Recombinant Human Albumin-Granulocyte Macrophage Colony Stimulating Factor Fusion Protein With Long-Lasting Biological Effects

Yu; Zailin ;   et al.

Patent Application Summary

U.S. patent application number 13/327621 was filed with the patent office on 2012-05-24 for recombinant human albumin-granulocyte macrophage colony stimulating factor fusion protein with long-lasting biological effects. Invention is credited to Yan Fu, Zailin Yu.

Application Number20120128624 13/327621
Document ID /
Family ID34192929
Filed Date2012-05-24

United States Patent Application 20120128624
Kind Code A1
Yu; Zailin ;   et al. May 24, 2012

Recombinant Human Albumin-Granulocyte Macrophage Colony Stimulating Factor Fusion Protein With Long-Lasting Biological Effects

Abstract

Compositions, kits and methods are provided for promoting general health or for prevention or treatment of diseases by using novel recombinant fusion proteins of human serum albumin (HSA) and bioactive molecules. The bioactive molecules may be a protein or peptide having a biological function in vitro or in vivo, and preferably, having a therapeutic activity when administered to a human. By fusing the bioactive molecule to HSA, stability of the bioactive molecule in vivo can be improved and the therapeutic index increased due to reduced toxicity and longer-lasting therapeutic effects in vivo. In addition, manufacturing processes are provided for efficient, cost-effective production of these recombinant proteins in yeast.


Inventors: Yu; Zailin; (New York, NY) ; Fu; Yan; (New York, NY)
Family ID: 34192929
Appl. No.: 13/327621
Filed: December 15, 2011

Related U.S. Patent Documents

Application Number Filing Date Patent Number
12203116 Sep 2, 2008 8084021
13327621
11825686 Jul 8, 2007 7442371
12203116
10609346 Jun 26, 2003 7244833
11825686
60392948 Jul 1, 2002

Current U.S. Class: 424/85.1 ; 435/252.3; 435/254.2; 435/320.1; 435/325; 435/348; 435/360; 435/419; 530/363; 536/23.4
Current CPC Class: Y02A 50/30 20180101; A61P 7/00 20180101; C07K 14/505 20130101; C07K 2319/00 20130101; C07H 21/04 20130101; Y02A 50/463 20180101; C07K 14/765 20130101; C07K 14/52 20130101; C07K 14/54 20130101; C07K 14/79 20130101; C07K 14/475 20130101; A61K 47/646 20170801; A61P 37/04 20180101; C07K 14/5431 20130101; A61K 47/643 20170801; A61P 43/00 20180101; C07K 14/5403 20130101; A61K 38/38 20130101; C07K 14/47 20130101; A61K 38/38 20130101; A61K 2300/00 20130101
Class at Publication: 424/85.1 ; 530/363; 536/23.4; 435/320.1; 435/325; 435/348; 435/419; 435/252.3; 435/254.2; 435/360
International Class: A61K 38/38 20060101 A61K038/38; C12N 15/62 20060101 C12N015/62; C12N 15/63 20060101 C12N015/63; C12N 1/19 20060101 C12N001/19; C12N 15/85 20060101 C12N015/85; C12N 15/81 20060101 C12N015/81; C12N 5/10 20060101 C12N005/10; C12N 1/21 20060101 C12N001/21; C07K 19/00 20060101 C07K019/00; C12N 15/82 20060101 C12N015/82

Claims



1. A human serum albumin-granulocyte macrophage colony stimulating factor (HSA-GM-CSF) fusion protein comprising: (a) the amino acid sequence set forth in SEQ ID NO: 10; (b) the amino acid sequence encoded by the polynucleotide set forth in SEQ ID NO: 9; or (c) the amino acid sequence encoded by the polynucleotide contained in the yeast strain designated as YZ-HSA/hGM-CSF in ATCC.RTM. Deposit No: PTA-4607, said HSA-GM-CSF fusion protein having a plasma half-life longer than a plasma half-life of granulocyte macrophage colony stimulating factor.

2. The HSA-GM-CSF fusion protein of claim 1, wherein said HSA-GM-CSF fusion protein has a shelf-life four times longer than a shelf-life of the granulocyte macrophage colony stimulating factor.

3. The HSA-GM-CSF fusion protein of claim 1, wherein said HSA-GM-CSF fusion protein has said plasma half-life four times longer than said plasma half-life of the granulocyte macrophage colony stimulating factor.

4. A composition comprising said HSA-GM-CSF fusion protein of claim 1 and a pharmaceutically acceptable excipient.

5. The composition of claim 4 further comprising a human serum albumin-cell proliferation stimulatory factor (HSA-CPSF) fusion protein different from said HSA-GM-CSF fusion protein.

6. A polynucleotide comprising a polynucleotide sequence encoding said HSA-GM-CSF fusion protein of claim 1.

7. A vector comprising said polynucleotide of claim 6.

8. The vector of claim 7, wherein said vector is an expression vector for expressing said fusion protein in a host organism comprising mammal, fish, insect, plant, yeast, or bacterium.

9. The vector of claim 8, wherein said host organism is said yeast.

10. The vector of claim 9, wherein a strain of said yeast is selected from the group consisting of Saccharomyces, Candida, Pichia, Kluyveromyces, Torulaspora, and Schinosaccharomyces.

11. The vector of claim 9, wherein a strain of said yeast is Pichia pastoris.

12. The vector of claim 7, wherein said vector is a yeast transfer vector comprising pPICZ A, pPICZ B, or pPICZ C.

13. A host cell comprising said vector of claim 7.

14. The host cell of claim 13, wherein said host cell is selected from the group consisting of mammalian, fish, insect, plant, yeast, and bacterial cells.

15. The host cell of claim 14, wherein said mammalian cells are Chinese hamster ovarian (CHO) cells.
Description



CROSS REFERENCE TO RELATED APPLICATION

[0001] This application is a Continuation Application of patent application Ser. No. 12/203,116, filed Sep. 2, 2008, which is a Continuation Application of patent application Ser. No. 11/825,686, filed Jul. 8, 2007, now issued as U.S. Pat. No. 7,442,371 B2, which is a Continuation Application of patent application Ser. No. 10/609,346, filed Jun. 26, 2003, now issued as U.S. Pat. No. 7,244,833 B2, which claims the priority of Provisional Application Ser. No. 60/392,948, filed Jul. 1, 2002. All parent applications are hereby incorporated by reference in their entirety.

BACKGROUND OF THE INVENTION

[0002] 1. Field of the Invention

[0003] This invention relates to the manufacture and use of recombinant albumin fusion proteins and combinations thereof, and particularly to yeast expressed fusion proteins formed between human albumin and bioactive molecules such as therapeutic proteins and peptides, and more particularly to yeast expressed fusion proteins formed between human albumin and cell proliferation stimulatory factor (CPSF), such as, blood cell-stimulatory factors, erythropoietin (EPO), interleukins (ILs), stem cell factor (SCF), thrombopoietin (TPO), granulocyte colony stimulating factor (G-CSF), and granulocyte macrophage colony stimulating factor (GM-CSF).

[0004] 2. Description of Related Art

1. Albumin

[0005] Albumin is a soluble, monomeric protein which comprises about one-half of the blood serum protein. Albumin functions primarily as a carrier protein for steroids, fatty acids, and thyroid hormones and plays a role in stabilizing extracellular fluid volume. Mutations in this gene on chromosome 4 result in various anomalous proteins. Albumin is a globular un-glycosylated serum protein of molecular weight 65,000. The human albumin gene is 16,961 nucleotides long from the putative `cap` site to the first poly(A) addition site. It is split into 15 exons which are symmetrically placed within the 3 domains that are thought to have arisen by triplication of a single primordial domain. Albumin is synthesized in the liver as pre-pro-albumin which has an N-terminal peptide that is removed before the nascent protein is released from the rough endoplasmic reticulum. The product, proalbumin, is in turn cleaved in the Golgi vesicles to produce the secreted albumin. HSA has 35 cysteins; in blood this protein monomer has 17 disulfide linkage (Brown, J. R. "Albumin structure, Function, and Uses" Pergamon, N.Y., 1977). HSA is misfolded when produced intracellularly in yeast without its amino terminal secretion peptide sequence. This conclusion is based on its insolubility, loss of great than 90% of its antigenicity (as compared to human-derived HSA), and formation of large protein aggregates. At present albumin for clinical use is produced by extraction from human blood. The production of recombinant albumin in microorganisms has been disclosed in EP 330 451 and EP 361 991.

[0006] Albumin is a stable plasma transporter function provided by any albumin variant and in particular by human albumin. HSA is highly polymorphic and more than 30 different genetic alleles have been reported (Weikamp L, R, et al., Ann. Hum. Genet., 37 219-226, 1973). The albumin molecule, whose three-dimensional structure has been characterized by X-ray diffraction (Carter D. C. et al., Science 244, 1195-1198, 1989), was chosen to provide the stable transporter function because it is the most abundant plasma protein (40 g per liter in human), it has a high plasma half-life (14-20 days in human, Waldmann T. A., in "Albumin Structure, Function and Uses", Rosenoer V. M. et al (eds), Pergamon Press, Oxford, 255-275,1977), and above all it has the advantage of being devoid of enzymatic function, thus permitting its therapeutic utilization at high dose.

2. Interleukin-11 (IL-11)

[0007] Human IL-11 (Paul et al. (1990), Pro. Natl. Acad. Sci. 87:7512) has been identified in medium conditioned by primate bone marrow-derived stromal cells. IL-11 is expressed in cells of mesenchymal origin, such as stromal fibroblasts, fetal lung fibroblasts and trophoblasts.

[0008] IL-11, also called adipogenesis inhibitory factor (AGIF), acts on hematopoietic progenitor cells and stromal cells (Kawashima, I., et al., Progress in Growth Factor Research, 4,191 1992). The mature molecule is a non-glycosylation protein, 178aa in length, and has an apparent molecular weight 23 KD (as determined by SDS-PAGE). Human IL-11 gene consists of five exons and four introns and was mapped on chromosome 19 at band 19q13.3-q13.4. IL-11 exhibits a primary structure unrelated to that of known cytokines, but it often acts similar to other cytokines, notably IL-6. IL-11 is a pleiotropic growth factor effecting hematopoietic and non-hematopoietic cells, often in synergy with interleukins, colony stimulating factors, or stem cell factor. In hematopoietic cells, IL-11 can enhance megakaryopoiesis, stimulate early and intermediate myeloid progenitor cells, initiate proliferation of dormant hematopoietic progenitor cells, and stimulate T-cell-dependent development of antibody-secreting B-cells. In non-hematopoietic cells, IL-11 can inhibit adipogenesis, and mediates the hepatic acute phase response. IL-11 stimulated the production of erythrocytes was reported only by Quesniaux, VFJ., et al., Blood, 80, 1218 (1992).

[0009] Human IL-11 (e.g., NEURMEGA.RTM., manufactured by America Home Products Company) has been approved for clinical trials in the United States for directly stimulating the proliferation of hematopoietic stem cells and megakaryocyte progenitor cells and inducing megakaryocyte maturation, resulting in increased platelet production. It has been used for the prevention of severe thrombocytopenia and the reduction of the need for platelet transfusion following myelosuppressive chemotherapy.

3. Erythropoietin (EPO)

[0010] Erythropoietin (EPO) is a glycoprotein that is the principle regulator of red blood cells growth and differentiation (U.S. Pat. No. 5,547,933). Erythropoiesis, the production of red blood cells, occurs continuously throughout the human life span to offset cell destruction. Erythropoiesis is a very precisely controlled physiological mechanism enabling sufficient numbers of red blood cells to be available in the blood for proper tissue oxygenation, but not so many that the cells would impede circulation. The formation of red blood cells occurs in the bone marrow and is under the control of the hormone EPO.

[0011] EPO is an acidic glycoprotein (.about.30,400 Daltons) produced primarily by the kidney and is the principal factor regulating red blood cell production in mammals. Renal production of EPO is regulated by changes in oxygen availability. Under conditions of hypoxia, the level of EPO in the circulation increases and this leads to increased production of red blood cells. The over-expression of EPO may be associated with certain pathophysiological conditions. Polycythemia exists when there is an overproduction of red blood cells (RBCs). Primary polycythemias, such as Polycythemia vera, are caused by EPO-independent growth of erythrocytic progenitors from abnormal stem cells and low to normal levels of EPO are found in the serum of affected patients.

[0012] On the other hand, various types of secondary polycythemias are associated with the production of higher than normal levels of EPO. The overproduction of EPO may be an adaptive response associated with conditions that produce tissue hypoxia, such as living at high altitude, chronic obstructive pulmonary disease, cyanotic heart disease, sleep apnea, high-affinity hemoglobinopathy, smoking, or localized renal hypoxia. In other instances, excessive EPO levels are the result of production by neoplastic cells. Cases of increased EPO production and erythrocytosis have been recorded for patients with renal carcinomas, benign renal tumors, Wilms' tumors, hepatomas liver carcinomas, cerebellar hemangioblastomas, adrenal gland tumors, smooth muscle tumors, and leiomyomas.

[0013] Deficient EPO production is found in conjunction with certain forms of anemias. These include anemia of renal failure and end-stage renal disease, anemias of chronic disorders [chronic infections, autoimmune diseases, rheumatoid arthritis, AIDS, malignancies], anemia of prematurity, anemia of hypothyroidism, and anemia of malnutrition. Many of these conditions are associated with the generation of IL-1 and TNF-, factors that have been shown to be inhibitors of EPO activity. Other forms of anemias, on the other hand, are due to EPO-independent causes and affected individuals show elevated levels of EPO. These forms include aplastic anemias, iron deficiency anemias, thalassemias, megaloblastic anemias, pure red cell aplasias, and myelodysplastic syndromes.

[0014] The amount of erythropoietin in the circulation is increased under conditions of hypoxia when oxygen transport by blood cells in the circulation is reduced. Hypoxia may be caused by loss of large amounts of blood through hemorrhage, destruction of red blood cells by over-exposure to radiation, reduction in oxygen intake due to high altitudes or prolonged unconsciousness, or various forms of anemia. In response to tissues undergoing hypoxic stress, erythropoietin will increase red blood cell production by stimulating the conversion of primitive precursor cells in the bone marrow into proerythroblasts which subsequently mature, synthesize hemoglobin and are released into the circulation as red blood cells. When the number of red blood cells in circulation is greater than needed for normal tissue oxygen requirements, erythropoietin in circulation is decreased.

[0015] EPO may occur in three forms: alpha-, beta and asialo-EPO. The alpha- and beta-forms differ slightly in carbohydrate components, but have the same potency, biological activity and molecular weight. The asialo-form is an alpha- or beta-form with the terminal carbohydrate (sialic acid) removed. EPO is present in very low concentrations in plasma when the body is in a healthy state wherein tissues receive sufficient oxygenation from the existing number of erythrocytes. This normal low concentration is enough to stimulate replacement of red blood cells which are lost normally through aging. See generally for references, Testa, et al., Exp. Hematol., 8(Supp. 8), 144-152 (1980); Tong, et al., J. Biol. Chem., 256(24), 12666-12672 (1981); Goldwasser, J. Cell. Physiol., 110(Supp 1), 133-135 (1982); Finch, Blood, 60(6), 1241-1246 (1982); Sytowski, et al., Exp. Hematol., 8(Supp 8), 52-64 (1980): Naughton, Ann. Clin. Lab. Sci., 13(5), 432-438 (1983); Weiss, et al., Am. J. Vet. Res., 44(10), 1832-1835 (1983); Lappin, et al., Exp. Hematol., 11(7), 661-666 (1983); Baciu, et al., Ann. N.Y. Acad. Sci., 414, 66-72 (1983); Murphy, et al., Acta. Haematologica Japonica, 46(7), 1380-1396 (1983); Dessypris, et al., Brit. J. Haematol, 56, 295-306 (1984); and, Emmanouel, et al., Am. J. Physiol., 247 (1 Pt 2), F168-76 (1984).

[0016] Because EPO is essential in the process of red blood cell formation, the hormone has potential useful application in both the diagnosis and the treatment of blood disorders characterized by low or defective red blood cell production. See, generally, Pennathur-Das, et al., Blood, 63(5), 1168-71 (1984) and Haddy, Am. Jour. Ped. Hematol./Oncol., 4, 191-196, (1982) relating to erythropoietin in possible therapies for sickle cell disease, and Eschbach, et al. J. Clin. Invest., 74(2), pp. 434-441, (1984), describing a therapeutic regimen for uremic sheep based on in vivo response to erythropoietin-rich plasma infusions and proposing a dosage of 10 U EPO/kg per day for 15-40 days as corrective of anemia of the type associated with chronic renal failure. See also, Krane, Henry Ford Hosp. Med. J., 31(3), 177-181 (1983).

[0017] Prior attempts to obtain erythropoietin in good yield from plasma or urine have proven relatively unsuccessful. Complicated and sophisticated laboratory techniques are necessary and generally result in the collection of very small amounts of impure and unstable extracts containing erythropoietin. Genetically engineered EPO (U.S. Pat. No. 5,547,933) has been expressed in Chinese Hamster Ovary cell line (CHO). It has been approved for administration on clinical trials by FDA and now manufactures by Amgen Inc. under the name of EPOGEN.

[0018] It has been estimated that the availability of erythropoietin in quantity would allow for treatment each year of anemias of 1,600,000 persons in the United States alone. See, e.g., Morrison, "Bioprocessing in Space--an Overview", pp. 557-571 in The World Biotech Report 1984, Volume 2:USA, (Online Publications, New York, N.Y. 1984). Recent studies have provided a basis for projection of efficacy of erythropoietin therapy in a variety of disease states, disorders and states of hematologic irregularity: Vedovato, et al., Acta. Haematol, 71, 211-213 (1984) (beta-thalassemia); Vichinsky, et al., J. Pediatr., 105(1), 15-21 (1984) (cystic fibrosis); Cotes, et al., Brit. J. Obstet. Gyneacol., 90(4), 304-311 (1983) (pregnancy, menstrual disorders); Haga, et al., Acta. Pediatr. Scand., 72, 827-831 (1983) (early anemia of prematurity); Claus-Walker, et al., Arch. Phys. Med. Rehabil., 65, 370-374 (1984) (spinal cord injury); Dunn, et al., Eur. J. Appl. Physiol., 52, 178-182 (1984) (space flight); Miller, et al., Brit. J. Haematol., 52, 545-590 (1982) (acute blood loss); Udupa, et al., J. Lab. Clin. Med., 103(4), 574-580 and 581-588 (1984); and Lipschitz, et al., Blood, 63(3), 502-509 (1983) (aging); and Dainiak, et al., Cancer, 51(6), 1101-1106 (1983) and Schwartz, et al., Otolaryngol., 109, 269-272 (1983) (various neoplastic disease states accompanied by abnormal erythropoiesis).

4. Granulocyte Colony Stimulating Factor (G-CSF)

[0019] Granulocyte colony stimulating factor (G-CSF) is produced by monocytes and fibroblasts. It stimulating granulocyte colony formation, activates neutrophils, differentiates certain myeloid leukemic cell lines and is a potent activator of mature granulocytes (Metcalf D., cell, 43, 5, 1985; Groopman, J. E., Cell., 50, 5 1987);. Nature human G-CSF is a 19.6 KD glycoprotein having 177 amino acids (Souza, L. M et al., Science, 232, 62, 1986). Human and murine GCSF share approximately 75% amino acid sequence homology and have biology cross-reactivity (Morstyn, G., and Burgess, A., Cancer Res., 48, 5624, 1988). The biological activity of recombinant human G-CSF was measured in a cell proliferation assay using NFS-60 cells (Shurafuji, N et al., Exp. Hematol., 17, 116, 1989). Human G-CSF has been brought to the market under the name of NEUPOGEN.RTM. by Amgen, Inc.

5. Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF)

[0020] Granulocyte-Macrophage colony stimulating factor (GM-CSF) induces myeloid progenitor cells from bone marrow to from colonies contains macrophages and granulocytes in semisolid media. GM-CSF also acts upon mature macrophages, eosinophils and nutrophils to stimulate various functional activities (Mazur, E., and Cohen, J., Clin Pharmacol. Ther., 46, 250, 1989; Morstyn, T G., and Burgess. A., Cancer Res., 48, 5624, 1988). GM-CSF is an acidic glycoprotein {18-22 KD human (Wong, G., et al., Science, 228, 810, 1986), 23 KD mouse (Metcalf, D., Blood, 67, 257, 1986)} which binds to high affinity receptors on GM-CSF sensitive cells. Although human and mouse GMCSF share 54% amino acid sequence homology, their biological actions are species-specific (Metcalf, D., Blood, 67, 257, 1986). Other growth factors and CSFs modulate receptor binding or actions of GM-CSF (Nicola, N., Immunol. Today, 8, 134, 1987). The proliferative activity of human GMCSF is tested in culture using human TF-1 cells (Kitamura, T., et al., J. cell Physiol., 140, 323, 1989). Human GM-CSF has been brought to the market under the name of LEUKINE.RTM. by Immunex, Inc

6. Macrophage Colony Stimulating Factor (M-CSF)

[0021] Macrophage Colony Stimulating Factor (M-CSF) is produced by monocytes, fibroblasts and endothelial cells. It stimulates the formation of macrophage colonies (Metcalf, D., Blood, 67, 257, 1986), enhances antibody-dependent cell mediated cytotoxicity by monocytes and macrophages (Mufson, R. A. et al., Cellular Immunol., 119, 182, 1989), and inhibits bone resorption by osteoclasts (Hattersley, G., et al., J. Cell Physiol., 137, 199, 1988). M-CSF is glycoprotein and appears in a few different molecular weight forms due to variation in glycosylation. The peptide has 159 amino acids (Kawasaki, E. S., et al., Science, 230, 291, 1985).

7. Thrombopoietin (TPO)

[0022] Thrombopoietin (TPO), the ligand for the receptor encoded by the c-Mpl proto-oncogene, acts as a stimulator of the development of megakaryocyte precursors of platelets. Similar to erythropoietin, TPO leads to an increase in number of circulating platelets. TPO affects the entire thrombopoietic process, with stronger effects in the later stages. Other thrombopoietic cytokines include Stem cell factor (SCF), IL-3, IL-6 and IL-11.

[0023] TPO is an approximately 35 KD polypeptide of 335 amino acid. However, due to glycosylation the protein has an apparent molecular weight of 75 KD in SDS-PAGE. The precursor form of TPO consists of 356 amino acids. To generate the mature TPO (335aa), the precursor cleaves a 21 amino acids signal peptide. Human, mouse and dog TPO shows 69-75% amino acid homology. The biological activities of recombinant human TPO was measured in a cell proliferation assay using MO7e cells.

8. Interleukin-3 (IL-3) Interleukin-3 (IL-3) is one of a large and growing group of growth factors which support the proliferation and differentiation of hematopoietic progenitors as well as cells committed to various myeloid lineages in vitro and in vivo. Human IL-3 has 133 amino acids in mature protein and the glycosylation is not necessary for biological activity in vitro and in vivo. The homology between human and murine IL-3 is considerably less. The initial studies on the biology and biochemistry of IL-3 shows that among the well characterized hematopoietic growth factors, IL-3, is the only factor to be predominantly, if not exclusively, produced by activated T cell in normal cells in mice (Ihele and Weinstein, 1986) as well as in human (Yang and Clark 1998). The structure of IL-3, and the structure and location of its gene, are very much like those of a number of the hematopoietic growth factors and suggest that IL-3 is a member of an evolutionarily related family of growth factors. In preclinical and clinical trials, the most prominent and consistent effect of 11-3 in vivo is a significant increase in the absolute neutrophil count (ANC). In vitro IL-3, in combination with other cytokines such as stem-cell factor, IL-6, IL-1, IL-11, G-CSF. GM-CSF, erythropoietin (EPO), or Thrombopoietin (TPO) induces the proliferation of colony-forming units granulocyte-macrophage (CFU-GM), CFU-Eo, CFU-Baso, burst-forming units-erythroid (BFU-E), colony-forming units-megakaryocyte (CFU-MK) and colony-forming units-granulocyte/erythroid/macrophage/megakaryocyte (CFU-GEMM) in semisolid medium, and it stimulates the proliferation of purified CD34+ cells in suspension culture (Eder, et al., Stem Cell, 15:327-333, 1997).

SUMMARY OF THE INVENTION

[0024] The present invention provides compositions, kits and methods for promoting general health or for prevention or treatment of diseases by using novel recombinant fusion proteins of human serum albumin (HSA) and bioactive molecules. The bioactive molecules may be a protein or peptide having a biological function in vitro or in vivo, and preferably, having a therapeutic activity when administered to a human. It is believed that by fusing the bioactive molecule to HSA, stability of the bioactive molecule in vivo may be improved and the therapeutic index increased due to reduced toxicity.

[0025] In one aspect of the invention, recombinant fusion proteins of human serum albumin (HSA) and a cell proliferation stimulatory factor (CPSF) are provided 1) to stimulate proliferation of multiple cell types, especially cells of various developmental lineages in the blood, 2) allow a slower release of the HSA-CPSF fusion in the body to maximize the therapeutic effects of the CPSF, and/or 3) to reduce potential side effects or toxicity associated with administration of CPSF alone. In addition, manufacturing processes are provided for efficient, cost-effective production for producing these recombinant proteins in yeast.

[0026] In another aspect of the invention, an isolated polynucleotide is provided that encodes a fusion protein formed between HSA and a CPSF, i.e., an HSA/CPSF fusion. The CPSF may include any protein that can stimulate cell proliferation and/or production, preferably selected from the group consisting of colony-stimulatory factors such as colony-stimulating factors such as G-CSF, GM-CSF, eosinophil (EOS)-CSF (i.e. Interleukin-5), macrophage (M)-CSF (CSF-1), multi-CSF(i.e. IL-3) and erythropoietin (EPO); interleukins such as IL-1; IL-2; IL-4; IL-6; IL-7; IL-9; IL-10; IL-11; IL-12; IL-13 and IL-18; Steel factors (SLF: c-kit ligand; Stem-cell factor (SCF); mast cell growth factor); erythroid potentiating activity (EPA), Lactoferrin (LF), H-subunit ferritin (i.e., acidic isoferritin), prostaglandin (PG) E1 and E2, tumor necrosis factor (TNF)-.alpha., -.beta. (i.e. lymphotoxin), interferon (IFN)-.alpha.(1b, 2a and 2b), -.beta., -.omega. and -.gamma.; transforming growth factor (TGF)-.beta., activin, inhibin, leukemic inhibitory factor, oncostatin M; and chemokines such as macrophage inflammatory protein (MIP)-1-.alpha. (i.e. Stem-cell inhibitor); macrophage inflammatory protein (MIP)-1.beta.; macrophage inflammatory protein (MIP)-2-.alpha. (i.e., GRO-.beta.); GRO-.alpha.; MIP-2-.beta. (i.e., GRO-.gamma.); platelet factor-4; IL-8; macrophage chemotactic and activating factor and IP-10.

[0027] The CPSF may be linked directly to the N-terminus or the C-terminus of HSA to form an HSA-CPSF fusion. Optionally, there is a peptide linker (L) linking HSA and CPSF together to form the fusion protein: HSA-L-CPSF, or CPSF-L-HSA. The length of peptide is preferably between 2-100 aa, more preferably between 5-50 aa, and most preferably between 14-30 aa. The peptide linker may be a flexible linker that minimizes steric hindrance imposed by the bulky HSA protein on CPSF, such as a (G.sub.4S).sub.3-4 linker.

[0028] In one embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-interleukin-11 fusion protein (HSA-IL-11). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 1 (FIG. 1). Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 1. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 2.

[0029] In one embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-interleukin-3 fusion protein (HSA-IL-3). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 3. Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 3. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 4.

[0030] In another embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-erythropoietin fusion protein (HSA-EPO). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 5. Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 5. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 6. [HSA-EPO].

[0031] In yet another embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-granulocyte colony stimulating factor fusion protein (HSA-GCSF). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 7. Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 7. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 8.

[0032] In yet another embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-granulocyte macrophage colony stimulating factor fusion protein (HSA-GMCSF). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 9. Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 9. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 10.

[0033] In yet another embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-CPSF fusion protein (HSA-CPSF). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 11. Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 11. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 12. Optionally, the polynucleotide further comprises a nucleotide sequence at least 90% identical to SEQ ID NOs. 13, 15, 17, 19, or 21. Preferably, the polynucleotide further comprises a nucleotide sequence encoding an amino acid sequence comprising SEQ ID NOs. 14, 16, 18, 20, or 22.

[0034] According to the embodiment, the CPSF may be selected from the group consisting of G-CSF, GM-CSF, (EOS)-CSF, CSF-1, EPO, IL-1; IL-2, IL-3, IL-4; IL-6; IL-7; IL-8, IL-9; IL-10; IL-11; IL-12; IL-13, IL-18, SLF, SCF, mast cell growth factor, EPA, Lactoferrin, H-subunit ferritin, prostaglandin (PG) E1 and E2, TNF-.alpha., TNF-.beta., IFN-.alpha., IFN-.beta., IFN-.alpha.), IFN -.gamma.; TGF-.beta., activin, inhibin, leukemic inhibitory factor, oncostatin M, MIP-1-.alpha., MIP-1.beta.; MIP-2-.alpha., GRO-.alpha.; MIP-2-.beta., platelet factor-4, macrophage chemotactic and activating factor and IP-10.

[0035] The fusion protein may be a secretory protein, which binds to a specific antibody of human albumin, and optionally, binds to a specific antibody of the CPSF in this fusion protein.

[0036] In another aspect of the invention, a recombinant vector is provided that comprises the sequence of the polynucleotide described above. The recombinant vectors can be an expression vector for expressing the fusion protein encoded by the polynucleotide, HSA-CPSF, HSA-L-CPSF, or CPSF-L-HSA in a host organism. The host organism includes, but is not limited to, mammalian (e.g., human, monkey, mouse, rabbit, etc.), fish, insect, plant, yeast, and bacterium.

[0037] In a preferred embodiment, the host organism belongs to a genus of yeast such as Saccharomyces (e.g., S. cerevisiae), Pichia, Kluyveromyces, Torulaspora, and Schinosaccharomyces. In a more preferred embodiment, the host organism is Pichia pastoris. In a particular embodiment, the recombinant vector is a pPICZ A, pPICZ B, or pPICZ C.

[0038] Depending upon the host organism employed in a recombinant process for producing the fusion proteins, the recombinant fusion proteins of the present invention may be glycosylated or may be non-glycosylated. Preferably, when expressed in a host organism, the fusion protein of HSA and CPSF is glycosylated to substantially the same extent as that when expressed in mammalian cells such as Chinese hamster ovarian (CHO) cells, or as that when expressed in Pichia pastoris.

[0039] In yet another aspect of the invention, a recombinant cell is provided that is capable of expressing the sequence of the polynucleotide described above. The recombinant cell may constitutively or be induced in the presence or absence of an agent to express the fusion protein encoded by the nucleic acid, HSA-CPSF, HSA-L-CPSF, or CPSF-L-HSA in a host organism. The type of the recombinant cell includes, but is not limited to, mammalian (e.g., human, monkey, mouse, rabbit, etc.), fish, insect, plant, yeast, and bacterial cells.

[0040] When stored at ambient temperature or a lower temperature, the fusion protein of HSA and CPSF may have a shelf-life 2 times longer, preferably 4 times longer, more preferably 6 times, and most preferably 10 times, longer than that of the CPSF alone (i.e., not fused to HSA) stored under the same condition.

[0041] The fusion protein of the present invention may be administered to a mammal, preferably a human, via a variety of routes, including but not limited to, orally, parenterally, intraperitoneally, intravenously, intraarterially, topically, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally. The antibody may also be delivered to the host locally (e.g., via stents or cathetors) and/or in a timed-release manner. In a particular embodiment, the fusion protein is delivered parenterally via injection.

[0042] When delivered in vivo to an animal, the fusion protein of HSA and CPSF may have a plasma half-life 2 times longer, preferably 4 times longer, more preferably 6 times, and most preferably 10 times, longer than that of the CPSF alone (i.e., not fused to HSA).

[0043] The HSA/CPSF fusion proteins of the present invention may also be administered in combination with a natural or recombinant human albumin, preferably a recombinant human serum albumin at a therapeutically effective dose and ratio.

[0044] In yet another aspect of the invention, combinations of different HSA/CPSF fusion proteins of are provided. The specific combinations of these fusion proteins may be administered to a patient to stimulate proliferation of multiple types of cells in the body or to synergistically enhance proliferation of a particular cell type.

[0045] In one embodiment, HSA/IL-11 fusion may be combined with HSA/EPO fusion and the resulting combination may be administered to a patient with a hematological disorder to simultaneously stimulate proliferation of erythrocytes and platelets.

[0046] In another embodiment, HSA/IL-3 fusion may be combined with HSA/EPO fusion and the resulting combination may be administered to a patient with a hematological disorder to enhance EPO-induced production of erythrocytes.

[0047] In yet another embodiment, HSA/IL-3 fusion may be combined with HSA/GCSF fusion and the resulting combination may be administered to a patient with a hematological disorder to increase the production of erythrocytes and neutrophils, as well as eosinophils.

[0048] Alternatively, an HSA/CPSF fusion may be co-administered with a different HSA/CPSF fusion simultaneously or sequentially to a patient in need thereof. This combination therapy may confer synergistic therapeutic effects on the patients.

[0049] In yet another aspect of the invention, a method is provided for treating a patient with a CPSF in need thereof. In one embodiment, the method comprises: administering a pharmaceutical formulation comprising a fusion protein of HSA and CPSF to the patient in a therapeutically effective amount. The pharmaceutical formulation may contain any pharmaceutically acceptable excipient and agents that stabilizes the HSA/CPSF fusion protein. The pharmaceutical formulation may further comprise natural or recombinant human serum albumin and/or another, different HSA/CPSF fusion protein.

[0050] The pharmaceutical formulation may contain any pharmaceutically acceptable excipient and agents that stabilizes the HSA/CPSF fusion protein. The pharmaceutical formulation may further comprise natural or recombinant human serum albumin and/or another, different HSA/CPSF fusion protein.

[0051] In another embodiment, the method comprises: administering a first pharmaceutical formulation comprising a first fusion protein of HSA and a first CPSF to the patient in a therapeutically effective amount; and administering to the patient a second pharmaceutical formulation comprising a second fusion protein of HSA and a second CPSF to the patient in a therapeutically effective amount. Such a combination therapy may confer synergistic therapeutic effects on the patient.

[0052] For example, HSA-IL-11 fusion protein may be administered to the patient first, followed by administration of HSA-EPO, HSA-GCSF and/or HSA-GMCSF at therapeutically effective doses and ratios to stimulate proliferation of different types of blood cells.

[0053] In yet another aspect of the invention, a kit is provided, comprising: a first fusion protein of HSA and a first CPSF, and a second fusion protein of HSA and a second CPSF. The first and second CPSFs may be the same or different. For example, the first CPSF is IL-11 and the second CPSF is EPO; the first CPSF is IL-3 and the second CPSF is EPO; or the first CPSF is IL-11 and the second CPSF is GCSF.

BRIEF DESCRIPTION OF THE FIGURES

[0054] FIG. 1 shows nucleotide and amino acid sequences of embodiments of HSA-CPSF fusion proteins, HSA, and examples of individual CPSFs.

[0055] FIG. 2 illustrates a plasmid DNA vector contains the HSA sequence and as a backbone vector for making HSA-CPSF fusion proteins.

[0056] FIG. 3 shows a Western blot detected using mouse monoclonal anti-human serum albumin (Sigma Cat# A6684). Each lane was load with equivalent of 100 ng of proteins. A), HSA (65 Kd) from blood plasma; B), HSA (65 Kd) from yeast; C), HSA/hG-CSF (84.2 Kd); D), HSA/hEPO (83.5 Kd); E), HSA/hIL-11 (84.5 Kd).

[0057] FIG. 4 shows a Western blot detected using goat polyclonal anti-hIL-11 antibody (R&D Systems, Cat# Ab-218-NA), each lane contains 100 ng proteins. A), human IL-11 expressed by E. coli; B), HSA/hIL-11 fusion protein expressed by yeast.

[0058] FIG. 5 shows a Western blot detected using monoclonal anti-hGMCSF antibody (R&D Systems), each lane contains 20 ng proteins. A), Human GMCSF expressed by E. coli; B), HSA/hGMCSF fusion protein expressed in yeast.

[0059] FIG. 6 is a bioassay for human IL-11 and HSA/hIL-11 fusion protein in stimulation of T1165 cell proliferation. A), with hIL-6 antibody in medium; B), without hIL-6 antibody in medium.

[0060] FIG. 7 is an ELISA bioassay for EPO and HSA fusion protein, HSA/hEPO.

[0061] FIG. 8 shows the results of a stability test of rHSA/hIL-11 fusion protein under different temperature and its cell proliferation activity. A), 37.degree. C.; B), 50.degree. C.

[0062] FIG. 9 shows the results of an in vivo animal test of synergistic effects of a combination of different HSA-CPSFs in stimulating multiple blood cell proliferation, as compared with those when single HSA-CPSFs or CPSFs were administered.

[0063] FIG. 10 shows a SDS-PAGE of purified HSA-CPSFs. Each lane was loaded with about 20 .mu.g of protein. A), HSA (65 Kd) from blood plasma; B), HSA (65 Kd) from yeast; C), HSA/hG-CSF (84.2 Kd); D), HSA/hEPO (83.5 Kd); E), HSA/hIL-11(84.5 Kd).

DETAILED DESCRIPTION OF THE INVENTION

[0064] The present invention provides innovative compositions, kits and methods for modulating cell proliferation in vivo and more particularly for promoting cell growth for enhancing general health or for treating diseases or undesirable conditions.

[0065] In general, recombinant fusion proteins of human serum albumin (HSA) and a cell proliferation stimulatory factor (CPSF) are provided in order to circumvent problems associated with conventional therapy using the CPSF protein itself. Generally, compared with the CPSF protein alone, the inventive fusion proteins of the present invention possess the following advantages: 1) being capable of stimulating proliferation of multiple cell types, especially cells of various developmental lineages in the blood; 2) allowing a slower release of the HSA-CPSF fusion in the body to maximize the therapeutic effects of the CPSF, and/or 3) reducing potential side effects or toxicity associated with administration of CPSF alone.

[0066] The present invention also provides a method for treating a patient with a CPSF in need thereof. In one embodiment, the method comprises: administering a pharmaceutical formulation comprising a fusion protein of HSA and CPSF to the patient in a therapeutically effective amount. The pharmaceutical formulation may contain any pharmaceutically acceptable excipient and agents that stabilizes the HSA/CPSF fusion protein. The pharmaceutical formulation may further comprise natural or recombinant human serum albumin and/or another, different HSA/CPSF fusion protein. The pharmaceutical formulation may contain any pharmaceutically acceptable excipient and agents that stabilizes the HSA/CPSF fusion protein. The pharmaceutical formulation may further comprise natural or recombinant human serum albumin and/or another, different HSA/CPSF fusion protein.

[0067] In addition, the present invention also provides manufacturing processes efficient, cost-effective production for producing these recombinant fusion proteins in yeast. In particular, fusion proteins of HSA with each of human IL-11, EPO, G-GCSF and GM-CSF have been expressed in a yeast strain of Pichia pastoria and shown to have superior stability in storage and in plasma, and when combined, to possess synergistic effects on the production and growth of multiple types of blood cells.

1. HSA/CPSF Fusion Proteins

[0068] In one aspect of the invention, isolated polynucleotides are provided that encode fusion proteins formed between HSA and a CPSF, i.e., HSA/CPSF fusion. It should be noted other types of albumin can also be employed to produce a fusion protein with a CPSF of the present invention.

[0069] The CPSF may include any protein that can stimulate cell proliferation and/or production. In a particular embodiment, the CPSF is a hematopoietically active cytokine Examples of such a CPSF are described in Aggarwal and Puri (1995) "Role of cytokines in immuno-regulation", in "Human Cytokines Their role in disease and therapy", edited by Aggarwal and Puri, Blackwell Science Inc., Cambridge, Mass., USA, pp 28, which is incorporated herein by reference in its entirety.

[0070] Specific examples of the CPSF include, but are not limited to, colony-stimulating factors such as G-CSF, GM-CSF, eosinophil (EOS)-CSF (i.e. Interleukin-5), macrophage (M)-CSF (CSF-1), multi-CSF(i.e. IL-3) and erythropoietin (EPO); interleukins such as IL-1; IL-2; IL-4; IL-6; IL-7; IL-9; IL-10; IL-11; IL-12; IL-13 and IL-18; Steel factors (SLF: c-kit ligand; Stem-cell factor (SCF); mast cell growth factor); erythroid potentiating activity (EPA), Lactoferrin (LF), H-subunit ferritin (i.e., acidic isoferritin), prostaglandin (PG) E1 and E2, tumor necrosis factor (TNF)-.alpha., -.beta. (i.e. lymphotoxin), interferon (IFN)-.alpha.(1b, 2a, and 2b), -.beta., -.omega. and -.gamma., transforming growth factor (TGF)-.beta., activin, inhibin, leukemic inhibitory factor, oncostatin M; and chemokines such as macrophage inflammatory protein (MIP)-1-.alpha. (i.e. Stem-cell inhibitor); macrophage inflammatory protein (MIP)-1.beta.; macrophage inflammatory protein (MIP)-2-.alpha. (i.e., GRO-.beta.); GRO-.alpha.; MIP-2-.beta. (i.e., GRO-.gamma.); platelet factor-4; IL-8; macrophage chemotactic and activating factor and IP-10.

[0071] Four distinct colony-stimulating factors (CSFs) that promote survival proliferation and differentiation of bone marrow precursor cells have been well characterized: GM-CSF, G-CSF, M-CSF and Interleukin-3 (IL-3, Multi-CSF). Both GM-CSF and IL-3 are multipotent growth factors, stimulating proliferation of progenitor cells from more than one hematopoietic lineage. In contrast, G-CSF and M-CSF are lineage-restricted hematopoietic growth factors, stimulating the final mitotic divisions and the terminal cellular maturation of partially differentiated hematopoietic progenitors. Erythropoietin is a hematopoietic growth factor that stimulated red blood cell proliferation. Growth factors, such as Stem Cell Factor, all the interleukins, and all the interferon, all are CPSF.

[0072] The CPSF may be linked directly to the N-terminus or the C-terminus of HSA to form an HSA-CPSF fusion. Optionally, there is a peptide linker (L) linking HSA and CPSF together to form the fusion protein: HSA-L-CPSF, or CPSF-L-HSA. The length of peptide is preferably between 2-100 aa, more preferably between 5-50 aa, and most preferably between 14-30 aa. The peptide linker may be a flexible linker that minimizes steric hindrance imposed by the bulk HA protein on CPSF, such as a (G.sub.4S).sub.3-4 linker. The linker addition may be good for CPSF binds to its receptor.

[0073] The fusion protein may be a secretory protein, which binds to a specific antibody of human albumin, and optionally, binds to a specific antibody of the CPSF in this fusion protein.

[0074] In one embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-interleukin-11 fusion protein (HSA-IL-11). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 1. Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 1. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 2. [HSA-IL-11].

[0075] In one embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-interleukin-3 fusion protein (HSA-IL-3). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 3. Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 3. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 4. [HSA-IL-3].

[0076] In another embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-erythropoietin fusion protein (HSA-EPO). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 5. Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 5. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 6 [HSA-EPO].

[0077] In yet another embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-granulocyte colony stimulating factor fusion protein (HSA-GCSF). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 7. Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 7. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 8 [HSA-GCSF].

[0078] In yet another embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-granulocyte macrophage colony stimulating factor fusion protein (HSA-GMCSF). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 9. Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 9. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 10 [HSA-GMCSF].

[0079] In yet another embodiment, an isolated polynucleotide is provided that encodes a human serum albumin-CPSF fusion protein (HSA-CPSF). The polynucleotide comprises a nucleotide sequence at least 90% identical to SEQ ID NO. 11. Preferably, the polynucleotide comprises a nucleotide sequence at least 95% identical to SEQ ID NO. 11. Preferably, the polynucleotide encodes an amino acid sequence comprising SEQ ID NO. 12. Optionally, the polynucleotide further comprises a nucleotide sequence at least 90% identical to SEQ ID NOs. 13, 15, 17, 19, or 21. Preferably, the polynucleotide further comprises a nucleotide sequence encoding an amino acid sequence comprising SEQ ID NOs. 14, 16, 18, 20, or 22.

[0080] According to the embodiment, the CPSF may be selected from the group consisting of G-CSF, GM-CSF, (EOS)-CSF, CSF-1, EPO, IL-1; IL-2, IL-3, IL-4; IL-6; IL-7; IL-8, IL-9; IL-10; IL-11; IL-12; IL-13, IL-18, SLF, SCF, mast cell growth factor, EPA, Lactoferrin, H-subunit ferritin, prostaglandin (PG) E1 and E2, TNF-.alpha., TNF-.beta., IFN-.alpha., IFN-.beta., IFN-.omega., IFN -.gamma.; TGF-.beta., activin, inhibin, leukemic inhibitory factor, oncostatin M, MIP-1-.alpha., MIP-1.beta.; MIP-2-.alpha., GRO-.alpha.; MIP-2-.beta., platelet factor-4, macrophage chemotactic and activating factor and IP-10.

[0081] The above-described polynucleotide with a sequence having a certain degree of sequence identity, for example at least 95% "identical" to a reference nucleotide sequence encoding a HSA/CPSF fusion protein, is intended that the polynucleotide sequence is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence encoding the HSA/CPSF fusion protein. In other words, to obtain a polynucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence, up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence. These mutations of the reference sequence may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence.

[0082] As a practical matter, whether any particular nucleic acid molecule is at least 90%, 95%, 96%, 97%, 98% or 99% identical to, for instance, the polynucleotide sequence encoding a HSA/CPSF fusion protein can be determined conventionally using known computer programs such as the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, Wis. 53711). Bestfit uses the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482-489 (1981), to find the best segment of homology between two sequences. When using Bestfit or any other sequence alignment program to determine whether a particular sequence is, for instance, 95% identical to a reference sequence according to the present invention, the parameters are set, of course, such that the percentage of identity is calculated over the full length of the reference nucleotide sequence and that gaps in homology of up to 5% of the total number of nucleotides in the reference sequence are allowed.

[0083] When stored at ambient temperature or a lower temperature, the fusion protein of HSA and CPSF may have a shelf-life 2 times longer, preferably 4 times longer, more preferably 6 times, and most preferably 10 times, longer than that of the CPSF alone stored under the same condition.

[0084] The present invention involves the utilization of albumin as a vehicle to carry a therapeutic protein such as a CPSF that can be used in the treatment of certain diseases such as cancers, or people in need of an increased blood cell proliferation in order to increase the blood cell numbers. The fusion protein of the present invention may be administered to a mammal, preferably a human, via a variety of routes, including but not limited to, orally, parenterally, intraperitoneally, intravenously, intraarterially, topically, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally. The HSA-CPSF may also be delivered to the host locally (e.g., via stents or cathetors) and/or in a timed-release manner. In a particular embodiment, the fusion protein is delivered parenterally via injection.

[0085] When delivered in vivo to an animal, the fusion protein of HSA and CPSF may have a plasma half-life 2 times longer, preferably 4 times longer, more preferably 6 times, and most preferably 10 times, longer than that of the CPSF alone.

[0086] The HSA/CPSF fusion proteins of the present invention may also be administered in combination with a natural or recombinant human albumin, preferably a recombinant human serum albumin at a therapeutically effective dose and ratio.

[0087] It is believed that after fusion with albumin, the CPSF protein can have a longer shelf-life and plasma half-life, which allows cost-effective storage and transportation, as well as reduces amount and/or frequency of drug administration.

[0088] It is noted that the same strategy may be used to develop stable fusion proteins with anticancer functions. For example, tumor suppressors, such as p53, pRb, pRb2/p130, and KLF6 may be fused with HSA. Optionally, insulin and insulin-like growth factor may be fused with HSA for therapeutic treatment of diabetes. The gene of antibody from human or other sources with therapeutic function also can be fused with albumin for easy delivery and therapeutic purpose. Antigens against which vaccines are developed can also be fused to albumin for in vivo delivery and increasing the antigenicity of the antigens. Antibody can be fused to albumin and then delivered into blood system to induce an immune response to the invaded foreign antigens such as bacteria, parasites and viruses. In addition anti-aging proteins may be fused with albumin and delivered to humans.

2. Expression of Fusion Proteins in Host Organisms

[0089] The polynucleotides encoding the inventive HSA/CPSF fusion proteins can be cloned by recombinant techniques into vectors which are introduced to host cells where the fusion proteins can be expressed.

[0090] Generally, host cells are genetically engineered (transduced or transformed or transfected) with the vectors of this invention which may be, for example, a cloning vector or an expression vector. The vector may be, for example, in the form of a plasmid, a viral particle, a phage, etc. The engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the polynucleotides encoding HSA/CPSF fusion proteins. The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.

[0091] According to the invention, a recombinant vector is provided that comprises the polynucleotide sequence encoding an HSA/CPSF fusion protein. The recombinant vectors can be an expression vector for expressing the fusion protein encoded by the nucleic acid, HSA-CPSF, HSA-L-CPSF, or CPSF-L-HSA in a host organism. The host organism includes, but is not limited to, mammalian (e.g., human, monkey, mouse, rabbit, etc.), fish, insect, plant, yeast, and bacterium.

[0092] Expression of the polynucleotide encoding an HSA/CPSF fusion protein is under the control of a suitable promoter. Suitable promoters which may be employed include, but are not limited to, adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV) promoter; inducible promoters, such as the MMT promoter, a tetracycline or tetracycline-like inducible promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAI promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral LTRs (including the modified retroviral LTRs hereinabove described); the .beta.-actin promoter; and human growth hormone promoters. The promoter also may be the native promoter which controls the polynucleotide encoding an HSA/CPSF fusion protein.

[0093] Also according to the invention, a recombinant cell is provided that is capable of expressing comprises the polynucleotide sequence encoding an HSA/CPSF fusion protein. The recombinant cell may constitutively or be induced in the presence or absence of an agent to express the fusion protein encoded by the nucleic acid, HSA-CPSF, HSA-L-CPSF, or CPSF-L-HSA in a host organism. The type of the recombinant cell includes, but is not limited to, mammalian (e.g., human, monkey, mouse, rabbit, etc.), fish, insect, plant, yeast, and bacterial cell.

[0094] In a preferred embodiment, the host organism belongs to a genus of yeast such as Saccharomyces (e.g., S. cerevisiae), Pichia, Kluyveromyces, Torulaspora, and Schinosaccharomyces. In a more preferred embodiment, the host organism is Pichia pastoris. In a particular embodiment, the recombinant vector is a pPICZ A, pPICZ B, or pPICZ C.

[0095] Depending upon the host employed in a recombinant process for producing the fusion proteins, the fusion proteins of the present invention may be glycosylated or may be non-glycosylated. Preferably, when expressed in a host organism, the fusion protein of HSA and CPSF may be glycosylated to substantially the same extent as that when expressed in mammalian cells such as Chinese hamster ovarian (CHO) cells, or as that when expressed in Pichia pastoris.

[0096] As indicated above, the albumin fusion proteins of the present invention are substantially preferably proteomic and can therefore be generated by the techniques of genetic engineering. The preferred way to obtain these fusion proteins is by the culture of cells transformed, transfected, or infected by vectors expressing the fusion protein. In particular, expression vectors capable of transforming yeasts, especially of the genus Pichia, for the secretion of proteins will be used.

[0097] It is particularly advantageous to express the HSA/CPSF fusion protein in yeast. Such an expression system allows for production of high quantities of the fusion protein in a mature form, which is secreted into the culture medium, thus facilitating purification.

[0098] The development of yeast genetic engineering has been made possible the expression of heterologous genes and the secretion of their protein products from yeast. The advantages of protein secretion (export) of yeast are including but not limited to, high expression level, soluble protein, corrected folding, easy to scale-up and easy for purification.

[0099] The HSA/CPSF fusion protein can be secreted into the media of yeast via an albumin natural secretion signal. The polypeptide sequence of HSA fusion protein can be preceded by a signal sequence which serves to direct the proteins into the secretory pathway. In a preferred embodiment the prepro-sequence of human albumin is used to secret the fusion protein out of yeast cells into the culture medium. Other secret signal peptides, such as the native Saccharomyces cerevisiae .alpha.-factor secretion signal, can also be used to make fusion protein of the present invention.

[0100] Yeast-expressed HSA is soluble and appears to have the same disulfide linkages as the human-blood derived counterpart. If used as a pharmaceutical, which may be potentially used in gram amounts in humans, a recombinant HSA will require a close identity with the natural HSA product. Secreting the HSA/CPSF fusion protein into the growth media of yeast, which is via prepro-amino-terminal processing (no initiator methionine residue), also circumvents the problems associated with preparing yeast extracts, such as the resistance of yeast cells to lysis. In addition, the purity of the product can be increased obtained by placing the product in an environment in which 0.5-1.0% of total yeast proteins is included and the lacks toxic proteins that would contaminate the product.

[0101] In a preferred embodiment, a particular species of yeast Pichia pastoris is used the system for expressing HSA/CPSF fusions of the present invention. Pichia pastoris was developed into an expression system by scientists at Salk Institute Biotechnology/Industry Association (SIBA) and Phillips Petroleum for high-level expression of recombinant proteins. The techniques related to Pichia are taught in, for example, U.S. Pat. Nos. 4,683,293, 4,808,537, and 4,857,467.

[0102] There are some advantages of using yeast Pichia pastoris to expression of HSA and HSA fusion proteins than using other systems. Pichia pastoris is a species of yeast genus, Pichia. Pichia has many of advantages of higher eukaryotic expression systems such as protein processing, protein folding, and posttranslational modification, while being as easy to manipulate as E. coli or Saccharomyces cerevisiae. It is faster, easier, and less expensive to use than other eukaryotic expression systems such as baculovirus or mammalian tissue culture, and generally gives higher expression levels. Pichia has an additional advantage which gives 10 to 100-fold higher heterologous protein expression levels. Those features make Pichia very useful as a protein expression system.

[0103] Owing to the similarity between Pichia and Saccharomyces, many techniques developed for Saccharomyces may be applied to Pichia. These include: transformation by complementation, gene disruption, gene replacement. In addition, the genetic nomenclature used for Sac has been applied to Pichia. For example, histidinol dehydrogenase is encoded by the HIS4 gene in both Sac and Pichia. The Pichia as a methylotrophic yeast is capable of metabolizing methanol as its sole carbon source. The first step in the metabolism of methanol is oxidation of methanol to formaldehyde using molecular oxygen by the enzyme alcohol oxidase. In addition to formaldehyde, this reaction generates hydrogen peroxide. To avoid hydrogen peroxide toxicity, methanol metabolism takes place within a specialized cell organelle, called the peroxisome, which sequesters toxic by-products away from the rest of the cell. Alcohol oxidase has a poor affinity for O.sub.2, and Pichia compensates by generating large amounts of this enzyme. The promoter regulating the production of alcohol oxidase is the one used to drive heterologous (HSA or HSA fused) protein expression in Pichia.

[0104] Compared with Saccharomyces cerevisiae, Pichia may have an advantage in the glycosylation of secreted proteins because it generally does not hyper-glycosylate. Both Saccharomyces and Pichia have a majority of N-linked glycosylation of the high-mannose type; however, the length of the oligosaccharide chains added post-translation ally to proteins in Pichia (average 8-14 mannose residues per side chain) is much shorter than those in Saccharomyces (50-150 mannose residues). Very little O-linked glycosylation has been observed in Pichia. In addition, Saccharomyces core oligosaccharide have terminal .alpha.-1,3 glycan linkages whereas Pichia does not. It is believed that the .alpha.-1,3 glycan linkages in glycosylated proteins produced from Saccharomyces are primarily responsible for the hyper-antigenic nature of those proteins making them particularly unsuitable for therapeutic use. Although not yet proven, this is predicted to be less of a problem for glycoprotein generated in Pichia, because it may resemble the glycoprotein structure of higher eukaryotes. Protein expressed as a secreted form for correctly refolding and easy to purification of HSA and HSA fusion proteins. Watanabe, et al. (2001) "In vitro and in vivo properties of recombinant human serum albumin from Pichia pastoris purified by a method of short processing time", Pharm Res 2001 December 18(12):1775; and Kobayashi, K et al. (1998) "The development of recombinant human serum albumin" Ther Apher, November 2(4):257-62.

[0105] There are many expression systems available for expressing in Pichia, such as EasySelect.TM. Pichia Expression Kit from Invitrogen, Inc. On this vector, an AOX1 promoter is used to allow methanol-inducible high level expression in Pichia and a Zeocin.TM. resistance as selective market for the recombinants from the transformation. Promoters (transcription initiation region) are very important in expression of fusion proteins in this invention.

[0106] AOX1 gene promoter is a very strong promoter in yeast system, especially in Pichia. Two Alcohol Oxidase Proteins are coded in Pichia for alcohol oxidase--AOX1 and AOX2. The AOX1 gene is responsible for the vast majority of alcohol oxidase activity in the cell. Expression of the AOX1 gene is tightly regulated and induced by methanol to very high levels, typically.gtoreq.30% of the total soluble protein in cells grown with methanol as the carbon source. The AOX1 gene has been isolated and a plasmid-bone version of the AOX1 promoter is used to drive expression of the gene of interest encoding the desired heterologous protein (Ellis et al., 1985; Koutz et al., 1989; Tschopp et al., 1987a). While AOX2 is about 97% homologous to AOX1, growth on methanol is much slower than with AOX1. This slow growth on methanol allows isolation of Mut.sup.s strains (aox 1). Except AOX1 gene promoter, other promoters can also be used to driver HSA fusion gene in yeast. They are including the promoter from, but not limited to, PGK1, GAPDH, Gal1, Gal10, CYC1, PH05, TRP1, ADH1, or ADH2 gene.

[0107] The expression plasmid can also take the form of shuttle vectors between a bacterial host such as E. coli, DH5a from GIBCO/Life Science and yeast; the antibiotic Zeocin are used to be a marker for HSA carrier vector in all the examples.

[0108] The expression vector contains the polynucleotide of HSA or HSA fusion therapeutic protein are introduced into yeast according to the protocols described in the kit from Invitrogen Inc. After selection of transformed yeast colonies, those cells expressing the HSA fusion protein of interest are inoculated into appropriate selective medium and then tested for their capacity to secrete the given fusion protein into the extracellular medium. The harvesting of the protein can be conducted during cell growth for continuous cultures, or at the end of the growth phase for batch cultures. The fusion proteins which are the subject of this invention are then further purified from the culture supernatant by methods which take into account the albumin purification methods and pharmacological activities.

[0109] It is noted that other expression systems may also be used to express rHSA and HSA/CPSF fusion proteins, including but not limited to E. coli, B. Subtitis, Saccharomyces, Kluyverornyces, Candida, Torulopsis, Torulaspora, Schizosaccharomyces, Citeromyces, Pachysolen, Debaromyces, Metschunikowia, Rhodosporidium, Leucosporidium, Botryoascus, Sporidiobolus, Endomycopsis, animals, plants, and insect cells.

3. Combination Therapy of HSA/CPSF Fusion Proteins

[0110] The present invention also provides combinations of different HSA/CPSF fusion proteins. The specific combinations of these fusion proteins may be administered to a patient to stimulate proliferation of multiple types of cells in the body or to synergistically enhance proliferation of a particular cell type. In particular, a combination of human albumin fusions with different hematopoietically active cytokines is used to effectively promoting proliferation of the multiple blood cells and platelets. By using a combination of HSA/CPSF fusion proteins targeting the signal transduction pathways of different types of blood cells, multiple blood functional cell production, such as platelets, erythrocytes and macrophages of white cells, can be increased after administration by just one injection.

[0111] In the present invention, the albumin's plasma transporter function and the therapeutic function of the CPSF are integrated into a fusion form. The presence of albumin may confer a superior stability to the CPSF by resisting degradation by proteases in the blood circulation, thus significantly prolonging the plasma half life of the CPSF. Due to the masking effect of a bulky albumin, a combination of different CPSFs fused with albumin may impose less interference with the biological function(s) among different albumin fused CPSFs than among a combination of the "naked" CPSFs. Further, a CPSF fused with albumin may be slowly released in the system over an extensive period of time, thereby reducing the toxicity associated with injection of the CPSF alone in abnormally high concentrations in the body. Such a slow release mode of action of the fusion protein combination can significantly reduce the amount and/or frequency of injections of the CPSF, thereby further reducing the side effects of CPSFs. Such combinations are particularly useful for stimulating multiple blood cell proliferation after or before the chemo- or radiation therapy of cancer patients whose tolerance for frequent, high dose injection of CPSF are seriously compromised.

[0112] For example, in animal testing human TPO, stem cell factor (SCF) and IL-3 have shown very strong toxicities in vivo. Due to the severe toxicity in vivo, SCF has been approved by the FDA for use in vitro only. The limitation on SCF has prevented it from being developed into useful therapeutics in the clinic. The side effects of IL-3 are dose-dependent. At a dose higher than 5 .mu.g/Kg, side effects have been observed in patients treated with IL-3, including fever, rash, fatigue, diarrhea, rigor, musculoskeletal pain, chills, headache, conjunctivitis, edema, chest pain, dyspnea, decrease in platelet counts, increasing in basophilic counts, marrow fibrosis, and pulmonary edema. Eder, M et al. (1997) "IL-3 in the Clinic", Stem Cells, 15:327-333.

[0113] According to the present invention, HSA fusion protein with this type of CPSF may remove above limitations by slowly releasing the drug into the patient's system. In addition, such fusion proteins may be combined with a relatively higher amount of albumin to further reduce the impact resulted from directly injecting the drug into the blood which causes a strong, adverse reaction of the central nervous system.

[0114] It is also known that "naked" cytokines (i.e., cytokines not fused to another protein such as HSA) are quite unstable when stored and have a short plasma half-life. Clearly, a therapeutic protein with such a weak stability in vivo constitutes a major handicap. In effect, repeated injections of the product, which are costly and inconvenient for patient, or an administration of product by perfusion, become necessary to attain an efficient concentration in plasma. Due to its extended plasma half-life and enhanced stability, the HSA/CPSF fusion proteins of the present invention and their combinations, e.g., HSA fusions with hIL-11, hEPO, hG-CSF and hGM-CSF, can be used to stimulate the production of multiple blood cells in plasma of humans.

[0115] In one embodiment, HSA/IL-11 fusion may be combined with HSA/EPO fusion and the resulting combination may be administered to a patient with a hematological disorder to simultaneously stimulate proliferation of erythrocytes and platelets. For example, cancer patients may be injected with a combination of HSA/IL-11 and HSA/EPO fusion proteins, before or after, chemotherapy treatment to avoid blood transfusion and to stimulate proliferation of erythrocytes and platelets.

[0116] In another embodiment, HSA/IL-3 fusion may be combined with HSA/EPO fusion and the resulting combination may be administered to a patient with a hematological disorder to enhance EPO-induced production of erythrocytes.

[0117] In yet another embodiment, HSA/IL-3 fusion may be combined with HSA/GCSF fusion and the resulting combination may be administered to a patient with a hematological disorder to increase the production of erythrocytes and neutrophils, as well as eosinophils.

Alternatively, an HSA/CPSF fusion may be co-administered with a different HSA/CPSF fusion simultaneously or sequentially to a patient in need thereof. This combination therapy may confer synergistic therapeutic effects on the patients. In one embodiment, the method is provided, comprising: administering a first pharmaceutical formulation comprising a first fusion protein of HSA and a first CPSF to the patient in a therapeutically effective amount; and administering to the patient a second pharmaceutical formulation comprising a second fusion protein of HSA and a second CPSF to the patient in a therapeutically effective amount. Such a combination therapy may confer synergistic therapeutic effects on the patient.

[0118] For example, HSA-IL-11 fusion protein may be administered to the patient first, followed by administration of HSA-EPO, HSA-GCSF and/or HSA-GMCSF at therapeutically effective doses and ratios to stimulate proliferation of different types of blood cells.

[0119] The present invention further provides a kit for use in the combination therapy described above. The kit comprises: a first fusion protein of HSA and a first CPSF, and a second fusion protein of HSA and a second CPSF. The first and second CPSFs may be the same or different. For example, the first CPSF is IL-11 and the second CPSF is EPO; the first CPSF is IL-3 and the second CPSF is EPO; or the first CPSF is IL-11 and the second CPSF is GCSF.

[0120] The HSA/CPSF fusion proteins and their combinations thereof may be used to treat a wide variety of diseases, including but not limited to the hematological disorders such as hypochromia, hypochromic microcytic anemia, and anemia, platelet-less, HIV infection, cancer, renal failure, and tissue/organ transplantation. These fusion proteins are preferred not to contain non-human sequences that may elicit adverse immunogenicity in the patient.

Examples

1. General Molecular Cloning Techniques

[0121] The classic methods of molecular cloning including, DNA preparative extractions, agarose and polyacrylamide electrophoresis, plasmid DNA purification by column or from gel, DNA fragment ligations, and restriction digestion, are described in detail in Maniatis T. et al., "Molecular cloning, a Laboratory Manual", Cold Spring Harbor laboratory, Cold Spring Harbor, N.Y., 1982 and will not be reiterated here.

[0122] Polymerase Chain Reaction (PCR) used through out all the examples is described by Saiki, R. K. et al, Science 230:1350-1354, 1985 and is carried out on a DNA thermal cycler (Perkin Elmer) according to the manufacturer's specification. DNA sequencing was performed by using standard facilities and following the method developed by Sanger et al., Proc. Natl. Acad. Sci. USA, 74:5463-5467, 1977. Oligonucleotides were synthesized by commercial facilities.

[0123] Transformation of E. coli was done by using DH5a competent cells from GIBCO/BRL. Qiagen plasmid DNA purification columns were used in the purification of plasmid DNAs. The transformation of yeast was carried out by electroporation following the instruction provided by the manufacturer or according to the manual of EasySelect.TM. Pichia Expression Kit (Invitrogen Inc). All yeast stains used in the examples are members of the family of Pichia, and in particular, the strain of Pichia pastoris (supplied by Invitrogen).

2. Construction of a Backbone Vector Expressing Human Serum Albumin

[0124] A total RNA isolated from human fetal liver was used in a reverse transcription polymerase chain reaction (RT-PCR) to generate the polynucleotide encoding human serum albumin. Briefly, 5 .mu.g of RNA was reverse transcribed by adding a poly(T).sub.18+N primer and the SuperScript.TM. II RNase H.sup.- reverse transcriptase (GIBCO/BRL) to make the complementary first strand of cDNA. The reaction was incubated at 45.degree. C. for 20 minutes, then at 55.degree. C. for 40 minutes.

[0125] The primers for cloning human serum albumin (HSA) are the following:

TABLE-US-00001 SEQ ID No. 23: 5'-GAATTCATGAAGTGGGTAACCTTTATTTCC-3' and SEQ ID No. 24: 5'-GAATTCTTATAAGCCTAAGGCAGCTTGACTTGC-3'.

[0126] These primers were designed based on the HSA sequence published by GenBank (Access# V00494). Two EcoR I (underline of primers) sites were created at the 5' end and 3' end for sub-cloning into an expression vector. After inactivating the reverse transcriptase at 94.degree. C. for 4 minutes, the DNA encoding of HSA was further amplified by Taq DNA PCR (Perkin Elmer) with 35 cycles of 94.degree. C./30 seconds and 58.degree. C./30 seconds and 72.degree. C./2 minutes 30 second, followed by a 72.degree. C./10 minutes incubation. The PCR product (1842 base pairs) was confirmed by 1% agarose gel electrophoresis. The product was subcloned into a pCR II TA cloning vector from Invitrogen. DNA sequencing confirmed that the plasmid DNA contained an insert whose polynucleotide sequence matches the DNA sequence published in GenBank (Access# V00494). FIG. 1, Seq ID No. 11 is a polynucleotide DNA sequence and Seq ID No 12 is the protein amino acid sequence of human serum albumin.

[0127] After restriction digestion of the PCR product with EcoR I, the gel purified HSA DNA fragment was inserted into a pPICZ-A vector (provided by Invitrogen) at the EcoR I site. After transformation of bacteria DH5a cells with this vector encoding HSA, a colony was selected from a low salt LB-agar plate contains 25 .mu.g/ml Zeocin. The direction of the insert was confirmed by restriction enzyme double digestion of plasmid DNA by Xho I/Nde I. The construct was designated as pYZ-HSA (Y: yeast vector; Z: Zeocin resistant) and its physical map is shown in FIG. 2.

[0128] There are some advantages associated with the vector constructed above. It confers resistance to the antibiotic Zeocin. Zeocin is isolated from Streptomyces and is structurally related to bleomycin/phleomycin-type antibiotics. Antibiotics in the family of bleomycin/phleomycin are broad spectrum antibiotics that act as strong antibacterial and anti-tumor drugs. They show strong toxicity against bacteria, fungi (including yeast), plants, and mammalian cells. However, Zeocin is not as toxic as bleomycin on fungi. A single antibiotic Zeocin could be used in selecting the recombinants in both bacteria and in yeast. Further, there are multiple cloning sites at the 3' end of HSA for conveniently subcloning a CPSF protein in frame to encode a HSA-CPSF. In addition, a myc epitope sequence and a polyhistidine tag can be fused to the C-terminal of the expressed fusion protein for easy detection and/or purification by using commercially available antibodies against myc or polyhistidine tags. This vector, as a backbone vector, was used in the construction of expression vectors for all the HSA fusion proteins described in the Example section.

3. Molecular Cloning of Human IL-11, EPO, G-CSF, and GM-CSF

3.1. Molecular Cloning Of Human IL-11 Gene

[0129] Human Il-11 was cloned from a total RNA preparation of human bone marrow-derived stromal cells by RT-PCR method described in Example 2. The oligonucleotide primers are

TABLE-US-00002 SEQ ID NO. 25: 5'-CATATGAACTGTGTTTGCCGCCTGGTCC-3' SEQ ID NO. 26: 5'-GATATGTATGACACATTTAATTCCC-3'

[0130] A polynucleotide having 1051 base pairs (bp) was amplified from RT-PCR reaction and subcloned into pCR II TA cloning vector from Invitrogen Inc. DNA sequencing confirmed the reading frame of human IL-11 and inclusion of a 448 bp 3'-end un-translation region of hIL-11. An Nde I restriction enzyme site was created at the 5' end. The ATG initiate start codon of hIL-11 was included in this site (underlined in SEQ ID NO. 25). The DNA sequence of hIL-11 (SEQ ID NO. 13) and its amino acid sequence (SEQ ID NO. 14) are shown in FIG. 1.

3.2. Molecular Cloning of Human Erythropoietin Gene

[0131] Human erythropoietin gene was obtained by RT-PCR from human fetal kidney mRNA from Clontech Laboratory. EPO specific primers were designed based on the sequence published by Lin, F K et al., "Cloning and expression of human erythropoietin gene", Proc. Natl. Acad. Sci. USA., 82(22):7580-7584 (1985). They are

TABLE-US-00003 SEQ ID NO. 27: 5'-GGATCCATGGGGGTGCACGAATGTCC-3', and SEQ ID NO. 28: 5'-GAATTCTCATCTGTCCCCTGTCCTGC-3'.

[0132] The PCR product was a full length reading frame of EPO with two newly created restriction enzymes in each end, Bam HI at the 5' end and EcoR I at 3' end. The product was inserted into pCR II vector and sequence confirmed. The human EPO DNA sequence (SEQ ID NO. 15) and amino acid sequence (SEQ ID NO. 16) are showed in FIG. 1.

3.3. Molecular Cloning of Human G-CSF

[0133] Primers used to clone the human G-CSF gene from a cDNA library of human fetal liver tissues are

TABLE-US-00004 SEQ ID NO. 29: 5'-GGATCCATGGCTGGACCTGCCACCC-3', and SEQ ID NO. 30: 5'-GAATTCTCAGGGCTGGGCAAGGTGGC-3'

[0134] These primers were designed based on Nagata, S et al., Molecular Cloning and Expression of cDNA for Human Granulocyte Colony-Stimulating Factor, Nature, 319:415-418, 1986. A Bam HI site at 5' end and an EcoR I site at 3' end of G-CSF were created. The PCR products were gel-purified and subcloned into pCR2.1 TA cloning vectors and DNA sequence was confirmed. The human G-CSF DNA sequence (SEQ ID NO. 17) and the amino acid sequence (SEQ ID NO. 18) are shown in FIG. 1.

3.4. Molecular Cloning of Human GM-CSF

[0135] Human GM-CSF was cloned from a total RNA sample prepared from human fetal liver based on Wong, G G et al., "Human GM-CSF: molecular cloning of the complementary DNA and purification of the nature and recombinant proteins" Science, 228:810-815, 1985. The Primers were:

TABLE-US-00005 SEQ ID NO. 31: 5'-GGATCCATGTGGCTGCAGAGCCTGCTGC-3', and SEQ ID NO. 32: 5'-GAATTCTCACTCCTGGACTGGCTCC-3'

[0136] The PCR products were gel-purified and inserted into pCR2.1 TA cloning vector and sequence confirmed. The human GM-CSF DNA sequence (SEQ ID NO. 19) and amino acid sequence (SEQ ID NO. 20) are shown in FIG. 1.

4. In Frame Fusion of HSA With Human IL-11, EPO, G-CSF or GM-CSF

[0137] There is a Bsu36 I site at the C'-terminus of HSA. All of the CPSFs described in the Example section were fused into this site by PCR primer extension to generate a restriction enzyme site of Bsu36 I at the N-terminus of the CPSF DNA sequence. The CPSF DNA fragments were amplified by PCR and then subcloned into Bsu36 I and Xho I sites of pYZ-HSA vector which had been double digested with Bsu36 I and Xho I to linearize the plasmid DNA

4.1. Construction of Vector Containing Hybrid Polynucleotide of HSA/hIL-11

[0138] IL-11 gene was fused to HSA C'-terminus by using the following PCR primers:

TABLE-US-00006 SEQ ID NO. 33: 5'-CTGCCTTAGGCTTACCTGGGCCACCACCTGGCC-3'. (Human IL-11 mature protein sequence is underlined), and SEQ ID NO. 34: 5'-TGTCGACTCACAGCCGAGTCTTCAGCAGC-3'.

[0139] A Sal I site (underlined in SEQ ID NO. 34) was created at the 3' end of hIL-11 gene because there is a Xho I site in the sequence of mature protein of IL-11. The Sal I site sequence is a cohesive sequence with Xho I site. After ligation, the Sal I and Xho I site were all gone.

[0140] The PCR products were digested with Bsu36I and Sal I, and the fragment was gel purified and inserted into pYZ-HSA between of Bsu36 I and Xho I sites to generate a new plasmid DNA, pYZ-HSA/hIL-11. The HSA-hIL-11 hybrid polynucleotide sequence (SEQ ID NO. 1) and its fusion protein amino acid sequence (SEQ ID NO. 2) are showed in FIG. 1.

4.2. Construction of Vector Containing Hybrid Polynucleotide of HSA/EPO

[0141] To make a HSA-EPO fusion protein, the following primers were designed

TABLE-US-00007 SEQ ID NO. 35: 5'-CTGCCTTAGGCTTAATCTGTGACAGCCGAGTCC-3' (human EPO mature protein sequence underlined), and SEQ ID NO. 36: 5'-CACTCGAGTCATCTGTCCCCTGTCCTGC-3' (Xho I site underlined)

and used to generate the modified human IL-11 DNA fragment. The PCR products were inserted between Bsu36I and Xho I sites of pYZ-HSA to generate a pYZ-HSA/hEPO. The HSA-EPO hybrid polynucleotide sequence (SEQ ID NO. 5) and its fusion protein amino acid sequence (SEQ ID NO. 6) are shown in FIG. 1.

4.3. Construction of Vector Containing Hybrid Polynucleotide of HSA/HG-CSF

[0142] Human G-CSF gene was fused with HSA DNA sequence by using two primers:

TABLE-US-00008 SEQ ID NO. 37: 5'-CTGCCTTAGGCTTAACCCCCCTGGGCCCTGCCAGC-3' (G-CSF mature protein sequence underlined), and SEQ ID NO. 38: 5'-CTCGAGTCAGGGCTGGGCAAGGTGG-3' (Xho I site at the 3'-teminus of G-CSF underlined).

[0143] The PCR products were gel purified and subcloned between Bsu36I and Xho I sites of pYZ-HSA to generate a pYZ-HSA/hG-CSF. The HSA-G-CSF hybrid polynucleotide sequence (SEQ ID NO. 7) and its amino acid sequence (SEQ ID NO. 8) are shown in FIG. 1.

4.4. Construction of Vector Containing Hybrid Polynucleotide of HSA/HGM-CSF

[0144] The following primers:

TABLE-US-00009 SEQ ID NO. 39: 5'-ACTCCTTAGGCTTAGCACCCGCCCGCTCGCCCAGC-3' (GM-CSF mature protein sequence underlined), and SEQ ID NO. 40: 5'-CTCGAGTCACTCCTGGACTGGCTCC-3' (Xho I site underlined)

[0145] were used to modify GM-CSF DNA sequence in order to subclone it into pYZ-HSA vector. PCR products were gel purified and double digested with Bsu36 I and Xho I and inserted between Bsu36 I and XhoI sites of pYZ-HSA to generate a pYZ-HSA/hGMCSF. The HSA/GM-CSF hybrid polynucleotide sequence (SEQ ID NO. 9) and its fusion protein amino acid sequence (SEQ ID NO. 10) are shown in FIG. 1.

5. Transformation of Yeasts

[0146] A yeast Pichia pastoris strain, GS115, colony was inoculated into 5 ml of YPD medium in a 50 ml conical tube at 30.degree. C. overnight with shaking at 250 rpm. 0.2 ml of the culture was inoculated into 500 ml of YPD medium continually shaking at 30.degree. C. for further 2-3 hours or until the cell density reach to OD.sub.600=1.3-1.5. The cells were collected by centrifuging. The cell pellet resuspend in 500 ml of ice-cold sterile water in order to wash the cells. After two rounds water washing, the cells were resuspended in 20 ml of ice-cold 1M sorbitol to wash again. The cells finally suspended in 1 ml of ice-cold 1M sorbitol. The plasmid DNA constructs from Example 2, pYZ-HSA and in Example 4, pYZ-HSA/IL-11, pYZ-HSA/hEPO, pYZ HSA/hG-CSF, and pYZ-HSA/hGM-CSF was linearized by Pmel restriction enzyme digestion first.

[0147] Five .mu.g of each linear plasmid DNA was used to transform 80 .mu.l of the freshly made yeast cells in an ice-cold 0.2 cm electroporation cuvette. The cells mixed with plasmid DNA were pulsed for 5-10 ms with field strength of 7500V/cm. After the pulse, 1 ml of ice-cold 1M sorbitol was immediately added into the cuvette and the content was transferred to a sterile 15 ml tube. The transformed cells were incubated in 30.degree. C. without shaking for 2 hours then spread on pre-made YPD-agar plates with 100 .mu.g/ml Zeocin. The colonies were identified with the insert and the expression level was determined by SDS-PAGE or western-blot with proper antibodies.

[0148] A consortium of 4 yeast strains produced above (collected referred to as YZ-HSA/CPSFs) of Pichia pastoris encoding HSA/IL-11, HSA/hEPO, HSA/hG-CSF, and HSA/hGM-CSF was deposited to the ATCC.RTM. (America Type Culture Collection) under the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. The 4 yeast strains are separately designated as YZ-HSA/IL-11, YZ-HSA/hEPO, YZ-HSA/hG-CSF, and YZ-HSA/hGM-CSF, respectively, and their consortium is deposited at the ATCC.RTM. under Patent Deposit Designation No: PTA-4607.

[0149] Different strains of Pichia, such as X-33, KM71 and proteinase deficient strains SMD1168 and ZY101 were tested for the expression and secretory of recombinant proteins.

6. Secretion and Characterization of HSA-CPSF Fusion Proteins Expressed by Pichia

[0150] Several colonies from each transformation of the HSA-CPSF were cultured with Zeocin in the buffered minimal medium with glycerol overnight or until OD.sub.600=2-6 at 30.degree. C. and shaking at 300 rpm. The cultured cells were collected by centrifuge at 1500 rpm for 5 minutes. Resuspend the cells into buffered minimal medium without glycerol and cell densities was keep in OD.sub.600=1.0. 100% methanol was added into each flask to a final concentration at 0.5% every 24 hours to induce the protein expression. The culture medium was collected at different time points and the expression of each fusion protein was confirmed by SDS-PAGE and western blot. The results showed that human albumin and HSA-CPSF fusion protein were expressed and secreted into the medium.

[0151] Mouse monoclonal anti-human serum albumin (Sigma) was used for immunoblotting on a SDS-PAGE gel. A typical Western blot experiment was carried on by electrophoresis transfer the protein from SDS-PAG to a nylon or nitrocellulose filter and incubated with a specific antibody (as the "first antibody"). Then an anti-first antibody was added to bind to the first antibody (as the "second antibody"). The second antibody was labeled with Fluorescence and the filter was exposed to an X-ray film. Protein molecular weight standard was used to determine the protein size. The results (FIG. 3) showed that the expressed recombinant proteins, HSA, HSA-CPSF therapeutic fusion protein, had an expected molecular weight and also had the same antigen as that of HSA prepared from a human blood plasma (Sigma). Using monoclonal anti-hIL-11 specific antibody as the first antibody, the HSA/hIL-11 fusion protein and human IL-11 (R&D System) had the same antigen and showed that the molar ratio of HSA to hIL-11 in the HSA/hIL-11 fusion protein is as expected (FIG. 4). Using monoclonal anti-hGMCSF specific antibody (R&D System) as the first antibody, the HSA-GMCSF fusion protein and human GMCSF (R&D System) had the same antigen and showed that the molar ratio of HSA to GMCSF in the HSA/GMCSF fusion protein is as expected (FIG. 5).

7. Purification and Molecular Characterization of HSA-CPSF

[0152] The cell culture medium (supernatant) containing the secreted protein of HSA or HSA-CPSF fusion protein produced from the recombinant Pichia was collected, the salt concentration reduced, and the pH was adjusted to above 7.5. The concentrated sample was passed through an Affi-Gel Blue-gel (50-100 mesh) (Bio-Rad). The albumin or albumin fusion protein was bound to the matrix and eluded by a gradient 1-5M NaCl. 75-85% pure albumin or albumin-CPSF can be recovered in this step. If further purification is necessary, a size exclusion chromatography is applied to give a 95-99% purity of proteins. The pyrogen was removed from the protein samples in order to meet the requirement for use in in vivo test. The Affi-Prep Polymyxin Support (BIO-Rad) column was used to remove endotoxin from the samples. The purified protein finally passed through 0.2 .mu.M filter to be sterilized and the protein concentration was measured by a standard method by using a Bio-Rad Protein Assay Kit.

8. Cell Proliferation Assay of Human IL-11

[0153] A murine plasmacytoma cell line T1165 which is an IL-6 depended cell line was used to carry out a bioassay for IL-11 according to Paul, S R et al., PNAS 87:7512-7516, 1990. The cells were maintained in RPMI medium supplemented with 10% heat-inactivated fetal calf serum, 2 mM glutamine, penicillin (100 U/ml) and streptomycin (100 .mu.g/ml) (Gibco/BRL), 50 .mu.M 2-meracaptoethanol (sigma), and recombinant human IL-6 (4-10 ng/ml or 20 U/ml in final concentration) (Gibco/BRL). Bioassay is preformed by 1.times.10.sup.4 cells/well were placed into 96-well tissue culture plate in 200 ul of IL-6 free medium for 48 hr in 37.degree. C. in the presence of multiple dilution of hIL-11 purified from Thioredoxin fusion protein in E. coli or from yeast purified HSA/IL-11 fusion protein. In the final 6 hours, the cells were pulse-labeled with 0.5 mCi of [.sup.3H]thymidine (1Ci=37 GBq, from Dupont) per well. After incubation, the cells were collected on to a glass filter, washed and assayed on a Beckman scintillation counter, Neutralizing goat anti-human IL-6 antibody was included to abrogate the effect of IL-6 as control in the testing of purified protein samples. The control proteins including with or without HSA (from human blood preparation), rHSA expressed in yeast (Pichia) in the lab, Thioredoxin (Trx) was a peptide purified from the enterokinase digested Trx/IL-11 fusion protein. The results are shown in FIG. 6 (A and B panels). The HSA/hIL-11 bioactivities were not affected by the presence of antibody of human IL-6. HSA fusion protein has about 1/3 of cell proliferation activity compared with that human IL-11 alone in the same amount of protein. Since HSA has a molecular weight about 3 times higher than that of hIL-11, it can be inferred that HSA-hIL-11 fusion protein has the same bioactivity as that of human IL-11 alone.

9. Bioassay of EPO by ELISA

[0154] Enzyme-linked immunosorbent assay (ELISA) kit from R&D Systems was used for the quantitative determination of erythropoietin (EPO) concentration and bioactivities comparison with a commercial EPO sample. The EPO ELISA is based on the double-antibody sandwich method. Microplate wells, precoated with monoclonal (murine) antibody specific for human EPO were incubated with samples or standard. Erythropoietin binds to the immobilized antibody on the plate. After removing excess protein sample or standard, wells were incubated with an anti-EPO polyclonal (rabbit) antibody conjugated to horseradish peroxidase. During the second incubation, the antibody-enzyme conjugate bound to the immobilized EPO. Excess conjugate was removed by washing. A chromogen was added into the wells and oxidized by the enzyme reaction to form a blue colored complex.

[0155] The reaction was stopped by the addition of acid, which turned the blue to yellow. The amount of color generated was directly proportional to the amount of conjugate bound to the EPO antibody complex, which, in turn, was directly proportional to the amount of EPO in the protein samples or standard. The absorbance of this complex was measured and a standard curve was generated by plotting absorbance versus the concentration of the EPO standards. The EPO concentration of the unknown sample was determined by comparing the optical density of the protein samples to the standard curve. The standards used in this assay were recombinant human EPO (with kit) calibrated against the Second International Reference Preparation (67/343), a urine-derived form of human erythropoietin. Human recombinant EPO expressed in CHO cells was used as a control to determine the rHSA/EPO bioassay sensitivity.

[0156] The results showed that in the bioassay hEPO fused to HSA had 1/10 of the sensitivity compared with the standard (FIG. 7). The size of HSA-EPO fusion protein molecule may be too large, which prevents the anti-EPO antibody from efficiently binding to the EPO molecule fused to HSA, thereby reducing the sensitivity of the detection in this bioassay.

10. Stability Testing of HSA-CPSF Fusion Proteins In Vitro

[0157] Using HSA/hIL-11 as an example, the stability of this HSA-CPSF fusion protein was tested at different time points at 37.degree. C. and 50.degree. C. 5 ng of human IL-11 from bacteria or 5 ng of rHSA/hIL-11 was put into 200 .mu.l thin-well PCR tube with 200 .mu.l of tissue culture medium RPM1 without fetal bovine serum and other components. The tubes were sealed and left in water bath. Samples were taken out at different time points and immediately put into -80.degree. C. for storage. After all of samples were collected, a cell proliferation test on T1165 cell line was carried out by incorporating .sup.3H-Thymidine into newly synthesized DNA of proliferating cells. The control of the test was set up in the same way as that in the bioassay of human IL-11 (See Paul et al., 1990). As shown in FIG. 8, after 5 weeks in 37.degree. C. (Panel A), the bioactivity of HSA/IL-11 still remained the same, but the "naked" human IL-11 lost almost all of its bioactivity after three weeks at 37.degree. C. At 50.degree. C. (Panel B), the "naked" human IL-11 lost its the bioactivity completely in 2 weeks. And the HSA/IL-11 fusion protein still retained at least half of its bioactivity. These results indicate that a CPSF fused to human albumin can have a longer storage time and more resistant to degradation in harsh environment such as high temperatures.

11. Synergistic Effects of Combination of HSA-CPSF Fusion Proteins in Stimulation of Multicell Proliferation

[0158] Rabbits (2.3-2.6 Kg) were injected with 200 .mu.l of recombinant proteins prepared above at day 1, day 3 and day 6. Rabbit A was injected with a mixture of 150 U/kg EPO (about 10 .mu.g of protein) and 100 .mu.g recombinant HSA (rHSA); Rabbit B with a mixture of 120 .mu.g IL-11 and 100 .mu.g rHSA; and Rabbit C with a mixture of 120 .mu.g rHSA/hIL-11 fusion (equivalent of 50 .mu.g of pure bacteria-expressed IL-11), 27 .mu.g rHSA/hEPO fusion (equivalent of 150 U of HSA-EPO determined by using the EPO-ELISA Kit, R&D System Inc.) and 50 .mu.g rHSA. Rabbit D was injected with 120 .mu.g of HSA-IL-11 and about 80 .mu.g rHSA.

[0159] Blood samples were collected and red blood cell and platelet numbers were counted by a hemacytometer. The results are shown in FIG. 9 (panels A, B, and C). The cell counts on the starting day of the experiment was treated as the base line. After the treatment with the proteins, the cell counts were compared with those on the starting day and the changes were plotted in the graphs in FIG. 9.

[0160] As shown in panel A of FIG. 9, both EPO and HSA/EPO fusion protein stimulated the production of erythrocytes in rabbit A and C, respectively. However, in rabbit A injected with the naked EPO, the level of erythrocytes reached a peak around day 35 post first injection and then declined quickly to reach near a baseline level around day 55. In contrast, in rabbit C injected with HSA/EPO fusion protein, the level of erythrocytes increased and reached a plateau around day 35 post first injection but remained high till the end of the experiment. These results demonstrated that HSA/EPO fusion protein has a much longer plasma half-life than the naked EPO and remains bioactive for a much longer time than the naked EPO in vivo. As also shown in this panel, IL-11 had little effect in stimulation of erythrocyte production in rabbit B.

[0161] As shown in panel B of FIG. 9, both IL-11 and HSA/IL-11 fusion protein stimulated the production of platelets in rabbit B and C, respectively. However, in rabbit B injected with the naked IL-11, the level of platelets reached a peak around day 20 post first injection and then declined quickly to reach near a baseline level around day 43. In contrast, in rabbit C injected with HSA/IL11 fusion protein, the level of platelets increased and reached a plateau around day 40 post first injection but remained high till the end of the experiment. These results demonstrated that HSA/IL-11 fusion protein has a much longer plasma half-life than the naked IL-11 and remains bioactive for a much longer time than the naked IL-11 in vivo. As also shown in this panel, EPO had little effect in stimulation of platelet production in rabbit A.

[0162] Panel C in FIG. 9 compares the effects of a combination of HSA/EPO and HSA/IL-11 with HSA/EPO and HSA/IL-11 individually in stimulation of the production of erythrocytes and platelets on day 35 post first injection. As shown in this panel, compared with individual HSA/EPO and HSA/IL-11, a combination of HSA/EPO and HSA/IL-11 had much stronger effects in stimulating the production of both erythrocytes and platelets. For example, the erythrocyte level in the rabbit C which was injected with 27 .mu.g of HSA/EPO in combination 120 .mu.g HSA/IL-11 with is higher than the total erythrocyte level combining that in rabbit A (injected with 10 .mu.g HSA/EPO) and rabbit D (120 .mu.g HSA/IL-11). Since the molar ratio of HSA to EPO in the HSA/EPO fusion is about 5:1 and the amount of HSA/EPO in rabbit C is only 2.7 folds of that in Rabbit A, it can be predicted that if the amount of HSA/EPO is increased 5 folds so that a equal mole of HSA/EPO in both the HSA/EPO+HSA/IL-11 combination and HSA/EPO alone is administered, the erythrocyte level in the animal should be even higher with the administration of the combination. Based on this analysis, it can be reasonably inferred that a combination of HSA/EPO+HSA/IL-11 fusion proteins has a synergistic effect in stimulating multiple blood cell production.

12. Expression and Scale-Up of HSA-CPSF Fusion Protein by Fermentation

[0163] In this example, it is shown that expression and scale-up are much easier by using a Pichia system than other currently available systems. After Pichia recombinants were isolated, expression of both Mut+ and Mut.sup.s recombinants was tested. This involved growing a small culture of each recombinant, inducing with methanol, and taking sample at different time points. For secretory expression, both the cell pellet and supernatant were analyzed from each time point. The samples were analyzed on SDS-PAGE gels by using both Coomassie staining and Western blot. Bioactivities of expressed samples were tested and the expression levels and purity were monitored in each step for production of HSA fusion proteins. FIG. 10 shows a SDS-PAGE of the purity of various HSA-CPSFs fusion proteins.

REFERENCES CITED

TABLE-US-00010 [0164] U.S. Patent Documents 4,683,293 July, 1987 Craig 4,847,201 July, 1989 Kaswasaki, et al. 5,292,646 March, 1994 McCoy, et al. 5,371,193 December, 1994 Bennett, et al. 5,457,038 October, 1995 Trinchieri, et al. 5,466,781 November, 1995 Dorin, et al. 5,470,569 November, 1995 Kaswasaki, et al. 5,547,933 August, 1996 Lin, et al. 5,686,263 November, 1997 Wurm 6,022,953 February, 2000 Ralph, et al. 6,160,089 December, 2000 Honjo, et al. 6,162,467 December, 2000 Baumann, et al. (*) 6,165,470 December, 2000 Becquart, et al. 6,207,802 March, 2001 Zsebo, et al. 6,254,870 July, 2001 Staten, et al. 6,258,559 July, 2001 Zamost 6,300,314 October, 2001 Wallner, et al. 6,309,632 October, 2001 Agosti 6,322,779 November, 2001 Halenbeck, et al. 6,326,198 December, 2001 Emerson, et al. Foreign Patent Documents WO01/79271 October, 2001 Balance et al. WO01/79442 October, 2001 Rosen and Haseltin

OTHER REFERENCES

[0165] Testa, et al., Exp. Hematol., 8(Supp. 8), 144-152 (1980). [0166] Tong, et al., J. Biol. Chem., 256(24), 12666-12672 (1981). [0167] Goldwasser, J. Cell. Physiol., 110(Supp 1), 133-135 (1982). [0168] Finch, Blood, 60(6), 1241-1246 (1982); Sytowski, et al., Exp. Hematol., 8(Supp 8), 52-64 (1980). [0169] Naughton, Ann. Clin. Lab. Sci., 13(5), 432-438 (1983). [0170] Weiss, et al., Am. J. Vet. Res., 44(10), 1832-1835 (1983). [0171] Lappin, et al., Exp. Hematol., 11(7), 661-666 (1983). [0172] Baciu, et al., Ann. N.Y. Acad. Sci., 414, 66-72 (1983). [0173] Murphy, et al., Acta. Haematologica Japonica, 46(7), 1380-1396 (1983). [0174] Dessypris, et al., Brit. J. Haematol, 56, 295-306 (1984). [0175] Emmanouel, et al., Am. J. Physiol., 247 (1 Pt 2), F168-76 (1984). [0176] Lin, F K et al., "Cloning and expression of human erythropoietin gene", Proc. Natl. Acad. Sci. USA., 82(22):7580-7584 (1985). [0177] Sanger et al., Proc. Natl. Acad. Sci. USA, 74:5463-5467, 1977) [0178] Saiki, R. K. et al, Science 230:1350-1354, 1985 [0179] Maniatis T et al., "Molecular cloning, a Laboratory Manual", Cold Spring Harbor laboratory, Cold Spring Harbor, N.Y., 1982 [0180] Ellis et al., 1985; Koutz et al., 1989; Tschopp et al., 1987a (Pichia manu) [0181] Watanabe, et al., Pharm Res 2001 December 18(12):1775. [0182] Kobayashi, K et al., Ther Apher, November 2(4):257-62, 1998. (Pichia) [0183] Buckholz and Gleeson, 1991; Cregg and Higgins, 1995 (Pichia) [0184] Metcalf, D., Blood, 67, 257, 1986 [0185] Mufson, R. A. et al., Cellular Immunol., 119, 182, 1989 [0186] Wong, G., et al., Science, 228, 810, 1986. [0187] Metcalf, D., Blood, 67, 257, 1986. [0188] Hattersley, G., et al., J. Cell Physiol., 137, 199, 1988. [0189] Nicola, N., Immunol. Today, 8, 134, 1987. [0190] Kitamura, T., et al., J. cell Physiol., 140, 323, 1989. [0191] Kawasaki, E. S., et al., Science, 230, 291, 1985. [0192] Morrison, "Bioprocessing in Space--an Overview", pp. 557-571 in The World Biotech [0193] REPOrt 1984, Volume 2:USA, (Online Publications, New York, N.Y. 1984). [0194] Vedovato, et al., Acta. Haematol, 71, 211-213 (1984) [0195] Vichinsky, et al., J. Pediatr., 105(1), 15-21 (1984) [0196] Cotes, et al., Brit. J. Obstet. Gyneacol., 90(4), 304-311 (1983) [0197] Haga, et al., Acta. Pediatr. Scand., 72, 827-831 (1983) [0198] Claus-Walker, et al., Arch. Phys. Med. Rehabil., 65, 370-374 (1984) [0199] Dunn, et al., Eur. J. Appl. Physiol., 52, 178-182 (1984) [0200] Miller, et al., Brit. J. Haematol., 52, 545-590 (1982) [0201] Udupa, et al., J. Lab. Clin. Med., 103(4), 574-580 and 581-588 (1984); [0202] Lipschitz, et al., Blood, 63(3), 502-509 (1983) [0203] Dainiak, et al., Cancer, 51(6), 1101-1106 (1983) [0204] Schwartz, et al., Otolaryngol., 109, 269-272 (1983) [0205] Pennathur-Das, et al., Blood, 63(5), 1168-71 (1984) [0206] Haddy, Am. Jour. Ped. Hematol./Oncol., 4, 191-196, (1982) [0207] Eschbach, et al. J. Clin. Invest., 74(2), pp. 434-441, (1984 [0208] Krane, Henry Ford Hosp. Med. J., 31(3), 177-181 (1983).

Sequence CWU 1

1

4012229DNAArtificial SequenceDNA of HSA-hIL-11 1atgaagtggg taacctttat ttcccttctt tttctcttta gctcggctta ttccaggggt 60gtgtttcgtc gagatgcaca caagagtgag gttgctcatc ggtttaaaga tttgggagaa 120gaaaatttca aagccttggt gttgattgcc tttgctcagt atcttcagca gtgtccattt 180gaagatcatg taaaattagt gaatgaagta actgaatttg caaaaacatg tgttgctgat 240gagtcagctg aaaattgtga caaatcactt catacccttt ttggagacaa attatgcaca 300gttgcaactc ttcgtgaaac ctatggtgaa atggctgact gctgtgcaaa acaagaacct 360gagagaaatg aatgcttctt gcaacacaaa gatgacaacc caaacctccc ccgattggtg 420agaccagagg ttgatgtgat gtgcactgct tttcatgaca atgaagagac atttttgaaa 480aaatacttat atgaaattgc cagaagacat ccttactttt atgccccgga actccttttc 540tttgctaaaa ggtataaagc tgcttttaca gaatgttgcc aagctgctga taaagctgcc 600tgcctgttgc caaagctcga tgaacttcgg gatgaaggga aggcttcgtc tgccaaacag 660agactcaagt gtgccagtct ccaaaaattt ggagaaagag ctttcaaagc atgggcagta 720gctcgcctga gccagagatt tcccaaagct gagtttgcag aagtttccaa gttagtgaca 780gatcttacca aagtccacac ggaatgctgc catggagatc tgcttgaatg tgctgatgac 840agggcggacc ttgccaagta tatctgtgaa aatcaagatt cgatctccag taaactgaag 900gaatgctgtg aaaaacctct gttggaaaaa tcccactgca ttgccgaagt ggaaaatgat 960gagatgcctg ctgacttgcc ttcattagct gctgattttg ttgaaagtaa ggatgtttgc 1020aaaaactatg ctgaggcaaa ggatgtcttc ctgggcatgt ttttgtatga atatgcaaga 1080aggcatcctg attactctgt cgtgctgctg ctgagacttg ccaagacata tgaaaccact 1140ctagagaagt gctgtgccgc tgcagatcct catgaatgct atgccaaagt gttcgatgaa 1200tttaaacctc ttgtggaaga gcctcagaat ttaatcaaac aaaattgtga gctttttgag 1260cagcttggag agtacaaatt ccagaatgcg ctattagttc gttacaccaa gaaagtaccc 1320caagtgtcaa ctccaactct tgtagaggtc tcaagaaacc taggaaaagt gggcagcaaa 1380tgttgtaaac atcctgaagc aaaaagaatg ccctgtgcag aagactatct atccgtggtc 1440ctgaaccagt tatgtgtgtt gcatgagaaa acgccagtaa gtgacagagt caccaaatgc 1500tgcacagaat ccttggtgaa caggcgacca tgcttttcag ctctggaagt cgatgaaaca 1560tacgttccca aagagtttaa tgctgaaaca ttcaccttcc atgcagatat atgcacactt 1620tctgagaagg agagacaaat caagaaacaa actgcacttg ttgagcttgt gaaacacaag 1680cccaaggcaa caaaagagca actgaaagct gttatggatg atttcgcagc ttttgtagag 1740aagtgctgca aggctgacga taaggagacc tgctttgccg aggagggtaa aaaacttgtt 1800gctgcaagtc aagctgcctt aggcttagct cccatgaccc agacaacgtc cttgaagaca 1860agctgggtta actgctctaa catgatcgat gaaattataa cacacttaaa gcagccacct 1920ttgcctttgc tggacttcaa caacctcaat ggggaagacc aagacattct gatggaaaat 1980aaccttcgaa ggccaaacct ggaggcattc aacagggctg tcaagagttt acagaacgca 2040tcagcaattg agagcattct taaaaatctc ctgccatgtc tgcccctggc cacggccgca 2100cccacgcgac atccaatcca tatcaaggac ggtgactgga atgaattccg gaggaaactg 2160acgttctatc tgaaaaccct tgagaatgcg caggctcaac agacgacttt gagcctcgcg 2220atcttttag 22292763PRTArtificial SequenceHSA-hIL-11 2Asp Ala His Lys Ser Glu Val Ala His Arg Phe Lys Asp Leu Gly Glu1 5 10 15Glu Asn Phe Lys Ala Leu Val Leu Ile Ala Phe Ala Gln Tyr Leu Gln 20 25 30Gln Cys Pro Phe Glu Asp His Val Lys Leu Val Asn Glu Val Thr Glu 35 40 45Phe Ala Lys Thr Cys Val Ala Asp Glu Ser Ala Glu Asn Cys Asp Lys 50 55 60Ser Leu His Thr Leu Phe Gly Asp Lys Leu Cys Thr Val Ala Thr Leu65 70 75 80Arg Glu Thr Tyr Gly Glu Met Ala Asp Cys Cys Ala Lys Gln Glu Pro 85 90 95Glu Arg Asn Glu Cys Phe Leu Gln His Lys Asp Asp Asn Pro Asn Leu 100 105 110Pro Arg Leu Val Arg Pro Glu Val Asp Val Met Cys Thr Ala Phe His 115 120 125Asp Asn Glu Glu Thr Phe Leu Lys Lys Tyr Leu Tyr Glu Ile Ala Arg 130 135 140Arg His Pro Tyr Phe Tyr Ala Pro Glu Leu Leu Phe Phe Ala Lys Arg145 150 155 160Tyr Lys Ala Ala Phe Thr Glu Cys Cys Gln Ala Ala Asp Lys Ala Ala 165 170 175Cys Leu Leu Pro Lys Leu Asp Glu Leu Arg Asp Glu Gly Lys Ala Ser 180 185 190Ser Ala Lys Gln Arg Leu Lys Cys Ala Ser Leu Gln Lys Phe Gly Glu 195 200 205Arg Ala Phe Lys Ala Trp Ala Val Ala Arg Leu Ser Gln Arg Phe Pro 210 215 220Lys Ala Glu Phe Ala Glu Val Ser Lys Leu Val Thr Asp Leu Thr Lys225 230 235 240Val His Thr Glu Cys Cys His Gly Asp Leu Leu Glu Cys Ala Asp Asp 245 250 255Arg Ala Asp Leu Ala Lys Tyr Ile Cys Glu Asn Gln Asp Ser Ile Ser 260 265 270Ser Lys Leu Lys Glu Cys Cys Glu Lys Pro Leu Leu Glu Lys Ser His 275 280 285Cys Ile Ala Glu Val Glu Asn Asp Glu Met Pro Ala Asp Leu Pro Ser 290 295 300Leu Ala Ala Asp Phe Val Glu Ser Lys Asp Val Cys Lys Asn Tyr Ala305 310 315 320Glu Ala Lys Asp Val Phe Leu Gly Met Phe Leu Tyr Glu Tyr Ala Arg 325 330 335Arg His Pro Asp Tyr Ser Val Val Leu Leu Leu Arg Leu Ala Lys Thr 340 345 350Tyr Glu Thr Thr Leu Glu Lys Cys Cys Ala Ala Ala Asp Pro His Glu 355 360 365Cys Tyr Ala Lys Val Phe Asp Glu Phe Lys Pro Leu Val Glu Glu Pro 370 375 380Gln Asn Leu Ile Lys Gln Asn Cys Glu Leu Phe Glu Gln Leu Gly Glu385 390 395 400Tyr Lys Phe Gln Asn Ala Leu Leu Val Arg Tyr Thr Lys Lys Val Pro 405 410 415Gln Val Ser Thr Pro Thr Leu Val Glu Val Ser Arg Asn Leu Gly Lys 420 425 430Val Gly Ser Lys Cys Cys Lys His Pro Glu Ala Lys Arg Met Pro Cys 435 440 445Ala Glu Asp Tyr Leu Ser Val Val Leu Asn Gln Leu Cys Val Leu His 450 455 460Glu Lys Thr Pro Val Ser Asp Arg Val Thr Lys Cys Cys Thr Glu Ser465 470 475 480Leu Val Asn Arg Arg Pro Cys Phe Ser Ala Leu Glu Val Asp Glu Thr 485 490 495Tyr Val Pro Lys Glu Phe Asn Ala Glu Thr Phe Thr Phe His Ala Asp 500 505 510Ile Cys Thr Leu Ser Glu Lys Glu Arg Gln Ile Lys Lys Gln Thr Ala 515 520 525Leu Val Glu Leu Val Lys His Lys Pro Lys Ala Thr Lys Glu Gln Leu 530 535 540Lys Ala Val Met Asp Asp Phe Ala Ala Phe Val Glu Lys Cys Cys Lys545 550 555 560Ala Asp Asp Lys Glu Thr Cys Phe Ala Glu Glu Gly Lys Lys Leu Val 565 570 575Ala Ala Ser Gln Ala Ala Leu Gly Leu Pro Gly Pro Pro Pro Gly Pro 580 585 590Pro Arg Val Ser Pro Asp Pro Arg Ala Glu Leu Asp Ser Thr Val Leu 595 600 605Leu Thr Arg Ser Leu Leu Ala Asp Thr Arg Gln Leu Ala Ala Gln Leu 610 615 620Arg Asp Lys Phe Pro Ala Asp Gly Asp His Asn Leu Asp Ser Leu Pro625 630 635 640Thr Leu Ala Met Ser Ala Gly Ala Leu Gly Ala Leu Gln Leu Pro Gly 645 650 655Val Leu Thr Arg Leu Arg Ala Asp Leu Leu Ser Tyr Leu Arg His Val 660 665 670Gln Trp Leu Arg Arg Ala Gly Gly Ser Ser Leu Lys Thr Leu Glu Pro 675 680 685Glu Leu Gly Thr Leu Gln Ala Arg Leu Asp Arg Leu Leu Arg Arg Leu 690 695 700Gln Leu Leu Met Ser Arg Leu Ala Leu Pro Gln Pro Pro Pro Asp Pro705 710 715 720Pro Ala Pro Pro Leu Ala Pro Pro Ser Ser Ala Trp Gly Gly Ile Arg 725 730 735Ala Ala His Ala Ile Leu Gly Gly Leu His Leu Thr Leu Asp Trp Ala 740 745 750Val Arg Gly Leu Leu Leu Leu Lys Thr Arg Leu 755 76032229DNAArtificial SequenceDNA of HSA-hIL-3 3atgaagtggg taacctttat ttcccttctt tttctcttta gctcggctta ttccaggggt 60gtgtttcgtc gagatgcaca caagagtgag gttgctcatc ggtttaaaga tttgggagaa 120gaaaatttca aagccttggt gttgattgcc tttgctcagt atcttcagca gtgtccattt 180gaagatcatg taaaattagt gaatgaagta actgaatttg caaaaacatg tgttgctgat 240gagtcagctg aaaattgtga caaatcactt catacccttt ttggagacaa attatgcaca 300gttgcaactc ttcgtgaaac ctatggtgaa atggctgact gctgtgcaaa acaagaacct 360gagagaaatg aatgcttctt gcaacacaaa gatgacaacc caaacctccc ccgattggtg 420agaccagagg ttgatgtgat gtgcactgct tttcatgaca atgaagagac atttttgaaa 480aaatacttat atgaaattgc cagaagacat ccttactttt atgccccgga actccttttc 540tttgctaaaa ggtataaagc tgcttttaca gaatgttgcc aagctgctga taaagctgcc 600tgcctgttgc caaagctcga tgaacttcgg gatgaaggga aggcttcgtc tgccaaacag 660agactcaagt gtgccagtct ccaaaaattt ggagaaagag ctttcaaagc atgggcagta 720gctcgcctga gccagagatt tcccaaagct gagtttgcag aagtttccaa gttagtgaca 780gatcttacca aagtccacac ggaatgctgc catggagatc tgcttgaatg tgctgatgac 840agggcggacc ttgccaagta tatctgtgaa aatcaagatt cgatctccag taaactgaag 900gaatgctgtg aaaaacctct gttggaaaaa tcccactgca ttgccgaagt ggaaaatgat 960gagatgcctg ctgacttgcc ttcattagct gctgattttg ttgaaagtaa ggatgtttgc 1020aaaaactatg ctgaggcaaa ggatgtcttc ctgggcatgt ttttgtatga atatgcaaga 1080aggcatcctg attactctgt cgtgctgctg ctgagacttg ccaagacata tgaaaccact 1140ctagagaagt gctgtgccgc tgcagatcct catgaatgct atgccaaagt gttcgatgaa 1200tttaaacctc ttgtggaaga gcctcagaat ttaatcaaac aaaattgtga gctttttgag 1260cagcttggag agtacaaatt ccagaatgcg ctattagttc gttacaccaa gaaagtaccc 1320caagtgtcaa ctccaactct tgtagaggtc tcaagaaacc taggaaaagt gggcagcaaa 1380tgttgtaaac atcctgaagc aaaaagaatg ccctgtgcag aagactatct atccgtggtc 1440ctgaaccagt tatgtgtgtt gcatgagaaa acgccagtaa gtgacagagt caccaaatgc 1500tgcacagaat ccttggtgaa caggcgacca tgcttttcag ctctggaagt cgatgaaaca 1560tacgttccca aagagtttaa tgctgaaaca ttcaccttcc atgcagatat atgcacactt 1620tctgagaagg agagacaaat caagaaacaa actgcacttg ttgagcttgt gaaacacaag 1680cccaaggcaa caaaagagca actgaaagct gttatggatg atttcgcagc ttttgtagag 1740aagtgctgca aggctgacga taaggagacc tgctttgccg aggagggtaa aaaacttgtt 1800gctgcaagtc aagctgcctt aggcttagct cccatgaccc agacaacgtc cttgaagaca 1860agctgggtta actgctctaa catgatcgat gaaattataa cacacttaaa gcagccacct 1920ttgcctttgc tggacttcaa caacctcaat ggggaagacc aagacattct gatggaaaat 1980aaccttcgaa ggccaaacct ggaggcattc aacagggctg tcaagagttt acagaacgca 2040tcagcaattg agagcattct taaaaatctc ctgccatgtc tgcccctggc cacggccgca 2100cccacgcgac atccaatcca tatcaaggac ggtgactgga atgaattccg gaggaaactg 2160acgttctatc tgaaaaccct tgagaatgcg caggctcaac agacgacttt gagcctcgcg 2220atcttttag 22294718PRTArtificial SequenceHSA-hIL-3 4Asp Ala His Lys Ser Glu Val Ala His Arg Phe Lys Asp Leu Gly Glu1 5 10 15Glu Asn Phe Lys Ala Leu Val Leu Ile Ala Phe Ala Gln Tyr Leu Gln 20 25 30Gln Cys Pro Phe Glu Asp His Val Lys Leu Val Asn Glu Val Thr Glu 35 40 45Phe Ala Lys Thr Cys Val Ala Asp Glu Ser Ala Glu Asn Cys Asp Lys 50 55 60Ser Leu His Thr Leu Phe Gly Asp Lys Leu Cys Thr Val Ala Thr Leu65 70 75 80Arg Glu Thr Tyr Gly Glu Met Ala Asp Cys Cys Ala Lys Gln Glu Pro 85 90 95Glu Arg Asn Glu Cys Phe Leu Gln His Lys Asp Asp Asn Pro Asn Leu 100 105 110Pro Arg Leu Val Arg Pro Glu Val Asp Val Met Cys Thr Ala Phe His 115 120 125Asp Asn Glu Glu Thr Phe Leu Lys Lys Tyr Leu Tyr Glu Ile Ala Arg 130 135 140Arg His Pro Tyr Phe Tyr Ala Pro Glu Leu Leu Phe Phe Ala Lys Arg145 150 155 160Tyr Lys Ala Ala Phe Thr Glu Cys Cys Gln Ala Ala Asp Lys Ala Ala 165 170 175Cys Leu Leu Pro Lys Leu Asp Glu Leu Arg Asp Glu Gly Lys Ala Ser 180 185 190Ser Ala Lys Gln Arg Leu Lys Cys Ala Ser Leu Gln Lys Phe Gly Glu 195 200 205Arg Ala Phe Lys Ala Trp Ala Val Ala Arg Leu Ser Gln Arg Phe Pro 210 215 220Lys Ala Glu Phe Ala Glu Val Ser Lys Leu Val Thr Asp Leu Thr Lys225 230 235 240Val His Thr Glu Cys Cys His Gly Asp Leu Leu Glu Cys Ala Asp Asp 245 250 255Arg Ala Asp Leu Ala Lys Tyr Ile Cys Glu Asn Gln Asp Ser Ile Ser 260 265 270Ser Lys Leu Lys Glu Cys Cys Glu Lys Pro Leu Leu Glu Lys Ser His 275 280 285Cys Ile Ala Glu Val Glu Asn Asp Glu Met Pro Ala Asp Leu Pro Ser 290 295 300Leu Ala Ala Asp Phe Val Glu Ser Lys Asp Val Cys Lys Asn Tyr Ala305 310 315 320Glu Ala Lys Asp Val Phe Leu Gly Met Phe Leu Tyr Glu Tyr Ala Arg 325 330 335Arg His Pro Asp Tyr Ser Val Val Leu Leu Leu Arg Leu Ala Lys Thr 340 345 350Tyr Glu Thr Thr Leu Glu Lys Cys Cys Ala Ala Ala Asp Pro His Glu 355 360 365Cys Tyr Ala Lys Val Phe Asp Glu Phe Lys Pro Leu Val Glu Glu Pro 370 375 380Gln Asn Leu Ile Lys Gln Asn Cys Glu Leu Phe Glu Gln Leu Gly Glu385 390 395 400Tyr Lys Phe Gln Asn Ala Leu Leu Val Arg Tyr Thr Lys Lys Val Pro 405 410 415Glu Val Ser Thr Pro Thr Leu Val Glu Val Ser Arg Asn Leu Gly Lys 420 425 430Val Gly Ser Lys Cys Cys Lys His Pro Glu Ala Lys Arg Met Pro Cys 435 440 445Ala Glu Asp Tyr Leu Ser Val Val Leu Asn Gln Leu Cys Val Leu His 450 455 460Glu Lys Thr Pro Val Ser Asp Arg Val Thr Lys Cys Cys Thr Glu Ser465 470 475 480Leu Val Asn Arg Arg Pro Cys Phe Ser Ala Leu Glu Val Asp Glu Thr 485 490 495Tyr Val Pro Lys Glu Phe Asn Ala Glu Thr Phe Thr Phe His Ala Asp 500 505 510Ile Cys Thr Leu Ser Glu Lys Glu Arg Gln Ile Lys Lys Gln Thr Ala 515 520 525Leu Val Glu Leu Val Lys His Lys Pro Lys Ala Thr Lys Glu Gln Leu 530 535 540Lys Ala Val Met Asp Asp Phe Ala Ala Phe Val Glu Lys Cys Cys Lys545 550 555 560Ala Asp Asp Lys Glu Thr Cys Phe Ala Glu Glu Gly Lys Lys Leu Val 565 570 575Ala Ala Ser Gln Ala Ala Leu Gly Leu Ala Pro Met Thr Gln Thr Thr 580 585 590Ser Leu Lys Thr Ser Trp Val Asn Cys Ser Asn Met Ile Asp Glu Ile 595 600 605Ile Thr His Leu Lys Gln Pro Pro Leu Pro Leu Leu Asp Phe Asn Asn 610 615 620Leu Asn Gly Glu Asp Gln Asp Ile Leu Met Glu Asn Asn Leu Arg Arg625 630 635 640Pro Asn Leu Glu Ala Phe Asn Arg Ala Val Lys Ser Leu Gln Asn Ala 645 650 655Ser Ala Ile Glu Ser Ile Leu Lys Asn Leu Leu Pro Cys Leu Pro Leu 660 665 670Ala Thr Ala Ala Pro Thr Arg His Pro Ile His Ile Lys Asp Gly Asp 675 680 685Trp Asn Glu Phe Arg Arg Lys Leu Thr Phe Tyr Leu Lys Thr Leu Glu 690 695 700Asn Ala Gln Ala Gln Gln Thr Thr Leu Ser Leu Ala Ile Phe705 710 71552313DNAArtificial SequenceDNA of HSA-hEPO 5atgaagtggg taacctttat ttcccttctt tttctcttta gctcggctta ttccaggggt 60gtgtttcgtc gagatgcaca caagagtgag gttgctcatc ggtttaaaga tttgggagaa 120gaaaatttca aagccttggt gttgattgcc tttgctcagt atcttcagca gtgtccattt 180gaagatcatg taaaattagt gaatgaagta actgaatttg caaaaacatg tgttgctgat 240gagtcagctg aaaattgtga caaatcactt catacccttt ttggagacaa attatgcaca 300gttgcaactc ttcgtgaaac ctatggtgaa atggctgact gctgtgcaaa acaagaacct 360gagagaaatg aatgcttctt gcaacacaaa gatgacaacc caaacctccc ccgattggtg 420agaccagagg ttgatgtgat gtgcactgct tttcatgaca atgaagagac atttttgaaa 480aaatacttat atgaaattgc cagaagacat ccttactttt atgccccgga actccttttc 540tttgctaaaa ggtataaagc tgcttttaca gaatgttgcc aagctgctga taaagctgcc 600tgcctgttgc caaagctcga tgaacttcgg gatgaaggga aggcttcgtc tgccaaacag 660agactcaagt gtgccagtct ccaaaaattt ggagaaagag ctttcaaagc atgggcagta 720gctcgcctga gccagagatt tcccaaagct gagtttgcag aagtttccaa gttagtgaca 780gatcttacca aagtccacac ggaatgctgc catggagatc tgcttgaatg tgctgatgac 840agggcggacc ttgccaagta tatctgtgaa aatcaagatt cgatctccag taaactgaag 900gaatgctgtg aaaaacctct gttggaaaaa tcccactgca ttgccgaagt ggaaaatgat 960gagatgcctg ctgacttgcc ttcattagct gctgattttg ttgaaagtaa ggatgtttgc 1020aaaaactatg ctgaggcaaa ggatgtcttc ctgggcatgt ttttgtatga atatgcaaga 1080aggcatcctg attactctgt cgtgctgctg ctgagacttg ccaagacata tgaaaccact 1140ctagagaagt gctgtgccgc tgcagatcct catgaatgct atgccaaagt gttcgatgaa 1200tttaaacctc ttgtggaaga gcctcagaat ttaatcaaac aaaattgtga gctttttgag 1260cagcttggag agtacaaatt ccagaatgcg ctattagttc gttacaccaa gaaagtaccc

1320caagtgtcaa ctccaactct tgtagaggtc tcaagaaacc taggaaaagt gggcagcaaa 1380tgttgtaaac atcctgaagc aaaaagaatg ccctgtgcag aagactatct atccgtggtc 1440ctgaaccagt tatgtgtgtt gcatgagaaa acgccagtaa gtgacagagt caccaaatgc 1500tgcacagaat ccttggtgaa caggcgacca tgcttttcag ctctggaagt cgatgaaaca 1560tacgttccca aagagtttaa tgctgaaaca ttcaccttcc atgcagatat atgcacactt 1620tctgagaagg agagacaaat caagaaacaa actgcacttg ttgagcttgt gaaacacaag 1680cccaaggcaa caaaagagca actgaaagct gttatggatg atttcgcagc ttttgtagag 1740aagtgctgca aggctgacga taaggagacc tgctttgccg aggagggtaa aaaacttgtt 1800gctgcaagtc aagctgcctt aggcttaatc tgtgacagcc gagtcctgga gaggtacctc 1860ttggaggcca aggaggccga gaatatcacg acgggctgtg ctgaacactg cagcttgaat 1920gagaatatca ctgtcccaga caccaaagtt aatttctatg cctggaagag gatggaggtc 1980gggcagcagg ccgtagaagt ctggcagggc ctggccctgc tgtcggaagc tgtcctgcgg 2040ggccaggccc tgttggtcaa ctcttcccag ccgtgggagc ccctgcagct gcatgtggat 2100aaagccgtca gtggccttcg cagcctcacc actctgcttc gggctctgcg agcccagaag 2160gaagccatct cccctccaga tgcggcctca gctgctccac tccgaacaat cactgctgac 2220actttccgca aactcttccg agtctactcc aatttcctcc ggggaaagct gaagctgtac 2280acaggggagg cctgcaggac aggggacaga tga 23136746PRTArtificial SequenceHSA-hEPO 6Asp Ala His Lys Ser Glu Val Ala His Arg Phe Lys Asp Leu Gly Glu1 5 10 15Glu Asn Phe Lys Ala Leu Val Leu Ile Ala Phe Ala Gln Tyr Leu Gln 20 25 30Gln Cys Pro Phe Glu Asp His Val Lys Leu Val Asn Glu Val Thr Glu 35 40 45Phe Ala Lys Thr Cys Val Ala Asp Glu Ser Ala Glu Asn Cys Asp Lys 50 55 60Ser Leu His Thr Leu Phe Gly Asp Lys Leu Cys Thr Val Ala Thr Leu65 70 75 80Arg Glu Thr Tyr Gly Glu Met Ala Asp Cys Cys Ala Lys Gln Glu Pro 85 90 95Glu Arg Asn Glu Cys Phe Leu Gln His Lys Asp Asp Asn Pro Asn Leu 100 105 110Pro Arg Leu Val Arg Pro Glu Val Asp Val Met Cys Thr Ala Phe His 115 120 125Asp Asn Glu Glu Thr Phe Leu Lys Lys Tyr Leu Tyr Glu Ile Ala Arg 130 135 140Arg His Pro Tyr Phe Tyr Ala Pro Glu Leu Leu Phe Phe Ala Lys Arg145 150 155 160Tyr Lys Ala Ala Phe Thr Glu Cys Cys Gln Ala Ala Asp Lys Ala Ala 165 170 175Cys Leu Leu Pro Lys Leu Asp Glu Leu Arg Asp Glu Gly Lys Ala Ser 180 185 190Ser Ala Lys Gln Arg Leu Lys Cys Ala Ser Leu Gln Lys Phe Gly Glu 195 200 205Arg Ala Phe Lys Ala Trp Ala Val Ala Arg Leu Ser Gln Arg Phe Pro 210 215 220Lys Ala Glu Phe Ala Glu Val Ser Lys Leu Val Thr Asp Leu Thr Lys225 230 235 240Val His Thr Glu Cys Cys His Gly Asp Leu Leu Glu Cys Ala Asp Asp 245 250 255Arg Ala Asp Leu Ala Lys Tyr Ile Cys Glu Asn Gln Asp Ser Ile Ser 260 265 270Ser Lys Leu Lys Glu Cys Cys Glu Lys Pro Leu Leu Glu Lys Ser His 275 280 285Cys Ile Ala Glu Val Glu Asn Asp Glu Met Pro Ala Asp Leu Pro Ser 290 295 300Leu Ala Ala Asp Phe Val Glu Ser Lys Asp Val Cys Lys Asn Tyr Ala305 310 315 320Glu Ala Lys Asp Val Phe Leu Gly Met Phe Leu Tyr Glu Tyr Ala Arg 325 330 335Arg His Pro Asp Tyr Ser Val Val Leu Leu Leu Arg Leu Ala Lys Thr 340 345 350Tyr Glu Thr Thr Leu Glu Lys Cys Cys Ala Ala Ala Asp Pro His Glu 355 360 365Cys Tyr Ala Lys Val Phe Asp Glu Phe Lys Pro Leu Val Glu Glu Pro 370 375 380Gln Asn Leu Ile Lys Gln Asn Cys Glu Leu Phe Glu Gln Leu Gly Glu385 390 395 400Tyr Lys Phe Gln Asn Ala Leu Leu Val Arg Tyr Thr Lys Lys Val Pro 405 410 415Glu Val Ser Thr Pro Thr Leu Val Glu Val Ser Arg Asn Leu Gly Lys 420 425 430Val Gly Ser Lys Cys Cys Lys His Pro Glu Ala Lys Arg Met Pro Cys 435 440 445Ala Glu Asp Tyr Leu Ser Val Val Leu Asn Gln Leu Cys Val Leu His 450 455 460Glu Lys Thr Pro Val Ser Asp Arg Val Thr Lys Cys Cys Thr Glu Ser465 470 475 480Leu Val Asn Arg Arg Pro Cys Phe Ser Ala Leu Glu Val Asp Glu Thr 485 490 495Tyr Val Pro Lys Glu Phe Asn Ala Glu Thr Phe Thr Phe His Ala Asp 500 505 510Ile Cys Thr Leu Ser Glu Lys Glu Arg Gln Ile Lys Lys Gln Thr Ala 515 520 525Leu Val Glu Leu Val Lys His Lys Pro Lys Ala Thr Lys Glu Gln Leu 530 535 540Lys Ala Val Met Asp Asp Phe Ala Ala Phe Val Glu Lys Cys Cys Lys545 550 555 560Ala Asp Asp Lys Glu Thr Cys Phe Ala Glu Glu Gly Lys Lys Leu Val 565 570 575Ala Ala Ser Gln Ala Ala Leu Gly Leu Ile Cys Asp Ser Arg Val Leu 580 585 590Glu Arg Tyr Leu Leu Glu Ala Lys Glu Ala Glu Asn Ile Thr Thr Gly 595 600 605Cys Ala Glu His Cys Ser Leu Asn Glu Asn Ile Thr Val Pro Asp Thr 610 615 620Lys Val Asn Phe Tyr Ala Trp Lys Arg Met Glu Val Gly Gln Gln Ala625 630 635 640Val Glu Val Trp Gln Gly Leu Ala Leu Leu Ser Glu Ala Val Leu Arg 645 650 655Gly Gln Ala Leu Leu Val Asn Ser Ser Gln Pro Trp Glu Pro Leu Gln 660 665 670Leu His Val Asp Lys Ala Val Ser Gly Leu Arg Ser Leu Thr Thr Leu 675 680 685Leu Arg Ala Leu Arg Ala Gln Lys Glu Ala Ile Ser Pro Pro Asp Ala 690 695 700Ala Ser Ala Ala Pro Leu Arg Thr Ile Thr Ala Asp Thr Phe Arg Lys705 710 715 720Leu Phe Arg Val Tyr Ser Asn Phe Leu Arg Gly Lys Leu Lys Leu Tyr 725 730 735Thr Gly Glu Ala Cys Arg Thr Gly Asp Arg 740 74572352DNAArtificial SequenceDNA of HSA-GCSF 7atgaagtggg taacctttat ttcccttctt tttctcttta gctcggctta ttccaggggt 60gtgtttcgtc gagatgcaca caagagtgag gttgctcatc ggtttaaaga tttgggagaa 120gaaaatttca aagccttggt gttgattgcc tttgctcagt atcttcagca gtgtccattt 180gaagatcatg taaaattagt gaatgaagta actgaatttg caaaaacatg tgttgctgat 240gagtcagctg aaaattgtga caaatcactt catacccttt ttggagacaa attatgcaca 300gttgcaactc ttcgtgaaac ctatggtgaa atggctgact gctgtgcaaa acaagaacct 360gagagaaatg aatgcttctt gcaacacaaa gatgacaacc caaacctccc ccgattggtg 420agaccagagg ttgatgtgat gtgcactgct tttcatgaca atgaagagac atttttgaaa 480aaatacttat atgaaattgc cagaagacat ccttactttt atgccccgga actccttttc 540tttgctaaaa ggtataaagc tgcttttaca gaatgttgcc aagctgctga taaagctgcc 600tgcctgttgc caaagctcga tgaacttcgg gatgaaggga aggcttcgtc tgccaaacag 660agactcaagt gtgccagtct ccaaaaattt ggagaaagag ctttcaaagc atgggcagta 720gctcgcctga gccagagatt tcccaaagct gagtttgcag aagtttccaa gttagtgaca 780gatcttacca aagtccacac ggaatgctgc catggagatc tgcttgaatg tgctgatgac 840agggcggacc ttgccaagta tatctgtgaa aatcaagatt cgatctccag taaactgaag 900gaatgctgtg aaaaacctct gttggaaaaa tcccactgca ttgccgaagt ggaaaatgat 960gagatgcctg ctgacttgcc ttcattagct gctgattttg ttgaaagtaa ggatgtttgc 1020aaaaactatg ctgaggcaaa ggatgtcttc ctgggcatgt ttttgtatga atatgcaaga 1080aggcatcctg attactctgt cgtgctgctg ctgagacttg ccaagacata tgaaaccact 1140ctagagaagt gctgtgccgc tgcagatcct catgaatgct atgccaaagt gttcgatgaa 1200tttaaacctc ttgtggaaga gcctcagaat ttaatcaaac aaaattgtga gctttttgag 1260cagcttggag agtacaaatt ccagaatgcg ctattagttc gttacaccaa gaaagtaccc 1320caagtgtcaa ctccaactct tgtagaggtc tcaagaaacc taggaaaagt gggcagcaaa 1380tgttgtaaac atcctgaagc aaaaagaatg ccctgtgcag aagactatct atccgtggtc 1440ctgaaccagt tatgtgtgtt gcatgagaaa acgccagtaa gtgacagagt caccaaatgc 1500tgcacagaat ccttggtgaa caggcgacca tgcttttcag ctctggaagt cgatgaaaca 1560tacgttccca aagagtttaa tgctgaaaca ttcaccttcc atgcagatat atgcacactt 1620tctgagaagg agagacaaat caagaaacaa actgcacttg ttgagcttgt gaaacacaag 1680cccaaggcaa caaaagagca actgaaagct gttatggatg atttcgcagc ttttgtagag 1740aagtgctgca aggctgacga taaggagacc tgctttgccg aggagggtaa aaaacttgtt 1800gctgcaagtc aagctgcctt aggcttaacc cccctgggcc ctgccagctc cctgccccag 1860agcttcctgc tcaagtgctt agagcaagtg aggaagatcc agggcgatgg cgcagcgctc 1920caggagaagc tgtgtgccac ctacaagctg tgccaccccg aggagctggt gctgctcgga 1980cactctctgg gcatcccctg ggctcccctg agcagctgcc ccagccaggc cctgcagctg 2040gcaggctgct tgagccaact ccatagcggc cttttcctct accaggggct cctgcaggcc 2100ctggaaggga tctcccccga gttgggtccc accttggaca cactgcagct ggacgtcgcc 2160gactttgcca ccaccatctg gcagcagatg gaagaactgg gaatggcccc tgccctgcag 2220cccacccagg gtgccatgcc ggccttcgcc tctgctttcc agcgccgggc aggaggggtc 2280ctagttgcct cccatctgca gagcttcctg gaggtgtcgt accgcgttct acgccacctt 2340gcccagccct ga 23528759PRTArtificial SequenceHSA-GCSF 8Asp Ala His Lys Ser Glu Val Ala His Arg Phe Lys Asp Leu Gly Glu1 5 10 15Glu Asn Phe Lys Ala Leu Val Leu Ile Ala Phe Ala Gln Tyr Leu Gln 20 25 30Gln Cys Pro Phe Glu Asp His Val Lys Leu Val Asn Glu Val Thr Glu 35 40 45Phe Ala Lys Thr Cys Val Ala Asp Glu Ser Ala Glu Asn Cys Asp Lys 50 55 60Ser Leu His Thr Leu Phe Gly Asp Lys Leu Cys Thr Val Ala Thr Leu65 70 75 80Arg Glu Thr Tyr Gly Glu Met Ala Asp Cys Cys Ala Lys Gln Glu Pro 85 90 95Glu Arg Asn Glu Cys Phe Leu Gln His Lys Asp Asp Asn Pro Asn Leu 100 105 110Pro Arg Leu Val Arg Pro Glu Val Asp Val Met Cys Thr Ala Phe His 115 120 125Asp Asn Glu Glu Thr Phe Leu Lys Lys Tyr Leu Tyr Glu Ile Ala Arg 130 135 140Arg His Pro Tyr Phe Tyr Ala Pro Glu Leu Leu Phe Phe Ala Lys Arg145 150 155 160Tyr Lys Ala Ala Phe Thr Glu Cys Cys Gln Ala Ala Asp Lys Ala Ala 165 170 175Cys Leu Leu Pro Lys Leu Asp Glu Leu Arg Asp Glu Gly Lys Ala Ser 180 185 190Ser Ala Lys Gln Arg Leu Lys Cys Ala Ser Leu Gln Lys Phe Gly Glu 195 200 205Arg Ala Phe Lys Ala Trp Ala Val Ala Arg Leu Ser Gln Arg Phe Pro 210 215 220Lys Ala Glu Phe Ala Glu Val Ser Lys Leu Val Thr Asp Leu Thr Lys225 230 235 240Val His Thr Glu Cys Cys His Gly Asp Leu Leu Glu Cys Ala Asp Asp 245 250 255Arg Ala Asp Leu Ala Lys Tyr Ile Cys Glu Asn Gln Asp Ser Ile Ser 260 265 270Ser Lys Leu Lys Glu Cys Cys Glu Lys Pro Leu Leu Glu Lys Ser His 275 280 285Cys Ile Ala Glu Val Glu Asn Asp Glu Met Pro Ala Asp Leu Pro Ser 290 295 300Leu Ala Ala Asp Phe Val Glu Ser Lys Asp Val Cys Lys Asn Tyr Ala305 310 315 320Glu Ala Lys Asp Val Phe Leu Gly Met Phe Leu Tyr Glu Tyr Ala Arg 325 330 335Arg His Pro Asp Tyr Ser Val Val Leu Leu Leu Arg Leu Ala Lys Thr 340 345 350Tyr Glu Thr Thr Leu Glu Lys Cys Cys Ala Ala Ala Asp Pro His Glu 355 360 365Cys Tyr Ala Lys Val Phe Asp Glu Phe Lys Pro Leu Val Glu Glu Pro 370 375 380Gln Asn Leu Ile Lys Gln Asn Cys Glu Leu Phe Glu Gln Leu Gly Glu385 390 395 400Tyr Lys Phe Gln Asn Ala Leu Leu Val Arg Tyr Thr Lys Lys Val Pro 405 410 415Glu Val Ser Thr Pro Thr Leu Val Glu Val Ser Arg Asn Leu Gly Lys 420 425 430Val Gly Ser Lys Cys Cys Lys His Pro Glu Ala Lys Arg Met Pro Cys 435 440 445Ala Glu Asp Tyr Leu Ser Val Val Leu Asn Gln Leu Cys Val Leu His 450 455 460Glu Lys Thr Pro Val Ser Asp Arg Val Thr Lys Cys Cys Thr Glu Ser465 470 475 480Leu Val Asn Arg Arg Pro Cys Phe Ser Ala Leu Glu Val Asp Glu Thr 485 490 495Tyr Val Pro Lys Glu Phe Asn Ala Glu Thr Phe Thr Phe His Ala Asp 500 505 510Ile Cys Thr Leu Ser Glu Lys Glu Arg Gln Ile Lys Lys Gln Thr Ala 515 520 525Leu Val Glu Leu Val Lys His Lys Pro Lys Ala Thr Lys Glu Gln Leu 530 535 540Lys Ala Val Met Asp Asp Phe Ala Ala Phe Val Glu Lys Cys Cys Lys545 550 555 560Ala Asp Asp Lys Glu Thr Cys Phe Ala Glu Glu Gly Lys Lys Leu Val 565 570 575Ala Ala Ser Gln Ala Ala Leu Gly Leu Thr Pro Leu Gly Pro Ala Ser 580 585 590Ser Leu Pro Gln Ser Phe Leu Leu Lys Cys Leu Glu Gln Val Arg Lys 595 600 605Ile Gln Gly Asp Gly Ala Ala Leu Gln Glu Lys Leu Cys Ala Thr Tyr 610 615 620Lys Leu Cys His Pro Glu Glu Leu Val Leu Leu Gly His Ser Leu Gly625 630 635 640Ile Pro Trp Ala Pro Leu Ser Ser Cys Pro Ser Gln Ala Leu Gln Leu 645 650 655Ala Gly Cys Leu Ser Gln Leu His Ser Gly Leu Phe Leu Tyr Gln Gly 660 665 670Leu Leu Gln Ala Leu Glu Gly Ile Ser Pro Glu Leu Gly Pro Thr Leu 675 680 685Asp Thr Leu Gln Leu Asp Val Ala Asp Phe Ala Thr Thr Ile Trp Gln 690 695 700Gln Met Glu Glu Leu Gly Met Ala Pro Ala Leu Gln Pro Thr Gln Gly705 710 715 720Ala Met Pro Ala Phe Ala Ser Ala Phe Gln Arg Arg Ala Gly Gly Val 725 730 735Leu Val Ala Ser His Leu Gln Ser Phe Leu Glu Val Ser Tyr Arg Val 740 745 750Leu Arg His Leu Ala Gln Pro 75592211DNAArtificial SequenceDNA of HSA-GMCSF 9atgaagtggg taacctttat ttcccttctt tttctcttta gctcggctta ttccaggggt 60gtgtttcgtc gagatgcaca caagagtgag gttgctcatc ggtttaaaga tttgggagaa 120gaaaatttca aagccttggt gttgattgcc tttgctcagt atcttcagca gtgtccattt 180gaagatcatg taaaattagt gaatgaagta actgaatttg caaaaacatg tgttgctgat 240gagtcagctg aaaattgtga caaatcactt catacccttt ttggagacaa attatgcaca 300gttgcaactc ttcgtgaaac ctatggtgaa atggctgact gctgtgcaaa acaagaacct 360gagagaaatg aatgcttctt gcaacacaaa gatgacaacc caaacctccc ccgattggtg 420agaccagagg ttgatgtgat gtgcactgct tttcatgaca atgaagagac atttttgaaa 480aaatacttat atgaaattgc cagaagacat ccttactttt atgccccgga actccttttc 540tttgctaaaa ggtataaagc tgcttttaca gaatgttgcc aagctgctga taaagctgcc 600tgcctgttgc caaagctcga tgaacttcgg gatgaaggga aggcttcgtc tgccaaacag 660agactcaagt gtgccagtct ccaaaaattt ggagaaagag ctttcaaagc atgggcagta 720gctcgcctga gccagagatt tcccaaagct gagtttgcag aagtttccaa gttagtgaca 780gatcttacca aagtccacac ggaatgctgc catggagatc tgcttgaatg tgctgatgac 840agggcggacc ttgccaagta tatctgtgaa aatcaagatt cgatctccag taaactgaag 900gaatgctgtg aaaaacctct gttggaaaaa tcccactgca ttgccgaagt ggaaaatgat 960gagatgcctg ctgacttgcc ttcattagct gctgattttg ttgaaagtaa ggatgtttgc 1020aaaaactatg ctgaggcaaa ggatgtcttc ctgggcatgt ttttgtatga atatgcaaga 1080aggcatcctg attactctgt cgtgctgctg ctgagacttg ccaagacata tgaaaccact 1140ctagagaagt gctgtgccgc tgcagatcct catgaatgct atgccaaagt gttcgatgaa 1200tttaaacctc ttgtggaaga gcctcagaat ttaatcaaac aaaattgtga gctttttgag 1260cagcttggag agtacaaatt ccagaatgcg ctattagttc gttacaccaa gaaagtaccc 1320caagtgtcaa ctccaactct tgtagaggtc tcaagaaacc taggaaaagt gggcagcaaa 1380tgttgtaaac atcctgaagc aaaaagaatg ccctgtgcag aagactatct atccgtggtc 1440ctgaaccagt tatgtgtgtt gcatgagaaa acgccagtaa gtgacagagt caccaaatgc 1500tgcacagaat ccttggtgaa caggcgacca tgcttttcag ctctggaagt cgatgaaaca 1560tacgttccca aagagtttaa tgctgaaaca ttcaccttcc atgcagatat atgcacactt 1620tctgagaagg agagacaaat caagaaacaa actgcacttg ttgagcttgt gaaacacaag 1680cccaaggcaa caaaagagca actgaaagct gttatggatg atttcgcagc ttttgtagag 1740aagtgctgca aggctgacga taaggagacc tgctttgccg aggagggtaa aaaacttgtt 1800gctgcaagtc aagctgcctt aggcttagca cccgcccgct cgcccagccc cagcacgcag 1860ccctgggagc atgtgaatgc catccaggag gcccggcgtc tcctgaacct gagtagagac 1920actgctgctg agatgaatga aacagtagaa gtcatctcag aaatgtttga cctccaggag 1980ccgacctgcc tacagacccg cctggagctg tacaagcagg gcctgcgggg cagcctcacc 2040aagctcaagg gccccttgac catgatggcc agccactaca agcagcactg ccctccaacc 2100ccggaaactt cctgtgcaac ccagattatc acctttgaaa gtttcaaaga gaacctgaag 2160gactttctgc ttgtcatccc ctttgactgc tgggagccag tccaggagtg a 221110712PRTArtificial SequenceHSA-GMCSF 10Asp Ala His Lys Ser Glu Val Ala His Arg Phe Lys Asp Leu Gly Glu1 5 10 15Glu

Asn Phe Lys Ala Leu Val Leu Ile Ala Phe Ala Gln Tyr Leu Gln 20 25 30Gln Cys Pro Phe Glu Asp His Val Lys Leu Val Asn Glu Val Thr Glu 35 40 45Phe Ala Lys Thr Cys Val Ala Asp Glu Ser Ala Glu Asn Cys Asp Lys 50 55 60Ser Leu His Thr Leu Phe Gly Asp Lys Leu Cys Thr Val Ala Thr Leu65 70 75 80Arg Glu Thr Tyr Gly Glu Met Ala Asp Cys Cys Ala Lys Gln Glu Pro 85 90 95Glu Arg Asn Glu Cys Phe Leu Gln His Lys Asp Asp Asn Pro Asn Leu 100 105 110Pro Arg Leu Val Arg Pro Glu Val Asp Val Met Cys Thr Ala Phe His 115 120 125Asp Asn Glu Glu Thr Phe Leu Lys Lys Tyr Leu Tyr Glu Ile Ala Arg 130 135 140Arg His Pro Tyr Phe Tyr Ala Pro Glu Leu Leu Phe Phe Ala Lys Arg145 150 155 160Tyr Lys Ala Ala Phe Thr Glu Cys Cys Gln Ala Ala Asp Lys Ala Ala 165 170 175Cys Leu Leu Pro Lys Leu Asp Glu Leu Arg Asp Glu Gly Lys Ala Ser 180 185 190Ser Ala Lys Gln Arg Leu Lys Cys Ala Ser Leu Gln Lys Phe Gly Glu 195 200 205Arg Ala Phe Lys Ala Trp Ala Val Ala Arg Leu Ser Gln Arg Phe Pro 210 215 220Lys Ala Glu Phe Ala Glu Val Ser Lys Leu Val Thr Asp Leu Thr Lys225 230 235 240Val His Thr Glu Cys Cys His Gly Asp Leu Leu Glu Cys Ala Asp Asp 245 250 255Arg Ala Asp Leu Ala Lys Tyr Ile Cys Glu Asn Gln Asp Ser Ile Ser 260 265 270Ser Lys Leu Lys Glu Cys Cys Glu Lys Pro Leu Leu Glu Lys Ser His 275 280 285Cys Ile Ala Glu Val Glu Asn Asp Glu Met Pro Ala Asp Leu Pro Ser 290 295 300Leu Ala Ala Asp Phe Val Glu Ser Lys Asp Val Cys Lys Asn Tyr Ala305 310 315 320Glu Ala Lys Asp Val Phe Leu Gly Met Phe Leu Tyr Glu Tyr Ala Arg 325 330 335Arg His Pro Asp Tyr Ser Val Val Leu Leu Leu Arg Leu Ala Lys Thr 340 345 350Tyr Glu Thr Thr Leu Glu Lys Cys Cys Ala Ala Ala Asp Pro His Glu 355 360 365Cys Tyr Ala Lys Val Phe Asp Glu Phe Lys Pro Leu Val Glu Glu Pro 370 375 380Gln Asn Leu Ile Lys Gln Asn Cys Glu Leu Phe Glu Gln Leu Gly Glu385 390 395 400Tyr Lys Phe Gln Asn Ala Leu Leu Val Arg Tyr Thr Lys Lys Val Pro 405 410 415Glu Val Ser Thr Pro Thr Leu Val Glu Val Ser Arg Asn Leu Gly Lys 420 425 430Val Gly Ser Lys Cys Cys Lys His Pro Glu Ala Lys Arg Met Pro Cys 435 440 445Ala Glu Asp Tyr Leu Ser Val Val Leu Asn Gln Leu Cys Val Leu His 450 455 460Glu Lys Thr Pro Val Ser Asp Arg Val Thr Lys Cys Cys Thr Glu Ser465 470 475 480Leu Val Asn Arg Arg Pro Cys Phe Ser Ala Leu Glu Val Asp Glu Thr 485 490 495Tyr Val Pro Lys Glu Phe Asn Ala Glu Thr Phe Thr Phe His Ala Asp 500 505 510Ile Cys Thr Leu Ser Glu Lys Glu Arg Gln Ile Lys Lys Gln Thr Ala 515 520 525Leu Val Glu Leu Val Lys His Lys Pro Lys Ala Thr Lys Glu Gln Leu 530 535 540Lys Ala Val Met Asp Asp Phe Ala Ala Phe Val Glu Lys Cys Cys Lys545 550 555 560Ala Asp Asp Lys Glu Thr Cys Phe Ala Glu Glu Gly Lys Lys Leu Val 565 570 575Ala Ala Ser Gln Ala Ala Leu Gly Leu Ala Pro Ala Arg Ser Pro Ser 580 585 590Pro Ser Thr Gln Pro Trp Glu His Val Asn Ala Ile Gln Glu Ala Arg 595 600 605Arg Leu Leu Asn Leu Ser Arg Asp Thr Ala Ala Glu Met Asn Glu Thr 610 615 620Val Glu Val Ile Ser Glu Met Phe Asp Leu Gln Glu Pro Thr Cys Leu625 630 635 640Gln Thr Arg Leu Glu Leu Tyr Lys Gln Gly Leu Arg Gly Ser Leu Thr 645 650 655Lys Leu Lys Gly Pro Leu Thr Met Met Ala Ser His Tyr Lys Gln His 660 665 670Cys Pro Pro Thr Pro Glu Thr Ser Cys Ala Thr Gln Ile Ile Thr Phe 675 680 685Glu Ser Phe Lys Glu Asn Leu Lys Asp Phe Leu Leu Val Ile Pro Phe 690 695 700Asp Cys Trp Glu Pro Val Gln Glu705 710111830DNAHomo sapiens 11atgaagtggg taacctttat ttcccttctt tttctcttta gctcggctta ttccaggggt 60gtgtttcgtc gagatgcaca caagagtgag gttgctcatc ggtttaaaga tttgggagaa 120gaaaatttca aagccttggt gttgattgcc tttgctcagt atcttcagca gtgtccattt 180gaagatcatg taaaattagt gaatgaagta actgaatttg caaaaacatg tgttgctgat 240gagtcagctg aaaattgtga caaatcactt catacccttt ttggagacaa attatgcaca 300gttgcaactc ttcgtgaaac ctatggtgaa atggctgact gctgtgcaaa acaagaacct 360gagagaaatg aatgcttctt gcaacacaaa gatgacaacc caaacctccc ccgattggtg 420agaccagagg ttgatgtgat gtgcactgct tttcatgaca atgaagagac atttttgaaa 480aaatacttat atgaaattgc cagaagacat ccttactttt atgccccgga actccttttc 540tttgctaaaa ggtataaagc tgcttttaca gaatgttgcc aagctgctga taaagctgcc 600tgcctgttgc caaagctcga tgaacttcgg gatgaaggga aggcttcgtc tgccaaacag 660agactcaagt gtgccagtct ccaaaaattt ggagaaagag ctttcaaagc atgggcagta 720gctcgcctga gccagagatt tcccaaagct gagtttgcag aagtttccaa gttagtgaca 780gatcttacca aagtccacac ggaatgctgc catggagatc tgcttgaatg tgctgatgac 840agggcggacc ttgccaagta tatctgtgaa aatcaagatt cgatctccag taaactgaag 900gaatgctgtg aaaaacctct gttggaaaaa tcccactgca ttgccgaagt ggaaaatgat 960gagatgcctg ctgacttgcc ttcattagct gctgattttg ttgaaagtaa ggatgtttgc 1020aaaaactatg ctgaggcaaa ggatgtcttc ctgggcatgt ttttgtatga atatgcaaga 1080aggcatcctg attactctgt cgtgctgctg ctgagacttg ccaagacata tgaaaccact 1140ctagagaagt gctgtgccgc tgcagatcct catgaatgct atgccaaagt gttcgatgaa 1200tttaaacctc ttgtggaaga gcctcagaat ttaatcaaac aaaattgtga gctttttgag 1260cagcttggag agtacaaatt ccagaatgcg ctattagttc gttacaccaa gaaagtaccc 1320caagtgtcaa ctccaactct tgtagaggtc tcaagaaacc taggaaaagt gggcagcaaa 1380tgttgtaaac atcctgaagc aaaaagaatg ccctgtgcag aagactatct atccgtggtc 1440ctgaaccagt tatgtgtgtt gcatgagaaa acgccagtaa gtgacagagt caccaaatgc 1500tgcacagaat ccttggtgaa caggcgacca tgcttttcag ctctggaagt cgatgaaaca 1560tacgttccca aagagtttaa tgctgaaaca ttcaccttcc atgcagatat atgcacactt 1620tctgagaagg agagacaaat caagaaacaa actgcacttg ttgagcttgt gaaacacaag 1680cccaaggcaa caaaagagca actgaaagct gttatggatg atttcgcagc ttttgtagag 1740aagtgctgca aggctgacga taaggagacc tgctttgccg aggagggtaa aaaacttgtt 1800gctgcaagtc aagctgcctt aggcttataa 183012609PRTHomo sapiens 12Met Lys Trp Val Thr Phe Ile Ser Leu Leu Phe Leu Phe Ser Ser Ala1 5 10 15Tyr Ser Arg Gly Val Phe Arg Arg Asp Ala His Lys Ser Glu Val Ala 20 25 30His Arg Phe Lys Asp Leu Gly Glu Glu Asn Phe Lys Ala Leu Val Leu 35 40 45Ile Ala Phe Ala Gln Tyr Leu Gln Gln Cys Pro Phe Glu Asp His Val 50 55 60Lys Leu Val Asn Glu Val Thr Glu Phe Ala Lys Thr Cys Val Ala Asp65 70 75 80Glu Ser Ala Glu Asn Cys Asp Lys Ser Leu His Thr Leu Phe Gly Asp 85 90 95Lys Leu Cys Thr Val Ala Thr Leu Arg Glu Thr Tyr Gly Glu Met Ala 100 105 110Asp Cys Cys Ala Lys Gln Glu Pro Glu Arg Asn Glu Cys Phe Leu Gln 115 120 125His Lys Asp Asp Asn Pro Asn Leu Pro Arg Leu Val Arg Pro Glu Val 130 135 140Asp Val Met Cys Thr Ala Phe His Asp Asn Glu Glu Thr Phe Leu Lys145 150 155 160Lys Tyr Leu Tyr Glu Ile Ala Arg Arg His Pro Tyr Phe Tyr Ala Pro 165 170 175Glu Leu Leu Phe Phe Ala Lys Arg Tyr Lys Ala Ala Phe Thr Glu Cys 180 185 190Cys Gln Ala Ala Asp Lys Ala Ala Cys Leu Leu Pro Lys Leu Asp Glu 195 200 205Leu Arg Asp Glu Gly Lys Ala Ser Ser Ala Lys Gln Arg Leu Lys Cys 210 215 220Ala Ser Leu Gln Lys Phe Gly Glu Arg Ala Phe Lys Ala Trp Ala Val225 230 235 240Ala Arg Leu Ser Gln Arg Phe Pro Lys Ala Glu Phe Ala Glu Val Ser 245 250 255Lys Leu Val Thr Asp Leu Thr Lys Val His Thr Glu Cys Cys His Gly 260 265 270Asp Leu Leu Glu Cys Ala Asp Asp Arg Ala Asp Leu Ala Lys Tyr Ile 275 280 285Cys Glu Asn Gln Asp Ser Ile Ser Ser Lys Leu Lys Glu Cys Cys Glu 290 295 300Lys Pro Leu Leu Glu Lys Ser His Cys Ile Ala Glu Val Glu Asn Asp305 310 315 320Glu Met Pro Ala Asp Leu Pro Ser Leu Ala Ala Asp Phe Val Glu Ser 325 330 335Lys Asp Val Cys Lys Asn Tyr Ala Glu Ala Lys Asp Val Phe Leu Gly 340 345 350Met Phe Leu Tyr Glu Tyr Ala Arg Arg His Pro Asp Tyr Ser Val Val 355 360 365Leu Leu Leu Arg Leu Ala Lys Thr Tyr Glu Thr Thr Leu Glu Lys Cys 370 375 380Cys Ala Ala Ala Asp Pro His Glu Cys Tyr Ala Lys Val Phe Asp Glu385 390 395 400Phe Lys Pro Leu Val Glu Glu Pro Gln Asn Leu Ile Lys Gln Asn Cys 405 410 415Glu Leu Phe Glu Gln Leu Gly Glu Tyr Lys Phe Gln Asn Ala Leu Leu 420 425 430Val Arg Tyr Thr Lys Lys Val Pro Glu Val Ser Thr Pro Thr Leu Val 435 440 445Glu Val Ser Arg Asn Leu Gly Lys Val Gly Ser Lys Cys Cys Lys His 450 455 460Pro Glu Ala Lys Arg Met Pro Cys Ala Glu Asp Tyr Leu Ser Val Val465 470 475 480Leu Asn Gln Leu Cys Val Leu His Glu Lys Thr Pro Val Ser Asp Arg 485 490 495Val Thr Lys Cys Cys Thr Glu Ser Leu Val Asn Arg Arg Pro Cys Phe 500 505 510Ser Ala Leu Glu Val Asp Glu Thr Tyr Val Pro Lys Glu Phe Asn Ala 515 520 525Glu Thr Phe Thr Phe His Ala Asp Ile Cys Thr Leu Ser Glu Lys Glu 530 535 540Arg Gln Ile Lys Lys Gln Thr Ala Leu Val Glu Leu Val Lys His Lys545 550 555 560Pro Lys Ala Thr Lys Glu Gln Leu Lys Ala Val Met Asp Asp Phe Ala 565 570 575Ala Phe Val Glu Lys Cys Cys Lys Ala Asp Asp Lys Glu Thr Cys Phe 580 585 590Ala Glu Glu Gly Lys Lys Leu Val Ala Ala Ser Gln Ala Ala Leu Gly 595 600 605Leu 13600DNAHomo sapiens 13atgaactgtg tttgccgcct ggtcctggtc gtgctgagcc tgtggccaga tacagctgtc 60gcccctgggc caccacctgg cccccctcga gtttccccag accctcgggc cgagctggac 120agcaccgtgc tcctgacccg ctctctcctg gcggacacgc ggcagctggc tgcacagctg 180agggacaaat tcccagctga cggggaccac aacctggatt ccctgcccac cctggccatg 240agtgcggggg cactgggagc tctacagctc ccaggtgtgc tgacaaggct gcgagcggac 300ctactgtcct acctgcggca cgtgcagtgg ctgcgccggg caggtggctc ttccctgaag 360accctggagc ccgagctggg caccctgcag gcccgactgg accggctgct gcgccggctg 420cagctcctga tgtcccgcct ggccctgccc cagccacccc cggacccgcc ggcgcccccg 480ctggcgcccc cctcctcagc ctgggggggc atcagggccg cccacgccat cctggggggg 540ctgcacctga cacttgactg ggccgtgagg ggactgctgc tgctgaagac tcggctgtga 60014199PRTHomo sapiens 14Met Asn Cys Val Cys Arg Leu Val Leu Val Val Leu Ser Leu Trp Pro1 5 10 15Asp Thr Ala Val Ala Pro Gly Pro Pro Pro Gly Pro Pro Arg Val Ser 20 25 30Pro Asp Pro Arg Ala Glu Leu Asp Ser Thr Val Leu Leu Thr Arg Ser 35 40 45Leu Leu Ala Asp Thr Arg Gln Leu Ala Ala Gln Leu Arg Asp Lys Phe 50 55 60Pro Ala Asp Gly Asp His Asn Leu Asp Ser Leu Pro Thr Leu Ala Met65 70 75 80Ser Ala Gly Ala Leu Gly Ala Leu Gln Leu Pro Gly Val Leu Thr Arg 85 90 95Leu Arg Ala Asp Leu Leu Ser Tyr Leu Arg His Val Gln Trp Leu Arg 100 105 110Arg Ala Gly Gly Ser Ser Leu Lys Thr Leu Glu Pro Glu Leu Gly Thr 115 120 125Leu Gln Ala Arg Leu Asp Arg Leu Leu Arg Arg Leu Gln Leu Leu Met 130 135 140Ser Arg Leu Ala Leu Pro Gln Pro Pro Pro Asp Pro Pro Ala Pro Pro145 150 155 160Leu Ala Pro Pro Ser Ser Ala Trp Gly Gly Ile Arg Ala Ala His Ala 165 170 175Ile Leu Gly Gly Leu His Leu Thr Leu Asp Trp Ala Val Arg Gly Leu 180 185 190Leu Leu Leu Lys Thr Arg Leu 19515582DNAHomo sapiens 15atgggggtgc acgaatgtcc tgcctggctg tggcttctcc tgtccctgct gtcgctccct 60ctgggcctcc cagtcctggg cgccccacca cgcctcatct gtgacagccg agtcctggag 120aggtacctct tggaggccaa ggaggccgag aatatcacga cgggctgtgc tgaacactgc 180agcttgaatg agaatatcac tgtcccagac accaaagtta atttctatgc ctggaagagg 240atggaggtcg ggcagcaggc cgtagaagtc tggcagggcc tggccctgct gtcggaagct 300gtcctgcggg gccaggccct gttggtcaac tcttcccagc cgtgggagcc cctgcagctg 360catgtggata aagccgtcag tggccttcgc agcctcacca ctctgcttcg ggctctgcga 420gcccagaagg aagccatctc ccctccagat gcggcctcag ctgctccact ccgaacaatc 480actgctgaca ctttccgcaa actcttccga gtctactcca atttcctccg gggaaagctg 540aagctgtaca caggggaggc ctgcaggaca ggggacagat ga 58216193PRTHomo sapiens 16Met Gly Val His Glu Cys Pro Ala Trp Leu Trp Leu Leu Leu Ser Leu1 5 10 15Leu Ser Leu Pro Leu Gly Leu Pro Val Leu Gly Ala Pro Pro Arg Leu 20 25 30Ile Cys Asp Ser Arg Val Leu Glu Arg Tyr Leu Leu Glu Ala Lys Glu 35 40 45Ala Glu Asn Ile Thr Thr Gly Cys Ala Glu His Cys Ser Leu Asn Glu 50 55 60Asn Ile Thr Val Pro Asp Thr Lys Val Asn Phe Tyr Ala Trp Lys Arg65 70 75 80Met Glu Val Gly Gln Gln Ala Val Glu Val Trp Gln Gly Leu Ala Leu 85 90 95Leu Ser Glu Ala Val Leu Arg Gly Gln Ala Leu Leu Val Asn Ser Ser 100 105 110Gln Pro Trp Glu Pro Leu Gln Leu His Val Asp Lys Ala Val Ser Gly 115 120 125Leu Arg Ser Leu Thr Thr Leu Leu Arg Ala Leu Arg Ala Gln Lys Glu 130 135 140Ala Ile Ser Pro Pro Asp Ala Ala Ser Ala Ala Pro Leu Arg Thr Ile145 150 155 160Thr Ala Asp Thr Phe Arg Lys Leu Phe Arg Val Tyr Ser Asn Phe Leu 165 170 175Arg Gly Lys Leu Lys Leu Tyr Thr Gly Glu Ala Cys Arg Thr Gly Asp 180 185 190Arg17630DNAHomo sapiens 17ggatccatgg ctggacctgc cacccagagc cccatgaagc tgatggccct gcagctgctg 60ctgtggcaca gtgcactctg gacagtgcag gaagccaccc ccctgggccc tgccagctcc 120ctgccccaga gcttcctgct caagtgctta gagcaagtga ggaagatcca gggcgatggc 180gcagcgctcc aggagaagct gtgtgccacc tacaagctgt gccaccccga ggagctggtg 240ctgctcggac actctctggg catcccctgg gctcccctga gcagctgccc cagccaggcc 300ctgcagctgg caggctgctt gagccaactc catagcggcc ttttcctcta ccaggggctc 360ctgcaggccc tggaagggat ctcccccgag ttgggtccca ccttggacac actgcagctg 420gacgtcgccg actttgccac caccatctgg cagcagatgg aagaactggg aatggcccct 480gccctgcagc ccacccaggg tgccatgccg gccttcgcct ctgctttcca gcgccgggca 540ggaggggtcc tagttgcctc ccatctgcag agcttcctgg aggtgtcgta ccgcgttcta 600cgccaccttg cccagccctg agccgaattc 63018204PRTHomo sapiens 18Met Ala Gly Pro Ala Thr Gln Ser Pro Met Lys Leu Met Ala Leu Gln1 5 10 15Leu Leu Leu Trp His Ser Ala Leu Trp Thr Val Gln Glu Ala Thr Pro 20 25 30Leu Gly Pro Ala Ser Ser Leu Pro Gln Ser Phe Leu Leu Lys Cys Leu 35 40 45Glu Gln Val Arg Lys Ile Gln Gly Asp Gly Ala Ala Leu Gln Glu Lys 50 55 60Leu Cys Ala Thr Tyr Lys Leu Cys His Pro Glu Glu Leu Val Leu Leu65 70 75 80Gly His Ser Leu Gly Ile Pro Trp Ala Pro Leu Ser Ser Cys Pro Ser 85 90 95Gln Ala Leu Gln Leu Ala Gly Cys Leu Ser Gln Leu His Ser Gly Leu 100 105 110Phe Leu Tyr Gln Gly Leu Leu Gln Ala Leu Glu Gly Ile Ser Pro Glu 115 120 125Leu Gly Pro Thr Leu Asp Thr Leu Gln Leu Asp Val Ala Asp Phe Ala 130 135 140Thr Thr Ile Trp Gln Gln Met Glu Glu Leu Gly

Met Ala Pro Ala Leu145 150 155 160Gln Pro Thr Gln Gly Ala Met Pro Ala Phe Ala Ser Ala Phe Gln Arg 165 170 175Arg Ala Gly Gly Val Leu Val Ala Ser His Leu Gln Ser Phe Leu Glu 180 185 190Val Ser Tyr Arg Val Leu Arg His Leu Ala Gln Pro 195 20019448DNAHomo sapiens 19atgtggctgc agagcctgct gctcttgggc actgtggcct gcagcatctc tgcacccgcc 60cgctcgccca gccccagcac gcagccctgg gagcatgtga atgccatcca ggaggcccgg 120cgtctcctga acctgagtag agacactgct gctgagatga atgaaacagt agaagtcatc 180tcagaaatgt ttgacctcca ggagccgacc tgcctacaga cccgcctgga gctgtacaag 240cagggcctgc ggggcagcct caccaagctc aagggcccct tgaccatgat ggccagccac 300tacaagcagc actgccctcc aaccccggaa acttcctgtg caacccagat tatcaccttt 360gaaagtttca aagagaacct gaaggacttt ctgcttgtca tcccctttga ctgctgggag 420ccagtccagg agtgagaccg gccagatg 44820144PRTHomo sapiens 20Met Trp Leu Gln Ser Leu Leu Leu Leu Gly Thr Val Ala Cys Ser Ile1 5 10 15Ser Ala Pro Ala Arg Ser Pro Ser Pro Ser Thr Gln Pro Trp Glu His 20 25 30Val Asn Ala Ile Gln Glu Ala Arg Arg Leu Leu Asn Leu Ser Arg Asp 35 40 45Thr Ala Ala Glu Met Asn Glu Thr Val Glu Val Ile Ser Glu Met Phe 50 55 60Asp Leu Gln Glu Pro Thr Cys Leu Gln Thr Arg Leu Glu Leu Tyr Lys65 70 75 80Gln Gly Leu Arg Gly Ser Leu Thr Lys Leu Lys Gly Pro Leu Thr Met 85 90 95Met Ala Ser His Tyr Lys Gln His Cys Pro Pro Thr Pro Glu Thr Ser 100 105 110Cys Ala Thr Gln Ile Ile Thr Phe Glu Ser Phe Lys Glu Asn Leu Lys 115 120 125Asp Phe Leu Leu Val Ile Pro Phe Asp Cys Trp Glu Pro Val Gln Glu 130 135 14021459DNAHomo sapiens 21atgagccgcc tgcccgtcct gctcctgctc caactcctgg tccgccccgg actccaagct 60cccatgaccc agacaacgtc cttgaagaca agctgggtta actgctctaa catgatcgat 120gaaattataa cacacttaaa gcagccacct ttgcctttgc tggacttcaa caacctcaat 180ggggaagacc aagacattct gatggaaaat aaccttcgaa ggccaaacct ggaggcattc 240aacagggctg tcaagagttt acagaacgca tcagcaattg agagcattct taaaaatctc 300ctgccatgtc tgcccctggc cacggccgca cccacgcgac atccaatcca tatcaaggac 360ggtgactgga atgaattccg gaggaaactg acgttctatc tgaaaaccct tgagaatgcg 420caggctcaac agacgacttt gagcctcgcg atcttttag 45922152PRTHomo sapiens 22Met Ser Arg Leu Pro Val Leu Leu Leu Leu Gln Leu Leu Val Arg Pro1 5 10 15Gly Leu Gln Ala Pro Met Thr Gln Thr Thr Ser Leu Lys Thr Ser Trp 20 25 30Val Asn Cys Ser Asn Met Ile Asp Glu Ile Ile Thr His Leu Lys Gln 35 40 45Pro Pro Leu Pro Leu Leu Asp Phe Asn Asn Leu Asn Gly Glu Asp Gln 50 55 60Asp Ile Leu Met Glu Asn Asn Leu Arg Arg Pro Asn Leu Glu Ala Phe65 70 75 80Asn Arg Ala Val Lys Ser Leu Gln Asn Ala Ser Ala Ile Glu Ser Ile 85 90 95Leu Lys Asn Leu Leu Pro Cys Leu Pro Leu Ala Thr Ala Ala Pro Thr 100 105 110Arg His Pro Ile His Ile Lys Asp Gly Asp Trp Asn Glu Phe Arg Arg 115 120 125Lys Leu Thr Phe Tyr Leu Lys Thr Leu Glu Asn Ala Gln Ala Gln Gln 130 135 140Thr Thr Leu Ser Leu Ala Ile Phe145 1502330DNAArtificial SequenceCloning primer 23gaattcatga agtgggtaac ctttatttcc 302433DNAArtificial SequenceCloning primer 24gaattcttat aagcctaagg cagcttgact tgc 332528DNAArtificial SequenceCloning primer 25catatgaact gtgtttgccg cctggtcc 282625DNAArtificial SequenceCloning primer 26gatatgtatg acacatttaa ttccc 252726DNAArtificial SequenceCloning primer 27ggatccatgg gggtgcacga atgtcc 262826DNAArtificial SequenceCloning primer 28gaattctcat ctgtcccctg tcctgc 262925DNAArtificial SequenceCloning primer 29ggatccatgg ctggacctgc caccc 253026DNAArtificial SequenceCloning primer 30gaattctcag ggctgggcaa ggtggc 263128DNAArtificial SequenceCloning primer 31ggatccatgt ggctgcagag cctgctgc 283225DNAArtificial SequenceCloning primer 32gaattctcac tcctggactg gctcc 253333DNAArtificial SequenceCloning primer 33ctgccttagg cttacctggg ccaccacctg gcc 333429DNAArtificial SequenceCloning primer 34tgtcgactca cagccgagtc ttcagcagc 293533DNAArtificial SequenceCloning primer 35ctgccttagg cttaatctgt gacagccgag tcc 333628DNAArtificial SequenceCloning primer 36cactcgagtc atctgtcccc tgtcctgc 283735DNAArtificial SequenceCloning primer 37ctgccttagg cttaaccccc ctgggccctg ccagc 353825DNAArtificial SequenceCloning primer 38ctcgagtcag ggctgggcaa ggtgg 253935DNAArtificial SequenceCloning primer 39actccttagg cttagcaccc gcccgctcgc ccagc 354025DNAArtificial SequenceCloning primer 40ctcgagtcac tcctggactg gctcc 25

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed