombinant DNA technology. 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).


Still further, an antibody or antigen-binding portion thereof may be part of larger immunoadhesion polypeptides, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides. Examples of such immunoadhesion polypeptides include use of the streptavidin core region to make a tetrameric scFv polypeptide (Kipriyanov, S. M. et al. (1995) Human Antibodies and Hybridomas 6:93-101) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv polypeptides (Kipriyanov, S. M. et al. (1994) Mol. Immunol. 31:1047-1058). Antibody portions, such as Fab and F(ab′)2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies. Moreover, antibodies, antibody portions and immunoadhesion polypeptides can be obtained using standard recombinant DNA techniques, as described herein.


Antibodies may be polyclonal or monoclonal; xenogeneic, allogeneic, or syngeneic; or modified forms thereof (e.g., humanized, chimeric, etc.). Antibodies may also be fully human. The terms “monoclonal antibodies” and “monoclonal antibody composition”, as used herein, refer to a population of antibody polypeptides that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope of an antigen, whereas the term “polyclonal antibodies” and “polyclonal antibody composition” refer to a population of antibody polypeptides that contain multiple species of antigen binding sites capable of interacting with a particular antigen. A monoclonal antibody composition typically displays a single binding affinity for a particular antigen with which it immunoreacts. In addition, antibodies can be “humanized,” which includes antibodies made by a non-human cell having variable and constant regions which have been altered to more closely resemble antibodies that would be made by a human cell. For example, by altering the non-human antibody amino acid sequence to incorporate amino acids found in human germline immunoglobulin sequences. The humanized antibodies encompassed by the present invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs. The term “humanized antibody,” as used herein, also includes antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.


A “blocking” antibody is one which inhibits or reduces at least one biological activity of the antigen(s) it binds. In certain embodiments, the blocking antibodies or fragments thereof described herein substantially or completely inhibit a given biological activity of the antigen(s). Blocking antibodies are alternatively referred to herein with the prefix “anti” with respect to a target of them (e.g., anti-IL-22 for an antibody that binds to IL-22 and down-regulates IL-22 signaling).


An “antagonist” is one which attenuates, decreases, or inhibits at least one biological activity of at least one protein, such as a receptor (e.g., AHR), described herein. In certain embodiments, the antagonist described herein substantially or completely attenuates or inhibits a given biological activity of at least one protein described herein.


Any one of the antibodies or fragments thereof disclosed herein may bind specifically to any one of the amino acid sequences disclosed in Table 1.


The term “biomarker” refers to a measurable entity of the present invention that has been determined to be indicative of elevated and/or activated IL-22 signaling. Representative, non-limiting examples are described herein, such as IL-22 itself (e.g., increased IL-22 mRNA or protein levels in a body fluid, such as blood, bone marrow fluid, peripheral blood Th22 T lymphocytes), and/or IL-22 pathway member, such as increased levels of alarmins (e.g., S100A8, S100A9, S100 A10, S100A11, Stat3 phosphorylation), IL-22 receptor (such as IL-22RA1, IL-10Rbeta, and heterodimers thereof), and other pathway members like Camp, Ngp, Ptgs2, Rab7A, and the like in a body fluid. Moreover, IL-22 itself is indicative of IL-22 signaling, such as blood, bone marrow fluid, peripheral blood Th22 T lymphocytes. IL-22 activation is downstream of the arylhydrocarbon receptor, which is also a relevant biomarker. Biomarkers can include, without limitation, nucleic acids and proteins, including those shown in the Tables, the Examples, the Figures, and otherwise described herein. As described herein, any relevant characteristic of a biomarker can be used, such as the copy number, amount, activity, location, modification (e.g., phosphorylation), and the like.









TABLE 1







Representative biomarkers useful according to methods encompassed


by the present invention; exemplary amino acid and nucleic


acid sequences of such biomarkers disclosed below.











Accession Numbers


Biomarker
Accession Numbers (Protein)
(cDNA; mRNA)





IL-22
NP_065386.1
NM_020525.5


IL-22 receptor, including IL-
AAG22073.1
AF286095.1


22RA1, and heterodimers thereof


IL-10Rbeta, and heterodimers
NP_000619.3
NM_000628.5


thereof


Arylhydrocarbon receptor
NP_001612.1
NM_001621.5


S100A8
NP_002955.2
NM_002964.5



NP_001306125.1
NM_001319196.1



NP_001306126.1
NM_001319197.1



NP_001306127.1
NM_001319198.2



NP_001306130.1
NM_001319201.2


S100A9
NP_002956.1
NM_002965.4


S100A10
NP_002957.1
NM_002966.3


S100A11
NP_005611.1
NM_005620.2


Phosphorylated Stat3
NP_644805.1
NM_139276.3



NP_003141.2
NM_003150.4



NP_998827.1
NM_213662.2



NP_001356441.1
NM_001369512.1



NP_001356442.1
NM_001369513.1



NP_001356443.1
NM_001369514.1



NP_001356446.1
NM_001369516.1



NP_001371913.1
NM_001369517.1



NP_001371914.1
NM_001369518.1



NP_001371915.1
NM_001369519.1



NP_001371916.1
NM_001369520.1



NP_001371917.1
NM_001384984.1



NP_001371918.1
NM_001384985.1



NP_001371919.1
NM_001384986.1



NP_001371920.1
NM_001384987.1



NP_001371921.1
NM_001384988.1



NP_001371922.1
NM_001384989.1



XP_016880462.1
NM_001384990.1



XP_024306664.1
NM_001384991.1




NM_001384992.1




NM_001384993.1




XM_017024973.2




XM_024450896.1


Camp
NP_004336.4
NM_004345.5


Ngp
NP_032720.2
NM_008694.2


Ptgs2
NP_000954.1
NM_000963.4


Rab7A
NP_004628.4
NM_004637.6

















SEQ ID NO: 8 Human Amino Acid Sequence IL-22 (NP_065386.1)



MAALQKSVSSFLMGTLATSCLLLLALLVQGGAAAPISSHCRLDKSNFQQPYITNRTFMLAKEASLADNNT





DVRLIGEKLFHGVSMSERCYLMKQVLNFTLEEVLFPQSDRFQPYMQEVVPFLARLSNRLSTCHIEGDDLH





IQRNVQKLKDTVKKLGESGEIKAIGELDLLFMSLRNACI





SEQ ID NO: 9 Human Nucleic Acid cDNA/mRNA Sequence IL-22 (NM_020525.5)


ACAAGCAGAATCTTCAGAACAGGTTCTCCTTCCCCAGTCACCAGTTGCTCGAGTTAGAATTGTCTGCAAT





GGCCGCCCTGCAGAAATCTGTGAGCTCTTTCCTTATGGGGACCCTGGCCACCAGCTGCCTCCTTCTCTTG





GCCCTCTTGGTACAGGGAGGAGCAGCTGCGCCCATCAGCTCCCACTGCAGGCTTGACAAGTCCAACTTCC





AGCAGCCCTATATCACCAACCGCACCTTCATGCTGGCTAAGGAGGCTAGCTTGGCTGATAACAACACAGA





CGTTCGTCTCATTGGGGAGAAACTGTTCCACGGAGTCAGTATGAGTGAGCGCTGCTATCTGATGAAGCAG





GTGCTGAACTTCACCCTTGAAGAAGTGCTGTTCCCTCAATCTGATAGGTTCCAGCCTTATATGCAGGAGG





TGGTGCCCTTCCTGGCCAGGCTCAGCAACAGGCTAAGCACATGTCATATTGAAGGTGATGACCTGCATAT





CCAGAGGAATGTGCAAAAGCTGAAGGACACAGTGAAAAAGCTTGGAGAGAGTGGAGAGATCAAAGCAATT





GGAGAACTGGATTTGCTGTTTATGTCTCTGAGAAATGCCTGCATTTGACCAGAGCAAAGCTGAAAAATGA





ATAACTAACCCCCTTTCCCTGCTAGAAATAACAATTAGATGCCCCAAAGCGATTTTTTTTAACCAAAAGG





AAGATGGGAAGCCAAACTCCATCATGATGGGTGGATTCCAAATGAACCCCTGCGTTAGTTACAAAGGAAA





CCAATGCCACTTTTGTTTATAAGACCAGAAGGTAGACTTTCTAAGCATAGATATTTATTGATAACATTTC





ATTGTAACTGGTGTTCTATACACAGAAAACAATTTATTTTTTAAATAATTGTCTTTTTCCATAAAAAAGA





TTACTTTCCATTCCTTTAGGGGAAAAAACCCCTAAATAGCTTCATGTTTCCATAATCAGTACTTTATATT





TATAAATGTATTTATTATTATTATAAGACTGCATTTTATTTATATCATTTTATTAATATGGATTTATTTA





TAGAAACATCATTCGATATTGCTACTTGAGTGTAAGGCTAATATTGATATTTATGACAATAATTATAGAG





CTATAACATGTTTATTTGACCTCAATAAACACTTGGATATCCTAA





SEQ ID NO: 10: Human Amino Acid Sequence IL-22 Receptor (AAG22073.1)


MRTLLTILTVGSLAAHAPEDPSDLLQHVKFQSSNFENILTWDSGPEGTPDTVYSIEYKTYGERDWVAKKG





CQRITRKSCNLTVETGNLTELYYARVTAVSAGGRSATKMTDRFSSLQHTTLKPPDVTCISKVRSIQMIVH





PTPTPIRAGDGHRLTLEDIFHDLFYHLELQVNRTYQMHLGGKQREYEFFGLTPDTEFLGTIMICVPTWAK





ESAPYMCRVKTLPDRTWTYSFSGAFLFSMGFLVAVLCYLSYRYVTKPPAPPNSLNVQRVLTFQPLRFIQE





HVLIPVEDLSGPSSLAQPVQYSQIRVSGPREPAGAPQRHSLSEITYLGQPDISILQPSNVPPPQILSPLS





YAPNAAPEVGPPSYAPQVTPEAQFPFYAPQAISKVQPSSYAPQATPDSWPPSYGVCMEGSGKDSPTGTLS





SPKHLRPKGQLQKEPPAGSCMLGGLSLQEVTSLAMEESQEAKSLHQPLGICTDRTSDPNVLHSGEEGTPQ





YLKGQLPLLSSVQIEGHPMSLPLQPPSGPCSPSDQGPSPWGLLESLVCPKDEAKSPAPETSDLEQPTELD





SLFRGLALTVQWES





SEQ ID NO: 11: Human Nucleic Acid cDNA/mRNA Sequence IL-22 Receptor


(AF286095.1)


GGGAGGGCTCTGTGCCAGCCCCGATGAGGACGCTGCTGACCATCTTGACTGTGGGATCCCTGGCTGCTCA





CGCCCCTGAGGACCCCTCGGATCTGCTCCAGCACGTGAAATTCCAGTCCAGCAACTTTGAAAACATCCTG





ACGTGGGACAGCGGGCCAGAGGGCACCCCAGACACGGTCTACAGCATCGAGTATAAGACGTACGGAGAGA





GGGACTGGGTGGCAAAGAAGGGCTGTCAGCGGATCACCCGGAAGTCCTGCAACCTGACGGTGGAGACGGG





CAACCTCACGGAGCTCTACTATGCCAGGGTCACCGCTGTCAGTGCGGGAGGCCGGTCAGCCACCAAGATG





ACTGACAGGTTCAGCTCTCTGCAGCACACTACCCTCAAGCCACCTGATGTGACCTGTATCTCCAAAGTGA





GATCGATTCAGATGATTGTTCATCCTACCCCCACGCCAATCCGTGCAGGCGATGGCCACCGGCTAACCCT





GGAAGACATCTTCCATGACCTGTTCTACCACTTAGAGCTCCAGGTCAACCGCACCTACCAAATGCACCTT





GGAGGGAAGCAGAGAGAATATGAGTTCTTCGGCCTGACCCCTGACACAGAGTTCCTTGGCACCATCATGA





TTTGCGTTCCCACCTGGGCCAAGGAGAGTGCCCCCTACATGTGCCGAGTGAAGACACTGCCAGACCGGAC





ATGGACCTACTCCTTCTCCGGAGCCTTCCTGTTCTCCATGGGCTTCCTCGTCGCAGTACTCTGCTACCTG





AGCTACAGATATGTCACCAAGCCGCCTGCACCTCCCAACTCCCTGAACGTCCAGCGAGTCCTGACTTTCC





AGCCGCTGCGCTTCATCCAGGAGCACGTCCTGATCCCTGTCTTTGACCTCAGCGGCCCCAGCAGTCTGGC





CCAGCCTGTCCAGTACTCCCAGATCAGGGTGTCTGGACCCAGGGAGCCCGCAGGAGCTCCACAGCGGCAT





AGCCTGTCCGAGATCACCTACTTAGGGCAGCCAGACATCTCCATCCTCCAGCCCTCCAACGTGCCACCTC





CCCAGATCCTCTCCCCACTGTCCTATGCCCCAAACGCTGCCCCTGAGGTCGGGCCCCCATCCTATGCACC





TCAGGTGACCCCCGAAGCTCAATTCCCATTCTACGCCCCACAGGCCATCTCTAAGGTCCAGCCTTCCTCC





TATGCCCCTCAAGCCACTCCGGACAGCTGGCCTCCCTCCTATGGGGTATGCATGGAAGGTTCTGGCAAAG





ACTCCCCCACTGGGACACTTTCTAGTCCTAAACACCTTAGGCCTAAAGGTCAGCTTCAGAAAGAGCCACC





AGCTGGAAGCTGCATGTTAGGTGGCCTTTCTCTGCAGGAGGTGACCTCCTTGGCTATGGAGGAATCCCAA





GAAGCAAAATCATTGCACCAGCCCCTGGGGATTTGCACAGACAGAACATCTGACCCAAATGTGCTACACA





GTGGGGAGGAAGGGACACCACAGTACCTAAAGGGCCAGCTCCCCCTCCTCTCCTCAGTCCAGATCGAGGG





CCACCCCATGTCCCTCCCTTTGCAACCTCCTTCCGGTCCATGTTCCCCCTCGGACCAAGGTCCAAGTCCC





TGGGGCCTGCTGGAGTCCCTTGTGTGTCCCAAGGATGAAGCCAAGAGCCCAGCCCCTGAGACCTCAGACC





TGGAGCAGCCCACAGAACTGGATTCTCTTTTCAGAGGCCTGGCCCTGACTGTGCAGTGGGAGTCCTGAGG





GGAATGGGAAAGGCTTGGTGCTTCCTCCCTGTCCCTACCCAGTGTCACATCCTTGGCTGTCAATCCCATG





CCTGCCCATGCCACACACTCTGCGATCTGGCCTCAGACGGGTGCCCTTGAGAGAAGCAGAGGGAGTGGCA





TGCAGGGCCCCTGCCATGGGTGCGCTCCTCACCGGAACAAAGCAGCATGATAAGGACTGCAGCGGGGGAG





CTCTGGGGAGCAGCTTGTGTAGACAAGCGCGTGCTCGCTGAGCCCTGCAAGGCAGAAATGACAGTGCAAG





GAGGAAATGCAGGGAAACTCCCGAGGTCCAGAGCCCCACCTCCTAACACCATGGATTCAAAGTGCTCAGG





GAATTTGCCTCTCCTTGCCCCATTCCTGGCCAGTTTCACAATCTAGCTCGACAGAGCATGAGGCCCCTGC





CTCTTCTGTCATTGTTCAAAGGTGGGAAGAGAGCCTGGAAAAGAACCAGGCCTGGAAAAGAACCAGAAGG





AGGCTGGGCAGAACCAGAACAACCTGCACTTCTGCCAAGGCCAGGGCCAGCAGGACGGCAGGACTCTAGG





GAGGGGTGTGGCCTGCAGCTCATTCCCAGCCAGGGCAACTGCCTGACGTTGCACGATTTCAGCTTCATTC





CTCTGATAGAACAAAGCGAAATGCAGGTCCACCAGGGAGGGAGACACACAAGCCTTTTCTGCAGGCAGGA





GTTTCAGACCCTATCCTGAGAATGGGGTTTGAAAGGAAGGTGAGGGCTGTGGCCCCTGGACGGGTACAAT





AACACACTGTACTGATGTCACAACTTTGCAAGCTCTGCCTTGGGTTCAGCCCATCTGGGCTCAAATTCCA





GCCTCACCACTCACAAGCTGTGTGACTTCAAACAAATGAAATCAGTGCCCAGAACCTCGGTTTCCTCATC





TGTAATGTGGGGATCATAACACCTACCTCATGGAGTTGTGGTGAAGATGAAATGAAGTCATGTCTTTAAA





GTGCTTAATAGTGCCTGGTACATGGGCAGTGCCCAATAAACGGTAGCTATTTAAAAAAAAAAAAA





SEQ ID NO: 12: Human Amino Acid Sequence IL-10 Receptor Beta (NP_000619.3)


MAWSLGSWLGGCLLVSALGMVPPPENVRMNSVNFKNILQWESPAFAKGNLTFTAQYLSYRIFQDKCMNTTLTE





CDFSSLSKYGDHTLRVRAEFADEHSDWVNITFCPVDDTIIGPPGMQVEVLADSLHMRFLAPKIENEYETWTMK





NVYNSWTYNVQYWKNGTDEKFQITPQYDFEVLRNLEPWTTYCVQVRGFLPDRNKAGEWSEPVCEQTTHDETVP





SWMVAVILMASVFMVCLALLGCFALLWCVYKKTKYAFSPRNSLPQHLKEFLGHPHHNTLLFFSFPLSDENDVE





DKLSVIAEDSESGKQNPGDSCSLGTPPGQGPQS





SEQ ID NO: 13: Human Nucleic Acid cDNA/mRNA Sequence IL-10 Receptor Beta


(NM_000628.5)


ATCTCCGCTGGTTCCCGGAAGCCGCCGCGGACAAGCTCTCCCGGGCGCGGGCGGGGGTCGTGTGCTTGGAGGA





AGCCGCGGAACCCCCAGCGTCCGTCCATGGCGTGGAGCCTTGGGAGCTGGCTGGGTGGCTGCCTGCTGGTGTC





AGCATTGGGAATGGTACCACCTCCCGAAAATGTCAGAATGAATTCTGTTAATTTCAAGAACATTCTACAGTGG





GAGTCACCTGCTTTTGCCAAAGGGAACCTGACTTTCACAGCTCAGTACCTAAGTTATAGGATATTCCAAGATA





AATGCATGAATACTACCTTGACGGAATGTGATTTCTCAAGTCTTTCCAAGTATGGTGACCACACCTTGAGAGT





CAGGGCTGAATTTGCAGATGAGCATTCAGACTGGGTAAACATCACCTTCTGTCCTGTGGATGACACCATTATT





GGACCCCCTGGAATGCAAGTAGAAGTACTTGCTGATTCTTTACATATGCGTTTCTTAGCCCCTAAAATTGAGA





ATGAATACGAAACTTGGACTATGAAGAATGTGTATAACTCATGGACTTATAATGTGCAATACTGGAAAAACGG





TACTGATGAAAAGTTTCAAATTACTCCCCAGTATGACTTTGAGGTCCTCAGAAACCTGGAGCCATGGACAACT





TATTGTGTTCAAGTTCGAGGGTTTCTTCCTGATCGGAACAAAGCTGGGGAATGGAGTGAGCCTGTCTGTGAGC





AAACAACCCATGACGAAACGGTCCCCTCCTGGATGGTGGCCGTCATCCTCATGGCCTCGGTCTTCATGGTCTG





CCTGGCACTCCTCGGCTGCTTCGCCTTGCTGTGGTGCGTTTACAAGAAGACAAAGTACGCCTTCTCCCCTAGG





AATTCTCTTCCACAGCACCTGAAAGAGTTTTTGGGCCATCCTCATCATAACACACTTCTGTTTTTCTCCTTTC





CATTGTCGGATGAGAATGATGTTTTTGACAAGCTAAGTGTCATTGCAGAAGACTCTGAGAGCGGCAAGCAGAA





TCCTGGTGACAGCTGCAGCCTCGGGACCCCGCCTGGGCAGGGGCCCCAAAGCTAGGCTCTGAGAAGGAAACAC





ACTCGGCTGGGCACAGTGACGTACTCCATCTCACATCTGCCTCAGTGAGGGATCAGGGCAGCAAACAAGGGCC





AAGACCATCTGAGCCAGCCCCACATCTAGAACTCCCAGACCCTGGACTTAGCCACCAGAGAGCTACATTTTAA





AGGCTGTCTTGGCAAAAATACTCCATTTGGGAACTCACTGCCTTATAAAGGCTTTCATGATGTTTTCAGAAGT





TGGCCACTGAGAGTGTAATTTTCAGCCTTTTATATCACTAAAATAAGATCATGTTTTAATTGTGAGAAACAGG





GCCGAGCACAGTGGCTCACGCCTGTAATACCAGCACCTTAGAGGTCGAGGCAGGCGGATCACTTGAGGTCAGG





AGTTCAAGACCAGCCTGGCCAATATGGTGAAACCCAGTCTCTACTAAAAATACAAAAATTAGCTAGGCATGAT





GGCGCATGCCTATAATCCCAGCTACTCGAGTGCCTGAGGCAGGAGAATTGCATGAACCCGGGAGGAGGAGGAG





GAGGTTGCAGTGAGCCGAGATAGCGGCACTGCACTCCAGCCTGGGTGACAAAGTGAGACTCCATCTCAAAAAA





AAAAAAAAAAAAAATTGTGAGAAACAGAAATACTTAAAATGAGGAATAAGAATGGAGATGTTACATCTGGTAG





ATGTAACATTCTACCAGATTATGGATGGACTGATCTGAAAATCGACCTCAACTCAAGGGTGGTCAGCTCAATG





CTACACAGAGCACGGACTTTTGGATTCTTTGCAGTACTTTGAATTTATTTTTCTACCTATATATGTTTTATAT





GCTGCTGGTGCTCCATTAAAGTTTTACTCTGTGTTGCACTATA





SEQ ID NO: 14 Human Amino Acid Sequence Arylhydrocarbon receptor


(NP_001612.1)


MNSSSANITYASRKRRKPVQKTVKPIPAEGIKSNPSKRHRDRINTELDRLASLLPFPQDVINKLDKLSVL





RLSVSYLRAKSFFDVALKSSPTERNGGQDNCRAANFREGLNLQEGEFLLQALNGFVLVVTTDALVFYASS





TIQDYLGFQQSDVIHQSVYELIHTEDRAEFQRQLHWALNPSQCTESGQGIEEATGLPQTVVCYNPDQIPP





ENSPLMERCFICRLRCLLDNSSGFLAMNFQGKLKYLHGQKKKGKDGSILPPQLALFAIATPLQPPSILEI





RTKNFIFRTKHKLDFTPIGCDAKGRIVLGYTEAELCTRGSGYQFIHAADMLYCAESHIRMIKTGESGMIV





FRLLTKNNRWTWVQSNARLLYKNGRPDYIIVTQRPLTDEEGTEHLRKRNTKLPFMFTTGEAVLYEATNPF





PAIMDPLPLRTKNGTSGKDSATTSTLSKDSLNPSSLLAAMMQQDESIYLYPASSTSSTAPFENNFFNESM





NECRNWQDNTAPMGNDTILKHEQIDQPQDVNSFAGGHPGLFQDSKNSDLYSIMKNLGIDFEDIRHMQNEK





FFRNDFSGEVDERDIDLTDEILTYVQDSLSKSPFIPSDYQQQQSLALNSSCMVQEHLHLEQQQQHHQKQV





VVEPQQQLCQKMKHMQVNGMFENWNSNQFVPFNCPQQDPQQYNVFTDLHGISQEFPYKSEMDSMPYTQNF





ISCNQPVLPQHSKCTELDYPMGSFEPSPYPTTSSLEDFVTCLQLPENQKHGLNPQSAIITPQTCYAGAVS





MYQCQPEPQHTHVGQMQYNPVLPGQQAFLNKFQNGVLNETYPAELNNINNTQTTTHLQPLHHPSEARPFP





DLTSSGFL





SEQ ID NO: 15 Human Nucleic Acid cDNA/mRNA Sequence Arylhydrocarbon


receptor (NM_001621.5)


AGTGGCTGGGGAGTCCCGTCGACGCTCTGTTCCGAGAGCGTGCCCCGGACCGCCAGCTCAGAACAGGGGC





AGCCGTGTAGCCGAACGGAAGCTGGGAGCAGCCGGGACTGGTGGCCCGCGCCCGAGCTCCGCAGGCGGGA





AGCACCCTGGATTTAGGAAGTCCCGGGAGCAGCGCGGCGGCACCTCCCTCACCCAAGGGGCCGCGGCGAC





GGTCACGGGGCGCGGCGCCACCGTGAGCGACCCAGGCCAGGATTCTAAATAGACGGCCCAGGCTCCTCCT





CCGCCCGGGCCGCCTCACCTGCGGGCATTGCCGCGCCGCCTCCGCCGGTGTAGACGGCACCTGCGCCGCC





TTGCTCGCGGGTCTCCGCCCCTCGCCCACCCTCACTGCGCCAGGCCCAGGCAGCTCACCTGTACTGGCGC





GGGCTGCGGAAGCCTGCGTGAGCCGAGGCGTTGAGGCGCGGCGCCCACGCCACTGTCCCGAGAGGACGCA





GGTGGAGCGGGCGCGGCTTCGCGGAACCCGGCGCCGGCCGCCGCAGTGGTCCCAGCCTACACCGGGTTCC





GGGGACCCGGCCGCCAGTGCCCGGGGAGTAGCCGCCGCCGTCGGCTGGGCACCATGAACAGCAGCAGCGC





CAACATCACCTACGCCAGTCGCAAGCGGCGGAAGCCGGTGCAGAAAACAGTAAAGCCAATCCCAGCTGAA





GGAATCAAGTCAAATCCTTCCAAGCGGCATAGAGACCGACTTAATACAGAGTTGGACCGTTTGGCTAGCC





TGCTGCCTTTCCCACAAGATGTTATTAATAAGTTGGACAAACTTTCAGTTCTTAGGCTCAGCGTCAGTTA





CCTGAGAGCCAAGAGCTTCTTTGATGTTGCATTAAAATCCTCCCCTACTGAAAGAAACGGAGGCCAGGAT





AACTGTAGAGCAGCAAATTTCAGAGAAGGCCTGAACTTACAAGAAGGAGAATTCTTATTACAGGCTCTGA





ATGGCTTTGTATTAGTTGTCACTACAGATGCTTTGGTCTTTTATGCTTCTTCTACTATACAAGATTATCT





AGGGTTTCAGCAGTCTGATGTCATACATCAGAGTGTATATGAACTTATCCATACCGAAGACCGAGCTGAA





TTTCAGCGTCAGCTACACTGGGCATTAAATCCTTCTCAGTGTACAGAGTCTGGACAAGGAATTGAAGAAG





CCACTGGTCTCCCCCAGACAGTAGTCTGTTATAACCCAGACCAGATTCCTCCAGAAAACTCTCCTTTAAT





GGAGAGGTGCTTCATATGTCGTCTAAGGTGTCTGCTGGATAATTCATCTGGTTTTCTGGCAATGAATTTC





CAAGGGAAGTTAAAGTATCTTCATGGACAGAAAAAGAAAGGGAAAGATGGATCAATACTTCCACCTCAGT





TGGCTTTGTTTGCGATAGCTACTCCACTTCAGCCACCATCCATACTTGAAATCCGGACCAAAAATTTTAT





CTTTAGAACCAAACACAAACTAGACTTCACACCTATTGGTTGTGATGCCAAAGGAAGAATTGTTTTAGGA





TATACTGAAGCAGAGCTGTGCACGAGAGGCTCAGGTTATCAGTTTATTCATGCAGCTGATATGCTTTATT





GTGCCGAGTCCCATATCCGAATGATTAAGACTGGAGAAAGTGGCATGATAGTTTTCCGGCTTCTTACAAA





AAACAACCGATGGACTTGGGTCCAGTCTAATGCACGCCTGCTTTATAAAAATGGAAGACCAGATTATATC





ATTGTAACTCAGAGACCACTAACAGATGAGGAAGGAACAGAGCATTTACGAAAACGAAATACGAAGTTGC





CTTTTATGTTTACCACTGGAGAAGCTGTGTTGTATGAGGCAACCAACCCTTTTCCTGCCATAATGGATCC





CTTACCACTAAGGACTAAAAATGGCACTAGTGGAAAAGACTCTGCTACCACATCCACTCTAAGCAAGGAC





TCTCTCAATCCTAGTTCCCTCCTGGCTGCCATGATGCAACAAGATGAGTCTATTTATCTCTATCCTGCTT





CAAGTACTTCAAGTACTGCACCTTTTGAAAACAACTTTTTCAACGAATCTATGAATGAATGCAGAAATTG





GCAAGATAATACTGCACCGATGGGAAATGATACTATCCTGAAACATGAGCAAATTGACCAGCCTCAGGAT





GTGAACTCATTTGCTGGAGGTCACCCAGGGCTCTTTCAAGATAGTAAAAACAGTGACTTGTACAGCATAA





TGAAAAACCTAGGCATTGATTTTGAAGACATCAGACACATGCAGAATGAAAAATTTTTCAGAAATGATTT





TTCTGGTGAGGTTGACTTCAGAGACATTGACTTAACGGATGAAATCCTGACGTATGTCCAAGATTCTTTA





AGTAAGTCTCCCTTCATACCTTCAGATTATCAACAGCAACAGTCCTTGGCTCTGAACTCAAGCTGTATGG





TACAGGAACACCTACATCTAGAACAGCAACAGCAACATCACCAAAAGCAAGTAGTAGTGGAGCCACAGCA





ACAGCTGTGTCAGAAGATGAAGCACATGCAAGTTAATGGCATGTTTGAAAATTGGAACTCTAACCAATTC





GTGCCTTTCAATTGTCCACAGCAAGACCCACAACAATATAATGTCTTTACAGACTTACATGGGATCAGTC





AAGAGTTCCCCTACAAATCTGAAATGGATTCTATGCCTTATACACAGAACTTTATTTCCTGTAATCAGCC





TGTATTACCACAACATTCCAAATGTACAGAGCTGGACTACCCTATGGGGAGTTTTGAACCATCCCCATAC





CCCACTACTTCTAGTTTAGAAGATTTTGTCACTTGTTTACAACTTCCTGAAAACCAAAAGCATGGATTAA





ATCCACAGTCAGCCATAATAACTCCTCAGACATGTTATGCTGGGGCCGTGTCGATGTATCAGTGCCAGCC





AGAACCTCAGCACACCCACGTGGGTCAGATGCAGTACAATCCAGTACTGCCAGGCCAACAGGCATTTTTA





AACAAGTTTCAGAATGGAGTTTTAAATGAAACATATCCAGCTGAATTAAATAACATAAATAACACTCAGA





CTACCACACATCTTCAGCCACTTCATCATCCGTCAGAAGCCAGACCTTTTCCTGATTTGACATCCAGTGG





ATTCCTGTAATTCCAAGCCCAATTTTGACCCTGGTTTTTGGATTAAATTAGTTTGTGAAGGATTATGGAA





AAATAAAACTGTCACTGTTGGACGTCAGCAAGTTCACATGGAGGCATTGATGCATGCTATTCACAATTAT





TCCAAACCAAATTTTAATTTTTGCTTTTAGAAAAGGGAGTTTAAAAATGGTATCAAAATTACATATACTA





CAGTCAAGATAGAAAGGGTGCTGCCACGGAGTGGTGAGGTACCGTCTACATTTCACATTATTCTGGGCAC





CACAAAATATACAAAACTTTATCAGGGAAACTAAGATTCTTTTAAATTAGAAAATATTCTCTATTTGAAT





TATTTCTGTCACAGTAAAAATAAAATACTTTGAGTTTTGAGCTACTGGATTCTTATTAGTTCCCCAAATA





CAAAGTTAGAGAACTAAACTAGTTTTTCCTATCATGTTAACCTCTGCTTTTATCTCAGATGTTAAAATAA





ATGGTTTGGTGCTTTTTATAAAAAGATAATCTCAGTGCTTTCCTCCTTCACTGTTTCATCTAAGTGCCTC





ACATTTTTTTCTACCTATAACACTCTAGGATGTATATTTTATATAAAGTATTCTTTTTCTTTTTTAAATT





AATATCTTTCTGCACACAAATATTATTTGTGTTTCCTAAATCCAACCATTTTCATTAATTCAGGCATATT





TTAACTCCACTGCTTACCTACTTTCTTCAGGTAAAGGGCAAATAATGATCGAAAAAATAATTATTTATTA





CATAATTTAGTTGTTTCTAGACTATAAATGTTGCTATGTGCCTTATGTTGAAAAAATTTAAAAGTAAAAT





GTCTTTCCAAATTATTTCTTAATTATTATAAAAATATTAAGACAATAGCACTTAAATTCCTCAACAGTGT





TTTCAGAAGAAATAAATATACCACTCTTTACCTTTATTGATATCTCCATGATGATAGTTGAATGTTGCAA





TGTGAAAAATCTGCTGTTAACTGCAACCTTGTTTATTAAATTGCAAGAAGCTTTATTTCTAGCTTTTTAA





TTAAGCAAAGCACCCATTTCAATGTGTATAAATTGTCTTTAAAAACTGTTTTAGACCTATAATCCTTGAT





AATATATTGTGTTGACTTTATAAATTTCGCTTCTTAGAACAGTGGAAACTATGTGTTTTTCTCATATTTG





AGGAGTGTTAAGATTGCAGATAGCAAGGTTTGGTGCAAAGTATTGTAATGAGTGAATTGAATGGTGCATT





GTATAGATATAATGAACAAAATTATTTGTAAGATATTTGCAGTTTTTCATTTTAAAAAGTCCATACCTTA





TATATGCACTTAATTTGTTGGGGCTTTACATACTTTATCAATGTGTCTTTCTAAGAAATCAAGTAATGAA





TCCAACTGCTTAAAGTTGGTATTAATAAAAAGACAACCACATAGTTCGTTTACCTTCAAACTTTAGGTTT





TTTTAATGATATACTGATCTTCATTACCAATAGGCAAATTAATCACCCTACCAACTTTACTGTCCTAACA





TGGTTTAAAAGAAAAAATGACACCATCTTTTATTCTTTTTTTTTTTTTTTTTTGAGAGAGAGTCTTACTC





TGCCGCCCAAACTGGAGTGCAGTGGCACAATCTTGGCTCACTGCAACCTCTACCTCCTGGGTTCAAGTGA





TTCTCTTGCCTCAGCCTCCCGAGTTGCTGGGATTACAGGCATGTGCCACCATGCCCAGCTAATTTTTGTA





TTTTTAGTAGAAACGGGTTTCACCATGTTGGCCAGACTGGTCTCAAACTCCTGACCTCAGGTGAGCCTCC





CACCTTGGCCTCCCAAAGTGCTGGGATTACAGGCGTGAGCCACTGCATTCAGCTCTTCTTTTCTTTAGAT





ATGAGAGCTGAAGAGCTTAGACACATTTTGCATGTATTATTTGAAAATCTGATGGAATCCCAAACTGAGA





TGTATTAAAATACAATTTTTGGCCGGGTGCAGTGGCTCACGCCTGTAATCCCAGCACTTGGGGAGGGCGA





GGAGGGTGGATCACGAGGTCAAGAGATGGAGACCATCCTGACCAACATGGTGAAACCCTGTCTCTACTAA





AAATACAGAAATTAGCTGGGCATGGTGGCGTGAGCCTGTAGTCCTAGCTACTCAGGAGGCTGAGGCAGGA





GAATAGCCTGAACCTGGGAATCGGAGGTTGCAGAGCCAAGATCGCCCCACTGCACTCCAGCCTGGCAATA





GACCGAGACTCCGTCTCCAAAAAAAAAAAAAATACAATTTTTATTTCTTTTACTTTTTTTAGTAAGTTAA





TGTATATAAAAATGGCTTCGGACAAAATATCTCTGAGTTCTGTGTATTTTCAGTCAAAACTTTAAACCTG





TAGAATCAATTTAAGTGTTGGAAAAAATTTGTCTGAAACATTTCATAATTTGTTTCCAGCATGAGGTATC





TAAGGATTTAGACCAGAGGTCTAGATTAATACTCTATTTTTACATTTAAACCTTTTATTATAAGTCTTAC





ATAAACCATTTTTGTTACTCTCTTCCACATGTTACTGGATAAATTGTTTAGTGGAAAATAGGCTTTTTAA





TCATGAATATGATGACAATCAGTTATACAGTTATAAAATTAAAAGTTTGAAAAGCAATATTGTATATTTT





TATCTATATAAAATAACTAAAATGTATCTAAGAATAATAAAATCACGTTAAACCAAATACACGTTTGTCT





GTATTGTTAAGTGCCAAACAAAGGATACTTAGTGCACTGCTACATTGTGGGATTTATTTCTAGATGATGT





GCACATCTAAGGATATGGATGTGTCTAATTTAGTCTTTTCCTGTACCAGGTTTTTCTTACAATACCTGAA





GACTTACCAGTATTCTAGTGTATTATGAAGCTTTCAACATTACTATGCACAAACTAGTGTTTTTCGATGT





TACTAAATTTTAGGTAAATGCTTTCATGGCTTTTTTCTTCAAAATGTTACTGCTTACATATATCATGCAT





AGATTTTTGCTTAAAGTATGATTTATAATATCCTCATTATCAAAGTTGTATACAATAATATATAATAAAA





SEQ ID NO: 16 Human Amino Acid Sequence S100A8 (NP_001306125.1)


MSLVSCLSEDLKVLFFRWGKSVGIMLTELEKALNSIIDVYHKYSLIKGNFHAVYRDDLKKLLETECPQYI





RKKGADVWFKELDINTDGAVNFQEFLILVI KMGVAAHKKSHEESHKE





SEQ ID NO: 17 Human Nucleic Acid cDNA/mRNA Sequence S100A8


(NM_001319196.1)


GAGAAACCAGAGACTGTAGCAACTCTGGCAGGGAGAAGCTGTCTCTGATGGCCTGAAGCTGTGGGCAGCT





GGCCAAGCCTAACCGCTATAAAAAGGAGCTGCCTCTCAGCCCTGCATGTCTCTTGTCAGCTGTCTTTCAG





AAGACCTGAAGGTTCTGTTTTTCAGGTGGGGCAAGTCCGTGGGCATCATGTTGACCGAGCTGGAGAAAGC





CTTGAACTCTATCATCGACGTCTACCACAAGTACTCCCTGATAAAGGGGAATTTCCATGCCGTCTACAGG





GATGACCTGAAGAAATTGCTAGAGACCGAGTGTCCTCAGTATATCAGGAAAAAGGGTGCAGACGTCTGGT





TCAAAGAGTTGGATATCAACACTGATGGTGCAGTTAACTTCCAGGAGTTCCTCATTCTGGTGATAAAGAT





GGGCGTGGCAGCCCACAAAAAAAGCCATGAAGAAAGCCACAAAGAGTAGCTGAGTTACTGGGCCCAGAGG





CTGGGCCCCTGGACATGTACCTGCAGAATAATAAAGTCATCAATACCTCAAAAAAAAAA





SEQ ID NO: 18 Human Amino Acid Sequence S100A9 (NP_002956.1)


MTCKMSQLERNIETIINTFHQYSVKLGHPDTLNQGEFKELVRKDLQNFLKKENKNEKVIEHIMEDLDTNA





DKQLSFEEFIMLMARLTWASHEKMHEGDEGPGHHHKPGLGEGTP





SEQ ID NO: 19 Human Nucleic Acid cDNA/mRNA Sequence S100A9 (NM_002965.4)


AAACACTCTGTGTGGCTCCTCGGCTTTGACAGAGTGCAAGACGATGACTTGCAAAATGTCGCAGCTGGAA





CGCAACATAGAGACCATCATCAACACCTTCCACCAATACTCTGTGAAGCTGGGGCACCCAGACACCCTGA





ACCAGGGGGAATTCAAAGAGCTGGTGCGAAAAGATCTGCAAAATTTTCTCAAGAAGGAGAATAAGAATGA





AAAGGTCATAGAACACATCATGGAGGACCTGGACACAAATGCAGACAAGCAGCTGAGCTTCGAGGAGTTC





ATCATGCTGATGGCGAGGCTAACCTGGGCCTCCCACGAGAAGATGCACGAGGGTGACGAGGGCCCTGGCC





ACCACCATAAGCCAGGCCTCGGGGAGGGCACCCCCTAAGACCACAGTGGCCAAGATCACAGTGGCCACGG





CCACGGCCACAGTCATGGTGGCCACGGCCACAGCCACTAATCAGGAGGCCAGGCCACCCTGCCTCTACCC





AACCAGGGCCCCGGGGCCTGTTATGTCAAACTGTCTTGGCTGTGGGGCTAGGGGCTGGGGCCAAATAAAG





TCTCTTCCTCCAA





SEQ ID NO: 20 Human Amino Acid Sequence S100A10 (NP_002957.1)


MPSQMEHAMETMMFTFHKFAGDKGYLTKEDLRVLMEKEFPGFLENQKDPLAVDKIMKDLDQCRDGKVGFQ





SFFSLIAGLTIACNDYFVVHMKQKGKK





SEQ ID NO: 21 Human Nucleic Acid cDNA/mRNA Sequence S100A10 (NM_002966.3)


ACCCACCCGCCGCACGTACTAAGGAAGGCGCACAGCCCGCCGCGCTCGCCTCTCCGCCCCGCGTCCAGCT





CGCCCAGCTCGCCCAGCGTCCGCCGCGCCTCGGCCAAGGCTTCAACGGACCACACCAAAATGCCATCTCA





AATGGAACACGCCATGGAAACCATGATGTTTACATTTCACAAATTCGCTGGGGATAAAGGCTACTTAACA





AAGGAGGACCTGAGAGTACTCATGGAAAAGGAGTTCCCTGGATTTTTGGAAAATCAAAAAGACCCTCTGG





CTGTGGACAAAATAATGAAGGACCTGGACCAGTGTAGAGATGGCAAAGTGGGCTTCCAGAGCTTCTTTTC





CCTAATTGCGGGCCTCACCATTGCATGCAATGACTATTTTGTAGTACACATGAAGCAGAAGGGAAAGAAG





TAGGCAGAAATGAGCAGTTCGCTCCTCCCTGATAAGAGTTGTCCCAAAGGGTCGCTTAAGGAATCTGCCC





CACAGCTTCCCCCATAGAAGGATTTCATGAGCAGATCAGGACACTTAGCAAATGTAAAAATAAAATCTAA





CTCTCATTTGACAAGCAGAGAAAGAAAAGTTAAATACCAGATAAGCTTTTGATTTTTGTATTGTTTGCAT





CCCCTTGCCCTCAATAAATAAAGTTCTTTTTTAGTTCCAAA





SEQ ID NO: 22 Human Amino Acid Sequence S100A11 (NP_005611.1)


MAKISSPTETERCIESLIAVFQKYAGKDGYNYTLSKTEFLSFMNTELAAFTKNQKDPGVLDRMMKKLDTN





SDGQLDFSEFLNLIGGLAMACHDSFLKAVPSQKRT





SEQ ID NO: 23 Human Nucleic Acid cDNA/mRNA Sequence S100A11 (NM_005620.2)


GAGGAGAGGCTCCAGACCCGCACGCCGCGCGCACAGAGCTCTCAGCGCCGCTCCCAGCCACAGCCTCCCG





CGCCTCGCTCAGCTCCAACATGGCAAAAATCTCCAGCCCTACAGAGACTGAGCGGTGCATCGAGTCCCTG





ATTGCTGTCTTCCAGAAGTATGCTGGAAAGGATGGTTATAACTACACTCTCTCCAAGACAGAGTTCCTAA





GCTTCATGAATACAGAACTAGCTGCCTTCACAAAGAACCAGAAGGACCCTGGTGTCCTTGACCGCATGAT





GAAGAAACTGGACACCAACAGTGATGGTCAGCTAGATTTCTCAGAATTTCTTAATCTGATTGGTGGCCTA





GCTATGGCTTGCCATGACTCCTTCCTCAAGGCTGTCCCTTCCCAGAAGCGGACCTGAGGACCCCTTGGCC





CTGGCCTTCAAACCCACCCCCTTTCCTTCCAGCCTTTCTGTCATCATCTCCACAGCCCACCCATCCCCTG





AGCACACTAACCACCTCATGCAGGCCCCACCTGCCAATAGTAATAAAGCAATGTCACTTTTTTAAAACAT





GAA





SEQ ID NO: 24 Human Amino Acid Sequence Homo sapiens signal transducer


and activator of transcription 3 (STAT3) (NP_644805.1)


MAQWNQLQQLDTRYLEQLHQLYSDSFPMELRQFLAPWIESQDWAYAASKESHATLVFHNLLGEIDQQYSR





FLQESNVLYQHNLRRIKQFLQSRYLEKPMEIARIVARCLWEESRLLQTAATAAQQGGQANHPTAAVVTEK





QQMLEQHLQDVRKRVQDLEQKMKVVENLQDDFDFNYKTLKSQGDMQDLNGNNQSVTRQKMQQLEQMLTAL





DQMRRSIVSELAGLLSAMEYVQKTLTDEELADWKRRQQIACIGGPPNICLDRLENWITSLAESQLQTRQQ





IKKLEELQQKVSYKGDPIVQHRPMLEERIVELFRNLMKSAFVVERQPCMPMHPDRPLVIKTGVQFTTKVR





LLVKFPELNYQLKIKVCIDKDSGDVAALRGSRKFNILGTNTKVMNMEESNNGSLSAEFKHLTLREQRCGN





GGRANCDASLIVTEELHLITFETEVYHQGLKIDLETHSLPVVVISNICQMPNAWASILWYNMLTNNPKNV





NFFTKPPIGTWDQVAEVLSWQFSSTTKRGLSIEQLTTLAEKLLGPGVNYSGCQITWAKFCKENMAGKGFS





FWVWLDNIIDLVKKYILALWNEGYIMGFISKERERAILSTKPPGTFLLRFSESSKEGGVTFTWVEKDISG





KTQIQSVEPYTKQQLNNMSFAEIIMGYKIMDATNILVSPLVYLYPDIPKEEAFGKYCRPESQEHPEADPG





SAAPYLKTKFICVTPTTCSNTIDLPMSPRTLDSLMQFGNNGEGAEPSAGGQFESLTFDMELTSECATSPM





SEQ ID NO: 25 Human Nucleic Acid cDNA/mRNA Sequence Homo sapiens signal


transducer and activator of transcription 3 (STAT3), transcript variant


1, cDNA/mRNA (NM_139276.3)


GTCGCAGCCGAGGGAACAAGCCCCAACCGGATCCTGGACAGGCACCCCGGCTTGGCGCTGTCTCTCCCCC





TCGGCTCGGAGAGGCCCTTCGGCCTGAGGGAGCCTCGCCGCCCGTCCCCGGCACACGCGCAGCCCCGGCC





TCTCGGCCTCTGCCGGAGAAACAGTTGGGACCCCTGATTTTAGCAGGATGGCCCAATGGAATCAGCTACA





GCAGCTTGACACACGGTACCTGGAGCAGCTCCATCAGCTCTACAGTGACAGCTTCCCAATGGAGCTGCGG





CAGTTTCTGGCCCCTTGGATTGAGAGTCAAGATTGGGCATATGCGGCCAGCAAAGAATCACATGCCACTT





TGGTGTTTCATAATCTCCTGGGAGAGATTGACCAGCAGTATAGCCGCTTCCTGCAAGAGTCGAATGTTCT





CTATCAGCACAATCTACGAAGAATCAAGCAGTTTCTTCAGAGCAGGTATCTTGAGAAGCCAATGGAGATT





GCCCGGATTGTGGCCCGGTGCCTGTGGGAAGAATCACGCCTTCTACAGACTGCAGCCACTGCGGCCCAGC





AAGGGGGCCAGGCCAACCACCCCACAGCAGCCGTGGTGACGGAGAAGCAGCAGATGCTGGAGCAGCACCT





TCAGGATGTCCGGAAGAGAGTGCAGGATCTAGAACAGAAAATGAAAGTGGTAGAGAATCTCCAGGATGAC





TTTGATTTCAACTATAAAACCCTCAAGAGTCAAGGAGACATGCAAGATCTGAATGGAAACAACCAGTCAG





TGACCAGGCAGAAGATGCAGCAGCTGGAACAGATGCTCACTGCGCTGGACCAGATGCGGAGAAGCATCGT





GAGTGAGCTGGCGGGGCTTTTGTCAGCGATGGAGTACGTGCAGAAAACTCTCACGGACGAGGAGCTGGCT





GACTGGAAGAGGCGGCAACAGATTGCCTGCATTGGAGGCCCGCCCAACATCTGCCTAGATCGGCTAGAAA





ACTGGATAACGTCATTAGCAGAATCTCAACTTCAGACCCGTCAACAAATTAAGAAACTGGAGGAGTTGCA





GCAAAAAGTTTCCTACAAAGGGGACCCCATTGTACAGCACCGGCCGATGCTGGAGGAGAGAATCGTGGAG





CTGTTTAGAAACTTAATGAAAAGTGCCTTTGTGGTGGAGCGGCAGCCCTGCATGCCCATGCATCCTGACC





GGCCCCTCGTCATCAAGACCGGCGTCCAGTTCACTACTAAAGTCAGGTTGCTGGTCAAATTCCCTGAGTT





GAATTATCAGCTTAAAATTAAAGTGTGCATTGACAAAGACTCTGGGGACGTTGCAGCTCTCAGAGGATCC





CGGAAATTTAACATTCTGGGCACAAACACAAAAGTGATGAACATGGAAGAATCCAACAACGGCAGCCTCT





CTGCAGAATTCAAACACTTGACCCTGAGGGAGCAGAGATGTGGGAATGGGGGCCGAGCCAATTGTGATGC





TTCCCTGATTGTGACTGAGGAGCTGCACCTGATCACCTTTGAGACCGAGGTGTATCACCAAGGCCTCAAG





ATTGACCTAGAGACCCACTCCTTGCCAGTTGTGGTGATCTCCAACATCTGTCAGATGCCAAATGCCTGGG





CGTCCATCCTGTGGTACAACATGCTGACCAACAATCCCAAGAATGTAAACTTTTTTACCAAGCCCCCAAT





TGGAACCTGGGATCAAGTGGCCGAGGTCCTGAGCTGGCAGTTCTCCTCCACCACCAAGCGAGGACTGAGC





ATCGAGCAGCTGACTACACTGGCAGAGAAACTCTTGGGACCTGGTGTGAATTATTCAGGGTGTCAGATCA





CATGGGCTAAATTTTGCAAAGAAAACATGGCTGGCAAGGGCTTCTCCTTCTGGGTCTGGCTGGACAATAT





CATTGACCTTGTGAAAAAGTACATCCTGGCCCTTTGGAACGAAGGGTACATCATGGGCTTTATCAGTAAG





GAGCGGGAGCGGGCCATCTTGAGCACTAAGCCTCCAGGCACCTTCCTGCTAAGATTCAGTGAAAGCAGCA





AAGAAGGAGGCGTCACTTTCACTTGGGTGGAGAAGGACATCAGCGGTAAGACCCAGATCCAGTCCGTGGA





ACCATACACAAAGCAGCAGCTGAACAACATGTCATTTGCTGAAATCATCATGGGCTATAAGATCATGGAT





GCTACCAATATCCTGGTGTCTCCACTGGTCTATCTCTATCCTGACATTCCCAAGGAGGAGGCATTCGGAA





AGTATTGTCGGCCAGAGAGCCAGGAGCATCCTGAAGCTGACCCAGGTAGCGCTGCCCCATACCTGAAGAC





CAAGTTTATCTGTGTGACACCAACGACCTGCAGCAATACCATTGACCTGCCGATGTCCCCCCGCACTTTA





GATTCATTGATGCAGTTTGGAAATAATGGTGAAGGTGCTGAACCCTCAGCAGGAGGGCAGTTTGAGTCCC





TCACCTTTGACATGGAGTTGACCTCGGAGTGCGCTACCTCCCCCATGTGAGGAGCTGAGAACGGAAGCTG





CAGAAAGATACGACTGAGGCGCCTACCTGCATTCTGCCACCCCTCACACAGCCAAACCCCAGATCATCTG





AAACTACTAACTTTGTGGTTCCAGATTTTTTTTAATCTCCTACTTCTGCTATCTTTGAGCAATCTGGGCA





CTTTTAAAAATAGAGAAATGAGTGAATGTGGGTGATCTGCTTTTATCTAAATGCAAATAAGGATGTGTTC





TCTGAGACCCATGATCAGGGGATGTGGCGGGGGGTGGCTAGAGGGAGAAAAAGGAAATGTCTTGTGTTGT





TTTGTTCCCCTGCCCTCCTTTCTCAGCAGCTTTTTGTTATTGTTGTTGTTGTTCTTAGACAAGTGCCTCC





TGGTGCCTGCGGCATCCTTCTGCCTGTTTCTGTAAGCAAATGCCACAGGCCACCTATAGCTACATACTCC





TGGCATTGCACTTTTTAACCTTGCTGACATCCAAATAGAAGATAGGACTATCTAAGCCCTAGGTTTCTTT





TTAAATTAAGAAATAATAACAATTAAAGGGCAAAAAACACTGTATCAGCATAGCCTTTCTGTATTTAAGA





AACTTAAGCAGCCGGGCATGGTGGCTCACGCCTGTAATCCCAGCACTTTGGGAGGCCGAGGCGGATCATA





AGGTCAGGAGATCAAGACCATCCTGGCTAACACGGTGAAACCCCGTCTCTACTAAAAGTACAAAAAATTA





GCTGGGTGTGGTGGTGGGCGCCTGTAGTCCCAGCTACTCGGGAGGCTGAGGCAGGAGAATCGCTTGAACC





TGAGAGGCGGAGGTTGCAGTGAGCCAAAATTGCACCACTGCACACTGCACTCCATCCTGGGCGACAGTCT





GAGACTCTGTCTCAAAAAAAAAAAAAAAAAAAAGAAACTTCAGTTAACAGCCTCCTTGGTGCTTTAAGCA





TTCAGCTTCCTTCAGGCTGGTAATTTATATAATCCCTGAAACGGGCTTCAGGTCAAACCCTTAAGACATC





TGAAGCTGCAACCTGGCCTTTGGTGTTGAAATAGGAAGGTTTAAGGAGAATCTAAGCATTTTAGACTTTT





TTTTATAAATAGACTTATTTTCCTTTGTAATGTATTGGCCTTTTAGTGAGTAAGGCTGGGCAGAGGGTGC





TTACAACCTTGACTCCCTTTCTCCCTGGACTTGATCTGCTGTTTCAGAGGCTAGGTTGTTTCTGTGGGTG





CCTTATCAGGGCTGGGATACTTCTGATTCTGGCTTCCTTCCTGCCCCACCCTCCCGACCCCAGTCCCCCT





GATCCTGCTAGAGGCATGTCTCCTTGCGTGTCTAAAGGTCCCTCATCCTGTTTGTTTTAGGAATCCTGGT





CTCAGGACCTCATGGAAGAAGAGGGGGAGAGAGTTACAGGTTGGACATGATGCACACTATGGGGCCCCAG





CGACGTGTCTGGTTGAGCTCAGGGAATATGGTTCTTAGCCAGTTTCTTGGTGATATCCAGTGGCACTTGT





AATGGCGTCTTCATTCAGTTCATGCAGGGCAAAGGCTTACTGATAAACTTGAGTCTGCCCTCGTATGAGG





GTGTATACCTGGCCTCCCTCTGAGGCTGGTGACTCCTCCCTGCTGGGGCCCCACAGGTGAGGCAGAACAG





CTAGAGGGCCTCCCCGCCTGCCCGCCTTGGCTGGCTAGCTCGCCTCTCCTGTGCGTATGGGAACACCTAG





CACGTGCTGGATGGGCTGCCTCTGACTCAGAGGCATGGCCGGATTTGGCAACTCAAAACCACCTTGCCTC





AGCTGATCAGAGTTTCTGTGGAATTCTGTTTGTTAAATCAAATTAGCTGGTCTCTGAATTAAGGGGGAGA





CGACCTTCTCTAAGATGAACAGGGTTCGCCCCAGTCCTCCTGCCTGGAGACAGTTGATGTGTCATGCAGA





GCTCTTACTTCTCCAGCAACACTCTTCAGTACATAATAAGCTTAACTGATAAACAGAATATTTAGAAAGG





TGAGACTTGGGCTTACCATTGGGTTTAAATCATAGGGACCTAGGGCGAGGGTTCAGGGCTTCTCTGGAGC





AGATATTGTCAAGTTCATGGCCTTAGGTAGCATGTATCTGGTCTTAACTCTGATTGTAGCAAAAGTTCTG





AGAGGAGCTGAGCCCTGTTGTGGCCCATTAAAGAACAGGGTCCTCAGGCCCTGCCCGCTTCCTGTCCACT





GCCCCCTCCCCATCCCCAGCCCAGCCGAGGGAATCCCGTGGGTTGCTTACCTACCTATAAGGTGGTTTAT





AAGCTGCTGTCCTGGCCACTGCATTCAAATTCCAATGTGTACTTCATAGTGTAAAAATTTATATTATTGT





GAGGTTTTTTGTCTTTTTTTTTTTTTTTTTTTTTTGGTATATTGCTGTATCTACTTTAACTTCCAGAAAT





AAACGTTATATAGGAACCGTC





SEQ ID NO: 26 Human Amino Acid Sequence cathelicidin antimicrobial peptide


preproprotein (STAT3 (NP_004336.4)


MKTQRDGHSLGRWSLVLLLLGLVMPLAIIAQVLSYKEAVLRAIDGINQRSSDANLYRLLDLDPRPTMDGD





PDTPKPVSFTVKETVCPRTTQQSPEDCDFKKDGLVKRCMGTVTLNQARGSFDISCDKDNKRFALLGDFFR





KSKEKIGKEFKRIVQRIKDFLRNLVPRTES





SEQ ID NO: 27 Human Nucleic Acid cDNA/mRNA Sequence cathelicidin


antimicrobial peptide (CAMP), mRNA (NM_004345.5)


AGGCAGACATGGGGACCATGAAGACCCAAAGGGATGGCCACTCCCTGGGGCGGTGGTCACTGGTGCTCCT





GCTGCTGGGCCTGGTGATGCCTCTGGCCATCATTGCCCAGGTCCTCAGCTACAAGGAAGCTGTGCTTCGT





GCTATAGATGGCATCAACCAGCGGTCCTCGGATGCTAACCTCTACCGCCTCCTGGACCTGGACCCCAGGC





CCACGATGGATGGGGACCCAGACACGCCAAAGCCTGTGAGCTTCACAGTGAAGGAGACAGTGTGCCCCAG





GACGACACAGCAGTCACCAGAGGATTGTGACTTCAAGAAGGACGGGCTGGTGAAGCGGTGTATGGGGACA





GTGACCCTCAACCAGGCCAGGGGCTCCTTTGACATCAGTTGTGATAAGGATAACAAGAGATTTGCCCTGC





TGGGTGATTTCTTCCGGAAATCTAAAGAGAAGATTGGCAAAGAGTTTAAAAGAATTGTCCAGAGAATCAA





GGATTTTTTGCGGAATCTTGTACCCAGGACAGAGTCCTAGTGTGTGCCCTACCCTGGCTCAGGCTTCTGG





GCTCTGAGAAATAAACTATGAGAGCAATTTC





SEQ ID NO: 28 Human Amino Acid Sequence neutrophilic granule protein [Mus


musculus] (ngp) (NP_032720.2)


MAGLWKTFVLVVALAVVSCEALPQLRYERIVDRAIEAYNQGRQGRPIFRLLSATPPSSONPATNIPLQER





IKETECTSTOEROPKDCDFLEDGEERNCTGKFERRROSTSLTLTCDRDCSREDTOETSENDKQDVSEKEK





FEDVPPHIRNIYEDAKYDIIGNILKNE





SEQ ID NO: 29 Human Nucleic Acid cDNA/mRNA Sequence neutrophilic granule


protein [Mus musculus] (ngp), mRNA (NM_008694.2)



AGTCTCAATATCATCTACATAAAAGGGGCCAAGAGTGGTAGTGTGTCAGAGACAATGGCAGGGCTGTGGA







AGACCTTTGTATTGGTGGTGGCCTTGGCTGTGGTCTCCTGTGAGGCCCTTCGACAACTAAGATATGAGGA







GATTGTTGATAGAGCCATAGAGGCATACAACCAAGGGCGGCAAGGAAGACCCCTCTTCCGCCTGCTAAGT







GCCACTCCGCCTTCTAGTCAGAATCCTGCTACCAATATCCCACTCCAGTTCAGGATTAAAGAGACAGAGT







GTACTTCCACCCAGGAGAGACAGCCTAAAGACTGCGACTTCCTGGAGGATGGGGAGGAGAGAAATTGCAC







AGGGAAATTCTTCAGAAGGCGGCAGTCAACCTCCCTGACCTTGACCTGCGACAGGGATTGCAGTCGAGAG







GATACCCAAGAAACCAGTTTTAATGATAAGCAAGACGTCTCTGAAAAGGAAAAGTTCGAAGATGTGCCCC







CTCACATCAGGAACATTTATGAAGATGCCAAGTATGATATCATCGGCAACATCCTGAAAAATTTCTAGGG







CTGGAAAGAGGAGGGAGGTGCTCCCTGCATACTATGACCTCCTCTTTACCTCCACTACCCATCTCCCCCT







GCTGCATTCAGGATCTGCCCCTCCTTCCTGCCCTTCCCAGGAACACCCCCTCTAGAGTAGCTCTAGCTCC







TAAAACATCCATACCTTIGTCCATTTGCTTCCTTCTGCTGGGCCTTCCTGCCTTACCCTCTATCTGAAAC







CCTTATTGATTCTTCAAGGCCCAAGTTCAAAAGTCCCCTCCAGCGGGAAGCCTCCTCATTCTCCCAGAGC







CAAAGTCCTGCCCACATCAGTTCACTCATAATCTTCAAACCACATTGGTATTACCTGCTGTGTCCCCAGC







CAGACAACCCTGTATCTATTCACAGCTGGGCCTCCCGGGCCAGTTGCAGGTAGAATGAATATTTCAATGA







TGTGTCCCTGGAATCCTGGGAGGACAGAACCCTGTAGACTCCTGCTCTCTGCCTAGTCACTGTGACACCA







AATGCCCCTTTACATACCCAGATCCCTTAATGGGGATGTGGCAGGTGGGTGTGGTCAGATCACCTTGTGA






GGCCTATAAGAGAGGTTCAATAAAAATGCTTCTGAGATTAAAAAAAAAAAAAAAAA





SEQ ID NO: 30 Human Amino Acid Sequence prostaglandin G/H synthase 2


precursor (NP_000954.1)


MLARALLLCAVLALSHTANPCCSHPCQNRGVCMSVGFDQYKCDCTRTGFYGENCSTPEFLTRIKLFLKPT





PNTVHYILTHFKGFWNVVNNIPFLRNAIMSYVLTSRSHLIDSPPTYNADYGYKSWEAFSNLSYYTRALPP





VPDDCPTPLGVKGKKQLPDSNEIVEKLLLRRKFIPDPQGSNMMFAFFAQHFTHQFFKTDHKRGPAFTNGL





GHGVDLNHIYGETLARQRKLRLFKDGKMKYQIIDGEMYPPTVKDTQAEMIYPPQVPEHLRFAVGQEVFGL





VPGLMMYATIWLREHNRVCDVLKQEHPEWGDEQLFQTSRLILIGETIKIVIEDYVQHLSGYHFKLKFDPE





LLFNKQFQYQNRIAAEFNTLYHWHPLLPDTFQIHDQKYNYQQFIYNNSILLEHGITQFVESFTRQIAGRV





AGGRNVPPAVQKVSQASIDQSRQMKYQSFNEYRKRFMLKPYESFEELTGEKEMSAELEALYGDIDAVELY





PALLVEKPRPDAIFGETMVEVGAPFSLKGLMGNVICSPAYWKPSTFGGEVGFQIINTASIQSLICNNVKG





CPFTSFSVPDPELIKTVTINASSSRSGLDDINPTVLLKERSTEL





SEQ ID NO: 31 Human Nucleic Acid cDNA/mRNA Sequence prostaglandin G/H


synthase 2 precursor (NM_000963.4)


AATTGTCATACGACTTGCAGTGAGCGTCAGGAGCACGTCCAGGAACTCCTCAGCAGCGCCTCCTTCAGCT





CCACAGCCAGACGCCCTCAGACAGCAAAGCCTACCCCCGCGCCGCGCCCTGCCCGCCGCTGCGATGCTCG





CCCGCGCCCTGCTGCTGTGCGCGGTCCTGGCGCTCAGCCATACAGCAAATCCTTGCTGTTCCCACCCATG





TCAAAACCGAGGTGTATGTATGAGTGTGGGATTTGACCAGTATAAGTGCGATTGTACCCGGACAGGATTC





TATGGAGAAAACTGCTCAACACCGGAATTTTTGACAAGAATAAAATTATTTCTGAAACCCACTCCAAACA





CAGTGCACTACATACTTACCCACTTCAAGGGATTTTGGAACGTTGTGAATAACATTCCCTTCCTTCGAAA





TGCAATTATGAGTTATGTGTTGACATCCAGATCACATTTGATTGACAGTCCACCAACTTACAATGCTGAC





TATGGCTACAAAAGCTGGGAAGCCTTCTCTAACCTCTCCTATTATACTAGAGCCCTTCCTCCTGTGCCTG





ATGATTGCCCGACTCCCTTGGGTGTCAAAGGTAAAAAGCAGCTTCCTGATTCAAATGAGATTGTGGAAAA





ATTGCTTCTAAGAAGAAAGTTCATCCCTGATCCCCAGGGCTCAAACATGATGTTTGCATTCTTTGCCCAG





CACTTCACGCATCAGTTTTTCAAGACAGATCATAAGCGAGGGCCAGCTTTCACCAACGGGCTGGGCCATG





GGGTGGACTTAAATCATATTTACGGTGAAACTCTGGCTAGACAGCGTAAACTGCGCCTTTTCAAGGATGG





AAAAATGAAATATCAGATAATTGATGGAGAGATGTATCCTCCCACAGTCAAAGATACTCAGGCAGAGATG





ATCTACCCTCCTCAAGTCCCTGAGCATCTACGGTTTGCTGTGGGGCAGGAGGTCTTTGGTCTGGTGCCTG





GTCTGATGATGTATGCCACAATCTGGCTGCGGGAACACAACAGAGTATGCGATGTGCTTAAACAGGAGCA





TCCTGAATGGGGTGATGAGCAGTTGTTCCAGACAAGCAGGCTAATACTGATAGGAGAGACTATTAAGATT





GTGATTGAAGATTATGTGCAACACTTGAGTGGCTATCACTTCAAACTGAAATTTGACCCAGAACTACTTT





TCAACAAACAATTCCAGTACCAAAATCGTATTGCTGCTGAATTTAACACCCTCTATCACTGGCATCCCCT





TCTGCCTGACACCTTTCAAATTCATGACCAGAAATACAACTATCAACAGTTTATCTACAACAACTCTATA





TTGCTGGAACATGGAATTACCCAGTTTGTTGAATCATTCACCAGGCAAATTGCTGGCAGGGTTGCTGGTG





GTAGGAATGTTCCACCCGCAGTACAGAAAGTATCACAGGCTTCCATTGACCAGAGCAGGCAGATGAAATA





CCAGTCTTTTAATGAGTACCGCAAACGCTTTATGCTGAAGCCCTATGAATCATTTGAAGAACTTACAGGA





GAAAAGGAAATGTCTGCAGAGTTGGAAGCACTCTATGGTGACATCGATGCTGTGGAGCTGTATCCTGCCC





TTCTGGTAGAAAAGCCTCGGCCAGATGCCATCTTTGGTGAAACCATGGTAGAAGTTGGAGCACCATTCTC





CTTGAAAGGACTTATGGGTAATGTTATATGTTCTCCTGCCTACTGGAAGCCAAGCACTTTTGGTGGAGAA





GTGGGTTTTCAAATCATCAACACTGCCTCAATTCAGTCTCTCATCTGCAATAACGTGAAGGGCTGTCCCT





TTACTTCATTCAGTGTTCCAGATCCAGAGCTCATTAAAACAGTCACCATCAATGCAAGTTCTTCCCGCTC





CGGACTAGATGATATCAATCCCACAGTACTACTAAAAGAACGTTCGACTGAACTGTAGAAGTCTAATGAT





CATATTTATTTATTTATATGAACCATGTCTATTAATTTAATTATTTAATAATATTTATATTAAACTCCTT





ATGTTACTTAACATCTTCTGTAACAGAAGTCAGTACTCCTGTTGCGGAGAAAGGAGTCATACTTGTGAAG





ACTTTTATGTCACTACTCTAAAGATTTTGCTGTTGCTGTTAAGTTTGGAAAACAGTTTTTATTCTGTTTT





ATAAACCAGAGAGAAATGAGTTTTGACGTCTTTTTACTTGAATTTCAACTTATATTATAAGAACGAAAGT





AAAGATGTTTGAATACTTAAACACTGTCACAAGATGGCAAAATGCTGAAAGTTTTTACACTGTCGATGTT





TCCAATGCATCTTCCATGATGCATTAGAAGTAACTAATGTTTGAAATTTTAAAGTACTTTTGGTTATTTT





TCTGTCATCAAACAAAAACAGGTATCAGTGCATTATTAAATGAATATTTAAATTAGACATTACCAGTAAT





TTCATGTCTACTTTTTAAAATCAGCAATGAAACAATAATTTGAAATTTCTAAATTCATAGGGTAGAATCA





CCTGTAAAAGCTTGTTTGATTTCTTAAAGTTATTAAACTTGTACATATACCAAAAAGAAGCTGTCTTGGA





TTTAAATCTGTAAAATCAGTAGAAATTTTACTACAATTGCTTGTTAAAATATTTTATAAGTGATGTTCCT





TTTTCACCAAGAGTATAAACCTTTTTAGTGTGACTGTTAAAACTTCCTTTTAAATCAAAATGCCAAATTT





ATTAAGGTGGTGGAGCCACTGCAGTGTTATCTTAAAATAAGAATATTTTGTTGAGATATTCCAGAATTTG





TTTATATGGCTGGTAACATGTAAAATCTATATCAGCAAAAGGGTCTACCTTTAAAATAAGCAATAACAAA





GAAGAAAACCAAATTATTGTTCAAATTTAGGTTTAAACTTTTGAAGCAAACTTTTTTTTATCCTTGTGCA





CTGCAGGCCTGGTACTCAGATTTTGCTATGAGGTTAATGAAGTACCAAGCTGTGCTTGAATAATGATATG





TTTTCTCAGATTTTCTGTTGTACAGTTTAATTTAGCAGTCCATATCACATTGCAAAAGTAGCAATGACCT





CATAAAATACCTCTTCAAAATGCTTAAATTCATTTCACACATTAATTTTATCTCAGTCTTGAAGCCAATT





CAGTAGGTGCATTGGAATCAAGCCTGGCTACCTGCATGCTGTTCCTTTTCTTTTCTTCTTTTAGCCATTT





TGCTAAGAGACACAGTCTTCTCATCACTTCGTTTCTCCTATTTTGTTTTACTAGTTTTAAGATCAGAGTT





CACTTTCTTTGGACTCTGCCTATATTTTCTTACCTGAACTTTTGCAAGTTTTCAGGTAAACCTCAGCTCA





GGACTGCTATTTAGCTCCTCTTAAGAAGATTAAAAGAGAAAAAAAAAGGCCCTTTTAAAAATAGTATACA





CTTATTTTAAGTGAAAAGCAGAGAATTTTATTTATAGCTAATTTTAGCTATCTGTAACCAAGATGGATGC





AAAGAGGCTAGTGCCTCAGAGAGAACTGTACGGGGTTTGTGACTGGAAAAAGTTACGTTCCCATTCTAAT





TAATGCCCTTTCTTATTTAAAAACAAAACCAAATGATATCTAAGTAGTTCTCAGCAATAATAATAATGAC





GATAATACTTCTTTTCCACATCTCATTGTCACTGACATTTAATGGTACTGTATATTACTTAATTTATTGA





AGATTATTATTTATGTCTTATTAGGACACTATGGTTATAAACTGTGTTTAAGCCTACAATCATTGATTTT





TTTTTGTTATGTCACAATCAGTATATTTTCTTTGGGGTTACCTCTCTGAATATTATGTAAACAATCCAAA





GAAATGATTGTATTAAGATTTGTGAATAAATTTTTAGAAATCTGATTGGCATATTGAGATATTTAAGGTT





GAATGTTTGTCCTTAGGATAGGCCTATGTGCTAGCCCACAAAGAATATTGTCTCATTAGCCTGAATGTGC





CATAAGACTGACCTTTTAAAATGTTTTGAGGGATCTGTGGATGCTTCGTTAATTTGTTCAGCCACAATTT





ATTGAGAAAATATTCTGTGTCAAGCACTGTGGGTTTTAATATTTTTAAATCAAACGCTGATTACAGATAA





TAGTATTTATATAAATAATTGAAAAAAATTTTCTTTTGGGAAGAGGGAGAAAATGAAATAAATATCATTA





AAGATAACTCAGGAGAATCTTCTTTACAATTTTACGTTTAGAATGTTTAAGGTTAAGAAAGAAATAGTCA





ATATGCTTGTATAAAACACTGTTCACTGTTTTTTTTAAAAAAAAAACTTGATTTGTTATTAACATTGATC





TGCTGACAAAACCTGGGAATTTGGGTTGTGTATGCGAATGTTTCAGTGCCTCAGACAAATGTGTATTTAA





CTTATGTAAAAGATAAGTCTGGAAATAAATGTCTGTTTATTTTTGTACTATTTAAAAATTGACAGATCTT





TTCTGAAGATAAACTTTGATTGTTTCTATA





SEQ ID NO: 32 Human Amino Acid Sequence ras-related protein Rab-7a


(NP_004628.4)


MTSRKKVLLKVIILGDSGVGKTSLMNQYVNKKFSNQYKATIGADFLTKEVMVDDRLVTMQIWDTAGQERF





QSLGVAFYRGADCCVLVFDVTAPNTFKTLDSWRDEFLIQASPRDPENFPFVVLGNKIDLENRQVATKRAQ





AWCYSKNNIPYFETSAKEAINVEQAFQTIARNALKQETEVELYNEFPEPIKLDKNDRAKASAESCSC





SEQ ID NO: 33 Human Nucleic Acid cDNA/mRNA Sequence ras-related protein


Rab-7a (NM_004637.6)


AGTCTTGGCCATAAAGCCTGAGGCGGCGGCAGCGGCGGAGTTGGCGGCTTGGAGAGCTCGGGAGAGTTCC





CTGGAACCAGAACTTGGACCTTCTCGCTTCTGTCCTCCGTTTAGTCTCCTCCTCGGCGGGAGCCCTCGCG





ACGCGCCCGGCCCGGAGCCCCCAGCGCAGCGGCCGCGTTTGAAGGATGACCTCTAGGAAGAAAGTGTTGC





TGAAGGTTATCATCCTGGGAGATTCTGGAGTCGGGAAGACATCACTCATGAACCAGTATGTGAATAAGAA





ATTCAGCAATCAGTACAAAGCCACAATAGGAGCTGACTTTCTGACCAAGGAGGTGATGGTGGATGACAGG





CTAGTCACAATGCAGATATGGGACACAGCAGGACAGGAACGGTTCCAGTCTCTCGGTGTGGCCTTCTACA





GAGGTGCAGACTGCTGCGTTCTGGTATTTGATGTGACTGCCCCCAACACATTCAAAACCCTAGATAGCTG





GAGAGATGAGTTTCTCATCCAGGCCAGTCCCCGAGATCCTGAAAACTTCCCATTTGTTGTGTTGGGAAAC





AAGATTGACCTCGAAAACAGACAAGTGGCCACAAAGCGGGCACAGGCCTGGTGCTACAGCAAAAACAACA





TTCCCTACTTTGAGACCAGTGCCAAGGAGGCCATCAACGTGGAGCAGGCGTTCCAGACGATTGCACGGAA





TGCACTTAAGCAGGAAACGGAGGTGGAGCTGTACAACGAATTTCCTGAACCTATCAAACTGGACAAGAAT





GACCGGGCCAAGGCCTCGGCAGAAAGCTGCAGTTGCTGAGGGGGCAGTGAGAGTTGAGCACAGAGTCCTT





CACAAACCAAGAACACACGTAGGCCTTCAACACAATTCCCCTCTCCTCTTCCAAACAAAACATACATTGA





TCTCTCACATCCAGCTGCCAAAAGAAAACCCCATCAAACACAGTTACACCCCACATATCTCTCACACACA





CACACACACGCACACACACACACACAGATCTGACGTAATCAAACTCCAGCCCTTGCCCGTGATGGCTCCT





TGGGGTCTGCCTGCCCACCCACATGAGCCCGCGAGTATGGCAGCAGGACAAGCCAGCGGTGGAAGTCATT





CTGATATGGAGTTGGCATTGGAAGCTTATTCTTTTTGTTCACTGGAGAGAGAGAGAACTGTTTACAGTTA





ATCTGTGTCTAATTATCTGATTTTTTTTATTGGTCTTGTGGTCTTTTTACCCCCCCTTTCCCCTCCCTCC





TTGAAGGCTACCCCTTGGGAAGGCTGGTGCCCCATGCCCCATTACAGGCTCACACCCAGTCTGATCAGGC





TGAGTTTTGTATGTATCTATCTGTTAATGCTTGTTACTTTTAACTAATCAGATCTTTTTACAGTATCCAT





TTATTATGTAATGCTTCTTAGAAAAGAATCTTATAGTACATGTTAATATATGCAACCAATTAAAATGTAT





AAATTAGTGTAAGAAATTCTTGGATTATGTGTTTAAGTCCTGTAATGCAGGCCTGTAAGGTGGAGGGTTG





AACCCTGTTTGGATTGCAGAGTGTTACTCAGAATTGGGAAATCCAGCTAGCGGCAGTATTCTGTACAGTA





GACACAAGAATTATGTACGCCTTTTATCAAAGACTTAAGAGCCAAAAAGCTTTTCATCTCTCCAGGGGGA





AAACTGTCTAGTTCCCTTCTGTGTCTAAATTTTCCAAAACGTTGATTTGCATAATACAGTGGTATGTGCA





ATGGATAAATTGCCGTTATTTCAAAAATTAAAATTCTCATTTTCTTTCTTTTTTTTCCCCCCTGCTCCAC





ACTTCAAAACTCCCGTTAGATCAGCATTCTACTACAAGAGTGAAAGGAAAACCCTAACAGATCTGTCCTA





GTGATTTTACCTTTGTTCTAGAAGGCGCTCCTTTCAGGGTTGTGGTATTCTTAGGTTAGCGGAGCTTTTT





CCTCTTTTCCCCACCCATCTCCCCAATATTGCCCATTATTAATTAACCTCTTTCTTTGGTTGGAACCCTG





GCAGTTCTGCTCCCTTCCTAGGATCTGCCCCTGCATTGTAGCTTGCTTAACGGAGCACTTCTCCTTTTTC





CAAAGGTCTACATTCTAGGGTGTGGGCTGAGTTCTTCTGTAAAGAGATGAACGCAATGCCAATAAAATTG





AACAAGAACAATGAT






In some embodiments, an agent disclosed herein or a down-regulator of IL-22 signaling includes any agent that specific binds to or decreases the activity or level of any one of the biomarkers listed in Table 1.


The term “body fluid” refers to fluids that are excreted or secreted from the body as well as fluids that are normally not (e.g. amniotic fluid, aqueous humor, bile, blood and blood plasma, cerebrospinal fluid, cerumen and earwax, cowper's fluid or pre-ejaculatory fluid, chyle, chyme, stool, female ejaculate, interstitial fluid, intracellular fluid, lymph, menses, breast milk, mucus, pleural fluid, pus, saliva, sebum, semen, serum, sweat, synovial fluid, tears, urine, vaginal lubrication, vitreous humor, vomit).


The term “coding region” refers to regions of a nucleotide sequence comprising codons which are translated into amino acid residues, whereas the term “noncoding region” refers to regions of a nucleotide sequence that are not translated into amino acids (e.g., 5′ and 3′ untranslated regions).


The term “complementary” refers to the broad concept of sequence complementarity between regions of two nucleic acid strands or between two regions of the same nucleic acid strand. It is known that an adenine residue of a first nucleic acid region is capable of forming specific hydrogen bonds (“base pairing”) with a residue of a second nucleic acid region which is antiparallel to the first region if the residue is thymine or uracil. Similarly, it is known that a cytosine residue of a first nucleic acid strand is capable of base pairing with a residue of a second nucleic acid strand which is antiparallel to the first strand if the residue is guanine. A first region of a nucleic acid is complementary to a second region of the same or a different nucleic acid if, when the two regions are arranged in an antiparallel fashion, at least one nucleotide residue of the first region is capable of base pairing with a residue of the second region. Preferably, the first region comprises a first portion and the second region comprises a second portion, whereby, when the first and second portions are arranged in an antiparallel fashion, at least about 50%, and preferably at least about 75%, at least about 90%, or at least about 95% of the nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion. More preferably, all nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion.


As used herein, the phrase “conjoint administration” refers to any form of administration of two or more different therapeutic agents such that the second agent is administered while the previously administered therapeutic agent is still effective in the body (e.g., the two agents are simultaneously effective in the subject, which may include synergistic effects of the two agents). For example, the different therapeutic agents can be administered either in the same formulation or in separate formulations, either concomitantly or sequentially. In certain embodiments, the different therapeutic agents can be administered within about one hour, about 12 hours, about 24 hours, about 36 hours, about 48 hours, about 72 hours, or about a week of one another. Thus, a subject who receives such treatment can benefit from a combined effect of different therapeutic agents.


The term “control” refers to any reference standard suitable to provide a comparison to the expression products in the test sample. In one embodiment, the control comprises obtaining a “control sample” from which expression product levels are detected and compared to the expression product levels from the test sample. Such a control sample may comprise any suitable sample, including but not limited to a sample from a control patient (can be stored sample or previous sample measurement) with a known outcome; normal tissue or cells isolated from a subject, such as a normal subject or a subject with an MDS and/or an anemia, cultured primary cells/tissues isolated from a subject, such as a normal subject or a subject with an MDS and/or an anemia, adjacent normal cells/tissues obtained from the same organ or body location of a normal subject or a subject with an MDS and/or an anemia, a tissue or cell sample isolated from a normal subject, or a primary cells/tissues obtained from a depository. In another preferred embodiment, the control may comprise a reference standard expression product level from any suitable source, including but not limited to housekeeping genes, an expression product level range from normal tissue (or other previously analyzed control sample), a previously determined expression product level range within a test sample from a group of patients, or a set of patients with a certain outcome (for example, reduced anemia for one, two, three, four years, etc.) or receiving a certain treatment (for example, standard of care therapy). It will be understood by those of skill in the art that such control samples and reference standard expression product levels can be used in combination as controls in the methods of the present invention. In one embodiment, the control may comprise normal or an MDS and/or an anemia cell/tissue sample. In another preferred embodiment, the control may comprise an expression level for a set of patients, such as a set of patients, or for a set of patients receiving a certain treatment, or for a set of patients with one outcome versus another outcome. In the former case, the specific expression product level of each patient can be assigned to a percentile level of expression, or expressed as either higher or lower than the mean or average of the reference standard expression level. In another preferred embodiment, the control may comprise normal cells or cells from subjects treated with combination therapy. In another embodiment, the control may also comprise a measured value for example, average level of expression of a particular gene in a population compared to the level of expression of a housekeeping gene in the same population. Such a population may comprise normal subjects, subjects having an MDS and/or an anemia who have not undergone any treatment (i.e., treatment naive), or subjects having an MDS and/or an anemia undergoing standard of care therapy. In another preferred embodiment, the control comprises a ratio transformation of expression product levels, including but not limited to determining a ratio of expression product levels of two genes in the test sample and comparing it to any suitable ratio of the same two genes in a reference standard; determining expression product levels of the two or more genes in the test sample and determining a difference in expression product levels in any suitable control; and determining expression product levels of the two or more genes in the test sample, normalizing their expression to expression of housekeeping genes in the test sample, and comparing to any suitable control. In particularly preferred embodiments, the control comprises a control sample which is of the same lineage and/or type as the test sample. In another embodiment, the control may comprise expression product levels grouped as percentiles within or based on a set of patient samples, such as all subjects in a cohort having an MDS and/or an anemia. In one embodiment a control expression product level is established wherein higher or lower levels of expression product relative to, for instance, a particular percentile, are used as the basis for predicting outcome. In another preferred embodiment, a control expression product level is established using expression product levels from control subjects with a known outcome, and the expression product levels from the test sample are compared to the control expression product level as the basis for predicting outcome. As demonstrated by the data below, the methods of the present invention are not limited to use of a specific cut-point in comparing the level of expression product in the test sample to the control.


The “copy number” of a biomarker nucleic acid refers to the number of DNA sequences in a cell (e.g., germline and/or somatic) encoding a particular gene product. Generally, for a given gene, a mammal has two copies of each gene. The copy number can be increased, however, by gene amplification or duplication, or reduced by deletion. For example, germline copy number changes include changes at one or more genomic loci, wherein said one or more genomic loci are not accounted for by the number of copies in the normal complement of germline copies in a control (e.g., the normal copy number in germline DNA for the same species as that from which the specific germline DNA and corresponding copy number were determined). Somatic copy number changes include changes at one or more genomic loci, wherein said one or more genomic loci are not accounted for by the number of copies in germline DNA of a control (e.g., copy number in germline DNA for the same subject as that from which the somatic DNA and corresponding copy number were determined).


The “normal” copy number (e.g., germline and/or somatic) of a biomarker nucleic acid or “normal” level of expression of a biomarker nucleic acid or protein is the activity/level of expression or copy number in a biological sample, e.g., a sample containing tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, and bone marrow, from a subject, e.g., a human, not afflicted with an MDS and/or an anemia, or from a corresponding non-affected tissue in the same afflicted subject.


The term “diagnosing” includes the use of the methods, systems, and code of the present invention to determine a level of IL-22 signaling in an individual, cell population, tissue, and the like. The term also includes methods, systems, and code for assessing the level of disease activity in an individual.


The term “down-regulate” includes the decrease, limitation, or blockage, of, for example a particular action, function, or interaction. In some embodiments, IL-22 signaling is “down-regulated” if at least one effect of IL-22 signaling is alleviated, terminated, slowed, or prevented. Similarly, a “down-regulator” of IL-22 signaling is an agent (e.g., a therapeutic agent) that down-regulates IL-22 signaling. The terms “promote” and “up-regulate” have the opposite meaning as compared to “down-regulate.”


A molecule is “fixed” or “affixed” to a substrate if it is covalently or non-covalently associated with the substrate such that the substrate can be rinsed with a fluid (e.g., standard saline citrate, pH 7.4) without a substantial fraction of the molecule dissociating from the substrate.


The term “mode of administration” includes any approach of contacting a desired target (e.g., cells, a subject) with a desired agent (e.g., a therapeutic agent). The route of administration, as used herein, is a particular form of the mode of administration, and it specifically covers the routes by which agents are administered to a subject or by which biophysical agents are contacted with a biological material.


The term “pre-determined” biomarker amount and/or activity measurement(s) may be a biomarker amount and/or activity measurement(s) used to, by way of example only, evaluate a subject that may be selected for a particular treatment, evaluate a response to a treatment such as modulation of one or more biomarkers described herein, and/or evaluate the disease state. A pre-determined biomarker amount and/or activity measurement(s) may be determined in populations of patients with or without an MDS and/or an anemia. The pre-determined biomarker amount and/or activity measurement(s) can be a single number, equally applicable to every patient, or the pre-determined biomarker amount and/or activity measurement(s) can vary according to specific subpopulations of patients. Age, weight, height, and other factors of a subject may affect the pre-determined biomarker amount and/or activity measurement(s) of the individual. Furthermore, the pre-determined biomarker amount and/or activity can be determined for each subject individually. In one embodiment, the amounts determined and/or compared in a method described herein are based on absolute measurements. In another embodiment, the amounts determined and/or compared in a method described herein are based on relative measurements, such as ratios (e.g., serum biomarker normalized to the expression of housekeeping or otherwise generally constant biomarker). The pre-determined biomarker amount and/or activity measurement(s) can be any suitable standard. For example, the pre-determined biomarker amount and/or activity measurement(s) can be obtained from the same or a different human for whom a patient selection is being assessed. In one embodiment, the pre-determined biomarker amount and/or activity measurement(s) can be obtained from a previous assessment of the same patient. In such a manner, the progress of the selection of the patient can be monitored over time. In addition, the control can be obtained from an assessment of another human or multiple humans, e.g., selected groups of humans, if the subject is a human. In such a manner, the extent of the selection of the human for whom selection is being assessed can be compared to suitable other humans, e.g., other humans who are in a similar situation to the human of interest, such as those suffering from similar or the same condition(s) and/or of the same ethnic group.


An “RNA interfering agent” as used herein, is defined as any agent which interferes with or inhibits expression of a target biomarker gene by RNA interference (RNAi). Such RNA interfering agents include, but are not limited to, nucleic acid molecules including RNA molecules which are homologous to the target biomarker gene of the present invention, or a fragment thereof, short interfering RNA (siRNA), and small molecules which interfere with or inhibit expression of a target biomarker nucleic acid by RNA interference (RNAi).


“RNA interference (RNAi)” is an evolutionally conserved process whereby the expression or introduction of RNA of a sequence that is identical or highly similar to a target biomarker nucleic acid results in the sequence specific degradation or specific post-transcriptional gene silencing (PTGS) of messenger RNA (mRNA) transcribed from that targeted gene (see Coburn and Cullen (2002) J. Virol. 76:9225), thereby inhibiting expression of the target biomarker nucleic acid. In one embodiment, the RNA is double stranded RNA (dsRNA). This process has been described in plants, invertebrates, and mammalian cells. In nature, RNAi is initiated by the dsRNA-specific endonuclease Dicer, which promotes processive cleavage of long dsRNA into double-stranded fragments termed siRNAs. “Short interfering RNA” (siRNA), also referred to herein as “small interfering RNA” is defined as an agent which functions to inhibit expression of a target biomarker nucleic acid, e.g., by RNAi. An siRNA may be chemically synthesized, may be produced by in vitro transcription, or may be produced within a host cell. In one embodiment, siRNA is a double stranded RNA (dsRNA) molecule of about 15 to about 40 nucleotides in length, preferably about 15 to about 28 nucleotides, more preferably about 19 to about 25 nucleotides in length, and more preferably about 19, 20, 21, or 22 nucleotides in length, and may contain a 3′ and/or 5′ overhang on each strand having a length of about 0, 1, 2, 3, 4, or 5 nucleotides. The length of the overhang is independent between the two strands, i.e., the length of the overhang on one strand is not dependent on the length of the overhang on the second strand. Preferably the siRNA is capable of promoting RNA interference through degradation or specific post-transcriptional gene silencing (PTGS) of the target messenger RNA (mRNA). In another embodiment, an siRNA is a small hairpin (also called stem loop) RNA (shRNA). In one embodiment, these shRNAs are composed of a short (e.g., 19-25 nucleotide) antisense strand, followed by a 5-9 nucleotide loop, and the analogous sense strand. Alternatively, the sense strand may precede the nucleotide loop structure and the antisense strand may follow. These shRNAs may be contained in plasmids, retroviruses, and lentiviruses and expressed from, for example, the pol III U6 promoter, or another promoter (see, e.g., Stewart, et al. (2003) RNA April; 9(4):493-501 incorporated by reference herein).


RNA interfering agents, e.g., siRNA molecules, may be administered to a patient having or at risk for having an MDS and/or an anemia, to inhibit expression of a biomarker gene which is overexpressed in an MDS and/or an anemia, and thereby treat, prevent, or inhibit the MDS and/or the anemia.


siRNAs are incorporated into a protein complex that recognizes and cleaves target mRNAs. RNAi can also be initiated by introducing nucleic acid molecules, e.g., synthetic siRNAs or RNA interfering agents, to inhibit or silence the expression of target biomarker nucleic acids. As used herein, “inhibition of target biomarker nucleic acid expression” or “inhibition of marker gene expression” includes any decrease in expression or protein activity or level of the target biomarker nucleic acid or protein encoded by the target biomarker nucleic acid. The decrease may be of at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 99% or more as compared to the expression of a target biomarker nucleic acid or the activity or level of the protein encoded by a target biomarker nucleic acid which has not been targeted by an RNA interfering agent.


RNAi agent disclosed herein may target any one of the nucleic acids listed in Table 1. In some embodiments, any one of the RNAi agents may be complementary to any one of the nucleic acid sequences in Table 1.


The term “sample” used for detecting or determining the presence or level of at least one biomarker is typically brain tissue, cerebrospinal fluid, whole blood, plasma, serum, saliva, urine, stool (e.g., feces), tears, and any other bodily fluid (e.g., as described above under the definition of “body fluids”), or a tissue sample (e.g., biopsy) such as a small intestine, colon sample, or surgical resection tissue. In certain instances, the method of the present invention further comprises obtaining the sample from the individual prior to detecting or determining the presence or level of at least one marker in the sample.


The term “subject” refers to any healthy animal, mammal or human, or any animal, mammal or human afflicted with a red blood cell disorder. The term “subject” is interchangeable with “patient.”


The term “therapeutic effect” refers to a local or systemic effect in animals, particularly mammals, and more particularly humans, caused by a pharmacologically active substance. The term thus means any substance intended for use in the diagnosis, cure, mitigation, treatment or prevention of disease or in the enhancement of desirable physical or mental development and conditions in an animal or human.


The terms “therapeutically-effective amount” and “effective amount” as used herein means that amount of a compound, material, or composition comprising a compound encompassed by the present invention which is effective for producing some desired therapeutic effect in at least a sub-population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment. Toxicity and therapeutic efficacy of subject compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 and the ED50.


Compositions that exhibit large therapeutic indices are preferred. In some embodiments, the LD50 (lethal dosage) can be measured and can be, for example, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1000% or more reduced for the agent relative to administration of a suitable control agent. Similarly, the ED50 (i.e., the concentration which achieves a half-maximal inhibition of symptoms) can be measured and can be, for example, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1000% or more increased for the agent relative to administration of a suitable control agent.


II. Subjects

In some embodiments, the subject is a mammal (e.g., mouse, rat, primate, non-human mammal, domestic animal, such as a dog, cat, cow, horse, and the like), and is preferably a human. In other embodiments, the subject is an animal model of a red blood cell disorder. In some embodiments, the subject is not limited to animals or humans with genetic mutation in Riok2 or other ribosomal or non-ribosomal protein mutations. For example, anti-IL-22 treatment of wild type (wt) mice undergoing acute anemia was determined herein to increase peripheral blood red blood cells as compared to mice treated with an isotype antibody.


In addition, cells can be used according to the methods described herein, whether in vitro, ex vivo, or in vivo, such as cells from such subjects. In some embodiments, the cells are a collection of erythroid progenitors and/or erythroid progenitors defined according to developmental stage (e.g., I, II, III, and IV, expression of biomarkers of interest such as IL-22 or an IL-22 receptor like IL-22RA1, IL-10Rbeta, and heterodimers thereof, and combinations thereof).


In some embodiments encompassed by the methods of the present invention, the subject has not undergone treatment, such as with lenalidomide, azacitidine, decitabine, or an erythropoiesis-stimulating agent. In other embodiments, the subject has undergone treatment, such as with lenalidomide, azacitidine, decitabine, or an erythropoiesis-stimulating agent.


The methods encompassed by the present invention can be used across many different red blood cell disorders in subjects such as those described herein. The red blood cell disorders that can be treated with the disclosed methods include myelodysplastic syndromes (MDS) and anemias, such as, without limitation, anemia caused by insufficiency of serine/threonine-protein kinase RIOK2, anemia caused by mutations or deletions on human chromosome 5, macrocytic anemia, anemia associated with increased levels of IL-22, chronic kidney disease (CKD), stress-induced anemia, Diamond Blackfan anemia, and Shwachman-Diamond syndrome.


The ordinarily skilled artisan will appreciate from the results of a wide variety of experimental models described herein that the methods encompassed by the present invention apply generally to a subject having an MDS and/or an anemia, such as those indicating increased IL-22 signaling and/or activation, and are not limited to individuals having particular genetic mutations. In some particular embodiments, subjects have an MDS and/or an anemia defined according to a genetic mutation, such as a del(5q)-mediated MDS. In some embodiments, MDS/anemia patients have increased IL-22 levels in serum, plasma, Th22 T lymphocytes, or bone marrow fluid.


The methods encompassed by the present invention can be used to stratify subjects and/or determine responsiveness of subjects described herein to IL-22 signaling pathway modulation.


III. Therapeutic Agents

In some embodiments, the agents used are therapeutic agents that are down-regulators of IL-22 signaling. These agents can block or neutralize, at least to some extent, the biological activity or function of IL-22 or the biological activity or function of an IL-22 receptor.


In some embodiments, the down-regulator of IL-22 signaling is an antibody (or an antigen-binding fragment thereof) that binds to IL-22. Recombinant IL-22 is available from multiple vendors including PeproTech (Cat #AF-210-22-250UG), and various antibodies can be generated against IL-22 using IL-22 (or a fragment thereof) as the antigen. In addition, certain anti-IL-22 antibodies are already commercially available. For example, a human/mouse anti-IL-22 neutralizing antibody is available from Thermo Fisher Scientific (Cat #16-7222-85).


Structural information for IL-22, its receptor, and the IL22/IL22R1 receptor-ligand complex is well-known in the art (Nagem et al. (2002) Structure 10(8):1051-62; Xu et al. (2005) Acta Crystallogr D Biol Crystallogr. 61 (Pt 7):942-50; Bleicher et al. (2008) FEBS Lett. 582(20):2985-92; Jones et al. (2008) Structure 16(9):1333-44), such that structure-function relationships between agents that block or neutralize IL-22, including anti-IL-22 agents and anti-IL-22 receptor agents, and the mechanism of action are well-known in the art (see, for example, human IL-22 crystal structure information at PubMed identifier PMID 12176383 and PMID 15983417, as well as human IL-22/IL22-R1 complex crystal structure information at PubMed identifier PMID 18675809 and PMID 18599299). Structure-function relationships among non-human orthologs of IL-22 and IL-22 receptor are similarly well-known in the art. Mouse and human IL-22 share 78% protein sequence identity. Mouse and human IL22ra1 share 72% protein sequence identity. IL-22BP shares a 34% sequence identity with the extracellular region of IL-22RA1.


The term “down-regulator of IL-22 signaling or signaling pathway” includes any natural or non-natural agent prepared, synthesized, manufactured, and/or purified by humans that is capable of reducing, inhibiting, blocking, preventing, and/or that inhibits the IL-22 signaling pathway, including inhibition of IL-22 polypeptide (and its fragments, domains, and/or motifs thereof, discussed herein) directly. In one embodiment, such inhibitors may reduce or inhibit the binding/interaction between IL-22 and its substrates or other binding partners. In another embodiment, such inhibitors may reduce or inhibit an upstream and/or downstream member of the IL-22 signaling pathway. In still another embodiment, such inhibitors may increase or promote the turnover rate, reduce or inhibit the expression and/or the stability (e.g., the half-life) of IL-22, resulting in at least a decrease in IL-22 levels and/or activity. Such inhibitors may be any molecule, including but not limited to small molecule compounds, antibodies or intrabodies, RNA interfering (RNAi) agents (including at least siRNAs, shRNAs, microRNAs (miRNAs), piwi, and other well-known agents). Such inhibitors may be specific to IL-22 or also inhibit at least one IL-22 signaling pathway member. RNA interference agents for IL-22 polypeptides are well-known and commercially available (e.g., human or mouse shRNA (Cat. #TL303948, TR502849, TL303948V, etc.) products, siRNA products (Cat. #SR323991, SR404300, etc.), and human or mouse gene knockout kit via CRISPR (Cat. #KN409995, KN209995, etc.) from Origene (Rockville, MD), siRNA/shRNA products (Cat. #sc-39664, sc-39665, etc.) and human or mouse gene knockout kit via CRISPR (Cat. #sc-403228) from Santa Cruz Biotechnology (Dallas, Texas), and siRNA/shRNA products (Cat. #ABIN5784850, ABIN5784849, etc.) from Genomics Online (Limerick, PA). Methods for detection, purification, and/or inhibition of IL-22 (e.g., by anti-IL-22 antibodies) are also well-known and commercially available (e.g., multiple anti-IL-22 antibodies from Origene (Cat. #PP1224B1, TA326688, TA328247, TA337159, TA338422, PP1226, TA328506, TA328507, etc.), Novus Biologicals (Littleton, CO, Cat. #AF582, AF782, NBP2-27339, NB100-737, MAB582, MAB7821, NBP2-31215, NBP2-11699, NBP2-41245, MAB7822, NBP2-27322, NBP2-27360, MAP782, MAP5821, NBP2-27320, NBP2-27321, MAB7822R, NB100-733, NB100-738, H00050616-D01P, etc.), abcam (Cambridge, MA, Cat. #ab18498, ab203211, ab134035, ab227033, ab263954, ab18564, ab181007, ab109819, ab267467, ab267789, ab228687, ab96341, ab133545, ab211756, ab18566, ab84033, ab84225, ab174534, ab193813, ab90937, ab222646, ab222645, ab206858, ab5984, ab18568, ab211675, ab167213, ab232925, ab5982, ab98917, etc.), patent literature (e.g., U.S. Pat. No. 7,901,684), and the like). IL-22 knockout human cell lines are also well-known and available at Horizon (Cambridge, UK, Cat. #HZGHC50626). Reagents and kits for assaying IL-22 are well-known in the art (see, for example SMC™ Human IL-22 High Sensitivity Immunoassay Kit; EMD Millipore, product #03-0162-00).


Similarly, compositions for modulation (e.g., down-regulation), as well as detection and purification of IL-22 signaling pathway members, such as IL-22RA1, alarmins (e.g., S100A8, S100A9, S100 A10, phosphorylated Stat3, etc.), Camp, Ngp, and the like, are also well-known in the art. For example, RNA interference agents for IL-22RA1 polypeptides are well-known and commercially available (e.g., human or mouse shRNA (Cat. #TL303947V, TR303947, TR506901, TL506901V, etc.) products, siRNA products (Cat. #SR324794, SR417732, etc.), and human or mouse gene knockout kit via CRISPR (Cat. #KN406447, KN206447, etc.) from Origene (Rockville, MD), siRNA/shRNA products (Cat. #sc-146217, sc-88174, etc.) and human or mouse gene knockout kit via CRISPR (Cat. #sc-404104, etc.) from Santa Cruz Biotechnology (Dallas, Texas), and siRNA/shRNA products (Cat. #ABIN4120152, ABIN4163636, ABIN4120153, ABIN4163637, ABIN5353047, ABIN5353048, ABIN5353045, ABIN5353044, ABIN3401988, ABIN3430223, ABIN3396755, ABIN3818550, ABIN4013677, ABIN4013676, ABIN3818551, etc.) from Genomics Online (Limerick, PA). Methods for detection, purification, and/or inhibition of IL-22RA1 (e.g., by anti-IL-22RA1 antibodies) are also well-known and commercially available (e.g., multiple anti-IL-22RA1 antibodies from Origene (Cat. #AP07312PU-N, TA306082, TA306086, TA322224, TA322225, TA338469, TA338470, etc.), Novus Biologicals (Littleton, CO, Cat. #MAB42941, MAB2770, NBP1-76724, AF2770, MAB4294, AF4294, NB100-740, etc.), Antibodies-Online (Limerick, PA, Cat. #ABIN2783836, ABIN2783835, ABIN4899326, ABIN4899325, ABIN4895922, ABIN6742083, ABIN4895924, ABIN4324949, ABIN748178, ABIN6743529, etc.), and the like). IL-22 knockout human cell lines are also well-known and available at Horizon (Cambridge, UK, Cat. #HZGHC58985).


In some embodiments, the down-regulator of IL-22 signaling includes IL22JOP™ monoclonal antibody, fezakinumab, or a combination thereof.


In some embodiments, the down-regulator of IL-22 signaling includes an antibody (or an antigen-binding fragment thereof) directed against IL-22RA1. In certain embodiments, the down-regulator of IL-22 signaling includes an antibody (or an antigen-binding fragment thereof) directed against IL-22RA1/IL-10R2-heterodimer.


In certain embodiments, the down-regulator of IL-22 signaling includes IL-22 binding protein (IL-22BP) or a fragment of IL-22BP that can bind IL-22 and down-regulate its signaling.


In some embodiments, the down-regulator of IL-22 signaling includes an antagonist of aryl hydrocarbon receptor (AHR). For example, such a down-regulator can include stemregenin 1, CH-223191, or 6,2′,4′-trimethoxyflavone.


In some embodiments, an agent disclosed herein or a down-regulator of IL-22 signaling includes any agent that specific binds to or decreases the activity or level of any one of the biomarkers listed in Table 1.


In certain embodiments, the down-regulator of IL-22 signaling can be conjointly administered (e.g., separately or together, at different times or at the same time) with another therapeutic agent. Such therapeutic agents for a combination therapy include lenalidomide, azacitidine, decitabine, or a combination thereof. Such therapeutic agents for a combination therapy also include erythropoiesis-stimulating agents, such as erythropoietin, epoetin alfa, epoetin beta, epoetin omega, epoetin zeta, darbepoetin alfa, or a combination thereof. In some embodiments, lenalidomide, azacitidine, or decitabine is not conjointly administered with one of the erythropoiesis-stimulating agents, for example if the two are contraindicated.


IV. Therapeutic Methods

One aspect encompassed by the present invention pertains to methods of treating one or more red blood cell disorders in a subject. Such methods include administering to the subject an effective amount of a down-regulator of interleukin-22 (IL-22) signaling.


As an example, a method of treating anemia in a subject, according to some of the disclosed embodiments herein, includes administering to the subject fezakinumab.


Another aspect encompassed by the present invention pertains to methods of promoting differentiation from an erythroid progenitor cell toward a mature red blood cell in a subject. Such methods include administering to the subject an effective amount of a down-regulator of interleukin-22 (IL-22) signaling.


As an example, in a method according to some of the disclosed embodiments herein, fezakinumab is administered to a subject, after which erythroid progenitor cells classified as RI differentiate toward mature red blood cells (e.g., by first differentiating into erythroid progenitor cells classified as RII).


In connection with the therapeutic methods described above, an aspect encompassed by the present invention relates to methods of selecting a subject for treatment with a down-regulator of interleukin-22 (IL-22) signaling. Such a method includes determining that a subject has a chromosome 5 that comprises a mutation in its long arm; and selecting the subject for treatment with a down-regulator of IL-22 signaling.


The mutation for these methods can be any mutation that has been or is associated with MDS or another red blood cell disorder, such as anemia. For example, the mutation can include deletions in the q33.1, q33.2, q33.3 regions of human chromosome 5. In addition, the mutation can include a deletion in q15 region of human chromosome 5. In particular, the mutation can be a mutation in the RIOK2 gene. In some embodiments, the mutation results in an I245T mutation in the RIOK2 protein, defined with respect to SEQ ID NO: 1 provided as follows.









| Q9BVS4|RIOK2_HUMAN Serine/threonine-protein


kinase RIOK2


SEQ ID NO: 1 


MGKVNVAKLRYMSRDDFRVLTAVEMGMKNHEIVPGSLIASIASLKHGGCN





KVLRELVKHKLIAWERTKTVQGYRLTNAGYDYLALKTLSSRQVVESVGNQ





MGVGKESDIYIVANEEGQQFALKLHRLGRTSFRNLKNKRDYHKHRHNVSW





LYLSRLSAMKEFAYMKALYERKFPVPKPIDYNRHAVVMELINGYPLCQIH





HVEDPASVYDEAMELIVKLANHGLIHGDFNEFNLILDESDHITMIDFPQM





VSTSHPNAEWYFDRDVKCIKDFFMKRFSYESELFPTFKDIRREDTLDVEV





SASGYTKEMQADDELLHPLGPDDKNIETKEGSEFSFSDGEVAEKAEVYGS





ENESERNCLEESEGCYCRSSGDPEQIKEDSLSEESADARSFEMTEFNQAL





EEIKGQVVENNSVTEFSEEKNRTENYNRQDGQRVQGGVPAGSDEYEDECP





HLIALSSLNREFRPERDEENVGAMNQYRTRTLSITSSGSAVSCSTIPPEL





VKQKVKRQLTKQQKSAVRRRLQKGEANIFTKQRRENMQNIKSSLEAASFW





GE






The I245T mutation or any of the other mutations can be identified by sequencing (e.g., via high-throughput DNA sequencing) a nucleic acid from the subject.


Similar chromosomal regions, mutations, and the like are well-known in orthologs of non-human mammals, such as mice, and are contemplated for use according to the present invention.


The therapeutic methods described herein can also be used in a variety of in vitro and in vivo applications, such as in analyzing cellular models of an MDS and/or an anemia, without treating a subject. Such methods involved contacting a cell, such as an erythroid progenitor, with a modulatory agent described herein.


V. Further Uses and Methods Encompassed by the Present Invention

The methods and compositions described herein can be used in a variety of screening, diagnostic, and prognostic applications in addition to therapeutic applications. In any method described herein, such as a diagnostic method, prognostic method, therapeutic method, or combination thereof, all steps of the method can be performed by a single actor or, alternatively, by more than one actor. For example, diagnosis can be performed directly by the actor providing therapeutic treatment. Alternatively, a person providing a therapeutic agent can request that a diagnostic assay be performed. The diagnostician and/or the therapeutic interventionist can interpret the diagnostic assay results to determine a therapeutic strategy. Similarly, such alternative processes can apply to other assays, such as prognostic assays.


a. Screening Methods


One aspect of the present invention relates to screening assays, including non-cell-based assays and animal model assays. In one embodiment, the assays provide a method for identifying whether an agent is useful for treating an MDS and/or an anemia condition, such as by identifying agents that modulate an IL-22 signaling pathway inhibitor (e.g., one or more biomarkers listed in Table 1).


In one embodiment, the present invention relates to assays for screening test agents which bind to, or modulate the biological activity of, at least one biomarker described herein (e.g., in the Tables, Figures, Examples, or otherwise in the specification). In one embodiment, a method for identifying such an agent entails determining the ability of the agent to modulate, e.g. inhibit, the at least one biomarker described herein.


In one embodiment, an assay is a cell-free or cell-based assay, comprising contacting at least one biomarker described herein, with a test agent, and determining the ability of the test agent to modulate (e.g., inhibit) the activity of the biomarker, such as by measuring direct binding of substrates or by measuring indirect parameters as described below, and optionally further determining the effect on treating an MDS and/or an anemia.


For example, in a direct binding assay, biomarker protein (or their respective target polypeptides or molecules) can be coupled with a radioisotope or enzymatic label such that binding can be determined by detecting the labeled protein or molecule in a complex. For example, the targets can be labeled with 125I, 35S, 14C or 3H, either directly or indirectly, and the radioisotope detected by direct counting of radioemission or by scintillation counting. Alternatively, the targets can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product. Determining the interaction between biomarker and substrate can also be accomplished using standard binding or enzymatic analysis assays. In one or more embodiments of the above described assay methods, it may be desirable to immobilize polypeptides or molecules to facilitate separation of complexed from uncomplexed forms of one or both of the proteins or molecules, as well as to accommodate automation of the assay.


Binding of a test agent to a target can be accomplished in any vessel suitable for containing the reactants. Non-limiting examples of such vessels include microtiter plates, test tubes, and micro-centrifuge tubes. Immobilized forms of the antibodies described herein can also include antibodies bound to a solid phase like a porous, microporous (with an average pore diameter less than about one micron) or macroporous (with an average pore diameter of more than about 10 microns) material, such as a membrane, cellulose, nitrocellulose, or glass fibers; a bead, such as that made of agarose or polyacrylamide or latex; or a surface of a dish, plate, or well, such as one made of polystyrene.


In an alternative embodiment, determining the ability of the agent to modulate the interaction between the biomarker and a substrate or a biomarker and its natural binding partner can be accomplished by determining the ability of the test agent to modulate the activity of a polypeptide or other product that functions downstream or upstream of its position within the signaling pathway (e.g., feedback loops). Such feedback loops are well-known in the art (see, for example, Chen and Guillemin (2009) Int. J. Tryptophan Res. 2:1-19).


The present invention further encompasses novel agents identified by the above-described screening assays. Accordingly, it is within the scope of the present invention to further use an agent identified as described herein, such as in an appropriate animal model. For example, an agent identified as described herein can be used in an animal model to determine the efficacy, toxicity, or side effects of treatment with such an agent. Alternatively, an agent, such as an antibody, identified as described herein can be used in an animal model to determine the mechanism of action of such an agent.


b. Diagnostic and Predictive Medicine


The present invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, and monitoring clinical trials are used for prognostic (predictive) purposes to thereby stratify subject populations and/or treat an individual prophylactically. Accordingly, one aspect of the present invention relates to diagnostic assays for determining the amount and/or activity level of a biomarker described herein in the context of a biological sample (e.g., blood, serum, cells, or tissue) to thereby determine whether an individual afflicted with an MDS and/or an anemia is likely to respond to biomarker inhibitor treatments. Such assays can be used for prognostic or predictive purpose alone or can be coupled with a therapeutic intervention to thereby prophylactically treat an individual prior to the onset or after recurrence of a disorder characterized by or associated with biomarker polypeptide, nucleic acid expression or activity. The ordinarily skilled artisan will appreciate that any method can use one or more (e.g., combinations) of biomarkers described herein, such as those in the Tables, Figures, Examples, and otherwise described in the specification.


Another aspect of the present invention pertains to monitoring the influence of agents (e.g., drugs, compounds, and small nucleic acid-based molecules) on the expression or activity of a biomarker described herein. These and other agents are described in further detail in the following sections.


The ordinarily skilled artisan will also appreciate that, in certain embodiments, the methods of the present invention may implement a computer program and computer system. For example, a computer program can be used to perform the algorithms described herein. A computer system can also store and manipulate data generated by the methods of the present invention which comprises a plurality of biomarker signal changes/profiles which can be used by a computer system in implementing the methods of this invention. In certain embodiments, a computer system receives biomarker expression data; (ii) stores the data; and (iii) compares the data in any number of ways described herein (e.g., analysis relative to appropriate controls) to determine the state of informative biomarkers from tissue of interest. In other embodiments, a computer system (i) compares the determined expression biomarker level to a threshold value; and (ii) outputs an indication of whether said biomarker level is significantly modulated (e.g., above or below) the threshold value, or a phenotype based on said indication.


In certain embodiments, such computer systems are also considered part of the present invention. Numerous types of computer systems can be used to implement the analytic methods of this invention according to knowledge possessed by a skilled artisan in the bioinformatics and/or computer arts. Several software components can be loaded into memory during operation of such a computer system. The software components can comprise both software components that are standard in the art and components that are special to the present invention (e.g., dCHIP software described in Lin et al. (2004) Bioinformatics 20, 1233-1240; radial basis machine learning algorithms (RBM) known in the art).


The methods encompassed by the present invention can also be programmed or modeled in mathematical software packages that allow symbolic entry of equations and high-level specification of processing, including specific algorithms to be used, thereby freeing a user of the need to procedurally program individual equations and algorithms. Such packages include, e.g., Matlab from Mathworks (Natick, Mass.), Mathematica from Wolfram Research (Champaign, Ill.) or S-Plus from MathSoft (Seattle, Wash.).


In certain embodiments, the computer comprises a database for storage of biomarker data. Such stored profiles can be accessed and used to perform comparisons of interest at a later point in time. For example, biomarker expression profiles of a sample derived from the tissue of a subject not afflicted with an MDS and/or an anemia and/or profiles generated from population-based distributions of informative loci of interest in relevant populations of the same species can be stored and later compared to that of a sample derived from tissue of instructed, such as tissue suspected of being relevant to an MDS and/or an anemia of the subject.


In addition to the exemplary program structures and computer systems described herein, other, alternative program structures and computer systems will be readily apparent to the skilled artisan. Such alternative systems, which do not depart from the above described computer system and programs structures either in spirit or in scope, are therefore intended to be comprehended within the accompanying claims.


As an additional aspect encompassed by the present invention, methods of detecting a level of interleukin-22 (IL-22) signaling are disclosed. Such methods include determining an expression level of one or more biomarkers listed in Table 1. For example, S100A8, an IL-22 target gene, contributes to the dyserythropoiesis seen in anemia and MDS. S100A8 and other biomarkers listed in the Tables, Figures, Examples, and otherwise described in the specification can be used as a measure of functional inhibition of IL-22 signaling.


Prognostic assay methods are also provided that may be used to identify subjects having or at risk of developing an MDS and/or an anemia that is likely or unlikely to be responsive to a modulator of IL-22 signaling. Assays described herein, such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with a dysregulation of the amount or activity of at least one biomarker described herein, such as in an MDS and/or an anemia. Alternatively, the prognostic assays can be utilized to identify a subject having or at risk for developing a disorder associated with a dysregulation of at least one biomarker described herein, such as in an MDS and/or an anemia. Furthermore, the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, polypeptide, peptide, nucleic acid, small molecule, or other drug candidate) to treat a disease or disorder associated with the aberrant biomarker expression or activity.


The present invention provides, in part, methods, systems, and code for accurately classifying whether a biological sample is associated with an MDS and/or an anemia that is likely to respond to a modulator (e.g., inhibitor) of IL-22 pathway signaling. In some embodiments, the present invention is useful for classifying a sample (e.g., from a subject) as associated with or at risk for responding to or not responding to IL-22 pathway signaling modulation (e.g., inhibition) using a statistical algorithm and/or empirical data (e.g., the amount or activity of a biomarker described herein, such as in the Tables, Figures, Examples, and otherwise described in the specification).


An exemplary method for detecting the amount or activity of a biomarker described herein, and thus useful for classifying whether a sample (e.g., a sample from a subject having an MDS and/or an anemia or an in vitro model of an MDS and/or an anemia) is likely or unlikely to respond to IL-22 pathway signaling modulation (e.g., inhibition) involves obtaining a biological sample from a test subject and contacting the biological sample with an agent, such as a protein-binding agent like an antibody or antigen-binding fragment thereof, or a nucleic acid-binding agent like an oligonucleotide, capable of detecting the amount or activity of the biomarker in the biological sample. In some embodiments, at least one antibody or antigen-binding fragment thereof is used, wherein two, three, four, five, six, seven, eight, nine, ten, or more such antibodies or antibody fragments can be used in combination (e.g., in sandwich ELISAs) or in serial. In certain instances, the statistical algorithm is a single learning statistical classifier system. For example, a single learning statistical classifier system can be used to classify a sample as a based upon a prediction or probability value and the presence or level of the biomarker. The use of a single learning statistical classifier system typically classifies the sample as, for example, a likely responder or non-responder with a sensitivity, specificity, positive predictive value, negative predictive value, and/or overall accuracy of at least about 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.


Other suitable statistical algorithms are well-known to those of ordinary skill in the art. For example, learning statistical classifier systems include a machine learning algorithmic technique capable of adapting to complex data sets (e.g., panel of markers of interest) and making decisions based upon such data sets. In some embodiments, a single learning statistical classifier system such as a classification tree (e.g., random forest) is used. In other embodiments, a combination of 2, 3, 4, 5, 6, 7, 8, 9, 10, or more learning statistical classifier systems are used, preferably in tandem. Examples of learning statistical classifier systems include, but are not limited to, those using inductive learning (e.g., decision/classification trees such as random forests, classification and regression trees (C&RT), boosted trees, etc.), Probably Approximately Correct (PAC) learning, connectionist learning (e.g., neural networks (NN), artificial neural networks (ANN), neuro fuzzy networks (NFN), network structures, perceptrons such as multi-layer perceptrons, multi-layer feed-forward networks, applications of neural networks, Bayesian learning in belief networks, etc.), reinforcement learning (e.g., passive learning in a known environment such as naive learning, adaptive dynamic learning, and temporal difference learning, passive learning in an unknown environment, active learning in an unknown environment, learning action-value functions, applications of reinforcement learning, etc.), and genetic algorithms and evolutionary programming. Other learning statistical classifier systems include support vector machines (e.g., Kernel methods), multivariate adaptive regression splines (MARS), Levenberg-Marquardt algorithms, Gauss-Newton algorithms, mixtures of Gaussians, gradient descent algorithms, and learning vector quantization (LVQ). In certain embodiments, the method of the present invention further comprises sending the sample classification results to a clinician, e.g., a hematologist.


In another embodiment, the diagnosis of a subject is followed by administering to the individual a therapeutically effective amount of a defined treatment based upon the diagnosis.


In one embodiment, the methods further involve obtaining a control biological sample (e.g., biological sample from a subject who does not have an MDS and/or an anemia of interest or a sample that is susceptible to biomarker inhibitor treatment), a biological sample from the subject during remission, or a biological sample timepoint during treatment for the condition.


c. Clinical Efficacy


Similarly, clinical efficacy can be measured by any method known in the art. For example, the benefit from a therapy with an agent that down-regulates IL-22 signaling, alone or in combination with a another agent, such as lenalidomide, azacitidine, decitabine, or an erythropoiesis-stimulating agent (e.g., erythropoietin, epoetin alfa, epoetin beta, epoetin omega, epoetin zeta, darbepoetin alfa, IL-9), relates to an increase in the level of healthy red blood cells so that adequate oxygen can be carried to the tissues of the subject. As another example, the benefit from an anti-IL-22 therapy can relate to the level of red blood cells in the blood (e.g., hematocrit) or the level of hemoglobin in the blood, both of which can be measured as part of a routine complete blood count.


The benefit from using agents encompassed by the present invention can be determined by measuring the level of cytotoxicity in a biological material. The benefit from using agents encompassed by the present invention can be assessed by measuring transcription profiles, viability curves, microscopic images, biosynthetic activity levels, redox levels, and the like. The benefit from using agents encompassed by the present invention can also be determined by measuring the presence and severity of side effects from the anti-IL-22 treatment such as autoimmune or allergic sequelae. IL-22 signaling normally function to maintain epithelial integrity in lungs, skin and GI tract, induction of antibacterial proteins, protection against cellular damage, liver progenitor cell proliferation


In some embodiments, clinical efficacy of the therapeutic treatments described herein can be determined by measuring the clinical benefit rate (CBR). The clinical benefit rate is measured by determining the sum of the percentage of patients who are in complete remission (CR), the number of patients who are in partial remission (PR) and the number of patients having stable disease (SD) at a time point at least 6 months out from the end of therapy. The shorthand for this formula is CBR=CR+PR+SD over 6 months. In some embodiments, the CBR for a particular therapeutic regimen is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, or more.


Additional criteria for evaluating the response to therapies are related to “survival,” which includes all of the following: survival until mortality, also known as overall survival (wherein said mortality can be either irrespective of cause or tumor related); “recurrence-free survival” (wherein the term recurrence shall include both localized and distant recurrence); disease free survival. The length of said survival can be calculated by reference to a defined start point (e.g., time of diagnosis or start of treatment) and end point (e.g., death, recurrence). In addition, criteria for efficacy of treatment can be expanded to include response to therapy, probability of survival, and probability of recurrence.


For example, in order to determine appropriate threshold values, a particular anti-IL-22 therapeutic regimen can be administered to a population of subjects and the outcome can be correlated to biomarker measurements detailed previously that were determined prior to administration of any therapy. The outcome measurement can be pathologic response to therapy. Alternatively, outcome measures, such as overall survival and disease-free survival can be monitored over a period of time for subjects following therapies for whom biomarker measurement values are known as detailed previously. In certain embodiments, the same doses of therapy agents, if any, are administered to each subject. In related embodiments, the doses administered are standard doses known in the art for those agents used in therapies. The period of time for which subjects are monitored can vary. For example, subjects can be monitored for at least 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, 55, or 60 months.


d. Biomarker Analyses


Methods analyzing biomarkers encompassed by the present invention may be performed according to well-known techniques in the art. In some embodiments, biomarker amount and/or activity measurement(s) in a sample from a subject is compared to a predetermined control (standard) sample. The sample from the subject is typically from a diseased tissue. The control sample can be from the same subject or from a different subject. The control sample is typically a normal, non-diseased sample. However, in some embodiments, such as for staging of disease or for evaluating the efficacy of treatment, the control sample can be from a diseased tissue. The control sample can be a combination of samples from several different subjects. In some embodiments, the biomarker amount and/or activity measurement(s) from a subject is compared to a pre-determined level. This pre-determined level is typically obtained from normal samples. As described herein, a “pre-determined” biomarker amount and/or activity measurement(s) may be a biomarker amount and/or activity measurement(s) used to, by way of example only, evaluate a subject that may be selected for treatment (e.g., based on the number of genomic mutations and/or the number of genomic mutations causing non-functional proteins for DNA repair genes), evaluate a response to a modulator (e.g., an inhibitor) of one or more biomarkers listed in Table 1 and/or evaluate a response to a modulator (e.g., an inhibitor) of one or more biomarkers listed in Table 1. A pre-determined biomarker amount and/or activity measurement(s) may be determined in populations of patients with or without an MDS and/or an anemia. The pre-determined biomarker amount and/or activity measurement(s) can be a single number, equally applicable to every patient, or the pre-determined biomarker amount and/or activity measurement(s) can vary according to specific subpopulations of patients. Age, weight, height, and other factors of a subject may affect the pre-determined biomarker amount and/or activity measurement(s) of the individual. Furthermore, the pre-determined biomarker amount and/or activity can be determined for each subject individually. In one embodiment, the amounts determined and/or compared in a method described herein are based on absolute measurements.


In another embodiment, the amounts determined and/or compared in a method described herein are based on relative measurements, such as ratios (e.g., biomarker copy numbers, level, and/or activity before a treatment vs. after a treatment, such biomarker measurements relative to a spiked or man-made control, such biomarker measurements relative to the expression of a housekeeping gene, and the like). For example, the relative analysis can be based on the ratio of pre-treatment biomarker measurement as compared to post-treatment biomarker measurement. Pre-treatment biomarker measurement can be made at any time prior to initiation of therapy for an MDS and/or an anemia. Post-treatment biomarker measurement can be made at any time after initiation of therapy. In some embodiments, post-treatment biomarker measurements are made 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 weeks or more after initiation of therapy, and even longer toward indefinitely for continued monitoring.


The pre-determined biomarker amount and/or activity measurement(s) can be any suitable standard. For example, the pre-determined biomarker amount and/or activity measurement(s) can be obtained from the same or a different human for whom a patient selection is being assessed. In one embodiment, the pre-determined biomarker amount and/or activity measurement(s) can be obtained from a previous assessment of the same patient. In such a manner, the progress of the selection of the patient can be monitored over time. In addition, the control can be obtained from an assessment of another human or multiple humans, e.g., selected groups of humans, if the subject is a human. In such a manner, the extent of the selection of the human for whom selection is being assessed can be compared to suitable other humans, e.g., other humans who are in a similar situation to the human of interest, such as those suffering from similar or the same condition(s) and/or of the same ethnic group.


In some embodiments of the present invention the change of biomarker amount and/or activity measurement(s) from the pre-determined level is about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, or 5.0 fold or greater, or any range in between, inclusive. Such cutoff values apply equally when the measurement is based on relative changes, such as based on the ratio of pre-treatment biomarker measurement as compared to post-treatment biomarker measurement.


Biological samples can be collected from a variety of sources from a patient including a body fluid sample, cell sample, or a tissue sample comprising nucleic acids and/or proteins. In a preferred embodiment, the subject and/or control sample is selected from the group consisting of cells, cell lines, histological slides, paraffin embedded tissues, biopsies, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, and bone marrow. In another preferred embodiment, the subject and/or control sample is selected from the group consisting of whole blood, serum, plasma, bone marrow fluid, and/or Th22 T lymphocytes in peripheral blood.


The samples can be collected from individuals repeatedly over a longitudinal period of time (e.g., once or more on the order of days, weeks, months, annually, biannually, etc.). Obtaining numerous samples from an individual over a period of time can be used to verify results from earlier detections and/or to identify an alteration in biological pattern as a result of, for example, disease progression, drug treatment, etc. For example, subject samples can be taken and monitored every month, every two months, or combinations of one, two, or three month intervals according to the present invention. In addition, the biomarker amount and/or activity measurements of the subject obtained over time can be conveniently compared with each other, as well as with those of normal controls during the monitoring period, thereby providing the subject's own values, as an internal, or personal, control for long-term monitoring.


Sample preparation and separation can involve any of the procedures, depending on the type of sample collected and/or analysis of biomarker measurement(s). Such procedures include, by way of example only, concentration, dilution, adjustment of pH, removal of high abundance polypeptides (e.g., albumin, gamma globulin, and transferrin, etc.), addition of preservatives and calibrants, addition of protease inhibitors, addition of denaturants, desalting of samples, concentration of sample proteins, extraction and purification of lipids.


The sample preparation can also isolate molecules that are bound in non-covalent complexes to other protein (e.g., carrier proteins). This process may isolate those molecules bound to a specific carrier protein (e.g., albumin), or use a more general process, such as the release of bound molecules from all carrier proteins via protein denaturation, for example using an acid, followed by removal of the carrier proteins.


Removal of undesired proteins (e.g., high abundance, uninformative, or undetectable proteins) from a sample can be achieved using high affinity reagents, high molecular weight filters, ultracentrifugation and/or electrodialysis. High affinity reagents include antibodies or other reagents (e.g., aptamers) that selectively bind to high abundance proteins. Sample preparation could also include ion exchange chromatography, metal ion affinity chromatography, gel filtration, hydrophobic chromatography, chromatofocusing, adsorption chromatography, isoelectric focusing and related techniques. Molecular weight filters include membranes that separate molecules on the basis of size and molecular weight. Such filters may further employ reverse osmosis, nanofiltration, ultrafiltration and microfiltration.


Ultracentrifugation is a method for removing undesired polypeptides from a sample. Ultracentrifugation is the centrifugation of a sample at about 15,000-60,000 rpm while monitoring with an optical system the sedimentation (or lack thereof) of particles. Electrodialysis is a procedure which uses an electromembrane or semipermable membrane in a process in which ions are transported through semi-permeable membranes from one solution to another under the influence of a potential gradient. Since the membranes used in electrodialysis may have the ability to selectively transport ions having positive or negative charge, reject ions of the opposite charge, or to allow species to migrate through a semipermable membrane based on size and charge, it renders electrodialysis useful for concentration, removal, or separation of electrolytes.


Separation and purification in the present invention may include any procedure known in the art, such as capillary electrophoresis (e.g., in capillary or on-chip) or chromatography (e.g., in capillary, column or on a chip). Electrophoresis is a method which can be used to separate ionic molecules under the influence of an electric field. Electrophoresis can be conducted in a gel, capillary, or in a microchannel on a chip. Examples of gels used for electrophoresis include starch, acrylamide, polyethylene oxides, agarose, or combinations thereof. A gel can be modified by its cross-linking, addition of detergents, or denaturants, immobilization of enzymes or antibodies (affinity electrophoresis) or substrates (zymography) and incorporation of a pH gradient. Examples of capillaries used for electrophoresis include capillaries that interface with an electrospray.


Capillary electrophoresis (CE) is preferred for separating complex hydrophilic molecules and highly charged solutes. CE technology can also be implemented on microfluidic chips. Depending on the types of capillary and buffers used, CE can be further segmented into separation techniques such as capillary zone electrophoresis (CZE), capillary isoelectric focusing (CLEF), capillary isotachophoresis (cITP) and capillary electrochromatography (CEC). An embodiment to couple CE techniques to electrospray ionization involves the use of volatile solutions, for example, aqueous mixtures containing a volatile acid and/or base and an organic such as an alcohol or acetonitrile.


Capillary isotachophoresis (cITP) is a technique in which the analytes move through the capillary at a constant speed but are nevertheless separated by their respective mobilities. Capillary zone electrophoresis (CZE), also known as free-solution CE (FSCE), is based on differences in the electrophoretic mobility of the species, determined by the charge on the molecule, and the frictional resistance the molecule encounters during migration which is often directly proportional to the size of the molecule. Capillary isoelectric focusing (CLEF) allows weakly-ionizable amphoteric molecules, to be separated by electrophoresis in a pH gradient. CEC is a hybrid technique between traditional high performance liquid chromatography (HPLC) and CE.


Separation and purification techniques used in the present invention include any chromatography procedures known in the art. Chromatography can be based on the differential adsorption and elution of certain analytes or partitioning of analytes between mobile and stationary phases. Different examples of chromatography include, but not limited to, liquid chromatography (LC), gas chromatography (GC), high performance liquid chromatography (HPLC), etc.


Biomarker nucleic acids and/or biomarker polypeptides can be analyzed according to the methods described herein and techniques known to the skilled artisan to identify such genetic or expression alterations useful for the present invention including, but not limited to, 1) an alteration in the level of a biomarker transcript or polypeptide, 2) a deletion or addition of one or more nucleotides from a biomarker gene, 4) a substitution of one or more nucleotides of a biomarker gene, 5) aberrant modification of a biomarker gene, such as an expression regulatory region, and the like.


i. Methods for Detection of Copy Number


Methods of evaluating the copy number of a biomarker nucleic acid are well-known to those of skill in the art. The presence or absence of chromosomal gain or loss can be evaluated simply by a determination of copy number of the regions or markers identified herein.


In one embodiment, a biological sample is tested for the presence of copy number changes in genomic loci containing the genomic marker. A copy number of at least 3, 4, 5, 6, 7, 8, 9, or 10 is predictive of poorer outcome of inhibitors of one or more biomarkers listed in Table 1 and immunotherapy combination treatments.


Methods of evaluating the copy number of a biomarker locus include, but are not limited to, hybridization-based assays. Hybridization-based assays include, but are not limited to, traditional “direct probe” methods, such as Southern blots, in situ hybridization (e.g., FISH and FISH plus SKY) methods, and “comparative probe” methods, such as comparative genomic hybridization (CGH), e.g., cDNA-based or oligonucleotide-based CGH. The methods can be used in a wide variety of formats including, but not limited to, substrate (e.g. membrane or glass) bound methods or array-based approaches.


In one embodiment, evaluating the biomarker gene copy number in a sample involves a Southern Blot. In a Southern Blot, the genomic DNA (typically fragmented and separated on an electrophoretic gel) is hybridized to a probe specific for the target region. Comparison of the intensity of the hybridization signal from the probe for the target region with control probe signal from analysis of normal genomic DNA (e.g., a non-amplified portion of the same or related cell, tissue, organ, etc.) provides an estimate of the relative copy number of the target nucleic acid. Alternatively, a Northern blot may be utilized for evaluating the copy number of encoding nucleic acid in a sample. In a Northern blot, mRNA is hybridized to a probe specific for the target region. Comparison of the intensity of the hybridization signal from the probe for the target region with control probe signal from analysis of normal RNA (e.g., a non-amplified portion of the same or related cell, tissue, organ, etc.) provides an estimate of the relative copy number of the target nucleic acid. Alternatively, other methods well-known in the art to detect RNA can be used, such that higher or lower expression relative to an appropriate control (e.g., a non-amplified portion of the same or related cell tissue, organ, etc.) provides an estimate of the relative copy number of the target nucleic acid.


An alternative means for determining genomic copy number is in situ hybridization (e.g., Angerer (1987) Meth. Enzymol 152: 649). Generally, in situ hybridization comprises the following steps: (1) fixation of tissue or biological structure to be analyzed; (2) prehybridization treatment of the biological structure to increase accessibility of target DNA, and to reduce nonspecific binding; (3) hybridization of the mixture of nucleic acids to the nucleic acid in the biological structure or tissue; (4) post-hybridization washes to remove nucleic acid fragments not bound in the hybridization and (5) detection of the hybridized nucleic acid fragments. The reagent used in each of these steps and the conditions for use vary depending on the particular application. In a typical in situ hybridization assay, cells are fixed to a solid support, typically a glass slide. If a nucleic acid is to be probed, the cells are typically denatured with heat or alkali. The cells are then contacted with a hybridization solution at a moderate temperature to permit annealing of labeled probes specific to the nucleic acid sequence encoding the protein. The targets (e.g., cells) are then typically washed at a predetermined stringency or at an increasing stringency until an appropriate signal to noise ratio is obtained. The probes are typically labeled, e.g., with radioisotopes or fluorescent reporters. In one embodiment, probes are sufficiently long so as to specifically hybridize with the target nucleic acid(s) under stringent conditions. Probes generally range in length from about 200 bases to about 1000 bases. In some applications it is necessary to block the hybridization capacity of repetitive sequences. Thus, in some embodiments, tRNA, human genomic DNA, or Cot-I DNA is used to block non-specific hybridization.


An alternative means for determining genomic copy number is comparative genomic hybridization. In general, genomic DNA is isolated from normal reference cells, as well as from test cells (e.g., tumor cells) and amplified, if necessary. The two nucleic acids are differentially labeled and then hybridized in situ to metaphase chromosomes of a reference cell. The repetitive sequences in both the reference and test DNAs are either removed or their hybridization capacity is reduced by some means, for example by prehybridization with appropriate blocking nucleic acids and/or including such blocking nucleic acid sequences for said repetitive sequences during said hybridization. The bound, labeled DNA sequences are then rendered in a visualizable form, if necessary. Chromosomal regions in the test cells which are at increased or decreased copy number can be identified by detecting regions where the ratio of signal from the two DNAs is altered. For example, those regions that have decreased in copy number in the test cells will show relatively lower signal from the test DNA than the reference compared to other regions of the genome. Regions that have been increased in copy number in the test cells will show relatively higher signal from the test DNA. Where there are chromosomal deletions or multiplications, differences in the ratio of the signals from the two labels will be detected and the ratio will provide a measure of the copy number. In another embodiment of CGH, array CGH (aCGH), the immobilized chromosome element is replaced with a collection of solid support bound target nucleic acids on an array, allowing for a large or complete percentage of the genome to be represented in the collection of solid support bound targets. Target nucleic acids may comprise cDNAs, genomic DNAs, oligonucleotides (e.g., to detect single nucleotide polymorphisms) and the like. Array-based CGH may also be performed with single-color labeling (as opposed to labeling the control and the possible tumor sample with two different dyes and mixing them prior to hybridization, which will yield a ratio due to competitive hybridization of probes on the arrays). In single color CGH, the control is labeled and hybridized to one array and absolute signals are read, and the possible tumor sample is labeled and hybridized to a second array (with identical content) and absolute signals are read. Copy number difference is calculated based on absolute signals from the two arrays. Methods of preparing immobilized chromosomes or arrays and performing comparative genomic hybridization are well-known in the art (see, e.g., U.S. Pat. Nos. 6,335,167; 6,197,501; 5,830,645; and 5,665,549 and Albertson (1984) EMBO J. 3: 1227-1234; Pinkel (1988) Proc. Natl. Acad. Sci. USA 85: 9138-9142; EPO Pub. No. 430,402; Methods in Molecular Biology, Vol. 33: In situ Hybridization Protocols, Choo, ed., Humana Press, Totowa, N.J. (1994), etc.) In another embodiment, the hybridization protocol of Pinkel, et al. (1998) Nature Genetics 20: 207-211, or of Kallioniemi (1992) Proc. Natl Acad Sci USA 89:5321-5325 (1992) is used.


In still another embodiment, amplification-based assays can be used to measure copy number. In such amplification-based assays, the nucleic acid sequences act as a template in an amplification reaction (e.g., Polymerase Chain Reaction (PCR)). In a quantitative amplification, the amount of amplification product will be proportional to the amount of template in the original sample. Comparison to appropriate controls, e.g. healthy tissue, provides a measure of the copy number.


Methods of “quantitative” amplification are well-known to those of skill in the art. For example, quantitative PCR involves simultaneously co-amplifying a known quantity of a control sequence using the same primers. This provides an internal standard that may be used to calibrate the PCR reaction. Detailed protocols for quantitative PCR are provided in Innis, et al. (1990) PCR Protocols, A Guide to Methods and Applications, Academic Press, Inc. N.Y.). Measurement of DNA copy number at microsatellite loci using quantitative PCR analysis is described in Ginzonger, et al. (2000) Cancer Research 60:5405-5409. The known nucleic acid sequence for the genes is sufficient to enable one of skill in the art to routinely select primers to amplify any portion of the gene. Fluorogenic quantitative PCR may also be used in the methods of the present invention. In fluorogenic quantitative PCR, quantitation is based on amount of fluorescence signals, e.g., TaqMan and SYBR green.


Other suitable amplification methods include, but are not limited to, ligase chain reaction (LCR) (see Wu and Wallace (1989) Genomics 4: 560, Landegren, et al. (1988) Science 241:1077, and Barringer et al. (1990) Gene 89: 117), transcription amplification (Kwoh, et al. (1989) Proc. Natl. Acad. Sci. USA 86: 1173), self-sustained sequence replication (Guatelli, et al. (1990) Proc. Nat. Acad. Sci. USA 87: 1874), dot PCR, and linker adapter PCR, etc.


Loss of heterozygosity (LOH) and major copy proportion (MCP) mapping (Wang, Z. C., et al. (2004) Cancer Res 64(1):64-71; Seymour, A. B., et al. (1994) Cancer Res 54, 2761-4; Hahn, S. A., et al. (1995) Cancer Res 55, 4670-5; Kimura, M., et al. (1996) Genes Chromosomes Cancer 17, 88-93; Li et al., (2008) MBC Bioinform. 9, 204-219) may also be used to identify regions of amplification or deletion.


ii. Methods for Detection of Biomarker Nucleic Acid Expression


Biomarker expression may be assessed by any of a wide variety of well-known methods for detecting expression of a transcribed molecule or protein. Non-limiting examples of such methods include immunological methods for detection of secreted, cell-surface, cytoplasmic, or nuclear proteins, protein purification methods, protein function or activity assays, nucleic acid hybridization methods, nucleic acid reverse transcription methods, and nucleic acid amplification methods.


In preferred embodiments, activity of a particular gene is characterized by a measure of gene transcript (e.g. mRNA), by a measure of the quantity of translated protein, or by a measure of gene product activity. Marker expression can be monitored in a variety of ways, including by detecting mRNA levels, protein levels, or protein activity, any of which can be measured using standard techniques. Detection can involve quantification of the level of gene expression (e.g., genomic DNA, cDNA, mRNA, protein, or enzyme activity), or, alternatively, can be a qualitative assessment of the level of gene expression, in particular in comparison with a control level. The type of level being detected will be clear from the context.


In another embodiment, detecting or determining expression levels of a biomarker and functionally similar homologs thereof, including a fragment or genetic alteration thereof (e.g., in regulatory or promoter regions thereof) comprises detecting or determining RNA levels for the marker of interest. In one embodiment, one or more cells from the subject to be tested are obtained and RNA is isolated from the cells. In a preferred embodiment, a sample of breast tissue cells is obtained from the subject.


In one embodiment, RNA is obtained from a single cell. For example, a cell can be isolated from a tissue sample by laser capture microdissection (LCM). Using this technique, a cell can be isolated from a tissue section, including a stained tissue section, thereby assuring that the desired cell is isolated (see, e.g., Bonner et al. (1997) Science 278: 1481; Emmert-Buck et al. (1996) Science 274:998; Fend et al. (1999) Am. J. Path. 154: 61 and Murakami et al. (2000) Kidney Int. 58:1346). For example, Murakami et al., supra, describe isolation of a cell from a previously immunostained tissue section.


It is also be possible to obtain cells from a subject and culture the cells in vitro, such as to obtain a larger population of cells from which RNA can be extracted. Methods for establishing cultures of non-transformed cells, i.e., primary cell cultures, are known in the art.


When isolating RNA from tissue samples or cells from individuals, it may be important to prevent any further changes in gene expression after the tissue or cells has been removed from the subject. Changes in expression levels are known to change rapidly following perturbations, e.g., heat shock or activation with lipopolysaccharide (LPS) or other reagents. In addition, the RNA in the tissue and cells may quickly become degraded. Accordingly, in a preferred embodiment, the tissue or cells obtained from a subject is snap frozen as soon as possible.


RNA can be extracted from the tissue sample by a variety of methods, e.g., the guanidium thiocyanate lysis followed by CsCl centrifugation (Chirgwin et al., 1979, Biochemistry 18:5294-5299). RNA from single cells can be obtained as described in methods for preparing cDNA libraries from single cells, such as those described in Dulac, C. (1998) Curr. Top. Dev. Biol. 36, 245 and Jena et al. (1996) J. Immunol. Methods 190:199. Care to avoid RNA degradation must be taken, e.g., by inclusion of RNAsin.


The RNA sample can then be enriched in particular species. In one embodiment, poly(A)+ RNA is isolated from the RNA sample. In general, such purification takes advantage of the poly-A tails on mRNA. In particular and as noted above, poly-T oligonucleotides may be immobilized within on a solid support to serve as affinity ligands for mRNA. Kits for this purpose are commercially available, e.g., the MessageMaker kit (Life Technologies, Grand Island, NY).


In a preferred embodiment, the RNA population is enriched in marker sequences. Enrichment can be undertaken, e.g., by primer-specific cDNA synthesis, or multiple rounds of linear amplification based on cDNA synthesis and template-directed in vitro transcription (see, e.g., Wang et al. (1989) Proc. Natl. Acad. Sci. U.S.A. 86: 9717; Dulac et al., supra, and Jena et al., supra).


The population of RNA, enriched or not in particular species or sequences, can further be amplified. As defined herein, an “amplification process” is designed to strengthen, increase, or augment a molecule within the RNA. For example, where RNA is mRNA, an amplification process such as RT-PCR can be utilized to amplify the mRNA, such that a signal is detectable or detection is enhanced. Such an amplification process is beneficial particularly when the biological, tissue, or tumor sample is of a small size or volume.


Various amplification and detection methods can be used. For example, it is within the scope of the present invention to reverse transcribe mRNA into cDNA followed by polymerase chain reaction (RT-PCR); or, to use a single enzyme for both steps as described in U.S. Pat. No. 5,322,770, or reverse transcribe mRNA into cDNA followed by symmetric gap ligase chain reaction (RT-AGLCR) as described by R. L. Marshall, et al., PCR Methods and Applications 4: 80-84 (1994). Real time PCR may also be used.


Other known amplification methods which can be utilized herein include but are not limited to the so-called “NASBA” or “3SR” technique described in PNAS USA 87: 1874-1878 (1990) and also described in Nature 350 (No. 6313): 91-92 (1991); Q-beta amplification as described in published European Patent Application (EPA) No. 4544610; strand displacement amplification (as described in G. T. Walker et al., Clin. Chem. 42: 9-13 (1996) and European Patent Application No. 684315; target mediated amplification, as described by PCT Publication WO9322461; PCR; ligase chain reaction (LCR) (see, e.g., Wu and Wallace, Genomics 4, 560 (1989), Landegren et al., Science 241, 1077 (1988)); self-sustained sequence replication (SSR) (see, e.g., Guatelli et al., Proc. Nat. Acad. Sci. USA, 87, 1874 (1990)); and transcription amplification (see, e.g., Kwoh et al., Proc. Natl. Acad. Sci. USA 86, 1173 (1989)).


Many techniques are known in the state of the art for determining absolute and relative levels of gene expression, commonly used techniques suitable for use in the present invention include Northern analysis, RNase protection assays (RPA), microarrays and PCR-based techniques, such as quantitative PCR and differential display PCR. For example, Northern blotting involves running a preparation of RNA on a denaturing agarose gel, and transferring it to a suitable support, such as activated cellulose, nitrocellulose or glass or nylon membranes. Radiolabeled cDNA or RNA is then hybridized to the preparation, washed and analyzed by autoradiography.


In situ hybridization visualization may also be employed, wherein a radioactively labeled antisense RNA probe is hybridized with a thin section of a biopsy sample, washed, cleaved with RNase and exposed to a sensitive emulsion for autoradiography. The samples may be stained with hematoxylin to demonstrate the histological composition of the sample, and dark field imaging with a suitable light filter shows the developed emulsion. Non-radioactive labels such as digoxigenin may also be used.


Alternatively, mRNA expression can be detected on a DNA array, chip or a microarray. Labeled nucleic acids of a test sample obtained from a subject may be hybridized to a solid surface comprising biomarker DNA. Positive hybridization signal is obtained with the sample containing biomarker transcripts. Methods of preparing DNA arrays and their use are well-known in the art (see, e.g., U.S. Pat. Nos: 6,618,6796; 6,379,897; 6,664,377; 6,451,536; 548,257; U.S. 20030157485 and Schena et al. (1995) Science 20, 467-470; Gerhold et al. (1999) Trends In Biochem. Sci. 24, 168-173; and Lennon et al. (2000) Drug Discovery Today 5, 59-65, which are herein incorporated by reference in their entirety). Serial Analysis of Gene Expression (SAGE) can also be performed (See for example U.S. Patent Application 20030215858).


To monitor mRNA levels, for example, mRNA is extracted from the biological sample to be tested, reverse transcribed, and fluorescently-labeled cDNA probes are generated. The microarrays capable of hybridizing to marker cDNA are then probed with the labeled cDNA probes, the slides scanned and fluorescence intensity measured. This intensity correlates with the hybridization intensity and expression levels.


Types of probes that can be used in the methods described herein include cDNA, riboprobes, synthetic oligonucleotides and genomic probes. The type of probe used will generally be dictated by the particular situation, such as riboprobes for in situ hybridization, and cDNA for Northern blotting, for example. In one embodiment, the probe is directed to nucleotide regions unique to the RNA. The probes may be as short as is required to differentially recognize marker mRNA transcripts, and may be as short as, for example, 15 bases; however, probes of at least 17, 18, 19 or 20 or more bases can be used. In one embodiment, the primers and probes hybridize specifically under stringent conditions to a DNA fragment having the nucleotide sequence corresponding to the marker. As herein used, the term “stringent conditions” means hybridization will occur only if there is at least 95% identity in nucleotide sequences. In another embodiment, hybridization under “stringent conditions” occurs when there is at least 97% identity between the sequences.


The form of labeling of the probes may be any that is appropriate, such as the use of radioisotopes, for example, 32P and 35S. Labeling with radioisotopes may be achieved, whether the probe is synthesized chemically or biologically, by the use of suitably labeled bases.


In one embodiment, the biological sample contains polypeptide molecules from the test subject. Alternatively, the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject.


In another embodiment, the methods further involve obtaining a control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting marker polypeptide, mRNA, genomic DNA, or fragments thereof, such that the presence of the marker polypeptide, mRNA, genomic DNA, or fragments thereof, is detected in the biological sample, and comparing the presence of the marker polypeptide, mRNA, genomic DNA, or fragments thereof, in the control sample with the presence of the marker polypeptide, mRNA, genomic DNA, or fragments thereof in the test sample.


iii. Methods for Detection of Biomarker Protein Expression


The activity or level of a biomarker protein can be detected and/or quantified by detecting or quantifying the expressed polypeptide. The polypeptide can be detected and quantified by any of a number of means well-known to those of skill in the art. Aberrant levels of polypeptide expression of the polypeptides encoded by a biomarker nucleic acid and functionally similar homologs thereof, including a fragment or genetic alteration thereof (e.g., in regulatory or promoter regions thereof) are associated with the likelihood of response of an MDS and/or an anemia to modulators (e.g., inhibitors) of the IL-22 signaling pathway. Any method known in the art for detecting polypeptides can be used. Such methods include, but are not limited to, immunodiffusion, immunoelectrophoresis, radioimmunoassay (RIA), enzyme-linked immunosorbent assays (ELISAs), immunofluorescent assays, Western blotting, binder-ligand assays, immunohistochemical techniques, agglutination, complement assays, high performance liquid chromatography (HPLC), thin layer chromatography (TLC), hyperdiffusion chromatography, and the like (e.g., Basic and Clinical Immunology, Sites and Terr, eds., Appleton and Lange, Norwalk, Conn. pp 217-262, 1991 which is incorporated by reference). Preferred are binder-ligand immunoassay methods including reacting antibodies with an epitope or epitopes and competitively displacing a labeled polypeptide or derivative thereof.


For example, ELISA and RIA procedures may be conducted such that a desired biomarker protein standard is labeled (with a radioisotope such as 125I or 35S, or an assayable enzyme, such as horseradish peroxidase or alkaline phosphatase), and, together with the unlabeled sample, brought into contact with the corresponding antibody, whereon a second antibody is used to bind the first, and radioactivity or the immobilized enzyme assayed (competitive assay). Alternatively, the biomarker protein in the sample is allowed to react with the corresponding immobilized antibody, radioisotope- or enzyme-labeled anti-biomarker protein antibody is allowed to react with the system, and radioactivity or the enzyme assayed (ELISA-sandwich assay). Other conventional methods may also be employed as suitable.


The above techniques may be conducted essentially as a “one-step” or “two-step” assay. A “one-step” assay involves contacting antigen with immobilized antibody and, without washing, contacting the mixture with labeled antibody. A “two-step” assay involves washing before contacting, the mixture with labeled antibody. Other conventional methods may also be employed as suitable.


In one embodiment, a method for measuring biomarker protein levels comprises the steps of: contacting a biological specimen with an antibody or variant (e.g., fragment) thereof which selectively binds the biomarker protein, and detecting whether said antibody or variant thereof is bound to said sample and thereby measuring the levels of the biomarker protein.


Enzymatic and radiolabeling of biomarker protein and/or the antibodies may be effected by conventional means. Such means will generally include covalent linking of the enzyme to the antigen or the antibody in question, such as by glutaraldehyde, specifically so as not to adversely affect the activity of the enzyme, by which is meant that the enzyme must still be capable of interacting with its substrate, although it is not necessary for all of the enzyme to be active, provided that enough remains active to permit the assay to be effected. Indeed, some techniques for binding enzyme are non-specific (such as using formaldehyde), and will only yield a proportion of active enzyme.


It is usually desirable to immobilize one component of the assay system on a support, thereby allowing other components of the system to be brought into contact with the component and readily removed without laborious and time-consuming labor. It is possible for a second phase to be immobilized away from the first, but one phase is usually sufficient.


It is possible to immobilize the enzyme itself on a support, but if solid-phase enzyme is required, then this is generally best achieved by binding to antibody and affixing the antibody to a support, models and systems for which are well-known in the art. Simple polyethylene may provide a suitable support.


Enzymes employable for labeling are not particularly limited, but may be selected from the members of the oxidase group, for example. These catalyze production of hydrogen peroxide by reaction with their substrates, and glucose oxidase is often used for its good stability, ease of availability and cheapness, as well as the ready availability of its substrate (glucose). Activity of the oxidase may be assayed by measuring the concentration of hydrogen peroxide formed after reaction of the enzyme-labeled antibody with the substrate under controlled conditions well-known in the art.


Other techniques may be used to detect biomarker protein according to a practitioner's preference based upon the present disclosure. One such technique is Western blotting (Towbin et al., Proc. Nat. Acad. Sci. 76:4350 (1979)), wherein a suitably treated sample is run on an SDS-PAGE gel before being transferred to a solid support, such as a nitrocellulose filter. Anti-biomarker protein antibodies (unlabeled) are then brought into contact with the support and assayed by a secondary immunological reagent, such as labeled protein A or anti-immunoglobulin (suitable labels including 125I, horseradish peroxidase and alkaline phosphatase). Chromatographic detection may also be used.


Immunohistochemistry may be used to detect expression of biomarker protein, e.g., in a biopsy sample. A suitable antibody is brought into contact with, for example, a thin layer of cells, washed, and then contacted with a second, labeled antibody. Labeling may be by fluorescent markers, enzymes, such as peroxidase, avidin, or radiolabeling. The assay is scored visually, using microscopy.


Anti-biomarker protein antibodies, such as intrabodies, may also be used for imaging purposes, for example, to detect the presence of biomarker protein in cells and tissues of a subject. Suitable labels include radioisotopes, iodine (125I, 121I), carbon (14C), sulphur (35S), tritium (3H), indium (112In), and technetium (99mTc), fluorescent labels, such as fluorescein and rhodamine, and biotin.


For in vivo imaging purposes, antibodies are not detectable, as such, from outside the body, and so must be labeled, or otherwise modified, to permit detection. Markers for this purpose may be any that do not substantially interfere with the antibody binding, but which allow external detection. Suitable markers may include those that may be detected by X-radiography, NMR or MM. For X-radiographic techniques, suitable markers include any radioisotope that emits detectable radiation but that is not overtly harmful to the subject, such as barium or cesium, for example. Suitable markers for NMR and MM generally include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the antibody by suitable labeling of nutrients for the relevant hybridoma, for example.


The size of the subject, and the imaging system used, will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of technetium-99. The labeled antibody or antibody fragment will then preferentially accumulate at the location of cells which contain biomarker protein. The labeled antibody or antibody fragment can then be detected using known techniques.


Antibodies that may be used to detect biomarker protein include any antibody, whether natural or synthetic, full length or a fragment thereof, monoclonal or polyclonal, that binds sufficiently strongly and specifically to the biomarker protein to be detected. An antibody may have a Kd of at most about 10−6 M, 10−7 M, 10−8 M, 10−9 M, 10−10 M, 10−11 M, 10−12 M. The phrase “specifically binds” refers to binding of, for example, an antibody to an epitope or antigen or antigenic determinant in such a manner that binding can be displaced or competed with a second preparation of identical or similar epitope, antigen or antigenic determinant. An antibody may bind preferentially to the biomarker protein relative to other proteins, such as related proteins.


Antibodies are commercially available or may be prepared according to methods known in the art.


Antibodies and derivatives thereof that may be used encompass polyclonal or monoclonal antibodies, chimeric, human, humanized, primatized (CDR-grafted), veneered or single-chain antibodies as well as functional fragments, i.e., biomarker protein binding fragments, of antibodies. For example, antibody fragments capable of binding to a biomarker protein or portions thereof, including, but not limited to, Fv, Fab, Fab′ and F(ab′) 2 fragments can be used. Such fragments can be produced by enzymatic cleavage or by recombinant techniques. For example, papain or pepsin cleavage can generate Fab or F(ab′) 2 fragments, respectively. Other proteases with the requisite substrate specificity can also be used to generate Fab or F(ab′) 2 fragments. Antibodies can also be produced in a variety of truncated forms using antibody genes in which one or more stop codons have been introduced upstream of the natural stop site. For example, a chimeric gene encoding a F(ab′) 2 heavy chain portion can be designed to include DNA sequences encoding the CH, domain and hinge region of the heavy chain.


Synthetic and engineered antibodies are described in, e.g., Cabilly et al., U.S. Pat. No. 4,816,567 Cabilly et al., European Patent No. 0,125,023 B1; Boss et al., U.S. Pat. No. 4,816,397; Boss et al., European Patent No. 0,120,694 B1; Neuberger, M. S. et al., WO 86/01533; Neuberger, M. S. et al., European Patent No. 0,194,276 B1; Winter, U.S. Pat. No. 5,225,539; Winter, European Patent No. 0,239,400 B1; Queen et al., European Patent No. 0451216 B1; and Padlan, E. A. et al., EP 0519596 A1. See also, Newman, R. et al., BioTechnology, 10: 1455-1460 (1992), regarding primatized antibody, and Ladner et al., U.S. Pat. No. 4,946,778 and Bird, R. E. et al., Science, 242: 423-426 (1988)) regarding single-chain antibodies. Antibodies produced from a library, e.g., phage display library, may also be used.


In some embodiments, agents that specifically bind to a biomarker protein other than antibodies are used, such as peptides. Peptides that specifically bind to a biomarker protein can be identified by any means known in the art. For example, specific peptide binders of a biomarker protein can be screened for using peptide phage display libraries.


iv. Methods for Detection of Biomarker Structural Alterations


The following illustrative methods can be used to identify the presence of a structural alteration in a biomarker nucleic acid and/or biomarker polypeptide molecule in order to, for example, identify STUB1, UBQLN1, HSP90B1, or other biomarkers used in the immunotherapies described herein that are overexpressed, overfunctional, and the like.


In certain embodiments, detection of the alteration involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Pat. Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al. (1988) Science 241:1077-1080; and Nakazawa et al. (1994) Proc. Natl. Acad. Sci. USA 91:360-364), the latter of which can be particularly useful for detecting point mutations in a biomarker nucleic acid such as a biomarker gene (see Abravaya et al. (1995) Nucleic Acids Res. 23:675-682). This method can include the steps of collecting a sample of cells from a subject, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers which specifically hybridize to a biomarker gene under conditions such that hybridization and amplification of the biomarker gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.


Alternative amplification methods include: self-sustained sequence replication (Guatelli, J. C. et al. (1990) Proc. Natl. Acad. Sci. USA 87:1874-1878), transcriptional amplification system (Kwoh, D. Y. et al. (1989) Proc. Natl. Acad. Sci. USA 86:1173-1177), Q-Beta Replicase (Lizardi, P. M. et al. (1988) Bio-Technology 6:1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well-known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.


In an alternative embodiment, mutations in a biomarker nucleic acid from a sample cell can be identified by alterations in restriction enzyme cleavage patterns. For example, sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA. Moreover, the use of sequence specific ribozymes (see, for example, U.S. Pat. No. 5,498,531) can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.


In other embodiments, genetic mutations in biomarker nucleic acid can be identified by hybridizing a sample and control nucleic acids, e.g., DNA or RNA, to high density arrays containing hundreds or thousands of oligonucleotide probes (Cronin, M. T. et al. (1996) Hum. Mutat. 7:244-255; Kozal, M. J. et al. (1996) Nat. Med. 2:753-759). For example, biomarker genetic mutations can be identified in two dimensional arrays containing light-generated DNA probes as described in Cronin et al. (1996) supra. Briefly, a first hybridization array of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear arrays of sequential, overlapping probes. This step allows the identification of point mutations. This step is followed by a second hybridization array that allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants or mutations detected. Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene. Such biomarker genetic mutations can be identified in a variety of contexts, including, for example, germline and somatic mutations.


In yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence a biomarker gene and detect mutations by comparing the sequence of the sample biomarker with the corresponding wild-type (control) sequence. Examples of sequencing reactions include those based on techniques developed by Maxam and Gilbert (1977) Proc. Natl. Acad. Sci. USA 74:560 or Sanger (1977) Proc. Natl. Acad Sci. USA 74:5463. It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays (Naeve (1995) Biotechniques 19:448-53), including sequencing by mass spectrometry (see, e.g., PCT International Publication No. WO 94/16101; Cohen et al. (1996) Adv. Chromatogr. 36:127-162; and Griffin et al. (1993) Appl. Biochem. Biotechnol. 38:147-159).


Other methods for detecting mutations in a biomarker gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes (Myers et al. (1985) Science 230:1242). In general, the art technique of “mismatch cleavage” starts by providing heteroduplexes formed by hybridizing (labeled) RNA or DNA containing the wild-type biomarker sequence with potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent which cleaves single-stranded regions of the duplex such as which will exist due to base pair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with SI nuclease to enzymatically digest the mismatched regions. In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, for example, Cotton et al. (1988) Proc. Natl. Acad. Sci. USA 85:4397 and Saleeba et al. (1992) Methods Enzymol. 217:286-295. In a preferred embodiment, the control DNA or RNA can be labeled for detection.


In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called “DNA mismatch repair” enzymes) in defined systems for detecting and mapping point mutations in biomarker cDNAs obtained from samples of cells. For example, the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches (Hsu et al. (1994) Carcinogenesis 15:1657-1662). According to an exemplary embodiment, a probe based on a biomarker sequence, e.g., a wild-type biomarker treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like (e.g., U.S. Pat. No. 5,459,039.)


In other embodiments, alterations in electrophoretic mobility can be used to identify mutations in biomarker genes. For example, single strand conformation polymorphism (SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids (Orita et al. (1989) Proc Natl. Acad. Sci USA 86:2766; see also Cotton (1993) Mutat. Res. 285:125-144 and Hayashi (1992) Genet. Anal. Tech. Appl. 9:73-79). Single-stranded DNA fragments of sample and control biomarker nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In a preferred embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al. (1991) Trends Genet. 7:5).


In yet another embodiment the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al. (1985) Nature 313:495). When DGGE is used as the method of analysis, DNA will be modified to ensure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum and Reissner (1987) Biophys. Chem. 265:12753).


Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al. (1986) Nature 324:163; Saiki et al. (1989) Proc. Natl. Acad. Sci. USA 86:6230). Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.


Alternatively, allele specific amplification technology which depends on selective PCR amplification may be used in conjunction with the instant invention. Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization) (Gibbs et al. (1989) Nucleic Acids Res. 17:2437-2448) or at the extreme 3′ end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner (1993) Tibtech 11:238). In addition it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection (Gasparini et al. (1992) Mol. Cell Probes 6:1). It is anticipated that in certain embodiments amplification may also be performed using Taq ligase for amplification (Barany (1991) Proc. Natl. Acad. Sci USA 88:189). In such cases, ligation will occur only if there is a perfect match at the 3′ end of the 5′ sequence making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification.


VI. Administration of Agents

The agents encompassed by the present invention (e.g., down-regulators of IL-22) are administered to subjects in a biologically compatible form suitable for pharmaceutical administration in vivo, to enhance their effects. By “biologically compatible form suitable for administration in vivo” is meant a form to be administered in which any toxic effects are outweighed by the therapeutic effects. The term “subject” is intended to include living organisms in which an immune response can be elicited, e.g., mammals. Examples of subjects include humans, dogs, cats, mice, rats, and transgenic species thereof. Administration of an agent as described herein can be in any pharmacological form including a therapeutically active amount of an agent alone or in combination with a pharmaceutically acceptable carrier.


Administration of a therapeutically active amount of the therapeutic composition encompassed by the present invention is defined as an amount effective, at dosages and for periods of time necessary, to achieve the desired result. For example, a therapeutically active amount of an agent can vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of peptide to elicit a desired response in the individual. Dosage regimens can be adjusted to provide the optimum therapeutic response. For example, several divided doses can be administered daily or the dose can be proportionally reduced as indicated by the exigencies of the therapeutic situation.


Agents encompassed by the present invention can be administered either alone or in combination with an additional therapy. In the combination therapy, a down-regulator of IL-22 encompassed by the present invention and another agent, such as lenalidomide, azacitidine, decitabine, or an erythropoiesis-stimulating agent (e.g., erythropoietin, epoetin alfa, epoetin beta, epoetin omega, epoetin zeta, darbepoetin alfa) can be delivered to the same or different cells and can be delivered at the same or different times. The agents encompassed by the present invention can be incorporated into pharmaceutical compositions suitable for administration. Such compositions can comprise one or more agents or one or more molecules that result in the production of such one or more agents (e.g., a nucleic acid that results in production of an antigen-binding fragment of an anti-IL-22 antibody) and a pharmaceutically acceptable carrier.


The therapeutic agents described herein can be administered using a mode or route of administration that delivers them to the particular locations in the body where IL-22 or IL-22RA1 expression can increase in red blood cell disorders, such as the kidney, liver, lung, gastrointestinal tract, brain, thymus skin, or pancreas.


The therapeutic agents described herein can be administered in a convenient manner such as by injection (subcutaneous, intravenous, etc.), oral administration, inhalation, transdermal application, or rectal administration. Depending on the route of administration, the active compound can be coated in a material to protect the compound from the action of enzymes, acids and other natural conditions which can inactivate the compound. For example, for administration of agents, by other than parenteral administration, it can be desirable to coat the agent with, or co-administer the agent with, a material to prevent its inactivation.


An agent can be administered to an individual in an appropriate carrier, diluent or adjuvant, co-administered with enzyme inhibitors or in an appropriate carrier such as liposomes. Pharmaceutically acceptable diluents include saline and aqueous buffer solutions. Adjuvant is used in its broadest sense and includes any immune stimulating compound such as interferon. Adjuvants contemplated herein include resorcinols, non-ionic surfactants such as polyoxyethylene oleyl ether and n-hexadecyl polyethylene ether. Enzyme inhibitors include pancreatic trypsin inhibitor, diisopropylfluorophosphate (DEEP) and trasylol. Liposomes include water-in-oil-in-water emulsions as well as conventional liposomes (Sterna et al. (1984) J. Neuroimmunol. 7:27).


As described in detail below, the pharmaceutical compositions encompassed by the present invention can be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, boluses, powders, granules, pastes; (2) parenteral administration, for example, by subcutaneous, intramuscular or intravenous injection as, for example, a sterile solution or suspension; (3) topical application, for example, as a cream, ointment or spray applied to the skin; (4) intra-vaginally or intra-rectally, for example, as a pessary, cream or foam; or (5) aerosol, for example, as an aqueous aerosol, liposomal preparation or solid particles containing the compound.


The phrase “pharmaceutically acceptable” is employed herein to refer to those agents, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.


The phrase “pharmaceutically-acceptable carrier” as used herein means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations.


The term “pharmaceutically-acceptable salts” refers to the relatively non-toxic, inorganic and organic acid addition salts of the agents that modulates (e.g., inhibits) biomarker expression and/or activity, or expression and/or activity of the complex encompassed by the present invention. These salts can be prepared in situ during the final isolation and purification of the therapeutic agents, or by separately reacting a purified therapeutic agent in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like (See, for example, Berge et al. (1977) “Pharmaceutical Salts”, J. Pharm. Sci. 66:1-19).


In other cases, the agents useful in the methods encompassed by the present invention can contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically-acceptable salts with pharmaceutically-acceptable bases. The term “pharmaceutically-acceptable salts” in these instances refers to the relatively non-toxic, inorganic and organic base addition salts of agents that modulates (e.g., inhibits) biomarker expression and/or activity, or expression and/or activity of the complex. These salts can likewise be prepared in situ during the final isolation and purification of the therapeutic agents, or by separately reacting the purified therapeutic agent in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation, with ammonia, or with a pharmaceutically-acceptable organic primary, secondary or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like. Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like (see, for example, Berge et al., supra).


Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.


Examples of pharmaceutically-acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.


Formulations useful in the methods encompassed by the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal, aerosol and/or parenteral administration. The formulations can conveniently be presented in unit dosage form and can be prepared by any methods well-known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient, which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.


Methods of preparing these formulations or compositions include the step of bringing into association an agent that modulates (e.g., inhibits) biomarker expression and/or activity, with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a therapeutic agent with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.


Formulations suitable for oral administration can be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a therapeutic agent as an active ingredient. A compound can also be administered as a bolus, electuary or paste.


In solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, acetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions can also comprise buffering agents. Solid compositions of a similar type can also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.


A tablet can be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets can be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets can be made by molding in a suitable machine a mixture of the powdered peptide or peptidomimetic moistened with an inert liquid diluent.


Tablets, and other solid dosage forms, such as dragees, capsules, pills and granules, can optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well-known in the pharmaceutical-formulating art. They can also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They can be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions, which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions can also optionally contain opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions, which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.


Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms can contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.


Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.


Suspensions, in addition to the active agent can contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.


Formulations for rectal or vaginal administration can be presented as a suppository, which can be prepared by mixing one or more therapeutic agents with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.


Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.


Dosage forms for the topical or transdermal administration of an agent that modulates (e.g., inhibits) biomarker expression and/or activity include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active component can be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which can be required.


The ointments, pastes, creams and gels can contain, in addition to a therapeutic agent, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.


Powders and sprays can contain, in addition to an agent that modulates (e.g., inhibits) biomarker expression and/or activity, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.


The agents disclosed herein can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound. A nonaqueous (e.g., fluorocarbon propellant) suspension could be used. Sonic nebulizers are preferred because they minimize exposing the agent to shear, which can result in degradation of the compound.


Ordinarily, an aqueous aerosol is made by formulating an aqueous solution or suspension of the agent together with conventional pharmaceutically acceptable carriers and stabilizers. The carriers and stabilizers vary with the requirements of the particular compound, but typically include nonionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols. Aerosols generally are prepared from isotonic solutions.


Transdermal patches have the added advantage of providing controlled delivery of a therapeutic agent to the body. Such dosage forms can be made by dissolving or dispersing the agent in the proper medium. Absorption enhancers can also be used to increase the flux of the peptidomimetic across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the peptidomimetic in a polymer matrix or gel.


Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention.


Pharmaceutical compositions encompassed by the present invention suitable for parenteral administration comprise one or more therapeutic agents in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which can be reconstituted into sterile injectable solutions or dispersions just prior to use, which can contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.


Examples of suitable aqueous and nonaqueous carriers which can be employed in the pharmaceutical compositions encompassed by the present invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.


These compositions can also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms can be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It can also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form can be brought about by the inclusion of agents that delay absorption such as aluminum monostearate and gelatin.


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


Injectable depot forms are made by forming microencapsule matrices of an agent that modulates (e.g., inhibits) biomarker expression and/or activity, in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions, which are compatible with body tissue.


When the therapeutic agents encompassed by the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.


Actual dosage levels of the active ingredients in pharmaceutical compositions encompassed by the present invention can be determined by the methods encompassed by the present invention to obtain an amount of the active ingredient, which is effective to achieve the desired therapeutic response for a particular subject, composition, and mode of administration, without being toxic to the subject.


The nucleic acid molecules encompassed by the present invention can be inserted into vectors and used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Pat. No. 5,328,470) or by stereotactic injection (see e.g., Chen et al. (1994) Proc. Natl. Acad. Sci. USA 91:3054-3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells that produce the gene delivery system.


In one embodiment, an agent encompassed by the present invention is an antibody. As defined herein, a therapeutically effective amount of antibody (i.e., an effective dosage) ranges from about 0.001 to 30 mg/kg body weight, preferably about 0.01 to 25 mg/kg body weight, more preferably about 0.1 to 20 mg/kg body weight, and even more preferably about 1 to 10 mg/kg, 2 to 9 mg/kg, 3 to 8 mg/kg, 4 to 7 mg/kg, or 5 to 6 mg/kg body weight. The skilled artisan will appreciate that certain factors can influence the dosage required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of an antibody can include a single treatment or, preferably, can include a series of treatments. In a preferred example, a subject is treated with antibody in the range of between about 0.1 to 20 mg/kg body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks. It will also be appreciated that the effective dosage of antibody used for treatment can increase or decrease over the course of a particular treatment. Changes in dosage can result from the results of diagnostic assays.


EXAMPLES
Example 1: Materials and Methods for Examples 2-5

a. Flow Cytometry and Cell Isolation


Whole bone marrow (BM) cells were isolated by crushing hind leg bones (femur and tibia) with mortar and pestle in staining buffer (PBS, Corning) supplemented with 2% heat inactivated fetal bovine serum (FBS, Atlanta Biologicals) and EDTA (GIBCO)). Whole bone marrow was lysed with 1× Pharm Lyse™ (BD Biosciences) for 90 seconds, and the reaction was terminated by adding an excess of staining buffer. Cells were labeled with fluorochrome-conjugated antibodies in staining buffer for 30 minutes at 4° C. For flow cytometric analysis, cells were incubated with combinations of fluorochrome-conjugated antibodies to the following cell surface markers: CD3 (17A2), CD5 (53-7.3), CD11b (M1/70), Gr1 (RB6-8C5), B220 (RA3-6B2), Ter119 (TER119), CD71 (C2), ckit (2B8), Sca1 (D7), CD16/32 (93), CD150 (TC15-12F12.2), CD48 (HM48-1). For sorting of lineage-negative cells, lineage markers included CD3, CD5, CD11b, Gr1 and Ter119. For sorting of erythroid progenitor cells, the lineage cocktail did not include Ter119. All reagents were acquired from BD Biosciences, Thermo Fisher Scientific, Novus Biologicals, R&D Biosystems, Tonbo Biosciences, or BioLegend. Identification of apoptotic cells was carried out using the Annexin V Apoptosis Detection Kit (BioLegend). Intracytoplasmic and intranuclear staining was performed using Foxp3/Transcription Factor Staining Kit (eBioscience). To increase the sorting efficiency, whole bone marrow samples were lineage-depleted using magnetic microbeads (Miltenyi Biotec) and autoMACS® Pro magnetic separator (Miltenyi Biotec). Cell sorting was performed on a FACSAria® flow cytometer (BD Biosciences), data acquisition was performed on a BD Fortessa™ X-20 instrument equipped with 5 lasers (BD Biosciences). Data were analyzed by FlowJo (Tree Star) version 9 software. Flow analyses were performed on viable cells by exclusion of dead cells using either DAPI or a fixable viability dye (Tonbo Biosciences).


b. In Vivo Measurement of Protein Synthesis


One hundred mL of a 20 mM solution of 0-Propargyl-Puromycin (OP-Puro; BioMol) was injected intraperitoneally in mice and mice were then rested for 1 hour (1 h). Mice injected with PBS were used as controls. BM was harvested after 1 h and stained with antibodies against cell surface markers, washed to remove excess unbound antibodies, fixed in 1% paraformaldehyde, and permeabilized in PBS with 3% fetal bovine serum and 0.1% saponin. The azide-alkyne cyclo-addition was performed using the Click-iT™ Cell Reaction Buffer Kit (Thermo Fisher Scientific) and azide conjugated to Alexa Fluor 647® (Thermo Fisher Scientific) at 5 μM final concentration for 30 minutes. Cells were washed twice and analyzed by flow cytometry.


c. Methylcellulose Assay


Cells at a population of 1,000-2,500 BM c-kit+cells were sorted and plated in semi-solid methylcellulose culture medium (M3434, StemCell Technologies) and incubated at 37° C. in a humidified atmosphere for 7-10 days. At the end of the incubation period, each well was triturated with staining buffer to collect cells. Collected cells were then processed for flow cytometry as described above.


d. Phenylhydrazine Treatment


Phenylhydrazine (PhZ) was purchased from Sigma and injected intraperitoneally on 2 consecutive days (days 0 and 1) at a dose of 25 mg/kg. Peripheral blood was collected 3-4 days before the start of treatment and at day 4, 7 and 11. PhZ treatment experiments were carried out in 8-16 week-old mice.


e. T Cell Polarization


Single cell suspensions of mouse spleens were prepared by pressing tissue through a 70 μm cell strainer followed by red blood cell lysis using Pharm Lyse™ (BD Biosciences). Total splenic CD4+ T cells were isolated using CD4 T cell isolation kit (Miltenyi Biotec). Enriched CD4 T cells were then incubated with fluorochrome-conjugated antibodies to CD4, CD8, CD25, CD62L, and CD44 to purify naïve CD4 T cells using fluorescence activated cell sorting (FACS). In the presence of 1 μg/mL plate-bound anti-CD3ε and soluble 1 μg/mL anti-CD28, T cell polarizations were carried out in IMDM supplemented with 10% FBS, 2 mM L-glutamine, 100 mg/mL penicillin-streptomycin, HEPES, non-essential amino acids, 100 μM β-mercaptoethanol, and sodium pyruvate as follows—Th1 (20 ng/mL IL-12, 10 μg/mL anti-IL-4), Th2 (20 ng/mL IL-4, 10 μg/mL anti-IFN-γ), Th17 (30 ng/mL IL-6, 20 ng/mL IL-23, 20 ng/mL IL-1b, 10 μg/mL anti-IL-4, 10 μg/mL anti-IFN-γ), Tregs (1 ng/mL TGF-β, 10 μg/mL anti-IL-4, 10 μg/mL anti-IFN-γ), and Th22 (1 ng/mL TGF-β, 30 ng/mL IL-6, 20 ng/mL IL-23, 20 ng/mL IL-1b, 10 μg/mL anti-IL-4, 10 μg/mL anti-IFN-γ, 200 nM FICZ).


f. IL-22 Neutralization and Reconstitution.


Monoclonal anti-IL-22 (Clone IL22JOP) blocking antibody and isotype control IgG2a (Clone eBR2a) were purchased from Thermo Fisher Scientific. Mice were administered anti-IL-22 (50 μg/mouse) or isotype intraperitoneally every 48 h until the conclusion of the experiment. For recombinant IL-22 treatment, mice were injected with recombinant IL-22 (500 ng/mouse; PeproTech) intraperitoneally every 24 h until the conclusion of the experiment.


g. Cytokine Quantitation


IL-22 in human samples was quantified using SMC™ Human IL-22 High Sensitivity Immunoassay Kit (EMD Millipore, 03-0162-00) according to manufacturers' instructions. The assay was read on a SMCxPro™ (EMD Millipore) instrument. The lower limit of quantification (LLOQ) of this immunoassay is 0.1 pg/mL. IL-22 in mouse samples was quantified using ELISA MAX™ Deluxe Set Mouse IL-22 (BioLegend, 436304). The LLOQ of this immunoassay is 3.9 pg/mL. Concentration of lineage-associated cytokines in cell culture supernatants of polarized T cells were quantified using a custom-made ProcartaPlex™ assay (Thermo Fisher Scientific) acquired on a Luminex® platform.


h. Statistical Tests


Data are presented as mean±s.e.m. Comparison of two groups was performed using unpaired two-tailed t-test, assuming normal distribution. For multiple group comparison, analysis of variance (ANOVA) with post hoc Tukey's correction was applied. Statistical analyses were performed using GraphPad Prism 8.0 (GraphPad Software Inc., San Diego, CA). A p-value of less than 0.05 was considered significant.


Example 2: Riok2 Haploinsufficiency Blocks Erythroid Differentiation Leading to Anemia

T cells lacking the endoplasmic reticulum stress transcription factor Xbp1 have reduced Riok2 expression (Song et al. (2018) Nature 562:423-428). RIOK2 lies on the long arm of chromosome 5q (5q15) in the human genome, between the breakpoints (5q13-5q35), which are known to occur in del(5q) syndromes, such as (del(5q)) myelodysplastic syndromes (MDS) (FIG. 1A). Studies in yeast (Ferreira-Cerca et al. (2012) Nat. Struct. Mol. Biol. 19:1316-1323) and human (Zemp et al. (2009) J. Cell. Biol. 185:1167-1180) cancer cell lines have shown that Riok2 plays an indispensable role in the maturation of the pre-40S ribosomal complex. GEXC analysis revealed that in mouse bone marrow (BM), Riok2 expression is highest in pCFU-E (primitive colony-forming-unit erythroid) cells, indicating that Riok2 may be involved in maintaining red blood cell (RBC) output (FIG. 1B). To further study the role of Riok2 in hematopoiesis, Vav1-Cre+ transgenic floxed Riok2 (Riok2f/+Vav1cre) mice were generated in which the Cre recombinase is under the control of the hematopoietic cell-specific Vav1 promoter. Riok2 expression in hematopoietic cells from Riok2f/+Vav1cre mice was approximately 50% compared to those from Vav1-Cre+controls (FIG. 1C). Interestingly, no Vav1-Cre foxed Riok2 homozygous knockout mice were recovered (FIG. 1D), indicating embryonic lethality from complete hematopoietic deletion of Riok2. However, Vav1 Cre Riok2 f/+ mice were viable with approximately 50% Riok2 expression levels in hematopoietic cells compared to that of Vav1-Cre+ controls (FIG. 1C). As is seen with other ribosomal protein haploinsufficiency mouse models (Schneider et al. (2016) Nat. Med. 22:288-297), BM cells from Riok2f/+Vav1cre mice showed reduced nascent protein synthesis in vivo compared to Vav1-cre controls (FIG. 1D), which is consistent with Riok2's role in maturation of the pre-40S ribosome. A recent study showed that ribosomal protein deficiency-mediated reduced protein synthesis significantly affects erythropoiesis over myelopoiesis (Khajuria et al. (2018) Cell 173:90-103).


Consistent with the high expression of Riok2 in pCFU-e cells in the BM, mice with heterozygous deletion of Riok2 in hematopoietic cells (Riok2f/+Vav1cre) displayed anemia with reduced peripheral blood red blood cell (RBC) numbers, hemoglobin (Hb), and hematocrit (HCT) (FIG. 2A). It was next determined whether Riok2 haploinsufficiency-mediated anemia was secondary to a defect in erythroid development in the BM. The states of erythropoiesis (referred to herein as RI, RII, RIII and RIV) were characterized by flow cytometry by using the expression of Ter119 and CD71 (FIG. 3A). Riok2f Vav1cre mice had impaired erythropoiesis in the BM (FIG. 2B). Moreover, Riok2 haploinsufficiency led to increased apoptosis in erythroid progenitors as compared to controls (FIG. 2C), Additionally, Riok2f/+Vav1cre erythroid progenitors showed a decrease in cell quiescence with cell cycle block at the GI stage (FIG. 313). A block in cell cycle is driven by a group of proteins known as cyclin-dependent kinase inhibitors (CM). The expression of p21 (a CM encoded by Cdkn1a) was increased in erythroid progenitors from Riok2f/+Vav1cre mice compared to Riok2+/+Vav1cre controls (FIG. 3C).


In addition, the effect of Riok2 haploinsufficiency on stress-induced erythropoiesis was determined by analyzing mice in which hemolysis was induced by non-lethal phenylhydrazine treatment (25 mg/kg on days 0 and 1). After acute hemolytic stress, Riok2f/+Vav1cre mice developed more severe anemia and had a delayed RBC recovery response, as compared to Riok2+/+ Vav1cre control mice (FIG. 2D). Riok2P Vav1cre mice succumbed faster to a lethal dose of phenylhydrazine (35 Ing/kg on days 0 and 1) as compared to Vav1-cre controls (FIG. 3D). To determine whether Riok2 haploinsufficiency in BM cells drives anemia, bone marrow (BM) chimeras were generated. Wild-type (WT) mice transplanted with Riok2f/+Vav1cre whole BM developed anemia as compared to Riok2+/+ Vav1cre BM transplanted WT mice (FIG. 3E).


In addition to the reduction in RBC numbers in peripheral blood (PB) from Riok2f/+Vav1″ mice, an increased percentage of monocytes (monocytosis) and decreased percentage of neutrophils (neutropenia) compared to controls was also observed (FIG. 2E). Granulocyte macrophage progenitors (GNPs) in the BM give rise to PB myeloid cells. The percentage of BM GMPs was increased in Riok2f/+Vav1″ mice as compared to Riok2+/+Vav1cre controls (FIG. 2F). To analyze the effect of Riok2 haploinsufficiency on myelopoiesis in the absence of in vivo compensatory mechanisms, LSK (lineage-Sca-1+Kit+) cells from the BM of Riok2f/+Vav1cre and Riok2+/+Vav1cre controls were cultured in a MethoCult™ assay supplemented with growth factors (IL-6, IL-3, and SCF, but devoid of erythropoietin). LSKs from Riok2f/+Vav1cre mice gave rise to an increased percentage of CD11b+ myeloid cells suggesting a cell-intrinsic myeloproliferative effect due to Riok2 haploinsufficiency (FIG. 2G).


Example 3: Riok2 Haploinsufficiency Induces Increased Levels of Immune-Related Proteins in Erythroid Progenitors

To elucidate a mechanism for the erythroid differentiation defect observed in Riok2f/+Vav1cre mice, quantitative proteomic analysis of purified erythroid progenitors using mass spectrometry was performed. Riok2 haploinsufficiency led to upregulation of 564 distinct proteins (adjusted p-value of <0.05) in erythroid progenitors compared to those from Vav1-cre controls (FIG. 4A). Interestingly, the most highly upregulated proteins in the dataset correlated significantly (p-val: 1.66×10−16) with those observed upon haploinsufficiency of Rps14 (Schneider et al. (2016) Nat. Med. 22:288-297), another component of the 40S ribosomal complex (FIG. 4B). Fourteen of the total 26 upregulated proteins in the Rps14 dataset were also upregulated in the Riok2 haploinsufficient dataset, revealing a largely common proteomic signature on deletion of distinct ribosomal proteins (FIG. 4C).


In Riok2f/+Vav1cre erythroid progenitor cells, the upregulated proteins with the highest fold-change (S100A8, S100A9 Camp, Ngp, and the like) are proteins with known immune functions, such as antimicrobial defense. This indicated a possible role for the immune system in driving the proteomic changes seen in the Riok2 haploinsufficient erythroid progenitors. To assess if Riok2 haploinsufficiency leads to changes in immune cell function, naïve T cells from Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls were subjected to in vitro polarization towards known T cell lineages (Th1 Th2, Th17, Th22, and Tregs). Secretion of IL-2, IFN-gamma, IL-13, IL-17A, and the frequency of Foxp3±Tregs was comparable between Riok2f/+Vav1cre and Riok2+/+Vav1cre T cells (FIGS. 5A-5G). However, an exclusive increase in IL-22 secretion from Riok2f/+Vav1cre naïve T cells polarized towards the Th22 lineage was observed (FIG. 6A, left panel). The frequency of IL-22±CD4+ T cells was also higher in Riok2f/+Vav1cre Th22 cultures as compared to Vav1-cre control Th22 cultures (FIG. 6A, right panel). The concentration of IL-22 in the serum and BM fluid of Riok2f/+Vav1cre mice was also significantly higher as compared to Vav1-cre controls (FIG. 6B). Rps14 haploinsufficient Th22 cells also secreted elevated levels of IL-22 compared to Vav1-cre control Th22. cells (FIG. 4D).


Phenylhydrazine (PhZ) administration to wild-type (C57BL/6J, C57) mice treated intraperitoneally with rIL-22 led to decreased PB RBCs, Mb, and HCT owing to decreased BM erythroid progenitor cells (FIGS. 7A and 7C). Recombinant IL-22 led to an increase in apoptosis of erythroid progenitors (FIG. 7D). It also led to an increase in PB and BM reticulocytes as an indication of increased erythropoiesis under stress (FIG. 7B). Recombinant IL-22 also dose-dependently decreased terminal erythropoiesis in an in vitro erythropoiesis assay (FIG. 7E).


Example 4: Neutralization of IL-22 Signaling Alleviates Anemia in Riok2 Haploinsufficient Mice and Increases Red Blood Cell (RBC) Numbers in Wild-Type Mice

Mice harboring a compound genetic deletion of IL-22 on the Riok2 haploinsufficient background (Riok2f/+Il22+/−Vav1cre) exhibited increased numbers of PB RBCs and HCT compared to IL-22 sufficient Riok2 haploinsufficient mice on day 7 after two treatments with 25 mg/kg PhZ treatment (FIG. 6C). Interestingly, an increase in PB RBCs in Riok2-sufficient mice heterozygous for IL-22 deletion (Riok2+/+Il22+/−) as compared to Riok2-sufficient IL-22 sufficient mice (Riok2+/+Il22+/+) was observed. Next, it was assessed whether the increase in PB RBCs in Riok2+/−Il22+/+ mice was due to increased erythropoiesis in the BM of these mice. An increase in RII and RIV erythroid progenitors in Riok2+/−Il22+/− as compared to Riok2+/−Il22+/+ mice was observed (FIG. 6D). Using a neutralizing IL-22 antibody in vivo, a similar anti-anemic effect on PB RBCs and HCT in Riok2f/+Vav1cre mice, as well as Riok2 sufficient (Riok2+/+Vav1cre) mice undergoing PhZ-induced anemia, was observed (FIG. 6E). Unexpectedly, IL-22 neutralization also reduced the frequency of apoptotic erythroid progenitors in both Riok2-sufficient, as well as Riok2f/+Vav1cre, mice (FIG. 6F). These data indicate that IL-22 neutralization reverses anemia, at least in part, by reducing apoptosis of erythroid progenitors. Recently, dampening IL-22 signaling in intestinal epithelial stem cells was shown to reduce apoptosis (Gronke et al. (2019) Nature 566:249-253). The effect of IL-22 deficiency (genetic as well as antibody-mediated) in alleviating anemia in genetically wild-type mice indicates a role for IL-22 in reversing anemia regardless of ribosomal haploinsufficiency. Accordingly, treatment of C57BL/6J mice undergoing PhZ-induced anemia with anti-IL-22 antibody significantly increased PB RBCs, Hb, and HCT compared to isotype antibody-treated mice (FIG. 8B). Of note, PB RBCs, Hb, and HCT did not differ in healthy, non-anemic wt mice injected with anti-IL-22 versus isotype matched antibodies (FIG. 8A).


IL-22 signals through a cell surface heterodimeric receptor composed of IL-10Rbeta and IL-22RA1 (encoded by Il22ra1) (Kotenko et al. (2001) J. Biol. Chem. 276:2725-2732). IL-22RA1 expression has been reported to be restricted to cells of non-hematopoietic origin (e.g., epithelial cells and mesenchymal cells) (Wolk et al. (2004) Immunity 21:241-254), However, it was unexpectedly discovered herein that erythroid progenitors in the BM also express IL-22RA1 (FIG. 4A and FIG. 10A). Moreover, it was observed that the majority of the IL-22RA1-expressing cells in the BM were erythroid progenitors (FIG. 10B). Using a second anti-IL-22RA1 antibody (targeting a different epitope than the antibody used in FIG. 9A), the presence of IL-22RA1 on erythroid progenitors was confirmed (FIG. 10C).


Deletion of IL-22RA1 in Riok2-haploinsufficient mice (Riok2f/+Il22ra1f/fVav1cre/+) led to improvement in PB RBCs, Hb, and HCT as compared to IL-22RA1-sufficient Riok2-haploinsufficient mice (Riok2f/+Il22ra1+/+Vav1cre/+) (FIG. 9B). This improvement could be attributed to the increase in RH and RIV erythroid progenitors in the BM of Riok2f/+Il22ra1f/fVav1cre/+ mice (FIG. 9C). Erythroid cell-specific deletion of IL-22RA1 (using cre recombinase expressed under the erythropoietin receptor (EpoR) promoter) also increased numbers of PB RBCs and HCT (FIG. 9D) due to the increase in RIII and RIV erythroid progenitors in the BM (FIG. 9E). These data reinforce the notion that IL-22 signaling plays a role in regulating erythroid development regardless of ribosomal haploinsufficiency. Thus, using three different approaches to neutralize IL-22 signaling, it is demonstrated herein that IL-22 plays an important role in inducing anemia by directly regulating erythropoiesis.


Example 5: IL-22 and its Downstream Targets are Increased in del(5q) MDS Patients

It was next assessed whether IL-22 expression is increased in human disorders that display dyserythropoiesis due to ribosomal protein haploinsufficiency. A significant increase in IL-22 levels in the BM fluid (BMF) of del(5q) MDS patients as compared to BMF from healthy controls was observed. A small but significant increase in IL-22 was also observed in non-del(5q) MDS patients (FIG. 11A). Interestingly, a strong negative correlation between cellular RIOK2 mRNA expression and BMF IL-22 level was evident in the del(5q) MDS cohort (FIG. 11B) indicating that a decrease in Riok2 expression is associated with increased IL-22 expression. The frequency of CD4+ T cells producing IL-22 among freshly isolated PBMCs was significantly higher in MDS patients compared to healthy controls (FIGS. 11C and 12).


An independent analysis of a large-scale microarray sequencing dataset of CD34+ cells performed herein from normal, del(5q), non-del(5q) MDS subjects showed that RIOK2 mRNA was significantly decreased in the del(5q) MDS cohort (78% (37/47)). Interestingly, expression of known IL-22 target genes, such as S100A10, S100A1 I, PTGS2, and RAB7A was specifically increased in the del(5q) MDS cohort compared to both the healthy control and non-del(5q) groups (FIG. 11D).


Anemia is a common feature seen in chronic kidney disease (CKD) patients and is associated with poor outcomes. Anemia of CKD is resistant to erythropoiesis-stimulating agents (ESAs) in 10-20% of the patients (KDOQI (2006) Am. J. Kidney Dis. 47:S11-S15) suggesting that pathogenic mechanisms other than erythropoietin deficiency are at play. An increase in IL-22 concentration in the plasma of CKD patients with secondary anemia compared to healthy controls and CKD patients without anemia was determined herein (FIG. 1E) Plasma IL-22 levels negatively correlated with hemoglobin concentration in CKD patients indicating a function for IL-2.2 in driving anemia occurrence in CKD. Thus, the results provided herein demonstrate that IL-22 overexpression is partly responsible for very common anemias, like the anemia observed in patients with chronic kidney disease.


Del(5q), either isolated or accompanied by additional cytogenetic abnormalities, is the most commonly detected chromosomal abnormality in MDS, reported in ˜15% of patients. Anemia is the most common hematologic manifestation of MDS, particularly in patients with del(5q) MDS. The severe anemia in del(5q) MDS patients has been linked to haploinsufficiency of ribosomal proteins such as RPS14 (Schneider et al. (2016) Nat. Med. 22:288-297) and RPS19 (Dutt et al. (2011) Blood 117:2567-2576). Genes lying outside of the 5q region most commonly deleted (5q33) have also been implicated in MDS (Lane et al. (2010) Blood 115:3489-3497; Sebert et al. (2019) Blood 134:1441-1444). While much research has focused on the effect of such gene deletions or mutations in hematopoietic stem cells and lineage-committed progenitors, the Immunobiology underlying this MDS subtype has remained largely unexplored, thus impeding development of immune-targeted therapies. With the exception of the TNF-alpha inhibitor, etanercept (Maciejewski et al. (2002) Br. J. Haematol. 117:119-126), which proved to be ineffective, no other therapies against immune cell-derived cytokines have been tested in MDS patients.


Based on the results described herein, two critical functions have been identified for an understudied kinase, Riok2, that synergize to induce dyserythropoiesis and anemia. The primary effect of Riok2 loss in erythroid progenitors is the intrinsic block in erythroid differentiation owing to its indispensable role in the maturation of the pre-40S ribosomal complex leading to increased apoptosis and cell cycle arrest. The secondary effect of Riok2 loss is the extrinsic induction of the erythropoiesis-suppressive cytokine, IL-22, in T cells, which then directly activates IL-22RA signaling in erythroid progenitors. The data described herein reveal a novel molecular link between haploinsufficiency of a ribosomal protein and induction of erythropoiesis-suppressive cytokine IL-22. While IL-22 has been shown to modulate RBC production by controlling the expression of iron-chelating proteins, such as hepcidin (Smith et al. (2013) J. Immunol. 191:1845-1855) and haptoglobin (Sakamoto et al. (2017) Sci. Immunol. 2:eaai8371), the results described herein elucidate a novel role for IL-22 in directly regulating erythropoiesis in the BM. Using banked and fresh MDS patient samples, it was also demonstrated that IL-22 is elevated in BMF and T cells of MDS patients.


IL-22 is known to play a pathogenic role in some autoimmune diseases (Cai et al. (2013) PLoS One 8:e59009; Ikeuchi et al. (2005) Arthritis Rheum. 52:1037-1046; Yamamoto-Furusho et al. (2010) Inflamm. Bowel Dis. 16:1823). Interestingly, autoimmune diseases, such as colitis, Behcet's disease, and arthritis, are common in MDS patients, with features of autoimmunity observed in up to 10% of patients (Dalamaga et al. (2010) J. Eur. Acad. Dermatol. Venereol. 22:543-548; Lee et al. (2016)Medicine (Baltimore) 95:e3091). Based on the results described herein, it is believed that IL-22 accounts both for the onset of MDS and autoimmunity in this subset of patients. Low-level exposure to benzene, a hydrocarbon, has been associated with an increased risk of MDS (Schnatter et al. (2012) J. Natl. Cancer Inst. 104:1724-1737). Hydrocarbons are known ligands for Ahr, the transcription factor that controls IL-22 production in T cells (Monteleone et al. (2011) Gastroenterology 141:237-248). Stemregenin 1, an Ahr antagonist, was shown to promote the ex vivo expansion of human HSCs, with the highest fold expansion seen in the erythroid lineage (Boitano et al. (2010) Science 329:1345-1348).


More importantly, evidence is provided herein that neutralization of IL-22 signaling effectively treats MDS and anemias, such as stress-induced anemias and the anemia of CKD, diseases that are very much in need of new therapeutic approaches. Ideally, IL-22-based therapeutics may be used not only as monotherapy, but also in conjunction with already existing therapeutics, such as erythropoietin, lenalidomide and azacytidine, which unfortunately presently provide only an average 3- to 5-year window of survival.


Example 6: Effects of IL-22 Neutralization on Riok2 Haploinsufficiency to Alleviate Anemia and Aberrant Myelopoiesis

As described herein, in a model of ribosomal protein (Riok2) haploinsufficiency (Riok2+/−)-induced anemia and myelodysplasia, an increased IL-22 secretion from T cells as compared to normal wild-type (wt, Riok2+/+) T cells was observed. Additionally, a compound genetic deletion of IL-22 on the Riok2 haploinsufficient background (Riok2+/− Il22+/+) partially reversed the anemia seen in IL-22 sufficient Riok2 haploinsufficient (Riok2+/−IL22+/+) mice. Moreover, wt mice undergoing phenylhydrazine-induced acute anemia treated with an anti-IL-22 neutralizing antibody repopulated their peripheral blood RBCs faster than isotype-treated controls. Similarly, anti-IL-22 mAb treatment of Riok2+/− mice partially reversed the anemia in these mice as compared to isotype-treated controls. It was further found that IL-22 levels are increased in the bone marrow fluid (BMF) of MDS patients as compared to healthy controls. Based on these data, disclosed herein are methods of using agents that downregulate IL-22 signaling (e.g., a neutralizing antibody against IL-22) to alleviate the red blood cell deficit seen in anemia and MDS patients.


The MDS-like phenotype in Riok2+/− mice described herein indicated that RIOK2 deletions or inactivating mutations exist in a subclass of MDS patients (with or without del(5q)). Accordingly, a RIOK2 mutation (I245T) was identified in an aplastic anemia patient that acts as a dominant negative to inhibit erythropoiesis. Additionally, independent analysis of a publicly available microarray dataset of MDS patients (Pellagatti et al. (2010) Leukemia 24(4):756-764) showed significant reduction in RIOK2 mRNA in del(5q) MDS patients as compared to healthy controls and to non-del(5q) MDS patients.


The IL-22 receptor, IL-22RA1, is specifically expressed on structural cells and cells of non-hematopoietic origin. It has been determined herein for the first time that erythroid progenitors in the bone marrow express IL-22RA1. Using an erythroid specific-cre recombinase (erythropoietin receptor-cre, EpoR-cre), IL-22RA1 was deleted on erythroid progenitor cells. Mice lacking IL-22RA1 on erythroid cells had significantly higher peripheral blood RBC numbers and hematocrit as compared to the cre alone controls. Thus, using an alternative approach of neutralizing IL-22 signaling in erythroid origin cells, it was proved herein that IL-22 signaling leads to inhibition of erythropoiesis. Based on these data, anti-IL22RA1 blocking antibodies can be used in alleviating red blood cell deficit seen in anemia and MDS.


IL-22RA1, apart from dimerizing with IL-10R2 to form the IL-22RA1/IL-10R2 heterodimer, also couples with IL-20R2 to form a signaling receptor for IL-24. IL-24 has been implicated in anti-tumor functions. Thus, to specifically target IL-22 and not IL-24 signaling via its receptor, antibodies against the IL-22RA1/IL-10R2 heterodimer can be beneficial. An alternative approach to the anti-IL-22 antibody approach is to use recombinant IL-22 binding protein (IL-22BP). IL-22BP is a soluble IL-22 receptor that lacks an intracellular domain and thus sequesters IL-22 to thereby act as an antagonist solely to IL-22 signaling.


As described herein, Riok2 haploinsufficiency increases T cell-derived IL-22, the production of which is controlled by aryl hydrocarbon receptor (AHR) (Monteleone et al. (2011) Gastroenterology 141(1):237-248). Haploinsufficient deletion of Il22 in Riok2+/− mice (Riok2+/−Il22+/−) reversed the erythroid differentiation defect in Riok2+/− mice and antibody-mediated neutralization of IL-22 in wt mice increases peripheral blood RBCs. To further confirm whether IL-22 depletion in Riok2+/− mice normalizes the anemic/25 myelodysplastic phenotype: (a) IL-22 is neutralized using an IL-22 antibody; (b) an AHR antagonist, stemregenin 1 (SR1), is used to abrogate AHR signaling and thus IL-22 production; and (c) the increased IL-22 seen in Riok2+/− mice is compared with examined IL-22 levels for other ribosomal protein haploinsufficiencies (e.g. Rps14, Rpl11, and the like).


Anti-IL-22 antibody (Clone IL22JOP, Thermo Fisher Scientific) has been shown to effectively neutralize IL-22 (Chan et al. (2017) Infect Immun. 85(2); Mielke et al. (2013) J Exp Med. 210(6):1117-1124). Riok2+/− mice (20-24 wk old) are treated intraperitoneally with anti-IL-22 antibody or isotype control (Rat IgG2aκ) at 100 μg/day/mouse twice every week for 8 weeks. After the 8-week treatment period, peripheral blood is analyzed for RBC numbers and other hematological parameters at 8-week intervals to ascertain the prolonged effect of IL-22 neutralization on alleviating anemia. For endpoint analysis at 16 weeks after the cessation of antibody treatment, BM is assessed for frequency and number of hematopoietic and erythroid progenitor cells using flow cytometry.


SR1 is an AHR antagonist that has been shown to maintain pluripotency of human CD34+ stem cells (Boitano et al. (2010) Science 329(5997):1345-1348). SR1-pretreated CD34+cells showed a 129-fold increase in erythroid colonies as compared to untreated cells. No study has yet tested the effect of SR1 in the treatment of myelodysplasia. To this end, six to eight 20-24 wk old Riok2+/− mice are treated intraperitoneally with 0.1 mg/mL SR1 once every week for 8 wks. At the end of 8 weeks, hematological parameters and BM architecture are studied as described above.


For analyzing IL-22 secretion from T cells of ribosomal protein haploinsufficient mouse models, splenic naïve T cells are isolated and cultured in the presence of anti-CD3, anti-CD28, IL-1β, IL-23, and IL-6 for 3 days. IL-22 production is analyzed using flow cytometry and ELISA.


Optionally, the dose of SR1 being used is lowered or other available AHR antagonists (e.g., CH223191, 2′,4′,6-Trimethoxyflavone, and the like) are tested. Alternatively, this regimen is potentiated with low dose lenalidomide or erythropoiesis stimulating agents, such as erythropoietin.


Example 7: Materials and Methods for Examples 8-16

a. Human Samples and Processing


MDS and CKD patient samples were collected under IRB-approved protocols at Dana-Farber Cancer Institute (DFCI) and Brigham and Women's Hospital (BWH), respectively. All samples were de-identified at the time of inclusion in the study. All patients provided informed consent and the data collection was performed in accordance with the Declaration of Helsinki.


Peripheral blood mononuclear cells (PBMCs) from EDTA-treated whole blood were isolated using density gradient centrifugation. PBMCs were then incubated in RPMI with 10% FBS and Cell Activation Cocktail (Tonbo Biosciences) for 4 h and then processed for flow cytometry as described below. Relevant clinical information of MDS samples is provided in Table 3. Adult CKD plasma samples were stored at −80° C. until further use. Relevant clinical information of CKD samples is provided in Table 4.


b. Generation of Riok2 Floxed Mice


Riok2f/f mice were generated using frozen sperm obtained from Mutant Mouse Resource and Research Centers (MMRRC) (Riok2tmla(KOMP)Wtsi) In brief, a floxed Riok2 allele was created by inserting an FRT-flanked IRES-LacZ-neon cassette into intron 4 of the Riok2 gene. LoxP sites were inserted to flank exons 5 and 6. After germline transmission, the FRT cassette was removed by crossing to FLPe deleter mice and resulting floxed mice were bred with individual cre driver strains to create conditional Riok2-deleted mice (FIG. 1A). Genotyping (FIG. 22B) was carried out using the following primers:











Forward primer: 



(SEQ ID NO: 2)



5′ GCATCAGTGATTTACAGACTAAAATGCC 3′







Reverse primer 1:



(SEQ ID NO: 3)



5′ GCTCTTACCCACTGAGTCATCTCACC 3′







Reverse primer 2:



(SEQ ID NO: 4)



5′ CCCAGACTCCTTCTTGAAGTTCTGC 3′






c. Mice


Wild-type C57BL/6J mice (Stock no. 000664), Vav-icre mice (Stock no. 008610), R26-CreErt2 mice (Stock no. 008463), Il22ra1-floxed (Stock no. 031003), CD45.1 C57BL/6J mice (Stock no. 002014), Trp53−/− (Stock no. 002101), Cd4-cre (Stock no. 022071), and Ape′ (Stock no. 002020) mice were purchased from The Jackson Laboratory. Il22−/− mice were provided by R. Caspi (National Institutes of Health, Bethesda, MD) with permission from Genentech (San Francisco, CA). Epor-cre mice were a gift from U. Klingmüller (Deutsches Krebsforschungszentrum (DFKZ), Germany). Il22-tdtomato (Catch-22) mice were a gift from R. Locksley (University of California at San Francisco, CA). Rps14-floxed mice were a gift from B. Ebert (Dana-Farber Cancer Institute, Boston, MA). Mice were housed in the Animal Research Facility (ARF) at DFCI under ambient temperature and humidity with 12 h light/12 h dark cycle.


Animal procedures and treatments were in compliance with the guidelines set forth by the Institutional Animal Care and Use Committee (IACUC) at DFCI. Age- and gender-matched mice were used within experiments.


d. Competitive Bone Marrow (BM) Transplantation


2×106 freshly isolated BM cells from CD45.2+ Riok2f/+Ert2cre or Riok+/+Ert2cre mice were transplanted in competition with 2×106 freshly isolated CD45.1+ wild-type (WT) BM cells via retro-orbital injection into lethally irradiated 8-10 week old CD45.1+ WT recipient mice. The donor cell chimerism was determined in the peripheral blood four weeks after transplantation before the excision of Riok2 was induced by tamoxifen injection as well as every four to eight weeks as indicated. Tamoxifen (75 mg/kg) (Cayman Chemical, Cat #13258) was administered for five consecutive days.


e. Flow Cytometry and Cell Isolation


Whole bone marrow (BM) cells were isolated by crushing hind leg bones (femur and tibia) with mortar and pestle in staining buffer (PBS (Corning) supplemented with 2% heat-inactivated fetal bovine serum (FBS, Atlanta Biologicals) and EDTA (GIBCO). Whole BM was lysed with 1× PharmLyse (BD Biosciences) for 90 s, and the reaction was terminated by adding an excess of staining buffer. Cells were labeled with fluorochrome-conjugated antibodies in staining buffer for 30 min at 4° C. For flow cytometric analysis, cells were incubated with combinations of fluorochrome-conjugated antibodies to the following cell surface markers: CD3 (17A2, 1:500), CD5 (53-7.3, 1:500), CD11b (M1/70, 1:500), Gr1 (RB6-8C5, 1:500), B220 (RA3-6B2, 1:500), Ter119 (TER119, 1:500), CD71 (C2, 1:500), c-kit (2B8, 1:500), Sca-1 (D7, 1:500), CD16/32 (93, 1:500), CD150 (TC15-12F12.2, 1:150), CD48 (HM48-1, 1:500). For sorting of lineage-negative cells, lineage markers included CD3, CD5, CD11b, Gr1 and Ter119. For sorting erythroid progenitor cells, the lineage cocktail did not include Ter119. All reagents were acquired from BD Biosciences, Thermo Fisher Scientific, Novus Biologicals, Tonbo Biosciences, or BioLegend. Identification of apoptotic cells was carried out using the Annexin V Apoptosis Detection Kit (BioLegend). Intracytoplasmic and intranuclear staining was performed using Foxp3/Transcription Factor Staining Kit (Thermo Fisher Scientific) or 0.1% saponin in PBS supplemented with 3% FBS. For staining performed with AF647 p53 antibody (Cell Signaling Technology, 1:50), cells were permeabilized with 90% ice-cold methanol. To increase the sorting efficiency, whole BM samples were lineage-depleted using magnetic microbeads (Miltenyi Biotec) and autoMACS Pro magnetic separator (Miltenyi Biotec). Cell sorting was performed on a FACS Aria flow cytometer (BD Biosciences), data acquisition was performed on a BD Fortessa X-20 instrument equipped with 5 lasers (BD Biosciences) employing FACSDiva software. Data were analyzed by FlowJo (Tree Star) version 9 software. Flow analyses were performed on viable cells by exclusion of dead cells using either DAPI or a fixable viability dye (Tonbo Biosciences). Gating for early and committed hematopoietic progenitors was performed as described elsewhere. ILCs and NKT cells were identified as LinCD45+CD90+CD12+ and CD3ε+ NK1.1+, respectively.


f. Complete Blood Count


Mice were bled via the submandibular facial vein to collect blood in EDTA-coated tubes (BD MICROTAINER™ Capillary Blood Collector, BD 365974). Complete blood counts were obtained using the HemaVet CBC Analyzer (Drew Scientific) or Advia 120 (Siemens Inc.,) instruments.


g. In Vivo Measurement of Protein Synthesis


100 μL of a 20 mM solution of 0-Propargyl-Puromycin (OP-Puro; BioMol) was injected intraperitoneally in mice and mice were then rested for 1 h. Mice injected with PBS were used as controls. BM was harvested after 1 h and stained with antibodies against cell surface markers, washed to remove excess unbound antibodies, fixed in 1% paraformaldehyde, and permeabilized in PBS with 3% FBS and 0.1% saponin. The azide-alkyne cyclo-addition was performed using the Click-iT Cell Reaction Buffer Kit (Thermo Fisher Scientific) and azide conjugated to Alexa Fluor 647 (Thermo Fisher Scientific) at 5 μM final concentration for 30 min. Cells were washed twice and analyzed by flow cytometry. ‘Relative rate of protein synthesis’ was calculated by normalizing OP-Puro signals to whole bone marrow after subtracting autofluorescence.


h. Methylcellulose Assay


250-500 BM Linc-kit+Sca-1+cells were flow sorted and plated in semi-solid methylcellulose culture medium (M3534, StemCell Technologies) and incubated at 37° C. in a humidified atmosphere for 7-10 days. At the end of the incubation period, each well was triturated with staining buffer to collect cells and then processed for flow cytometry as described above. Enumeration of colonies in MethoCult media was performed with StemVision instrument StemCell Technologies).


i. Phenylhydrazine Treatment


Phenylhydrazine (PhZ) was purchased from Sigma and injected intraperitoneally on 2 consecutive days (days 0 and 1) at the dose of 25 mg/kg (sublethal model) or 35 mg/kg (lethal model). Peripheral blood was collected 3-4 days before the start of treatment and at day 4, 7, and 11. PhZ treatment experiments were carried out in 8-12 week old mice.


j. T Cell Polarization


Single-cell suspensions of mouse spleens were prepared by pressing tissue through a 70-μm cell strainer followed by red blood cell lysis using PharmLyse. Total splenic CD4+ T cells were isolated using CD4 T cell isolation kit (Miltenyi Biotec). Enriched CD4+ T cells were then incubated with fluorochrome-conjugated antibodies to CD4, CD8, CD25, CD62L, and CD44 to purify naïve CD4+ T cells using fluorescence activated cell sorting (FACS). In the presence of 1 μg/mL plate-bound anti-CD3ε and soluble 1 μg/mL anti-CD28, T cell polarizations were carried out in IMDM supplemented with 10% FBS, 2 mM L-glutamine, 100 mg/mL penicillin-streptomycin, HEPES (pH 7.2-7.6), non-essential amino acids, 100 μM β-mercaptoethanol (BME), and sodium pyruvate as follows—TH1 (20 ng/mL IL-12, 10 μg/mL anti-IL-4), TH2 (20 ng/mL IL-4, 10 μg/mL anti-IFN-γ), TH17 (30 ng/mL IL-6, 20 ng/mL IL-23, 20 ng/mL IL-1β, 10 μg/mL anti-IL-4, 10 μg/mL anti-IFN-γ), Treg cells (1 ng/mL TGF-β, 10 μg/mL anti-IL-4, 10 μg/mL anti-IFN-γ), and TH22 (30 ng/mL IL-6, 20 ng/mL IL-23, 20 ng/mL IL-1β, 10 μg/mL anti-IL-4, 10 μg/mL anti-IFN-γ, 400 nM FICZ). Pifithrin-α, p-Nitro and Nutlin-3a were purchased from Santa Cruz Biotechnology and Tocris Biosciences, respectively.


k. IL-22 Neutralization and Reconstitution


Monoclonal anti-IL-22 (Clone IL22JOP) blocking antibody and isotype control IgG2aκ (Clone eBR2a) were purchased from Thermo Fisher Scientific. Mice were administered anti-IL-22 (50 μg/mouse) or isotype intraperitoneally every 48 h until the conclusion of the experiment. For recombinant IL-22 treatment, mice were injected with recombinant IL-22 (500 ng/mouse; PeproTech) intraperitoneally every 24 h until the conclusion of the experiment. Mice were administered these reagents at least five times before inducing PhZ-mediated anemia.


l. Cytokine Quantitation


IL-22 in human samples was quantified using either Human IL-22 Quantikine ELISA Kit (D2200, R&D Systems) or SMCTM Human IL-22 High Sensitivity Immunoassay Kit (EMD Millipore, 03-0162-00) according to manufacturers' instructions. The SMC assay was read on a SMC Pro (EMD Millipore) instrument. The lower limit of quantification (LLOQ) of this immunoassay is 0.1 pg/mL. IL-22 in mouse samples was quantified using ELISA MAX™ Deluxe Set Mouse IL-22 (BioLegend, 436304). The LLOQ of this immunoassay is 3.9 pg/mL. S100A8 in human samples was quantified using Human S100A8 DuoSet ELISA (DY4570, R&D Systems).


Concentration of lineage-associated cytokines in cell culture supernatants of polarized T cells were quantified using a custom-made ProCarta Plex assay (Thermo Fisher Scientific) acquired on a Luminex platform. Hepcidin in mouse serum was quantified using a colorimetric assay from Hepcidin MURINE-COMPETE™ ELISA Kit from Intrinsic LifeSciences (HMC-001).


m. mRNA Quantitation


Cells were flow sorted directly into the lysis buffer provided with the CELLS-TO-CT 1-Step TAQMAN™ Kit (A25605, Thermo Fisher Scientific) and processed according to the manufacturer's instructions. Pre-designed TaqMan gene expression assays were used to quantify mRNA expression by qPCR using QuantStudio 6 (Thermo Fisher Scientific). Hprt was used as housekeeping control. Relative expression was calculated using the ΔCt method. Details on primers are indicated in Table 5 for primers details.


n. Chromatin Immunoprecipitation (ChIP)


ChIP was performed using EZ-ChIP Kit (EMD Millipore) according to manufacturer's instructions. Briefly, cells were fixed and cross-linked with 1% formaldehyde at 25° C. for 10 min and quenched with 125 mM glycine for an additional 10 min. Cells pellet was resuspended in lysis buffer and shearing was carried out Diagenode Bioruptor sonication system for a total of 40 cycles beads. Pre-cleared lysates were incubated with control Mouse IgG or anti-p53 (Santa Cruz Biotechnology) antibodies. Il22 promoter-specific primer pair was designed using Primer 3.0 Input and were as follows:











Forward Primer:



(SEQ ID NO: 5)



5′ CCAAACTTAACTTGACCTTGGC 3′







Reverse Primer:



(SEQ ID NO: 6)



5′ TTCTTCACAGCTCCCA TTGC 3′






o. In Vitro Erythroid Differentiation


Whole BM cells were labeled with biotin-conjugated lineage antibodies (cocktail of anti-CD3ε, anti-CD11b, anti-CD45R/B220, anti-Gr1, anti-CD5, and anti-TER-119) (BD Pharmingen) and purified using anti-biotin beads and negative selection on the AutoMACS Pro (Miltenyi). Purified cells were then seeded in fibronectin-coated (2 μg/cm2) tissue-culture treated polystyrene wells (CORNING® BIOCOAT™ Cellware) at a cell density of 105/mL. Erythroid differentiation was carried out according to modified published protocols. The erythropoietic medium was IMDM supplemented with erythropoietin at 10 U/mL, 10 ng/mL stem cell factor SCF, PeproTech), 10 μM Dexamethasone (Sigma-Aldrich), 15% FBS, 1% detoxified BSA (StemCell Technologies), 200 μg/mL holotransferrin (Sigma-Aldrich), 10 mg/mL human insulin (Sigma-Aldrich), 2 mM L-glutamine, 0.1 mM β-mercaptoethanol, and penicillin-streptomycin. After 48 h, the medium was replaced by IMDM medium containing 20% FBS, 2 mM L-glutamine, 0.1 mM β-mercaptoethanol, and penicillin-streptomycin. 50% of the culture media was replaced after 48 h and cell density was maintained at 0.5×106/mL. Total culture period for the assay was 6 days. Recombinant mouse IL-22 (PeproTech/Cell Signaling) was used where indicated. RII-RIV populations were gated as shown in FIG. 23A.


p. Proteomic Profiling


Proteomic profiling of sort-purified erythroid progenitors was performed as described elsewhere. Briefly, cells were captured in collection microreactors and stored at −80° C. Cell lysis was performed by adding 10 μL of 8 M urea, 10 mM TCEP and 10 mM iodoacetamide in 50 mM ammonium bicarbonate (ABC) to the cell pellet of 1×106 erythroid progenitors and incubated at room temperature for 30 min, shaking in the dark. 50 mM ABC was used to dilute the urea to less than 2 M and the appropriate amount of trypsin for a 1:100 enzyme to substrate ratio was added and allowed to incubate at 37° C. overnight. Once digestion is completed, the lysate is spun through the glass mesh directly onto a C18 Stage tip (Empore) at 3500×g until the entire digest passes through the C18 resin. 75 μL 0.1% formic acid (FA) is then used to ensure transfer of peptides to the C18 resin from the mesh while washing away the lysis buffer components. C18-bound peptides were immediately subjected to on-column TMT labeling.


On-column TMT Labeling—Resin was conditioned with 50 μL methanol (MeOH), followed by 50 μL 50% acetonitrile (ACN)/0.1% FA, and equilibrated with 75 μL 0.1% FA twice. The digest was loaded by spinning at 3500×g until the entire digest passed through. One μL of TMT reagent in 100% ACN was added to 100 μL freshly made HEPES, pH 8, and passed over the C18 resin at 350×g until the entire solution passed through. The HEPES and residual TMT was washed away with two applications of 75 μL 0.1% FA and peptides were eluted with 50 μL 50% ACN/0.1% FA followed by a second elution with 50% ACN/20 mM ammonium formate (NH4HCO2), pH 10. Peptide concentrations were estimated using an absorbance reading at 280 nm and checking of label efficiency was performed on 1/20th of the elution. After using 1/20th of the elution to test for labeling efficiency, the samples are mixed before fractionation and analysis.


Stage tip bSDB Fractionation—200 μL pipette tips were packed with two punches of sulfonated divinylbenzene (SDB-RPS, Empore) with a 16-gauge needle. After loading ˜20 μg peptides total, a pH switch was performed using 25 μL 20 mM NH4HCO2, pH 10, and was considered part of fraction one. Then, step fractionation was performed using ACN concentrations of 5, 7.5, 10, 12.5, 15, 17.5, 20, 25, 42, and 50%. Each fraction was transferred to autosampler vials and dried via vacuum centrifugation and stored at 80° C. until analysis. Data Acquisition—Chromatography was performed using a Proxeon UHPLC at a flow rate of 200 nl/min. Peptides were separated at 50° C. using a 75 μm i.d. PicoFrit (New Objective) column packed with 1.9 μm AQ-C18 material (Dr. Maisch, Germany) to 20 cm in length over a 110 min run. The on-ine LC gradient went from 6% B at 1 min to 30% B in 85 mins, followed by an increase to 60% B by minute 94, then to 90% by min 95, and finally to 50% B until the end of the run. Mass spectrometry was performed on a Thermo Scientific Lumos Tribrid mass spectrometer. After a precursor scan from 350 to 1800 m/z at 60,000 resolution, the topmost intense multiply charged precursors in a 2 second window were selected for higher energy collisional dissociation (HCD) at a resolution of 50,000. Precursor isolation width was set to 0.7 m/z and the maximum MS2 injection time was 110 msecs for an automatic gain control of 6e4. Dynamic exclusion was set to 45 s and only charge states two to six were selected for MS2. Half of each fraction was injected for each data acquisition run.


Data Processing—Data were searched all together with Spectrum Mill (Agilent) using the Uniprot Mouse database (28 Dec. 2017), containing common laboratory contaminants and 553 smORFs. A fixed modification of carbamidomethylation of cysteine and variable modifications of N-terminal protein acetylation, oxidation of methionine, and TMT-11plex labels were searched. The enzyme specificity was set to trypsin and a maximum of three missed cleavages was used for searching. The maximum precursor-ion charge state was set to six. The MS1 and MS2 mass tolerance were set to 20 ppm. Peptide and protein FDRs were calculated to be less than 1% using a reverse, decoy database. Proteins were only reported if they were identified with at least two distinct peptides and a Spectrum Mill score protein level score ˜20. TMT11 reporter ion intensities in each MS/MS spectrum were corrected for isotopic impurities by the Spectrum Mill protein/peptide summary module using the afRICA correction method which implements determinant calculations according to Cramer's Rule and general correction factors obtained from the reagent manufacturer's certificate of analysis. Differential Protein Abundance Analysis—The median normalized, median absolute deviation-scale data set was subjected to a moderated F-test, followed by Benjamini-Hochberg Procedure correcting for multiple hypothesis testing. An arbitrary cutoff were drew at adj. p val <0.05.


q. RNA Sequencing (RNA-Seq)


5000 IL-22+(CD4+IL-22(tdtomato)+) cells were FACS sorted directly into TLC Butter (Qiagen) with 1% β-mercaptoethanol. For the preparation of libraries, cell lysates were thawed and RNA was purified with 12×RNAClean SPRI beads (Beckman Coulter Genomics) without final elution. The RNA captured beads were air-dried. and processed immediately for RNA secondary structure denaturation (72° C. for 3 min) and cDNA synthesis. SMART-seq2 was performed on the resultant samples following the published protocol with minor modifications in the reverse transcription (RI) step. A 15 μL reaction mix was used for subsequent PCR and performed 10 cycles for cDNA amplification. The amplified cDNA. from this reaction was purified with 0.8× Ampure SPRI beads (Beckman Coulter Genomics) and eluted in 21 μL TIE buffer. 0.2 ng cDNA and one-eighth of the standard Illwnina NexteraXT (Illumina FC-131-1096) reaction volume were used to perform both the tagmentation and PCR indexing steps. Uniquely indexed libraries were pooled and sequenced with NextSeq 500 high output V2 7:5 cycle kits (Illutnina FC-404-2005) and 38×38 paired-end reads on the NextSeq 500 instrument. Reads were aligned to the mouse mm10 transcriptome using Bowtie62, and expression abundance TPM estimates were obtained using RSEM.


r. Pathway Analysis


Gene set enrichment analysis (GSEA) was performed with Broad Institute's GSEA Software. The ‘IL-22 Signature’ and ‘Rps14 Increased’ gene sets were created from the literature (FIG. 15A, D). Full lists of genes in the individual gene sets can be found in Table 2. Other reference gene sets are available from MSigDB. For GSEA analyses, mouse UniProt IDs were converted to their orthologous human gene symbols using MSigDB 7.1 CHIP file mappings. Pathway enrichment (FIG. 15C) was performed using Clarivate Analytics' METACORE™ software.


s. Microarray Data Analysis


Microarray data of CD34+cells from healthy, del(5q), and non-del(5q) MDS subjects was obtained from a previously published study submitted in Gene Expression Omnibus accessible under GSE19429.


t. Statistical Tests


Data are presented as mean±s.e.m unless otherwise indicated. Comparison of two groups was performed using paired or unpaired two-tailed t-test. For multiple group comparisons, analysis of variance (ANOVA) with Tukey's correction or Kruskal-Wallis test with Dunn's correction was depending on data requirements. Statistical analyses were performed using GraphPad Prism v8.0 (GraphPad Software Inc.). A P-value of less than 0.05 was considered significant.









TABLE 2







Signatures used for GSEA analyses.













Rps14



IL-22 Signature

Signature















Bcl1
Ccl20
S100a8



Cldn1
Muc1
Pglyrp1



Lbp
Muc3
Retnlg



Il1
Muc10
Mgp



Il6
Muc13
S100a9



Il8
Defb2
Ltf



Il11
Defb3
Lcn2



Gcsf
Lcn2
Chi3l1



Gmcsf
Mmp1
Camp



Ccl2
Mmp3
Ngp



Rankl
Il20
Apobr



Cxcl1
Saal1
Lrg1



Cxcl2
Hp
Padi4



Cxcl5
Hamp
Hp



Cxcl8
Cdk4
Lyz2



Reg3g
Myc
Anxa1



Reg3b
p21
Gys1



Bcl2
Il10
Gda



Bclxl
Serpina3
Nfrkb



S100a7
Cdkn1a
Aldh3b1




Defb



S100a8
Prdx5
S100a11



S100a9
Saa
Itgam



S100a10
Saa1
Itgb2



S100a11

Ncf2





Pygl





Cnn2

















TABLE 3







Karyotype of MDS patients. ISCN = International System for Human Cytogenetic Nomenclature








Patient
Karyotype (ISCN nomenclature)





non-del(5q)-1
46, XY[15]


non-del(5q)-2
46, XX[20]


non-del(5q)-3
45, XX, del(1)(p22), der(2)del(2)(p12)t(1; 2)(p22; q31), add(11)(p15), −13, 21



pstk+, add(22)(q11.2)[cp3]/46, XX[6]


non-del(5q)-4
42, X, −Y, −4, add(5)(q3?1), add(7)(q3?1), −13,



add(15)(p11.2), −16, −18, −20, +2mar[17]/46, XY[3}


del(5q)-1
43, X, −Y[4], del(1)(q11)[2], add(4)(q24), del(5)(q1?5q33), −7, −9, −12, add(16)(p11.2),



der(19)t(12; 19)(q13; p13.1), −22, +1-2r, +1-2mar[cp6]/46, XY[2]


non-del(5q)-5
46, XY[20]


non-del(5q)-6
46, XY[20]


non-del(5q)-7
46, XY[20]


non-del(5q)-8
46, XY[20]


non-del(5q)-9
46, XY, del(20)(q11.2q13.3)[cp8]/46, XY, +1, der(1)t(1; 13)(p12; q11), −13,



del(20)[cp12].nuc ish(D7Z1, D7S486)x2[100]


non-del(5q)-10
46, XY[20]


non-del(5q)-11
K1: 47, XY, +12[9]/47, idem, del(14)(q21q31)[4]/46, XY[cp7]



K2: 47, XY, +14[2]/46, XY[18].nuc ish(IGHx3)[14/100]


non-del(5q)-12
45, XX, −7[4]/46, XX[12].nuc ish(D7Z1, D7S486)x1[32/100]


non-del(5q)-13
46, XX[20]


non-del(5q)-14
46, XY, del(3)(q13q24)[7]/46, XY[13]


non-del(5q)-15
46, XY[20]


non-del(5q)-16
44, XY, add(3)(p21), add(5)(q11.2), der(6)t(3; 6)(p13; q13), −7,



del(12)(p11.2p13), −18, −20, add(20)(p12), +r[cp20]


del(5q)-2
41~45, Y, t(X; 2)(q?24; ? q31), add(2)(p13), −5, add(8)(p21), ?inv(9)(p22q12),



del(11)(p13), −12, −13, −15, −15, −16, add(17)(p11.2), del(18)(q12.2), del(18)(q21), −add(19)(p13.3), −21,



add(21)(q22), add(22)(q13), +4-6 mar[12]/32~35,idem, −3, −4, −7, −10,



add(17), −del(18)(q21), −20, −add(22), +2-4mar[5]/44, XY, add(2)(q31), −5, add(8),



adel(11), −13, add(15)(p11.2), add(17), add(19)(p13), add(20)(q13.3), +mar[3]/46, XY[1]


non-del(5q)-17
46, XX[20]


non-del(5q)-18
47, XY, +8[16]/46, XY[4]


non-del(5q)-19
46, XX[10]


non-del(5q)-20
45, X, −Y[6]/46, XY[14]


non-del(5q)-21
46, XX[20]


non-del(5q)-22
46, XY[20]


del(5q)-3
de15q (full report not available)


non-del(5q)-23
46, XY, del(8)(q2?2q2?4), add(14)(q32)[1]/46, XY[cp19


non-del(5q)-24
46, XY[20]
















TABLE 4







Clinical Information for CKD samples. eGFR =


Estimated glomerular filtration rate, Hgb - Hemoglobin.










eGFR
Hgb














29
15.2



25
13.5



25
14.9



34
9.9



16
14.7



18
13.9



15
10.3



26
12.6



29
14.5



23
13.7



28
9.2



19
14.4



26
14



21
9.4



21
11.9



19
9.9



27
13.3



12
9.8



15
12.6



13
10.5



17
9.6



27
14.9



17
7.7



33
9.7



58
13.3



10
13.8

















TABLE 5







qRT-PCR Taqman primers used in this study.










Gene
Catalog #







Mouse Riok2
Mm00482415_m1



Human RIOK2
Hs01084566_m1



Mouse S100a8
Mm00496696_g1



Mouse S100a9
Mm00656925_m1



Mouse Hprt
Mm03024075_m1



Human HPRT1
Hs02800695_m1



Mouse Cdkn1a
Mm00432448_m1



Mouse Il22ral
Mm01192943_m1



Mouse Trp53
Mm01731290_g1



Mouse Gadd45a
Mm00432802_m1



Mouse Bbc3
Mm00519268_m1










Example 8: Riok2 Haploinsufficiency Leads to Anemia

Myelodysplastic syndromes (MDS) are a group of cancers characterized by failure of blood cells in the bone marrow to mature. About 7 out of 100,000 people are affected and the typical survival time following diagnosis is less than three years. While a sizable percentage of MDS cases progress to acute myelogenous leukemia (AML), most of the morbidity and mortality associated with MDS results not from transformation to AML but rather from hematological cytopenias.


Anemia is the most common hematologic manifestation of MDS, particularly in the subset of patients with del(5q) MDS. Del(5q), either isolated or accompanied by additional cytogenetic abnormalities, is the most commonly detected chromosomal abnormality in MDS, reported in 10-15% of patients. The severe anemia in del(5q) MDS patients has been linked to haploinsufficiency of ribosomal proteins such as RPS14 and RPS19. Previous studies using mice with haploinsufficient 5q gene deletions revealed diminished erythroid progenitor frequency but the mechanisms underlying this phenotype are incompletely understood. Right open-reading-frame kinase 2 (RIOK2) encodes an atypical serine-threonine protein kinase with an indispensable function as a component of the pre-40S ribosome subunit.


There is growing evidence for the role of activated innate immunity and inflammation as well as immune dysregulation in the pathogenesis of MDS. Abnormal expression of numerous cytokines has been reported in MDS. Chronic immune stimulation in both hematopoietic stem and progenitor cells (HSPCs) and the bone marrow (BM) microenvironment was suggested to be central to the pathogenesis of MDS. In patients with chronic inflammation, cytokines in the BM have been associated with inhibition of erythropoiesis. Despite growing evidence for a link between the immune system and MDS pathogenesis, no study has identified the mechanism by which the immune microenvironment may initiate or contribute to the MDS phenotype. Further it remains unclear how ribosomal protein haploinsufficiency is connected with the immune system in MIDS.


As disclosed herein, Riok2 expression was reduced in T cells lacking the endoplasmic reticulum stress transcription factor Xbp1. RIOK2 is a little-studied atypical serine-threonine protein kinase encoded by RIOK2 at 5q15 in the human genome (FIG. 1A), adjacent to the 5q commonly deleted regions in MDS and frequently lost in MDS and acute myeloid leukemia. Gene expression commons (GEXC) analysis revealed that in mouse BM, Riok2 expression is highest in primitive colony-forming-unit erythroid (pCFU-E) cells, suggesting that RIOK2 may be involved in maintaining red blood cell (RBC) output (FIG. 1B). To further study the role of RIOK2 in hematopoiesis, Vav1-Cre transgenic foxed Riok2 (Riok2f/+Vav1cre) mice were generated in which the Cre recombinase is under the control of the hematopoietic cell—specific Vav1 promoter. Riok2 foxed mice were generated with exons 5 and 6 flanked by loxP sites (FIGS. 22A and 22B). Interestingly, no Vav1-Cre foxed Riok2 homozygous-deficient mice (Riok2 f Vav1cre) were recovered (FIG. 22D), indicating embryonic lethality from complete hematopoietic deletion of Riok2. However, heterozygous Riok2f/+Vav1cre mice were viable with approximately 50% Riok2 mRNA expression in hematopoietic cells compared to that of Vav1cre controls (FIG. 22C). As seen with other ribosomal protein haploinsufficiency mouse models, BM cells from Riok2f/+Vav1cre mice showed reduced nascent protein synthesis in vivo compared to Vav1cre controls (FIG. 22E), consistent with a RIOK2 role in maturation of the pre-40S ribosome. A recent study showed that ribosomal protein deficiency-mediated reduced protein synthesis predominantly affects erythropoiesis over myelopoiesis.


Consistent with the high expression of Riok2 in pCFU-e cells in the BM, aged (>60 wks) mice with heterozygous deletion of Riok2 in hematopoietic cells (Riok2f/+Vav1cre) displayed anemia with reduced peripheral blood (PB) RBC numbers, hemoglobin (Hb), and hematocrit (HCT) (FIG. 14A). Next, it was determined whether Riok2 haploinsufficiency-mediated anemia was secondary to a defect in erythroid development in the BM, the major site of erythropoiesis. The stages (referred to here as RI, RII, RIII and MV) of erythropoiesis were characterized by flow cytometry using the expression of Ter119 and CD71 (FIG. 23A). Riok2f/+Vav1cre mice had impaired erythropoiesis in the BM (FIG. 14B, FIG. 23B), Moreover, Riok2 haploinsufficiency led to increased apoptosis in erythroid precursors compared to controls (FIG. 14C). Additionally, Riok2f/+Vav1cre erythroid precursors showed a decrease in cell quiescence with cell cycle block at the G1 phase (FIG. 23C). A block in cell cycle is driven by a group of proteins known as cyclin-dependent kinase inhibitors (CM). The expression of p21 (a CKI encoded by Cdkn1a) was increased in erythroid precursors from Riok2f/+Vav1cre mice compared to Riok2+/+Vav1cre controls (FIG. 23D).


The effect of Riok2 haploinsufficiency on stress-induced erythropoiesis were examined using 8-12 wk old mice in which hemolysis was induced by non-lethal phenylhydrazine treatment (25 mg/kg on days 0 and 1). After acute hemolytic stress, Riok2f/+Vav1cre mice developed more severe anemia and had a delayed RBC recovery response, as compared to Riok2+/+Vav1cre control mice (FIG. 14D) and succumbed faster to a lethal dose of phenylhydrazine (35 mg/kg on days 0 and 1) compared to Vav1cre controls (FIG. 23E). The anemia in young phenylhydrazine-administered Riok2f/+Vav1cre mice seen on day 7 was preceded by a reduction in BM RIII and RI erythroid precursor frequency on day 6, highlighting an erythroid differentiation defect in Riok2 haploinsufficient mice (FIG. 14E, FIG. 23F). In line with a role for RIOK2 in driving erythroid differentiation, fewer CFU-e colonies were observed in erythropoietin-containing MethoCult cultures from Riok2 haploinsufficient Linc-kit+CD71+ cells compared to Riok2 sufficient cells (FIG. 14F). To determine whether Riok2 haploinsufficiency in BM cells drives anemia, BM chimeras were generated. Wild-type (WO mice transplanted with Riok2f/+Vav1cre whole BM developed anemia compared to Riok2+/+Vav1cre BM transplanted wt mice (FIG. 23G). In addition, tamoxifen-inducible deletion of Riok2 in Riok2f/+Ert2cre mice led to reduction in PB RBCs, Hb, and HCT compared to Riok2+/+Ert2cre controls (FIG. 23H). Taken together, these data show that Riok2 haploinsufficiency leads to anemia owing to defective bone marrow erythroid differentiation.


Example 9: Riok2 Haploinsufficiency Increases Myelopoiesis

In addition to the reduction in RBC numbers in PB from aged Riok2f/+Vav1cre mice, an increased percentage of monocytes (monocytosis) and decreased percentage of neutrophils (neutropenia) were also observed compared to controls (FIG. 14G, FIG. 23I). Granulocyte macrophage progenitors (GMPs) in the BM give rise to PB myeloid cells. The percentage of proliferating (Ki67+) GMPs in the BM was increased in Riok2f/+Vav1cre mice compared to Riok2+/+Vav1cre controls (FIG. 14H, FIG. 23J). To analyze the effect of Riok2 haploinsufficiency on myelopoiesis in the absence of in vivo compensatory mechanisms, LSK (lineageSca-1+Kit+) cells from the BM of Riok2f/+Vav and Riok2+/+Vav1cre controls were cultured in a MethoCult assay supplemented with growth factors (interleukin 6 (IL-6), IL-3, stem cell factor (SCF) but devoid of erythropoietin). LSKs from Riok2f/+Vav1cre mice gave rise to an increased percentage of CD11b+ myeloid cells (FIG. 14I, FIG. 23K), suggesting a cell-intrinsic myeloproliferative effect due to Riok2 haploinsufficiency consistent with a myelodysplasia phenotype.


It was also evaluated whether Riok2 haploinsufficiency affects early hematopoietic progenitors. Frequency and numbers of early hematopoietic progenitors were comparable between young Riok2f/+Vav1cre and Riok2+/+Vav1cre mice (FIG. 24A), however, long-term hematopoietic stem cells (LT-HSCs) were increased in the BM of aged Riok2f/+Vav1cre mice (FIG. 24A). To further corroborate this data, the capacity of Riok2 haploinsufficient cells were analyzed in a competitive transplantation assay. Starting at 8 weeks after tamoxifen treatment to induce Riok2 deletion, Riok2 haploinsufficient cells out-competed CD45.1+competitor cells, while Ert2cre control cells had no competitive advantage (FIG. 24B). Similar to non-transplanted mice (FIG. 24A), in competitive transplant experiments the frequency of Riok2-haploinsufficient LT-HSCs was significantly higher than Riok2-sufficient LT-HSCs in relation to competitor CD45.1+ cells (FIG. 24C). Thus, in addition to its effect on erythroid differentiation, Riok2 haploinsufficiency increases myelopoiesis and affects early hematopoietic progenitor differentiation.


Example 10: Reduced Riok2 Induces Alarmins in Erythroid Precursors

To elucidate a mechanism for the erythroid differentiation defect observed in Riok2f/+Vav1cre mice, quantitative proteomic analysis of purified erythroid precursors were performed using mass spectrometry. Riok2 haploinsufficiency led to upregulation of 564 distinct proteins (adjusted p-value <0.05) in erythroid precursors compared to those from Vav/cre controls (FIG. 4A). Interestingly, Riok2 haploinsufficiency resulted in down-regulation of other ribosomal proteins, loss of some of which (RPS5, PRL11) has been implicated in driving anemias (FIG. 25A). The alarmins including S100A8, S100A9, CAMP, NGP, and others were the most highly upregulated proteins in our dataset and interestingly, correlated significantly with those observed upon haploinsufficiency of Rps14, another component of the 40S ribosomal complex (FIG. 4B). Using the 26 upregulated proteins in the Rps14 haploinsufficient dataset as an ‘Rps14 signature’ (Table 4, gene set enrichment analysis (GSEA) revealed a marked enrichment for the Rps14 signature in the Riok2 haploinsufficient dataset, suggesting a shared proteomic signature upon deletion of distinct ribosomal proteins (FIG. 15A). The increased expression of S100A8 and S100. A9 in Riok2f/+Vav1cre mice was confirmed by flow cytometry and qRT-PCR (FIGS. 25B-E).


In Riok2f/+Vav1cre erythroid precursors cells, the upregulated proteins with the highest fold-change (S100A8, S100A9, CAMP, NGP) are proteins with known immune functions such as antimicrobial defense. GSEA analysis of the proteomics data indicated a possible role for the immune system in driving the proteomic changes seen in Riok2 haploinsufficient erythroid precursors (FIG. 15B). An independent analysis of the Riok2 proteomics dataset using MetaCore pathway analysis software showed immune response as the top differentially regulated pathway in Riok2f/+Vav1cre mice (FIG. 15C). To assess if Riok2 haploinsufficiency leads to changes in immune cell function, naïve T cells from Riok2f/+Vav1cre mice and Riok2+/+Vav1cre controls were subjected to in vitro polarization towards known CD4+ T helper cell lineages (TH1, TH2, TH17, TH22) and regulatory T cells (Treg). Secretion of interferon-γ (IFN-γ), IL-2, IL-4, IL-5, IL-13, IL-17A, and the frequency of Foxp3+ Treg cells was similar between Riok2f/+Vav1cre and Riok2+/+Vav1cre T cells (FIGS. 26A-G). Strikingly, however, an exclusive increase in IL-22 secretion were observed from Riok2f/+Vav1cre naïve T cells polarized towards the TH22 lineage (FIG. 16A). The frequency of IL-22+CD4+ T cells was higher in Riok2f/+Vav1cre TH22 cultures compared to Vav/cre control TH22 cultures (FIG. 16B). The concentration of IL-22 in the serum and BM fluid (BMF) of aged Riok2f/+Vav1cre mice was also significantly higher as compared to age-matched Vav/cre controls (FIG. 16C). Using known IL-2.2 target genes from the literature, an ‘IL-22 signature’ (Table 2) gene set were curated, which showed a statistically significant enrichment in the Riok2-haploinsufficient proteomics dataset using GS. EA (FIG. 15D), further suggesting that IL-22-induced inflammation is a contributing factor for Riok2 haploinsufficiency-mediated ineffective erythropoiesis and anemia. Increased numbers of splenic IL-22+CD4+ T, natural killer T (INKT), and innate lymphoid cells (ILCs) were observed in aged Riok2f/+Vav1cre mice compared to Riok2+/+Vav1cre mice (FIG. 16D, FIG. 26H, I). Interestingly, mild anemia was observed in mice lacking Riok2 only in T cells (FIG. 26K). Expression of IL-23, required for IL-22 production, was enhanced in Riok2-haploinsufficient dendritic cells (FIG. 26J). Rps14 haploinsufficient TH22 cells also secreted elevated concentrations of IL-22 compared to Vav1cre control TH22 cells (FIG. 26L). Mutation(s) in the gene adenomatosis polyposis coli (Ape), also found on human chromosome 5q, lead to anemia in addition to adenomas. in vitro generated TH22 cells from ApeMin mice secreted elevated IL-22 compared to littermate controls (FIG. 26M). In total, our analysis of three distinct heterozygous deletions of genes found on human chromosome 5q suggests that increased IL-22 is a generalized phenomenon observed upon heterozygous loss of genes found on chromosome 5q leading to anemia.


Example 11: p53 Upregulation Drives Increased IL-22 Secretion Upon Riok2 Loss

To identify cell-intrinsic molecular mechanism(s) driving the increase in IL-22 secretion upon Riok2 haploinsufficiency, RNA-sequencing (RNA-Seq) were performed on in vitro polarized TH22) cells purified by flow cytometry from Riok2+/+Il22tdtomato/+Vav1cre and Riok2f/+Il22tdtomato/+Vav1cre mice (FIG. 16E). GSEA analysis of the RNA-Seg dataset identified activation of the p53 pathway in Riok2f/+Vav1cre mice (FIGS. 16F, G). p53 increase in TH22 cells from Riok2f/+Vav1cre mice was confirmed by flow cytometry (FIGS. 16H, I). p53 upregulation was also observed in Riok2f/+Vav1cre erythroid precursors (FIGS. 26F, G). The p53 pathway is activated by decreased expression of ribosomal protein genes, however, its involvement in IL-22 regulation has not been known.


p53 is a transcription factor with well-defined consensus binding sites. To assess whether p53 drives Il22 transcription, the Il22 promoter for potential p53 binding sites were analyzed using LASAGNA algorithm and found putative p53 consensus binding sequences in the Il22 promoter (FIG. 16J). Chromatin immunoprecipitation (ChIP) confirmed the presence of p53 on the Il22 promoter (FIG. 16K). In line with the ChIP data, p53 inhibition by pifithrin-α, p-nitro decreased IL-22 concentrations while p53 activation by nutlin-3 increased IL-22 from in vitro polarized wild-type TH22 cells (FIG. 16L, M). Treatment with either pifithrin-α, p-nitro or nutlin-3 did not decrease cell viability (FIG. 26N). Accordingly, genetic deletion of Trp53 blunted the increase in IL-22 secretion observed upon Riok2 haploinsufficiency (FIG. 16N). A significant decrease in IL-22 secretion was also observed upon Trp53 deletion in Riok2-sufficient cells further suggesting a homeostatic role for p53 in controlling IL-22 production (FIG. 16N). Taken together, these data show that Riok2 haploinsufficiency-mediated p53 upregulation drives increased IL-22 secretion in Riok2f/+Vav1cre mice.


Example 12: IL-22 Neutralization Alleviates Stress-Induced Anemia

Mice with compound genetic deletion of Il22 on the Riok2 haploinsufficient background (Riok2f/+Il22+/−Vav1cre) exhibited increased numbers of PB RBCs compared to IL-22 sufficient Riok2 haploinsufficient mice on day 7 after two treatments with 25 mg/kg phenylhydrazine treatment (FIG. 17A). Interestingly, an increase was also evidenced in PB RBCs in Riok2 sufficient mice heterozygous for Il22 deletion (Riok2+/+Il22+/+Vav1cre) compared to Riok2 sufficient IL-22 sufficient mice (Riok2+/+Il22+/+Vav1cre). PB Hb and HCT also were increased in Il22 haploinsufficient mice, regardless of Riok2 background, however, this difference did not reach statistical significance (FIG. 17A). Next, it was assessed whether the increase in PB RBCs in Riok2f/+Il22+/− Vav1cre mice was due to increased erythropoiesis in the BM of these mice. An increase were observed in RII and RIV erythroid precursors in Riok2f/+Il22+/−Vav1cre compared to Riok2f/+Il22+/+Vav1cre mice (FIG. 17B, FIG. 27A). Treatment of mice with a neutralizing IL-22 antibody in vivo, also reversed phenylhydrazine-induced anemia as evidenced by increase in PB RBCs and HCT in Riok2f/+Vav1cre mice as well as Riok2 sufficient (Riok2+/+Vav1cre) mice (FIG. 17C). Unexpectedly, IL-22 neutralization also reduced the frequency of apoptotic erythroid precursors in both Riok2 sufficient as well as Riok2f/+Vav1cre mice (FIG. 17D). These data indicate that IL-22 neutralization reverses anemia, at least in part, by reducing apoptosis of erythroid precursors. Recently, dampening IL-22 signaling in intestinal epithelial stem cells was shown to reduce apoptosis. The effect of IL-22 deficiency (genetic as well as antibody-mediated) in alleviating anemia in genetically wild-type mice indicated a role for IL-22 in reversing anemia regardless of ribosomal haploinsufficiency. Accordingly, treatment of C57BL/6J mice undergoing phenylhydrazine-induced anemia with anti-IL-22 significantly increased PB RBCs, Hb, and HCT compared to isotype antibody-treated mice (FIG. 28B). This increase in PB RBCs could be attributed to the increased frequency of RIII and RIV erythroid precursors in the BM of mice treated with anti-IL-22 compared to isotype-administered controls (FIG. 28C). Of note, PB RBCs, Hb, and HCT did not differ in healthy, non-anemic wild-type mice injected with anti-IL-22 versus isotype-matched antibody (FIG. 28A). Thus, IL-22 neutralization, either by genetic deletion or antibody blockade, alleviates stress-induced anemia in Riok2f/+Vav1cre as well as wild-type mice.


Example 13: IL-22 Worsens Stress-Induced Anemia in Wild-Type Mice

Phenylhydrazine administration to wild-type C57BL/6J mice treated intraperitoneally with recombinant IL-22 (rIL-22) led to decreased PB RBCs, Hb, and 1-ICT owing to decreased BM erythroid precursor cell frequency and number (FIG. 18A, C, FIG. 27B). rIL-22 treatment led to increased apoptosis of erythroid precursors (FIG. 5d). This treatment also led to an increase in PB reticulocytes, an indication of increased erythropoiesis under stress (FIG. 18B). Recombinant IL-22 also dose-dependently decreased terminal erythropoiesis in an in vitro erythropoiesis assay (FIG. 18E, F). Importantly, IL-22-mediated inhibition of in vitro erythropoiesis led to induction of p53 suggesting a feedback loop between IL-22 and p53 in driving dyserythropoiesis (FIG. 18G). Overall, these data show that exogenous recombinant IL-22 exacerbates stress-induced anemia in wild-type mice.


Example 14: Erythroid Precursors Express IL-22RA1 Receptors

IL-22 signals through a cell surface heterodimeric receptor composed of IL-10Rβ and IL-22RA1 (encoded by Il22ra1). IL-22RA1 expression has been reported to be restricted to cells of non-hematopoietic origin (e.g., epithelial cells and mesenchymal cells). It was discovered, however, that erythroid precursors in the BM also express IL-22RA1 (FIG. 19A, FIG. 29A). Moreover, among BM hematopoietic progenitors, IL-22RA1-expressing cells were exclusively of the erythroid lineage (FIG. 29B). Using a second IL-22RA1-specific antibody (targeting a different epitope than the antibody used in FIG. 4A), the presence of IL-22RA1 on erythroid precursors were confirmed (FIG. 29C), Il22ra1 mRNA expression was detected exclusively in erythroid precursors among all lineage-negative cells in the BM (FIG. 29D).


Deletion of Il22ra1 in Riok2 haploinsufficient mice (Riok2f/+Il22ra1f/fVav1cre) led to improvement in PB RBCs and HCT as compared to IL-22RA1 sufficient Riok2 haploinsufficient mice (Riok2f/+Il22ra1+/+Vav1cre) (FIG. 1913). This improvement could be attributed to the increase in RIII and RIV erythroid precursors in the BM of Riok2f/+Il22ra1f/fVav1cre mice (FIG. 19C, FIG. 27C).


In accordance with the upregulation of p53 upon in vitro IL-22 stimulation in an in vitro erythropoiesis assay (FIG. 18G) and p53 upregulation in erythroid precursors upon Riok2 haploinsufficiency (FIG. 25F, G), a synergistic effect of Riok2 haploinsufficiency was observed in IL-22-responsive (IL-22RA1+) erythroid precursors in Riok2f/+Vav1cre mice (FIG. 19D, E). p53 target genes such as Gadd45a and Cdkn1a1 were also increased in IL-22RA1+ Riok2 haploinsufficient erythroid precursors compared to IL-22RA1+ Riok2 sufficient erythroid precursors (FIG. 19F). Given that rIL-22 induced apoptosis in erythroid precursors in vivo (FIG. 18D), it was sought to determine if Riok2 haploinsufficiency-mediated p53 upregulation played an independent role in apoptosis induction. In an IL-22-free in vitro erythropoiesis assay, p53 inhibition by pifithrin-α, p-nitro inhibited the apoptosis induced by Riok2 haploinsufficiency (FIG. 19G).


Erythroid cell-specific deletion of IL-22RA1 (using cre recombinase driven by the erythropoietin receptor (Epor) promoter) also increased numbers of PB RBCs and HCT (FIG. 7a) due to the increase in RIII and RIV erythroid precursors in the BM (FIG. 20B, FIG. 27D). Additionally, rIL-22 failed to exacerbate phenylhydrazine-induced anemia in Il22ra1f/fEporcre mice lacking the IL-22 receptor only on erythroid cells (FIG. 20C). These data reinforce our view that IL-22 signaling plays an important role in regulating erythroid development regardless of ribosomal haploinsufficiency. Thus, using three different approaches to neutralize IL-22 signaling, it is demonstrated herein that IL-22 plays a critical role in controlling RBC production by directly regulating early stages of erythropoiesis.


Example 15: IL-22 is Increased in del(5q) MDS Patients

Next, it was assessed whether IL-22. expression is increased in human disorders that display dyserythropoiesis due to ribosomal protein haploinsufficiencies. Given the localization of RIOK2 on human chromosome 5, it was focused on MDS with 5q deletion and compared it to MDS without 5q deletion and healthy controls. A significant increase was observed in IL-22 levels in the BM fluid (BMF) of del(5q) MDS patients compared to BMF from healthy controls and non-del(5q) MDS patients (FIG. 21A). Interestingly, a strong negative correlation between cellular RIOK2 mRNA expression and BMF IL-22 concentration was evident in the del(5q) MDS cohort (FIG. 21B) indicating that a decrease in RIOK2 expression is associated with increased IL-22 expression. In the MDS cohort, S100A8 concentrations were found to be higher than healthy controls regardless of del(5q) status (FIG. 21C). However, IL-22 positively correlated with S100A8 concentrations only in the del(5q) MDS group (FIG. 21D). Of note, S100A8 concentrations were higher in BMF from non-del(5q) MDS patients compared to MDS patients with del(5q) (FIG. 21C). These data suggest that the regulation of S100A8 expression may be IL-22-mediated in del(5q) MDS patients, but IL-22-independent in other subtypes of MDS. In a second cohort of MDS patients, the frequency of CD4+ T cells producing IL-22 (TH22 cells) among freshly isolated peripheral blood mononuclear cells (PBMCs) was significantly higher in MDS patients with 5q deletion compared to healthy controls (FIG. 21E—cumulative data of representative flow plots shown in FIG. 30A). As disclosed herein, the independent analysis of a large-scale microarray sequencing dataset of CD34+cells from normal, del(5q) MDS, and non-del(5q) MDS subjects showed that RIOK2 mRNA was significantly decreased in the del(5q) MDS cohort (78% (37/47)). Additionally, expression of known IL-22 target genes such as S100A10, 5100411, PTGS2, RAB7A, and LCN2 was specifically increased in the del(5q) MDS cohort compared to both the healthy control and non-del(5q) groups (FIG. 30B). Using differentially expressed proteins (adjusted p-value <0.01) from the Riok2 haploinsufficient proteomics dataset as a reference set, GSEA analyses of the CD34+ microarray dataset revealed significant enrichment scores (FIG. 31A, B) further suggesting that the mouse model of Riok2 haploinsufficiency faithfully recapitulates the molecular changes seen in patients with del(5q) MDS.


Example 16: High IL-22 in Anemic Chronic Kidney Disease Patients

Anemia is frequently observed in CKD patients and is associated with poor outcomes. Anemia of CKD is resistant to erythropoiesis-stimulating agents (ESAs) in 10-20% of patients, suggesting that pathogenic mechanisms other than erythropoietin deficiency are at play. A significant increase was found in IL-22 concentration in the plasma of CKD patients with secondary anemia compared to healthy controls and to CKD patients without anemia (FIG. 21F). Plasma IL-22 concentration negatively correlated with hemoglobin in CKD patients (FIG. 21G), suggesting a function for IL-22 in driving anemia in some patients with CKD.


As described herein, IL-22 signaling directly controls bone marrow erythroid differentiation and that its neutralization is a potential therapeutic approach for anemias and MDS. By exploring the function of a little-studied atypical kinase Riok2 in mammalian biology, the erythroid precursors were identified as a novel target for IL-22 action via the IL-22RA1. IL-22 were further identified as a disease biomarker for the del(5q) subtype of MDS and lastly, identify IL-22 signaling blockade as a potential therapeutic for stress-induced anemias irrespective of the genetic background. Interestingly, elevated levels of IL-22 were also detected in patients with anemia secondary to chronic kidney disease (CKD) suggesting that IL-22 signaling blockade may be therapeutic in reversing anemia in a much wider patient population.


Del(5q), either isolated or accompanied by additional cytogenetic abnormalities, is the most commonly detected chromosomal abnormality in MDS, reported in 10-15% of patients and enriched in therapy-related MDS. The severe anemia in MDS patients with isolated del(5q) has been linked to haploinsufficiency of ribosomal proteins such as RPS14 and RPS19. While much research has focused on the effect of such gene deletions or mutations in hematopoietic stem cells and lineage-committed progenitors, the immunobiology underlying this MDS subtype has remained largely unexplored, thus impeding the development of immune-targeted therapies. With the exception of the TNF-α inhibitor etanercept which proved to be ineffective, the only other therapy against immune cell-derived cytokines, is luspatercept, a recombinant fusion protein derived from human activin receptor type IIb which has just been approved for use in anemia in lower risk MDS patients. Here, two critical and independent functions of an understudied atypical kinase, RIOK2, that synergize to induce dyserythropoiesis and anemia were identified. One effect of Riok2 loss in erythroid precursors is an intrinsic block in erythroid differentiation owing to its indispensable role in the maturation of the pre-40S ribosomal complex, leading to increased apoptosis and cell cycle arrest. The second effect of Riok2 loss is the induction of the erythropoiesis-suppressive cytokine IL-22 in T cells which then directly acts on the IL-22RA1 on erythroid precursors (FIG. 31C). While IL-22RA1 is known to be widely expressed on epithelial cells and hepatocytes, its expression has also been recently reported on specialized cells such as retinal Müller glial cells and now described here, on erythroid precursors. Data disclosed herein reveals a novel molecular link between haploinsufficiency of a ribosomal protein and induction of erythropoiesis-suppressive cytokine IL-22. While IL-22 has been shown to modulate RBC production by controlling the expression of iron-chelating proteins such as hepcidin and haptoglobin, a novel role for IL-22 were uncovered in directly binding to the previously unknown IL-22R on erythroid precursors leading to their apoptosis. Diminished expression of ribosomal proteins has been shown to increase p53 levels. Data disclosed herein shows that Riok2 haploinsufficiency leads to p53 upregulation in T cells which drives the increase in IL-22 secretion. Additionally, it also shows that IL-22-responsive erythroid precursors express elevated p53 further suggesting a role for p53 downstream of IL-22 signaling in driving dyserythropoiesis. Using banked and fresh del(5q) MDS and CKD patient samples, the data disclosed herein shows that IL-22 is elevated in these human diseases.


The role of inflammatory cytokines in directly regulating various aspects of BM hematopoiesis in steady-state and diseased conditions is increasingly being recognized. IL-22 is known to play a pathogenic role in some autoimmune diseases. Interestingly, autoimmune diseases such as colitis, Behçet's disease, and arthritis are common in MDS patients, with features of autoimmunity observed in up to 10% of patients. It is intriguing to hypothesize that IL-22 may account both for the onset of MDS and autoimmunity in this subset of patients. Studies have reported that in patients with co-existence of MDS and autoimmunity, treatment for one can alleviate the symptoms of the other. Low-level exposure to benzene, a hydrocarbon, has been associated with an increased risk of MDS. Hydrocarbons are known ligands for aryl hydrocarbon receptor (AHR), the transcription factor that controls IL-22 production in T cells. Stemregenin 1, an AHR antagonist, was shown to promote the ex vivo expansion of human HSCs, with the highest fold expansion seen in the erythroid lineage. Overall, data disclosed herein suggests that inhibition of the AHR-IL-22 axis may be an attractive approach for treating red blood cell disorders that arise from dyserythropoiesis.


Further, data disclosed herein provides that neutralization of IL-22 signaling may be effective not only in the treatment of MDS and other stress-induced anemias, but also in the anemia of chronic diseases such as CKD, which are very much in need of new therapeutic approaches. With currently approved MDS therapies (lenalidomide and other hypomethylating agents, erythropoiesis-stimulating agents), the survival time of MDS patients after diagnosis is only 2.5-3 yrs. Patients also develop resistance to these therapies thus intensifying the need for additional therapeutic modalities. IL-22-based therapies could be used in conjunction with already existing therapeutics or after first-line therapies have failed due to acquisition of resistance.


Example 17: Anti-IL-22 Inhibits Recombinant IL-22-Induced IL-10 Production

IL-22 has been shown to induce IL-10 production from COLO-205 cells. To measure the effectiveness of anti-IL-22 in neutralizing IL-22 biological activity, COLO-205 cells were treated with recombinant mouse IL-22 (FIG. 32A) or recombinant human IL-22 (FIG. 32B), each in the presence of either isotype control antibody or anti-IL-22 antibody (Clone F0025, which blocks the interaction between IL-22 and IL-22 receptor or a heterodimeric complex of IL-22 receptor and IL-10 receptor beta subunit).


Briefly, in vitro IL-22 neutralization assays using anti-IL-22 antibodies were performed as follows. COLO-205 cells were purchased from American Type Culture Collection (ATCC) and cultured in complete medium (RPMI supplemented with 10% Fetal Bovine Serum (FBS)). 30,000 COLO-205 cells were cultured per well of a 96-well plate overnight in 100 uL complete medium. On the next day, cells were stimulated with human or mouse recombinant IL-22 (Cell Signaling Technology, Inc.) in the presence of isotype or IL-22 antibody for 24 hrs. Cell-free supernatant was collected at the end of the 24 hr period and subjected to IL-10 measurement using Human IL-10 Quantikine ELISA Kit (R&D Systems, Inc.).


Anti-IL-22 effectively neutralized the biological activity of both mouse and human IL-22, as seen by the observed decrease in IL-10 secretion from COLO-205 cells (FIGS. 32A and 32B).


Similarly, in vivo IL-22 neutralization assays using anti-IL-22 antibodies were performed. Neutralizing anti-IL-22 (Clone F0025, which blocks the interaction between IL-22 and IL-22 receptor) and isotype control IgG1 (purchased from BioXCell). 8-10 week old C57BL/6 mice were administered anti-IL-22 (700 μg/mouse/dose) or isotype intraperitoneally every 48 hours until the conclusion of the experiment. For induction of stress-induced anemia, mice were administered 25 mg/kg phenylhydrazine on days 0 and 1. Blood was collected from mice via the submandibular vein on days 4 and 7 post-phenylhydrazine administration for quantifying RBC numbers, hemoglobin, and hematocrit.


Treatment of C57BL/6J mice undergoing PhZ-induced anemia with the anti-IL-22 antibody significantly increased PB RBCs, Hb, and HCT compared to isotype antibody-treated mice (FIG. 33).









TABLE 6





Sequence of the anti-IL-22 antibody used in Example 17
















Heavy chain
QVQLVQSGAE VKKPGASVKV SCKASGYTFT NYYMHWVRQA PGQGLEWVGW



INPYTGSAFY



AQKFRGRVTM TRDTSISTAY MELSRLRSDD TAVYYCAREP EKFDSDDSDV



WGRGTLVTVS



SASTKGPSVF PLAPSSKSTS GGTAALGCLV KDYFPEPVTV SWNSGALTSG



VHTFPAVLQS



SGLYSLSSVV TVPSSSLGTQ TYICNVNHKP SNTKVDKKVE PKSCDKTHTC



PPCPAPELLG



GPSVFLFPPK PKDTLMISRT PEVTCVVVDV SHEDPEVKFN WYVDGVEVHN



AKTKPREEQY



NSTYRVVSVL TVLHQDWLNG KEYKCKVSNK ALPAPIEKTI SKAKGQPREP



QVYTLPPSRE



EMTKNQVSLT CLVKGFYPSD IAVEWESNGQ PENNYKTTPP VLDSDGSFFL



YSKLTVDKSR



WQQGNVFSCS VMHEALHNHY TQKSLSLSPG





Light chain
QAVLTQPPSV SGAPGQRVTI SCTGSSSNIG AGYGVHWYQQ LPGTAPKLLI



YGDSNRPSGV



PDRFSGSKSG TSASLAITGL QAEDEADYYC QSYDNSLSGY VFGGGTQLTV



LGQPKAAPSV



TLFPPSSEEL QANKATLVCL ISDFYPGAVT VAWKADSSPV KAGVETTTPS



KQSNNKYAAS



SYLSLTPEQW KSHRSYSCQV THEGSTVEKT VAPTECS









INCORPORATION BY REFERENCE

All publications, patents, and patent applications mentioned herein are hereby incorporated by reference in their entirety as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference. In case of conflict, the present application, including any definitions herein, will control.


EQUIVALENTS

Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims
  • 1-43. (canceled)
  • 44. A method of treating one or more red blood cell disorders in a human subject, the method comprising administering to the human subject an effective amount of an antibody means for inhibiting the binding of IL-22 to IL-22RA1 and a pharmaceutically acceptable carrier, wherein the red blood cell disorder is selected from a myelodysplastic syndrome, an anemia caused by a myelodysplastic syndrome, acute myelogenous leukemia that has progressed from a myelodysplastic syndrome, an anemia caused by insufficiency of serine/threonine-protein kinase RIOK2, an anemia caused by one or more mutations and/or deletions on human chromosome 5 or in an ortholog thereof, a macrocytic anemia, Diamond Blackfan anemia, Schwachman-Diamond syndrome, and an anemia associated with chronic kidney disease.
  • 45. The method of claim 44, wherein the antibody means for inhibiting the binding of IL-22 to IL-22RA1 is an anti-IL-22 antibody or an antigen binding fragment thereof or an anti-IL-22RA1 antibody or an antigen binding fragment thereof.
  • 46. The method of claim 44, wherein the antibody means for inhibiting the binding of IL-22 to IL-22RA1 is fezakinumab.
  • 47. The method of claim 44, wherein the one or more red blood cell disorders comprise one or more myelodysplastic syndromes.
  • 48. The method of claim 47, wherein the one or more myelodysplastic syndromes are mediated by a deletion in the long arm of human chromosome 5.
  • 49. The method of claim 44, further comprising conjointly administering to the human subject an effective amount of erythropoietin, epoetin alfa, epoetin beta, epoetin omega, epoetin zeta, IL-9, or darbepoetin alfa.
  • 50. A method of treating one or more red blood cell disorders in a human subject, the method comprising administering to the human subject an effective amount of fezakinumab, wherein the red blood cell disorder is selected from a myelodysplastic syndrome, an anemia caused by a myelodysplastic syndrome, acute myelogenous leukemia that has progressed from a myelodysplastic syndrome, an anemia caused by insufficiency of serine/threonine-protein kinase RIOK2, an anemia caused by one or more mutations and/or deletions on human chromosome 5 or in an ortholog thereof, a macrocytic anemia, Diamond Blackfan anemia, Schwachman-Diamond syndrome, and an anemia associated with chronic kidney disease.
  • 51. The method of claim 50, wherein the one or more red blood cell disorders comprise one or more myelodysplastic syndromes.
  • 52. The method of claim 51, wherein the one or more myelodysplastic syndromes are mediated by a deletion in the long arm of human chromosome 5.
  • 53. The method of claim 50, further comprising conjointly administering to the human subject an effective amount of erythropoietin, epoetin alfa, epoetin beta, epoetin omega, epoetin zeta, IL-9, or darbepoetin alfa.
  • 54. A method of treating one or more red blood cell disorders in a human subject, the method comprising administering to the human subject an effective amount of a non-antibody means for down-regulating IL-22 signaling and a pharmaceutically acceptable carrier, wherein the red blood cell disorder is selected from a myelodysplastic syndrome, an anemia caused by a myelodysplastic syndrome, acute myelogenous leukemia that has progressed from a myelodysplastic syndrome, an anemia caused by insufficiency of serine/threonine-protein kinase RIOK2, an anemia caused by one or more mutations and/or deletions on human chromosome 5 or in an ortholog thereof, a macrocytic anemia, Diamond Blackfan anemia, Schwachman-Diamond syndrome, and an anemia associated with chronic kidney disease.
  • 55. The method of claim 54, wherein the non-antibody means for down-regulating IL-22 signaling is an antagonist of aryl hydrocarbon receptor (AHR) selected from stemregenin 1, CH-223191, and 6,2′,4′-trimethoxyflavone.
  • 56. The method of claim 55, wherein the one or more red blood cell disorders comprise one or more myelodysplastic syndromes.
  • 57. The method of claim 54, wherein the one or more myelodysplastic syndromes are mediated by a deletion in the long arm of human chromosome 5.
  • 58. The method of claim 54, further comprising conjointly administering erythropoietin, epoetin alfa, epoetin beta, epoetin omega, epoetin zeta, IL-9, or darbepoetin alfa.
  • 59. A composition for treating one or more red blood cell disorders in a human subject in need thereof, comprising an antibody means for inhibiting the binding of IL-22 to IL-22RA1 and a pharmaceutically acceptable carrier, wherein said composition is suitable for use in the treatment of one or more red blood cell disorders selected from a myelodysplastic syndrome, an anemia caused by a myelodysplastic syndrome, acute myelogenous leukemia that has progressed from a myelodysplastic syndrome, an anemia caused by insufficiency of serine/threonine-protein kinase RIOK2, an anemia caused by one or more mutations and/or deletions on human chromosome 5 or in an ortholog thereof, a macrocytic anemia, Diamond Blackfan anemia, Schwachman-Diamond syndrome, and an anemia associated with chronic kidney disease.
  • 60. The composition for treating of claim 59, wherein the antibody means for inhibiting the binding of IL-22 to IL-22RA1 is an anti-IL-22 antibody or an antigen binding fragment thereof or an anti-IL-22RA1 antibody or an antigen binding fragment thereof.
  • 61. The composition for treating of claim 59, wherein the antibody means for inhibiting the binding of IL-22 to IL-22RA1 is fezakinumab.
  • 62. The composition for treating of claim 59, wherein the one or more red blood cell disorders comprise one or more myelodysplastic syndromes.
  • 63. The composition for treating of claim 62, wherein the one or more myelodysplastic syndromes are mediated by a deletion in the long arm of human chromosome 5.
  • 64. The composition for treating of claim 59, further comprising conjointly administering to the human subject an effective amount of erythropoietin, epoetin alfa, epoetin beta, epoetin omega, epoetin zeta, IL-9, or darbepoetin alfa.
  • 65. A method of promoting differentiation of an erythroid progenitor cell toward a mature red blood cell in a human subject, the method comprising administering to the human subject an effective amount of an antibody means for inhibiting the binding of IL-22 to IL-22RA1 and a pharmaceutically acceptable carrier.
  • 66. A method of promoting differentiation of an erythroid progenitor cell toward a mature red blood cell in a human subject, the method comprising administering to the human subject an effective amount of a non-antibody means for down-regulating IL-22 signaling and a pharmaceutically acceptable carrier.
  • 67. A method of promoting differentiation of an erythroid progenitor cell toward a mature red blood cell in a human subject, the method comprising administering to the human subject an effective amount of fezakinumab.
RELATED APPLICATIONS

This application claims the benefit of priority from U.S. Provisional Application Ser. No. 63/155,430, filed Mar. 2, 2021; the entire contents of said application are incorporated herein in their entirety by this reference.

PCT Information
Filing Document Filing Date Country Kind
PCT/US22/18538 3/2/2022 WO
Provisional Applications (1)
Number Date Country
63155430 Mar 2021 US