CA2242384A1 - Exodus chemokine materials and methods - Google Patents

Exodus chemokine materials and methods Download PDF

Info

Publication number
CA2242384A1
CA2242384A1 CA002242384A CA2242384A CA2242384A1 CA 2242384 A1 CA2242384 A1 CA 2242384A1 CA 002242384 A CA002242384 A CA 002242384A CA 2242384 A CA2242384 A CA 2242384A CA 2242384 A1 CA2242384 A1 CA 2242384A1
Authority
CA
Canada
Prior art keywords
exodus
hiv
cells
dna
chemokine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002242384A
Other languages
French (fr)
Inventor
Robert Hromas
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Indiana University Research and Technology Corp
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2242384A1 publication Critical patent/CA2242384A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • C07K14/523Beta-chemokines, e.g. RANTES, I-309/TCA-3, MIP-1alpha, MIP-1beta/ACT-2/LD78/SCIF, MCP-1/MCAF, MCP-2, MCP-3, LDCF-1, LDCF-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Abstract

The present invention provides purified and isolated polynucleotide sequences encoding a human macrophage-derived C-C chemokine designated Exodus. Also provided are purified and isolated chemokine protein, fragments and polypeptide analogs thereof, antibodies thereto, and materials and methods for the recombinant production thereof. These products are useful in therapeutic, diagnostic and medical imaging applications.

Description

W O98/21330 PCTAUS97/20662 _ EXODUS CHEMOKIN~E MATERIALS AND ME'l~lODS
. .

The present invention relates generally to chemokines and more particularly to purified and isolated polynucleotides encoding a human C-C chemokine ~l~sign~tec~ Exodus and analogs thereof, to puri~led and isolated chemokine polypeptides cncoded by the polynucleotides, to materials and methods for the recombinant production of these polypeptides, and therapeutic uses of these polypeptides, particularly in myeloprotection during chemotherapy, in treatment of myeloproliferative diseases, and for ac~luired immunodeficiency syndrome (AIDS).

BACKGROUND OF T~E INVENTION
Chetnokines, also known as "intercrines" and "SIS
cytokines", comprise a superfamily of small secreted proteins (approximately 70-100 amino acids and 8-12 kilodaltons in size) which primarily regulate leukocyte migration and activation, and thereby aid in 1~ the stim~ tion and regulation of the immllne system. The name "chemokine" is derived from the term chemotactic cytokine, and refers to the ability of these proteins to stimulate chemotaxis of leukocytes. Tn~leefl, chemokines may comprise the ma~in attractants for infl~mm~tory cells into pathological tissues. [See generally, Baggiolini et al., Advances in Im~l7lunology, 55:97-179 (1994); Oppenhein1, The Chemokines, Lindley et al., eds., pages 183-186, Plenum Press~ NY (1993))]. While chemokines are generally secreted by leukocytes, several chemokines are expressed in a c multitude of tissues. Baggiolini et al., supra, Table II. Some chemokines also activate or attract a variety of cell types in addition to leukocytes, suchas endothelial cells and ~Ibroblasts.

W O 98/21330 PCTrUS97/20662 _ Previously identified chemokines generally exhibit 20-70%
amino acid identity to each other and contain four highly-conserved cysteine residues. Based on the relative position of the first two of these _ cysteine residues, chemokines have been further classified into two 5 subfamilies. In the "C-X-C" or "(x" subfamily, encoded by genes localized to human chromosome 4, the first two cysteines are separated by one amino acid. In the "C-C" or ",~" subfamily, encoded by genes which have been mapped to human chromosome 17, thc first two cysteines are adjacent. X-ray crystallography and NMR studies of several chemokines lO llavc indicated that, in each family, the first and third cysteines form a ~lrst disulfide bridge, and the second and fourth cysteines form a second disulfide bridgc, strongly influencing the native conformation of the proteins. In humans alone, nearly ten distinct sequences have been described for each chemokine subfamily. Chemokines of both subfamilies 15 havc characteristic leader sequences of twenty to twenty-five amino acids.

The C-X-C chemokines, which include IL-8, GROo~/~B/ y, platelct basic protein, Platelet Factor 4 (PF43, neutrophil-activating peptide-2 (NAP-2), macrophage chemotactic and activating factor (MCAF), IP-10, and others, share approximately 25% to 60% identity when any two amino 20 acid sequences are compared (except for the GROo~ / y members, which are 84-88% identical with each other). Most of the subfamily members (excluding IP-10 and Platelet Factor 4) share a common E-L-R tri-peptide motif upstream of the ~lrst two cysteine residues. The C-X-C chemokines are generally potent sti-n~ n~ of neutrophils, causing rapid shape change, 25 chemotaxis, rcspiratory bursts, and degranulation. Speci~lc truncation of the N-terminal amino acid sequence of certain C-X-C chemokines, including IL-8, is associated with marked increases in activity.
The C-C chemokines, which include Macrophage Inflammatory Proteins MIP~ [Nakao et al., Mol. Cell Biol., 10:3646 W O 98/21330 PCT~US97/20662 _ (1990)3 and MIP-I~ [Brown et al., J. Immunol., 142:679 (1989)], Monocyte Chemotactic Proteins MCP~ t~llshim~ et al., J. Exp. Med., 16g:1485 (1989)], MCP-2 [Van Damme et al., J. Exp. Med., 176:59 (1992) and Chang et al., Int. Immunol., 1.-388 (1989)~, and MCP-3 [Van Damme etal., supra], RA~S [Schall etal., J. Immunol., 141:1018 (1988)~ 309 [Miller et al., J. Immunol., 143:2907 (1989~], eotaxin ~Rothenberg et al., J. Exp. Med., 181:1211-1216 (1995)] and others, share 25% to 70% amino acid identity with each other. The C-C chemokines generally activate monocytes, Iymphocytes, basophils and eosinophils, but not neutrophils. Most of the rel~orted C-C chemokines activate monocytes, causing calcium flux and chemotaxis. More selective effects are seen on Iymphocytes, for example, T-lymphocytes, which respond most strongly to RAN~ES .
C-C chemokines can be further subdivided according to stmctural homologies and similar activities. MIP~ , MIP-l,B and RAN~ have closer homology and range of biological activities than the other members of the family. A.nother subfamily within the C-C
chemokine family are the monocyte chemotactic proteins (MCP), which are structurally more similar to each other than to other members of the C-C
chemokine family, and which preferentially stimulate monocytes to migr~te and respond to infl~mm~1ory stimuli.
Studies with deletion and substitution analogs have revealed that the critical receptor binding regions appear to be primarily in the amino-terminal residues of the chemokines, followed by a second region in the loop following the second cysteine. These general requirements for ~ function appear to be common to all chemokines. [Clark-Lewis et al., J.
~eukocyte Bio., 57:703 (1995).]
~ The chemokine receptors are seven-transmembrane-domain rhodopsin-like G protein-coupled receptors. A receptor specific for IL-8 has been cloned by Holmes et al., Science, 253:1278-83 (1991), while a W O 98/21330 PCTrUS97/20662 _ similar receptor (77% identity) which recognizes IL-8, GRO and NAP-2 has been cloned by Murphy and Tiffany, Science, 253:1280-83 (1991).
Five of the C-C chemokine receptors have been cloned to date: a C-C
chemokine receptor-l (CCR-l) which recognizes MIP-lo~ and RANTES
S [Neoteetal., Cell, 72:415-425 (1993)], areceptor(CCR-4)forMIP-l~, RANTES and MCP-1 [Power et al., J. Biol. Chem., 270:19495-19500 (1995)], an MCP-l receptor (CCR-2B) [Charo et al., Proc. Nat. Acad.
Sci., 91.-2752-56 (1994)], an eotaxin receptor (CCR-3) ~Combadiere et al., J. Biol. Che711. 270:16491-16494 (l99S)], and a receptor (CCR-S) for MIP-l o~, MIP- l ~ and RANTES [Raport et al ., J. Biol. Chem., 2 71: 17161 - 17166 (1996)] .
These receptors tend to be multifunctional, and may bind a nul1lber of different chemokines. The receptors themselves may play a role in human diseasc. For example, the Duffy antigen on human red blood cells (also known as the erythrocyte chemokine receptor), which binds avidly to chemokines including IL-8, NAP-2, GRO~, RANT~S, MCP-l, is an invasion receptor for a malaria-causing parasite, Plasmodium knowlesi.
Two herpesviridae, Herpesvirus saimiri and human cytomegalovims, also appear to encode functional chemokine receptor homologs. [Ahuja et al., Immlunol. Today, 15:281- (1994); Murphy, Ann. Rev. Immnol., 12:593-633 (1994); Horuk, TIPS, 15:159 (1994).~
Because of their pro-infl~mmzltory activities, chemokines are believed to play a role in a wide variety of diseases involving infi~mm~toly tissue destruction, such as rheum~toid arthritis, myocardial infarction, and adult respiratory distress syndrome. The role of a number of chemokines, particularly the C-X-C chemokine IL-8, has been well documented in various pathological conditions. See generally Baggiolini et al., supra, Table VII. For example, several studies have observed high levels of IL-8 in the synovial fluid of inflamed joints of patients suffering from rheumatic W O 98/21330 PCT~US97/20662_ diseases, osteoarthritis, and gout. Psoriasis has also been linked to over-production of IL-8.
The role of C-C chemokines in pathological conditions has also been documented. For example, the concentration of MCP-l is higher in the synovial fluid of patients suffering from rhellm~oid arthritis than that of patients suffering from other arthritic diseases. The MCP-1 dependent influx of mononuclear phagocytes may be an important event in the development of idiopathic pulmonary fibrosis. The role of C-C
chemokines in the recruitment of monocytes into atherosclerotic areas is currently of intense interest, with enhanced MCP-I expression having been detected in macrophage-rich arterial wall areas but not in normal ar~erial tissue. MCPs may also be involved in induction of angiogenesis and tumor growth or met~ . ~xpression of MCP-1 in m~ n~nt cells has been shown to suppress the ability of such cells to form tumors in vivo. (See U.S. Patent No. 5,179,07~, incorporated herein by reference.) Other chemokine activities include the ability to inhibit the proliferation of bone marrow progenitor cells. Recombinant MIP-lo~, but not MIP-1~, has been shown to suppress myelopoiesis of stem and progenitor cells, and ~ea~ to be selective in its ability to suppress growth factor-stimulated proliferation of multipotential progenitor cells (colony forming units of granulocyte-erythroid-macrophage-megakaryocytes, CFU-G~M[M) and subpopulations of burst-forming units of erythroid (BFU-E) and colony-forming units of granulocytes-- _ macrophages (CFU-GM) progenitor cells. [Broxmeyer et al., Blood, 76: 1110-l l 16 (1990~.] These effects are not a cytotoxic effect, but rather a cell cycle arrest. MIP-2O~, IL-8, PF4 and MCAF also have been reported to be suppressors of hemopoietic stem/progenitor cell proliferation.
~ [Broxmeyer et al., J. Immunol., 150:3448-3458 (1993); Broxmeyer et al., Am~. Hematol., 71:235-246 (1995).1 These chemokines appear to act directly at the level of the myeloid progenitors. Some reports indicate that W O 98/21330 PCTrUS97/20662 _ MIP-l(x has the potential to protect multipotent hematopoietic cells from the cytotoxic effects of chemotherapeutic agents. [Dunlop et al., Blood, 79:2221-2225 (1992) and Lord et al., Blood, 79:2605-2609 (1992).]
Clinical trials are reportedly under way for the use of a MIP-lol analog 5 (~lecign~ted BB10010, British Biotechnology) as a myeloprotective agent with Cytoxan~D (cyclophosphamide from Bristol-Myers Squibb Oncology).
Recently, there have been several reports that some C-C
chemokines, MIP-l~, MIP-1~ and RANTES, inhibit human immunode~lciency virus (HrV) production. [Cocchi et al., Science, 270:1811 (1996); Fauci, Nature, 378:561 (1996).] One study has reported that CD4~ Iymphocytes of individuals who have been exposed to HIV but remain HIV-negative express vely high levels of these C-C chemokines.
[Paxton et al., Nature Med., 2:412 (1996).] ~ potential mechanism for this inhibition has been suggested by the isolation and identiflcation of HIV co-receptors as members of the chemokine receptor families. The CCR-5 receptor which binds RANTES, MIP~ and MIP-l,B has been identified as the main co-receptor for most macrophage-tropic ~IV strains [Deng et al., Nature, 381:661 (1996); Dragic et al., Nature, 381:667 (1996); Alkhatib et al., Science, 272:1955 (1996)]. It has been reported that occasional primary HIV-l macrophage-tropic strains interact with the CCR-3 and CCR-2B receptors in vitro [Choe et al., Cell, 85:1135 (1996); Doranz et al., Cell, 85:1149(1996)~. Achemokinereceptor-lesign~t~d"Fusin" (now known as the C-~-C chemokine receptor CXCR-4) has been identified as a receptor for T-cell tropic strains of HIV [Feng et al., Science, 272:872 (1996)]. These HIV co-receptors are in the chemokine receptor families, and appear to be cofactors with CD4 for the fusion and entry of HIV
viruses into human target cells.
A need therefore exists for the identification and characterization of additional C-C chemokines, to further elucidate the role of this important family of molecules in pathological conditions and to ~~

L

W O 98/21330 PCT~US97/20662_ develop improved treatments for such conditions ~Itili7in~ chemokine-derived products.
~ Of interest to the present invention is International Publication No. WO 96/05856 published February 29, 1996, which reports the identification of two chemokines termed human chemokine beta-4 (Ck~-4) and human chemokine beta-10 (Ck,B-10) from cDNA libraries derived from human gall bladder and nine week human fetal tissue, respectively.
Ck,B-4 is very similar in both DNA and amino acid sequence to the Exodus chemokine described herein (the differences being that Ck~-4 has an additional alanine after residue 4 of the mature Exodus chemokine and that the reported deduced leader sequence of Ck~-4 is 24 amino acids, compared to the 22 amino acid leader sequence of Exodus). No biological activities of either chemokine Ck,B-4 or Ck,B-10 were determined. In particular, the publication does not mention any potential role for these chemokines in the pathogenesis of HIV infection, not does it specifically describe use of these chemokines for treating myeloproliferative diseases.
Also of interest is the cloning of another C-C chemokine, ~lesignzlted Exodus-2, that appears to be closely related to Exodus/MIP-3~-LARC, sharing 31 % amino acid identity and the same unique Asp-Cys-Cys-Leu motif seen around the first two cysteines. [Hromas et al., J.
Immlmol., 1S9:2554-2558 (1997).]
Chemokines of the C-C subfamily have been shown to - possess utility in medical im~ging, e.g., for im~ging the site of infection, infl~mm~tion, and other sites having C-C chemokine receptor molecules.
See, e.g., Kunkel et al., U.S. Patent No. 5,413,778, incorporated herein - by reference. Such methods involve chemical ~tt~chment of a labelling agent (e.g., a radioactive isotope) to the C-C chemokine using art recognized techniques (see, e.g., U.S. Patent Nos. 4,965,392 and 5,037,630, incorporated herein by reference), ~lmini~tration of the labelled chemokine to a subject in a pharmaceutically acceptable carrier, allowing W O 98121330 PCT~US~7/20662 _ the labelled chemokine to accumulate at a target site, and im~ging the Iabelled chemokine in vivo at the target site. A need in the art exists for additional new C-C chemokines to increase the available arsenal of medical im~gin~ tools.
More generally, due to the importance of chemokines as mediators of chemotaxis and infl~mm~tion, a need exists for the identification and isolation of new members of the chemokine family to facilitate modulation of infl~mm~tory and immune responses. For example, substances that promote the immune response may promote the healing of wounds or the speed of recovery from infectious diseases such as pneumonia. Substances that inhibit the pro-inflammatory effects of chemokines may be useful for treating pathological conditions mediated by inff~mm~tion, such as arthritis, Crohn's d;sease, and other autoimmune diseases.
1~ Additionally, the established correlation between chemokine expression and infl:~mm~tory conditions and disease states provides diagnostic and prognostic indications for the use of chemokines, as well as for antibody substances that are specifically immunoreactive with chemokines; a need exists for the identification and isolation of new chemokines to facilitate such diagnostic and prognostic indications.
For all of the aforementioned reasons, a need exists for recombinant methods of production of newly discovered chemokines, which methods facilitate clinical applications involving the chemokines and/or chemokine inhibitors.

SUMMARY OF T~IE INVENTION
The present invention fulfills one or more of the needs outlined above by providing purirled and isolated polynucleotides encoding a human C-C chemokine designated Exodus, and fragments and analogs thereof; purified and isolated Exodus polypeptides, fragments and analogs W O 98/21330 PCTrUS97/20662 _ thereof; n1aterials and methods for the recombinant production of these polypeptides, fragments, and analogs thereof; antibodies to such Exodus polypeptides and analogs; pharm~.e.lltic~l compositions comprising these polypeptides, fragments, analogs, or antibodies; and treatments using these S polypeptides, fragments, analogs, or antibodies, including prophylactic and therapeutic treatment.
Exodus is a member of the C-C chemokine family that is expressed preferentially in Iymphocytes and monocytes, and is markedly up-regulated by infl~mmzltory stimuli. The deduced amino acid sequence 10 of the cDNA encoding Exodus is ninety-five amino acids in length, of which the first twenty-two N-terrninal residues comprise a signal sequence.
Its biological activities as demonstrated herein are expected to render it useful in a number of different clinical applications. Like other C-C
chemokines, it stimulates chemotaxis of mononuclear cells. Significantly, 15 Exodus inhibits hematopoietic progenitor cell proliferation and also inhibits HIV prodllction in infected cells.
The invention specifically provides: purified polynucleotides (i.e., DNA and RNA, both sense and antisense strands) encoding the Exodus amino acid sequence of SEQ ID NO: 2, particularly a DNA
20 comprising a nucleotide sequence consisting of the Exodus protein-coding portion (nucleotides 43 to 327) of the nucleotide sequence of SEQ ID NO:
1; purified polynucleotides encoding amino acids 1 to 73 of SEQ ID NO:
2, particularly a DNA comprising a nucleotide sequence con.~ in~ of nucleotides 109 to 327 of SEQ ID NO: 1; and purified polynucleotides 25 encoding a full-length Exodus selected from the group consisting of: (a) nucleotides 43 to 327 of the D~A of SEQ ID NO: 1; ~b) a polynucleotide which hybridizes under stringeni conditions to the complementary strand of - nucleotides 43 to 327 of the DNA of SEQ ID NO: 1 or which would hy~ridize thereto under stringent conditions but for the degeneracy of the 30 genetic code; and (c) a polynucleotide which encodes the same Exodus W O 98/21330 PCTrUS97/20662 _ polypeptide as nucleotides 43 to 327 of the DNA of SEQ ID NO: 1. The invention also provides vectors comprising such polynucleotides, -particularly expression vectors where DNA encoding Exodus is operatively linked to an expression control DNA se~uence, host ce11s stably 5 transformed or transfected with such polynucleotide DNA, and corresponding methods for producing Exodus by culturing these host cells and isolating the ~xodus from the host cells or their nutrient medium. The invention further provides purified Exodus polypeptides, particularly a polypeptide comprising the amino acid sequence of SEQ ID NO: 2, or a 10 polypeptide comprising amino acids 1 to 73 of SEQ ID NO: 2. Another aspect of the invention provides antibodies specifically reactive with Exodus, including monoclonal antibodies and hybridoma cell lines producing such monoclonal antibodies.
Yet a further aspect of the invention provides a method of 15 increasing resistance to HIV infection by aflministering to a subject an amount of Exodus protein product effective to inhibit HIV proliferation, particularly where the subject is at risl~ of exposure to HIV, or has been exposed to HIV, or has been infected with ~V. This aspect of the invention also provides a method of treating HIV infection comprising 20 ~lmini~tering to a subject infected with HIV an amount of Exodus protein product effective to inhibit HIV proliferation. A further aspect of the invention provides a method of protecting bone marrow progenitor cells from cytotoxic effects comprising ~c~mini~t~ring an amount of Exodus protein product effective to suppress bone marrow progenitor cell 25 proliferation, particularly where the subject is undergoing chemotherapy or radiotherapy. Yet a further aspect of the invention provides a tnethod of treating myeloproliferative diseases comprising ~lmini~tering an amount of Exodus protein product effective to suppress m~lign~nt bone marrow progenitor cell proliferation. The invention is described more fully below.

W O 98/21330 PCTrUS97/20662 _ The invention provides purified and isolated polynucleotides (i.e., DNA and RNA, both sense and antisense strands) encoding Exodus.
Preferred DNA sequences of the invention include genomic and cDNA
sequences and chemically syn~hesi7e:1 DNA sequences.
The nucleotide sequence of a cDNA encoding this Exodus chemokine, including 5' and 3' non-coding sequences, is set forth in SEQ
ID NO: 1. Nucleotides 43 to 327 comprise the Exodus protein coding portion of this DNA of SEQ ID NO: 1, and a preferred DNA of the present invention comprises nucleotides 109 to 327 of SEQ ID NO: 1, which comprise the putative coding se~uence of the mature, secreted Exodus protein without its signal sequence.
The amino acid sequence of chemokine Exodus is set folth in SEQ ID NO: 2. Preferred polynucleotides of the present invention include, in addition to those polynucleotides described above, polynucleotides that encode the amino acid sequence set forth in SEQ ID NO: 2, and that differ from the polynucleotides described in the preceding paragraphs only due to the well-known degeneracy of the genetic code.
Similarly, sincc twenty-two amino acids (positions -22 to -1) of SEQ ID NO: 2 comprise a signal peptide that is cleaved to yield the mature Exodus chemokine, lJIerell~d polynucleotides include those which encode amino acids 1 to 73 of S.~Q ID NO: 2. Thus, a preferred polynucleotide is a purified polynucleotide encoding a polypeptide having an amino acid sequence comprising amino acids I to 73 of SEQ ID NO: 2.
Among the uses for the polynucleotides of the present invention is the use as hybridization probes, to identify and isolate genomic DNA encoding human Exodus, which gene is likely to have a three exon/two intron structure characteristic of C-C chemokines genes (See Baggiolini et al., supra); to identify and isolate non-human genes encoding proteins homologous to Exodus; to identify human and non-human W O 98/21330 PCTrUS97t20662 _ chemokines having similarity to Exodus; and to identify those cells which express Exodus and the conditions under wh;ch this protein is expressed.
Thus, in another aspect, the invention provides a puri~led polynucleotide which hybridizes under stringent conditions to the 5 complementary strand of the Exodus coding portion of the DNA of SEQ ID
NO: 1. Similarly, the invention provides a purified polynucleotide which, but for the re-1uncl~ncy of the genetic code, would hybridize under stringent conditions to the complementary strand of the Exodus coding portion of the DNA of SEQ ID NO: 1. Exemplary stringent hybridization conditions are as follows: hybridization at 42~C in 5X SSC, 20 mM NaPO4, pH 6.8, 50%
formamide; and washing at 42~C in 0.2X SSC. Those skilled in the art understand that it is desirable to vary these conditions empirically based on the length and the GC nucleotide base content of the sequences to by hybridized, and that formulas for determining such variation exist. [See, 15 e.g., Sambrook et al., Molecular Cloning.- a Laboratory Manual. Second Edition, Cold Spring ~arbor, New York: Cold Spring Harbor ~aboratory (1989)-]
In another aspect, the invention includes plasmid and viral DNA vectors incorporating DNAs of the invention, including any of the 20 DNAs described above. Preferred vectors include expression vectors in which the incorporated Exodus-encoding cDNA is operatively linked to an endogenous or heterologous expression control sequence. Such expression vectors may further includc polypeptide-encoding DNA se~uences operably linked to the Exodus-encoding DNA sequences, which vectors may be 25 expressed to yield a fusion protein comprising the E~xodus polypeptide of interest.
In another aspect, the invention includes a prokaryotic or eukaryotic host cell stably transfected or transformed with a DNA or vector of the present invention. In preferred host cells, the E~xodus polypeptide 30 encoded by the DNA or vector of the invention is expressed. The DNAs, CA 02242384 l998-07-09 W O 98/21330 PCT~US97/20662 ~_ vectors, and host cells of the present invention are useful, e.g., in methods for the recombinant production of large quantities of Exodus polypeptides of the present invention. Such rnethods are themselves aspects of the invention. For example, the invention includes a method for producing Exodus wherein a host cell of the invention is grown in a suitable nutrient medium and Exodus protein is isolated from the cell or the medium.
In yet another aspect, the invention includes purified and isolated Exodus polypeptides. A preferred peptide is a purified chemokine polypeptide having an amino acid sequence comprising amino acids I to 73 of SEQ ID NO: 2. The polypeptides of the present invention may be purified from natural sources, but are preferably produced by recombinant procedures, using the DNAs, vectors, and/or host cells of the present invention, or are chemically synthesized. Purif1ed polypeptides of the invention may be glycosylated (e.g., O-linked or N-linked) or non-glycosylated, water soluble or insoluble, oxidized, red~ce~l, etc., depending on the host cell selected, recombinant production method, isolation method, processing, storage buffer, and the like. Alternatively, Exodus polypeptides may be prepared by chemical peptide synthesis using techni~ues that have been used successfully for the production of other chemokines such as IL-8 [Clark-Lewis el al., J. Biol Cher~. 7 266:23128-34 (1991)] and MCP-l.
The invention also contemplates Exodus polypeptide - fragments, wherein one or more N-terminal or C-terminal amino acid residues are deleted, and which retain one or more of the biological activities characteristic of the C-C chemokines - Another aspect of the invention includes Exodus polypeptide analogs wherein one or more amino acid residues is added, deleted, or replaced from the Exodus of the present invention, and which retain one or more of the biological activities characteristic of the C-C chemokines.
Such analogs are usefill for, e.g., the medical im~ging methods described W O 98/21330 PCTrUS97120662 _ above or the treatment methods described below. They may be prepared by any recombinant or synthetic methods known in the art, including those deseribed below in Example 7.
E~cemplary analogs include substitutions in the E~codus amino 5 acid sequence designed to effect greater homology with the chemokines to which it is most closely related. Substitutions designed to effect greater homology with the C-C chemokine family include replacing the alanine at position 3-1 in the mature protein sequence with a threonine, or replaeing the phenylalanine at position 26 with a tyrosine. Other substitutions that 10 would effect greater homology with MIP-l~, MIP-1~ and RANT~S inelude replacing residues 1-8 of Exodus with residues 1-10 of MIP-l~x or residues 1-9 of RANTES, replacing the leucine at position 11 with a phenyl~l~ninP, replacing the glycine at position 12 with a serine, replaeing the glycine at position 25 with a glutamic acid, replacing the glutamic acid at position 36 15 with a serine, replacing the serine at position 46 with a gl~lt~mine, replacing the isoleucine at position 60 with a tyrosine, and replacing the serine at position 67 with an aspartic acid. These substitutions may be made singly or in all combinations, and are expected to have a potential for enhancing the activity of Exodus in myelosuppression or inhibition of HIV
20 productiom Other substitutions designed to enhance the properties of an amino acid at a given position (e.g., if an amino aeid is hydrophobie, the replacement is to be more hydrophobic~ may also enhance the activities of E~xodus: replacing the asparagine at position 6 with an aspartic aeid, 2~ replacing the leueine at position 18 with an isoleucine, replacing the glut~mine at position 29 with a glutamic acid, replacing the asparagine at position 38 with aspartic acid, replacing the valine at position 50 with isoleueine, and replacing the gh~ ninP at position 56 with glutamic acid.
These substitutions may be made singly or in all combinations.

w o 98nl330 PCTAUS97/20662 _ - 15 ~
A related aspect of the invention includes analogs which lack the biological activities of Exodus, but which are capable of competitively or non-competitively inhibiting the binding of C-C chemokines with their receptor(s). Such analogs are useful, e.g., in therapeutic compositions or 5 methods for inhibiting the biological activity of endogenous Exodus or other C-C chemokines in a host. Such Exodus polypeptide analogs are specifically contemplated to modulate the l~inding characteristics of Exodus to chemokine receptors and/or other molecules (e.g., heparin, glycosaminoglycans, erythrocyte chemokine receptors) that are considered lV ~o be important in presenting Exodus to its receptor.
In related aspects, the invention provides purified and isolated polynucleotides encoding such E~xodus polypeptide analogs, which polynucleotides are useful for, e.g., recombinantly producing the Exodus polypeptide analogs; plasmid and viral vectors incorporating such 15 polynucleotides; and prokaryotic and eukaryotic host cells stably transformed with such DNAs or vectors.
In another aspect, the invention includes antibody substances (e.g., monoclonal and polyclonal antibodies, single chain antibodies, chimeric or hum:~ni7ed antibodies, and the like) which are specifically 20 immunoreactive~with Exodus polypeptides and polypeptide analogs of the inventiom The invention further includes hybridoma cell lines that produce antibody substances of the invention. Such antibodies are useful, for example, for purifying polypëptides of the present invention, for detection or quantitative measurement of Exodus in fluid or tissue samples, e.g., 25 using well-k:nown ELISA techniques, and for morl~ ting binding of Exodus to its receptor(s). Some chemokine antibodies (e.g., anti-IL-8 antibodies) have been shown to have dramatic anti-infl~mm?,t--ry effects.
- Recombinant Exodus polypeptides and polypeptide analogs ofthe invention may be utilized in place of antibodies in binding reactions, to 30 identify cells expressing receptor(s) of Exodus and in standard expression W O 98/21330 PCTrUS97/20662 _ cloning techniques to isolate polynucieotides encoding the receptor(s). See, e.g., Example 16 below and the cloning of the IL-8 and MCP-l receptors in Holmes et al., supra, and Charo et al., s~pra~ respectively. Such Eixodus polypeptides, Exodus polypeptide analogs, and Exodus receptor S polypeptides are useful for modulation of Exodus chemokine activity, and for identification of polypeptide and chemical (e.g., small molecule) ~xodus agonists and antagonists.
As used herein, "Exodus protein product" includes Exodus polypeptides, fragments, or analogs thereof, including alternatively spliced 10 variants of Exodus, such as the chemokine Ck,~-4 described in International Publication No. WO 96/05856, supra, that retain the relevant biological activities of Exodus. We have demonstrated that the extra alanine found in CkB-4 (after residue 4 of Exodus) falls at an intron-exon boundary.
Sequencing across this region suggests that these two forms of Exodus arise 15 by alternative splicing.
The invention also contemplates pharmaceutical compositions comprising Exodus protein products for use in methods for enhancing the immune response in a m~mm~l suffering from a wound or an infectious disease. Also contemplated are pharm~cel~tical compositions comprising 20 Exodus protein products or antibodies thereto, for use in methods for reducing inFl~mm?~ion in infl~mm~tion-mediated pathological conditions, such as arthritis, Crohn's disease, or other autoimmune diseases. Further contemplated are pharmaceutical compositions for use in reducing atherosclerosis, angiogenesis or tumor growth or me~t~
Particularly contemplated are pharm~celltical compositions for use in suppressing proliferation of hematopoietic stem or progenitor cells. Such myelosuppression may protect stem/progenitor cells against cytotoxic effects during chemotherapy or radiotherapy. ~lso contemplated is use of an Exodus protein product for manufacture of a medicament for 30 suppressing bone marrow progenitor cell proliferation, said medicament W O 98/21330 PCTrUS97/20662._ being particularly desirable for ~lmini~ration to a subject undergoing chemotherapy or radiotherapy.
- Also particularly contemplated are pharm~ce-ltic~l compositions for use in treating myeloproliferative diseases, and use of an S Exodus protein product for manufacture of a medicament for treating myeloproliferative diseases.
E~urther particularly contemplated are pharm~ce--tical compositions for use in the treatment of patients recently exposed to HIV, but not yet tested for or confirmed to be HIV-positive by standard 10 diagnostic procedures (e.g., neonates from HIV-positive mothers, medical personnel exposed to ~IIV-positive blood), patients at risk of exposure to H[V, or patients already infccted with HIV, i.e., HIV-positive patients.
Also contemplated is use of an Exodus protein product for m~nllf~cture of a medicament for inhibiting HIV proliferation, said medicament being 15 particularly desirable for ~mini~tration to subjects at risk of exposure to ~V, or exposed to ~V, or infected with HIV. Further contemplated is use of an Exodus protein product for manufacture of a medicament for treating HIV infection.
Such pharmaceutical compositions comprise Exodus protein 20 product, or an antibody thereto, witll a physiologically acceptable diluent or carrier, and may optionally include other appropriate therapeutic agents depending on the clinical application, e.g., anti-infl~mmz~tory agents or anti-HIV agents. Dosages of Exodus protein product wi~l vary between about 1 ,ug to 100 mg/kg body weight, preferably 5 to 100 ~glkg body 25 weight, depending on the pathological condition to be treated. Such - pharmaceutical compositions may be ~lminist~red by a variety of routes depending on the condition to be treated, including via subcutaneous, intramuscu~ar, intravenous, intrapulmonary, transdermal, intrathecal, oral, or suppository ~imini~tration CA 02242384 l998-07-09 W O 98/21330 PCTrUS97/20662 _ The doses of the Exodus protein product may be increased or decreased, and the duration of treatment may be shortened or lengthened as determined by the treating physician. The frequency of dosing will depend on the pharmacokinetic parameters of the agents and the route of 5 ~lminictration. The optimal pharmaceutical formulation will be determined by one skilled in the art depending upon the route of ~-~minictratjon and desired dosage. See for example, Remington's Pharmflcel~ic~l Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, PA 18042) pages 1435-1712, the disclosure of which is hereby incorporated by reference. Such 10 fonnulations may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the ;~lmini~tered agents.
Those of ordinary skill in the art will readily optimize effective dosages and concurrent ~(lmini~tration regimens as determined by good medical practice and the clinical condition of the individual patient.
15 Regardless of the manner of ~lminictration, the specific dose may be calculated according to body weight, body surface area or organ size.
Further refinement of the calculations necessary to determine the a~,u~liate dosage for treatment involving each of the above mentioned formlllations is routinely made by those of ordinary skill in the art without 20 undue experimentation, especially in light of the dosage information and assays disclosed herein, as well as the pharmacokinetic data observed in the human clinical trials discussed above. Appropriate dosages may be ascertained through use of established assays for delel-lli"il-g blood levels dosages in conjunction with appluplialt; dose-response data. The final 25 dosage regiMen will be determined by the attending physician, considering various factors which modify the action of drugs, e.g. the drug's specific activity, the severity of the damage and the responsiveness of the patient, thc age, condition, body weight, sex and diet of the patient, the severity of any infection, time of ~dministration and other clinical factors. As studies W O 98/21330 PCTrUS97/20662 _ are conducted, further information will emerge regarding the appropriate ~ dosage levels for the treatment of various ~licç~çc and conditions.
The Exodus materials and methods described above may be employed in several clinical applications. First, as chemokines attract and S activate monocytes and macrophages (13aggiolini et al., supra), Exodus expression in a pathogenic infl~mm~tory setting may exacerbate the disease by recruiting additional monocytes and macrophages or other leukocytes to the disease site, by activating the leukocytes that are already there, or by inducing leukocytes to remain at the site. Thus, inhibiting the chemoattractant activity of Exodus may be expected to alleviate deleterious infl~mm~tclry processes. Significantly, the potential benefits of such an approach have been directly demonstrated in experiments involving IL-8, a C-X-C chemokine that attracts and activates neutrophils. Antibodies directed against IL-8 have a profound ability to inhibit infl~mm~tory disease mediated by neutrophils [Harada et al., J. Leukoc. Biol., 56:559 (1994)]. Inhibition of Exodus is expected to have a similar effect in ~lise~es in which monocytes or macrophages are presumed to play a role, e.g., Crohn's disease, rhenm~toid arthritis, atherosclerosis, myocardial infarction, or acute respiratory distress syndrome (Al~DS).
Alternatively, augmenting the effect of Exodus may have a beneficial role in diseases, as chemokines have also been shown to have a positive effect in wound healing and angiogenesis. Thus, exogenous Exodus protein products or Exodus agonists may be beneficial in promoting recovery from such diseases.
Exodus protein products or Exodus agonists may also prove - to be clinically important in the treatment of tumors, as suggested by the ability of the C-C chemokine TCA3 to inhibit tumor formation in mice (see Laning et al., supra). Exodus may act directly or indirectly to inhibit tumor formation, e.g., by attracting and activating various non-specific CA 02242384 l998-07-09 W O 98/21330 PCTrUS97/20662 _ effector cells to the tumor site or by stimulat;ng a specific anti-tumor immunity.
~ n addition, the myelosuppressive effeet demonstrated herein_ for Exodus indicates that Exodus protein products or Exodus agonists may 5 yield substantial benefits for patients reeeiving ehemotherapy or radiation therapy, reducing the deleterious effects of the therapy on the patient's myeloid stem or progenitor eells. For example, treatment with Exodus protein product before or during (e.g. a day before, immediately before, or at the same time as) ~-lmini~tration of cel} cycle-speeifie chemotherapeutie 10 agents may proteet Lhe bone marrow against the eytotoxic effects of the agents. Sueh eell eycle-speeific chemotherapeutic agents include vinblastine, etoposide, daunorubicin, doxorubicin, idarubicin, methotrexate, hydroxyurea, fluorouracil, cytosine arabinoside, mercaptopurine, thioguanine, pentostatin, fludarabine, and 2-chlorodeoxyadenosine (2-CDA). As discussed above, a MIP-l~x analog (de~ign~ d BB10010, British Biotechnology) is currently in elinieal trials as a myeloprotective agent in Cytoxan~ therapy (eyelophosphamide from Bristol-Myers Squibb Oneology).
The ability of Exodus to inhibit proliferation of eytokine-20 dependent myeloid cell lines, as shown herein, indicates that Exodusprotein product will also be useful in treating myeloproliferative dise~es7 including but not limited to chronic myelogenous leukemia, essential thrombocytosis, myelofibrosis, and polycythemia vera. ~lminictration of Exodus protein product for this purpose may be eoncurrent with 25 ~(lmini~tration of other chemotherapeutic agents or other cytokines, such as interferon.
~ Furthe~nore, the C-C chemokines RANTES, MIP-lo~ and MIP-I~ llave been shown to suppress replication of human immunodeficiency virus HIV-l ~Cocchi et ~l., Science, 270:1811-1815 30 (1995)], implicating them as possible therapeutic agents in the prevention W O98/21330 PCT~US97/20662 _ or treatment of AIDS. The ability of Exodus to inhibit HIV proliferation, as demonstrated herein, in~ljc~tes that Exodus protein product will also be beneficial for treating AIDS patients, in preventing onset or progression of AIDS, or in promoting reSi~t~n~e to H~V infection after HlV exposure.
S Full-blown AIDS does not appear ;mme~ ly upon infection with HIV;
there is a variable period of time during which the patient remains healthy but exhibits viremia. This viremia is sustained by continuous rounds of viral replication and reinfection of blood cells. One study has ~ound that measurements of plasma viral load (as well as CD4 lymphocyte counts) can 1~ predict the subsequent risk of AIlDS or death. [Ho, Science~ 272:112~
1125 (1996).] In~elre~ ce with the continuous cycle of viral replication may therefore result in an improved prognosis.
Additionally, the established correlation between chemokine expression and infl~mm~Qry con~itions and disease states provides 15 diagnostic and prognostic indications for the use of Exodus protein products, including antibody substances that are specifically immunoreactive with Exodus. Such Exodus materials are useful in methods for diagnosing and assessing the prognosis of infl~mm~ory conditions and disease states, as well as for medical im~ging of areas 20 involved in such conditions and disease states.
- Numerous additional aspects and advantages of the invention will become a~ alc;nt to those skilled in the art upon consideration of the following detailed description of the invention which describes presently preferred embodiments thereof.

25 BRIEF DESCRIPTION OF T~IE DRAWINGS
Figure 1 shows the effect of varying concentrations of Exodus on mononuclear cell chemotaxis.
Figure 2 shows the effect of untreated and ACN-treated Exodus on hematopoiesis in mice.

W O 98/21330 PCTrUS97/20662 _ Figure 3 shows the effect of Exodus alone or with MCP-l or MIG on cell cycling of hematopoietic progenitors in mice.
Figure 4 shows the effect of Exodus on the proliferation of the myeloid ceII line MO7E.
Figure 5 shows the effect of Exodus on the proliferation of the myeloid cell line TF~-1.
Figure 6 shows the effect of purified synthetic Exodus on the prolifieration of the myeloid cell line MO7E.
Figure 7 shows the effect of Exodus on the release of HIV
10 p24 protein by mononuclear cells after infection with HIV.
Figure 8 shows the effect of purified synthetic Exodus protein product on the release of HI~I p24 protein by mononuclear cells after infection with HIV.

DETAILED DESCRIPTION OF THE INVENTION
The invention is based upon the identification of a cDNA
sequence encoding Exodus and characterization of the activities of Exodus.
The complete cDNA of the chemokine Exodus is 821 nucleotides in Iength. There is a consensus polyadenylation site at 786.
The 3' untr~n~ ec~ sequence has a number of AAAU sequences that 20 mediate mRNA stability in many cytokine genes. These sequences promote message degradation, and contribute to the short half life of many cytokine transcripts, including chemokines. There is a short 5' untran~ ecl region of ~3 nucleotides.
There are 95 amino acids in the (1ednced amino acid 25 sequence of Exodus. This is consistent with the C-C chemokine family, where the length of family members ranges from 91 to 99. The first 22 amino acids of Exodus constitute a strongly hydrophobic signal peptide.
The four cysteines that participate in the disulfide bonds that define this family are also conserved in Exodus. Exodus is most closely related to W O98/21330 PC~US97/20662 MIP-lo~ and RANTES at the amino acid level, with 26-2g% identity, and about 75 % similarity when conservative changes are taken into accouIIt.
Exodus is especially similar to RAN~ from amino acids 24 to 46 and again from 58 to 75, where between these positions there are only six non-5 conservative changes.
While Exodus has many of the conserved amino acid features ----of the other human C-C chemokines, there are several llnusll~l characteristics of Exodus that are worth noting. Exodus has a highly basic carboxy-tenninus, more consistent with the MCP sub-family than 10 RA~I~S. In addition, Exodus lacks a conserved tyrosine and a threonine at position 47 and 51, respectively, that are present in all other human C-C
chemokines, including RANTE~S. It is not clear if these two highly conserved amino acids play a role in C-C chemokine activity since they are not predicted to contact the receptor.

Various aspects and advantages of the present invention will be understood upon consideration of the following illustrative examples.
Example I describes the identi~lcation of Exodus cDNA. Example 2 describes experiments e~mining the pattern of Exodus gene expression in various human cell lines and tissues. ~Example 3 describes the recombinant 20 expression of the Exodus gene in m~mm~ n cells. Example 4 describes another method for recombinant expression of the Exodus gene in m~mm~ n cells, and purification of the resulting protein. Example 5 provides a protocol for expression of the Exodus gene in prokaryotic cells and purification of the resulting protein. Example 6 provides a protocol for 25 the recombinant production of Exodus in yeast or invertebrate cells.
Example 7 describes production of Exodus and Exodus analogs by peptide synthesis or recombinant production methods. Example 8 provides a protocol for generating monoclonal antibodies that are specifically --- immunoreactive with Exodus. Example 9 addresses the effect of Exodus W O 98121330 PCTnJS97/20662 _ on monocyte chen1otaxis in vitro. Example 10 addresses the effect of Exodus on the proliferation of myeloid progenitor cells, myeloid cell lines and chronic myelogenous leukemia progenitor cells. Examp1e 11 addresses the effect of Exodus on HIV p24 protein production. Examples 12, 13, 14 5 and 15 provide in Yi~o assays of chemoattractant and leukocyte activation, tumor growth inhibition, and leukocyte activation after hltl~eliLoneal or subcutaneous injection. ~xample 16 describes cloning of an Exodus receptor.

Identification of the cDNA Sequence Encoding Exodus As described in Takeda et al., Hu~7lan Mol.. Genetics, 2:1793-1798 ~1993), messenger RNA was prepared from dissected normal adult human pancreatic islet cells, and first strand cDNA was synthesi7Pd by oligo(dT) priming using a primer that contained an XhoI site. After 15 second strand cDNA synthesis and blunting, EcoRI adapters were ligated to the cDNA, which was then size-fractionated to remove products of less than lO00 base pairs in size. After XhoI digestion, the products were cloned into lambda ZAP II, and amplified in XL1-Blue MRF' cells (Stratagene, La Jolla, CA). The library was converted to plasmids by 20 rescuing pBluescript SK- according to the manufacturer's instructions.
Partial sequences of 1000 of these randomly isolated pancreatic islet cDNAs were detennined by single-pass automated sequencing. These sequences deposited in GenBank (Takeda et al., supra) and were compared with other seql~ences in the National Center for Biotechnology Infonnation 25 (NCBI) ~l~t~ e. The average length of thc cDNA sequences used for comparison was approximately 200 bp. This work was published in Takeda et al., supra.
Subsequent to the work of Takeda et al., a clone encoding Exodus was identified among these 1000 pancreatic islet Epressed Sequence W O98/21330 PCTAUS97120662 _ Tags (ESTs) as follows Comparison of a consensus chemokine sequence against these ESTs using the BLAST service of NCBI revealed that one of the clones possessed a distant homology to the C-C chemokine family.
This homology to the chemokine family increased after several sequencing S errors from the original automated pass were identified by manual dideoxy double stranded sequencing, and the coding region and reading frame was properly characterized. This clone, originally designated HBC2850 by Takeda et al., was not identical to any other known chemokine. The cDNA in this clone consisted of 821 nucleotides, which contain the entire 10 open reading frame of the chemokine protein. This chemokine was designated Exodus.
The differences bctween the Exodus cDNA sequence, set forth in SEQ ID NO: 1, and the EST sequence of TAkeda et al. are as follows (with references to nucleotide numbering according to SEQ ID NO:
15 1): at nucleotide 64 ("C" in Exodus), the EST nucleotide was "N"; at mlcleotide 71 ("C" in ~xodus), the EST nucleotidc was "N"; between nucleotides 130 and 131, the BST contained an extra "G" base which caused a shift in the reading fralne; between nucleotides 150 and 151, the EST contained an extra "T" base which caused a shift in the reading frame;
20 at nucleotide 193 ("C" in Exodus), the EST nucleotide was "N"; at nucleotide 196 ("C" in Exodus), the EST nucleotide was "N"; at nucleotide 271 ("A" in Exodus), the EST nucleotide was "N"; and at nucleotidc 30g ("T" in Exodus), the EST nucleotides were "GC".

~XAMPLE 2 25 Exodus Gene Expression Pattern in Cell Lines and Tissues The pattern of Exodus mRNA expression was examinedl through Northern blotting of mRNA extracted from vanous human tissues and cell lines. The probe used was the cDNA contAining the complete coding region of Exodus, isolated by agarose gel electrophoresis, and W O 98/21330 PCT~US97/20662 _ labeled with 32P-dCTP and 32P-dl~P (DuPont-NEN, Boston, MA) by random priming according to the manufacturer's instructions (BMB, Tnfli~n~polis, IN).

A. Exodus Gene Expression in Human Tissues RNA was isolated from cell lines and cultured monocytes ---using RNA STAT-60 (Tel-Test B Inc., Friendswood, TX~ according to the manufacturer's instructions. Total RNA (20 ~g) was fractionated on 0.8%
formaldehyde agarose gels, transferred to nitrocellulose, hybridized and washed under stringent conditions. The films were exposed for one day 10 with an intensifying screen at -80~ C.
A Human Multiple Tissue Northern blot and a Human Immune System Multiple Tissue Northern (Clontech, Palo Alto, CA) were also probed with the Exodus cDNA and washed under stringent conditions according to the manufacturer's instructions. The autoradiograph was lS exposed as above for 1-4 days.
~xodus appeared to have a very restricted pattern of expression. It was not expressed in a number of cell lines tested, including TMR323 neuroblastoma, MDA breast carcinoma, K562 erythroleukemia, Jurkat T-cell leukemia, HL60 promyelocytic le~lkemiz~ HL60 cells 20 ~ ~i~relr~ te~1 to granulocytes with retinoic acid, 3T3 cmbryonic fibroblasts, or 293 embryonic kidney cells. When a commercially-prepared Northern blot of a variety of normal human tissues was analyzed for Exodus expression, expression was detectecl in the lung, and not in heart, brain, placenta, adult liver, skeletal muscle, kidney, pancreas, spleen or bone 25 marrow. The size oi the transcript was approximately 0.9 ld3, consistent with the size of the cDNA reported here, given the addition of a poly A
tail.
However, when a commercially-prepared Northern blot of lymphoid tissues was e~mined for Exodus expression, it was found to be -W O98/21330 PCTrUS97/20662 _ highly expressed in several different Iymphoid organs. Exodus was highly expressed in peripherai Iymph nodes, appendix, peripheral blood - mononuclear cells, and fetal liver. It was less highly expressed in thethymus, and there was no detectable expression in the spleen or marrow.
S This expression paLttern is typical of many chemokines.
Exodus was expressed mainly in lymphoid tissue, especially in lymph nodes, the appendix, and peripheral blood. It is possible that the Iymphoid tissue used in this Northern blot analysis may have been activated by some immunologic stimulus, thus causing a higher than usual level of expression 10 of Exodus. The poor expression of Exodus in bone marrow as opposed to peripheral blood may be due to the fact that the bone marrow is mainly composed of imm~hlre myeloid and erythroid precursors, while there are far more mature mononuclear cells in the peripheral blood.

B. Exodus Gene ~xpression After Infl~mm~tory Stimulus Since the expression of many chemokines is inflllcecl in mononuclear cells by infl~mm~ory stimuli, the expression of Exodus after exposure of various cell lines to LPS, TNF-alpha, or PMA was analyzed by Northern blot analysis.
The monocytic cell line TEP-l was obtained from American Type Culture Collection (Rockville, MD). Cells were m~int~ined in RPMI
1640 media (Biowhitaker, WaLkersville, MD) supplemented with 10% fetal calf serum (FCS, Eyclone Laboratories, Inc., Logan, Utah), 25 mM
HEPES, 100 U/ml penicillin and 100 ,ug/ml ~ll~lolllycin (tissue culture antibiotics, Life Technologies, Gaithersburg, MD). For stimulation experiments cells were cultured at a density of one million cells per ml in the presence of phorbol ester (PMA, Sigma, St. Louis, MO).
The immortalized human umbilical vein endothelia} cell line I-EUVEC was obtained from Dr. Jay Nelson, University of Oregon, and cultured in RPMI 1640 supplemented with 10% FCS (Hyclone), 400 ug/ml W O 98/21330 PCT~US97/20662 _ - 2~ -G418 (Life Teclmologies, Grand Island, NY), 1 u/ml heparin (Sigma), and 30 ,ug/ml endothelial cell growth factor (Collaborative Biomedical Products, Bedford, MA) to a confluency of 70-80%, then cultured in the presence or absence of 10 ng/ml tumor necrosis factor-alpha (TNF-~, Peprotech, NJ) for various periods of time.
Peripheral blood mononuclear cells werc purified on ~istopaque gradients ~Sigma) and monocytes were isolated by plastic adherence. Monocytes were cultured for 6 days, with media being replaced every two days to allow for diffelklllialion into macrophages.
10 Cells were stimulated with 100 ng/ml lipopolysacharide (LPS, Sigma) for various time periods.
Exodus expression was highly induced when peripheral blood mononuclear cells were exposed to EPS for 8 or 12 hours. Exodus expression was again highly induced when umbilical vein endothelial cells 15 were exposed to TN~-~x for only three hours. Significantly, Exodus expression stayed high as long as there was the infl~mm~tory stimuli present, When the monocytic leukemia cell line l~p-l was treated with PMA the expression of Exodus was also induced, reaching its peak at 48 hours after exposure, and declining slightly thereafter.
These results indicated that Exodus was poorly expressed unless infl~mm~tory stimuli were present. However, once such a stimulus was present Exodus was rapidly and stably up-regulated. The nature of the stimulus itself also seemed unrestricted, with LPS, TNF-cY, and PMA all up-regulating Exodus. Exodus production thus appears to be a function of 25 a mature Iymphophagocytic cell, especially after infl~mm~tory stimuli, and not of imm~ture myeloid cells.

CA 02242384 l998-07-09 W O 98/21330 PCTrUS97/20662 _ E~A~PT.E 3 Production of Reco[nbinant Exodus in COS Cells Recombinant Exodus was produced by transiently transfecting the Exodus cDNA into COS cells. The full length Exodus 5 cDNA was subcloned using com[non restriction sites into the polylinker site of pECE [~illis et al., Cell, 45:721 (1986)] an SV-40-driven expression vector, in sense orientation. Log phase COS cells (American Type Culture Collection (ATCC) No. CRL 1651) were plated in DMI~M with 10% F'CS
(Hyclone) and 100 U/ml penicillin and 100 ,ug/ml streptomycin-(tissue 10 culture antibiotics, Life Technologies, C~aithersburg, MD) at a density of one million cells per 100 mm culture dish and incubated overnight.
Twenty ~bg of purified pECE-Exodus plasmid DNA per plate was used for transfection of the COS cells with Lipofectin per the manufacturer's instructions (Life Technologies, Bethesda, MD). Purified pECE plasmid 15 (without Exodus DNA) was transfected identically into COS cells to serve as a control. An expression vector with the reporter gene beta-galactosidase (SV40/beta-Gal, Pharmacia, Piscataway, NJ) was co-transfected to control for transfection effllciencies. Seventy-two hours later, the supernatant of the COS cell culture was filtered through 0.2 ,um filters 20 and stored at -70~C. After supernatant removal, cell Iysates were made and beta galactosidase activity assayed as previously described in Rosenthal, Met~. Enzymol., 152:704 (1987). When pECE and pECE-Exodus transfections were performed, side-by-side tranfection efficiencies as detennined by beta-galactosidase activity were within 10% of each 25 other.
-Production of l~ecombinant Exodus in CHO Cells and Purification Thereof PCR was used to amplify bases 30 to 330 of the Exodus 30 cDNA (shown in SEQ ID NO: 1~, which includes 13 bp of 5' non-coding W O98/21330 PCTnUS97/20662 _ and 3 bp of 3' non-coding sequence. The sequences of the PCR primers were: 5'-GGCGAAGC77TGAGCTAAAAACCATG(SEQID NO: 3) and 5'-GCGGGAATTCTTACATGTTCTTGACT(SEQ ~ NO:4). To facilitate cloning, these primers include ~in~lm and EcoR~ restriction sites, 5 respectively (shown in italics~. The fragment was cloned into the vector pDCl (described in co-owned, co-pending U.S. Patent Application Serial No. 08/558,658 filed November 16, 1995, hereby incorporated by reference), which is a pBR322 derivative which contains the CMV
immediate early promoter adjacent to the cloning site to facilitate 10 expression of the insert, and the also contains the bacterial beta-lactamase gene and the murine dihydrofolate reductase (DHFR) gene to allow selection of the plasmid in bacterial and m~mm~ n cells, respectively (Sambrook el al., supra). The construct cont~ining the Exodus insert was linearized by restriction digestion with PvuI (BMB, Tndi~n~rlolis, IN), 15 which cleaves within the vector sequence. The linearized plasmid was precipitated with ethanol and redissolved in HBS (20 mM HEPES-NaOH, pH 7.0; 137 mM NaCl, 5 mM KCl; 0.7 mM Na2HPO4; 6mM Dextrose).
For electroporation, 107 cells of the CHO cell line DG44 ~Urlab et al., Cell, 33:405 (1983)~, were washed, resuspended in l ml PBS, mixed with 20 10 micrograms of linearized plasmid, and transferred to a 0.4 cm electroporation cuvette. The suspension was electroporated with a Biorad Gene Pulser (Richmond, CA) at 290 volts, 960 ,uFarad. Transformants were selected by growth in DMEM/F12 medium (Gibco) Cont~ining 10%
dialyzed FCS (Hyclone, Logan, UT) and lacking hypoxanthine and 25 thymidine. Cells from several hundred transformed colonies were pooled and replated in DMEM/F12 medium cont~ining 20 nM methotrexate (Sigma, St. Louis, MO). Colonies surviving this round of selection were isolated and expanded to obtain individual clones. The level of Exodus expression was determined as follows.

W O 98/21330 PCTfUS97/20662 _ Clones were grown Oll tissue culture plates to approximate1y 90% confluence in DMEM/F12 medium cont~inin~ 10% dialyzed FCS, at ~ which time the medium was replaced. The cells were allowed to grow for 4 days in DMEM/F12 me lilnn Cont~inin~ 1~ dialyzed FCS. The supernatant was loaded onto a column of Heparin Sepharose CL-6B
(Pharmacia, Piscataway, NJ). The column was washed with 0.2 M NaCl in 20 mM Tris, pH 7.5, and the chemokine was eluted with 0.6 M NaCI in 20 mM Tris, pH 7.5. The eluted Exodus was fractionated by SDS-PAGE
through an 18~ Tris glycine ge} (NOVEX, San Diego, CA) and transferred to a PVDF membrane (Millipore, Bedford, MA). The Exodus band migrating at approximately 7 kD was confirmed by detection with rabbit polyclonal antisera speci~lc for Exodus (prepared as described in Example 8 below).
Clones expressing the highest level of Exodus chemokine may be e~q)~n(11~.d for large scale protein production. The rçslllting recombinant Exodus is produced and purified from the supernatant as follows. The Exodus band migrating at approximately 7 kD is excised and the N-terminus sequenced on an automated sequencer (Applied Biosystems~
Model 473A, Foster City, CA).
As a further puri~lcation step, the Exodus eluted from the Heparin-Sepharose column is brought to 1.6 M NaCI and loaded onto a column of HI-Propyl 40 micron resin (J.T. Baker, Phillipsburg, NJ). The column is washed with 1.6 M NaCI in 20 mM Tris, pH 7.5, and the Exodus is eluted with 20 mM Tris, pH 7.5.
The integrity of the eluted Exodus is veri~ied by amino acid analysis to confirm the ratio of the amino acids predicted by the protein sequence and by mass spectrophotometry to confirm the predicted size.

W O 98/21330 PCT~US97/20662_ EXA~PLE 5 Production of Recombinant Exodus in Bacteria Exemplary protocols for the recombinant expression of Exodus in bacteria and purification of the resulting product follow.
The DNA sequence encoding the mature form of the protein is amplified by PCR and cloned into the vector pGEX-3X (Pharmacia, Piscataway, NJ). The pGEX vector is designed to produce a filsion protein comprising glutathione-S-transferase (GST), encoded by the vector, and a protein encoded by a DNA ~ragment inserted into the vector's cloning site.
10 The primers for the PCR are SEQ ID NO: 4 and 5'-TAT CGG ATC CTG
Gl~ CCG CGT GAA TCA GAA GCA AGC AAC T-3', which includes a BamHI restriction site, a thrombin cleavage site [Chang, Eur. J. Biochem., 151:217 (1985)], and nucleotides 109 to 127 of SEQ ID NO: 1. The resultant PCR product is digested with BamHI: and EcoRI and inserted into 15 a pGEX-3X plasmid digested with BglII and EcoRI.
Treatment of the recombinant fusion protein with thrombin or ~actor Xa (Pharmacia, Piscataway, NJ) is expected to cleave the fusion protein, releasing the chemokine from the GST portion. The pGEX-3X/Exodus constmct is transformed into E. coli XL-1 Blue cells 20 (Stratagene, La Jolla CA), and individual transformants were isolated and grown. Plasmid DNA from individual transformants is purified and par~ially sequenced using an automated sequencer to confirm the presence of tbe desired Exodus gene insert in the proper orientation.
Induction of the GST/Exodus filsion protein is achieved by 25 growing the transfonned XL-1 Blue culture at 37~C in LB medium (supplemented with carbenicillin) to an optical density at wavelength 600 nm of 0.4, followed by further incubation for 4 hours in the presence of 0.5 mM Isopropyl I~-D-Thiogalactopyranoside (Sigma Chemical Co., St.
Louis MO).

W O 98/21330 PCT~US97/20662 _ The fusion protein, expected to be produced as an insoluble inclusion body ;n the bacteria, may be purified as follows. Cells are harvested by centrifugation; washed in 0.15 M NaCI, 10 mM Tris, pH 8, 1 mM EDTA; and treated with 0.1 mg/ml Iysozyme (Sigma Chemical Co.) for 15 minutes at room temperature. The lysate is cleared by sonication, and cell debris is pelleted by centrifugation for 10 minutes at 12,000 X g.
The fusion protein-conf~inin~ pellet is resuspended in 50 mM Tris, pH 8, and 10 mM EDTA, layered over 50% glycerol, and centrifuged for 30 min. at 6000 X g. The pellet is resuspended in standard phosphate 10 buffered saline so~ution (PBS) free of Mg+ + and Ca~ t . The fusion protein is further purified by fractionating the resuspended pellet in a denaturing ~DS polyacrylamide gel (Sambrook et al., supra). The gel is soaked in 0.4 M KCl to visualize the protein, which is excised and electroeluted in gel-running bu~fer lacking SDS. If the GST/Exodus ~usion 1~ protein is produced in bacteria as a soluble protein, it may be purified using the GST Purification Module (Pharmacia Biotech).
The fusion protein may be subjected to thrombin digestion to cleave the GST from the mature Exodus protein. The digestion reaction (20-40 ,ug fusion protein, 20-30 units human thrombin (4000 U/mg (Sigma) 20 in 0.5 ml PBS) is incubated 16-48 hrs. at room temperature and loaded on a denaturing-SDS-PAGE gel to fractionate the reaction products. The gel is soaked in 0.4 M KCI to visualize the protein bands. The identity of the protein band corresponding to the expected molecular weight of Exodus may be confirmed by partiaT amino acid sequence analysis using an 25 automated sequencer (Applied Biosystems Model 473A, Foster City, CA).
Alternatively, the DNA sequence encoding the predicted mature Exodus protein may be cloned into a plasmid cont~ining a desired promoter and, optionally, a leader sequence [seel e.g., Better et al., ~cience, 240:1041-43 (1988)~. The sequence of this construct may be 30 confirmed by automated sequencing. The plasmid is then transformed into W O 98/21330 PCT~US97/20662__ ~ 34 -E. coli strain MC1061 using standard procedures employing CaC12 _ incubation and heat shock treatment of the bacteria (Sambrook et al., supra). The transformed bacteria are grown in LB medium supplemented with carbenicillin, and production of the expressed protein is inrlucecl by growth in a suitable medium. If present, the leader sequence will effect secretion of the mature Exodus protein and be cleaved during secretion.
The secreted recombinant protein is purified from the bacterial culture media by the method described above in Example 4 or, e.g., by adapting methods previously described for the purification of 10 recombinantly produced RANTES chemokine [Kuna et al., J. Immunol., 149:636-642 (1992)], MGSA chemokine [Horuk et al., J. BioE. Chem.
268:541~6 (1993)~, and IP-10 chemokine (expressed in insect cells) ~Sarris etal., J. Exp. Med., 178:1127-1132 (1993)].

Recombinant Productioll of Exodus in Yeast or Invertebrate Cells Exemplary protocols for the recombinant expression of Exodus in yeast or invertebrate cells, and for the purification of the resulting recombinant protein follow.
The coding region of the Exodus cDNA is amplified by 20 PCR. A DNA encoding the yeast pre-pro-alpha leader sequence is amplified from yeast genomic DNA in a PCR reaction using one primer cont~ining nucleotides 1-20 of the alpha mating factor gene and another primer complementary to nucleotides 255-235 of this gene ~Kurjan and Herskowitz, Cell, 30:g33-943 (1982)]. The pre-pro-alpha leader coding 25 sequence and Exodus coding sequence fragments are ligated into a plasmid cont~ining the yeast alcohol dehydrogenase (ADH2) promoter, such that the promoter directs expression of a fusion protein consisting of the pre-pro-alpha factor fused to the mature Exodus polypeptide. As taught by Rose and Broach, Met~l. Enz. 185:234-279, D. Goeddel, ed., Acadennic Press, W O98/21330 PCT~US97/2V~f~

Inc., San Diego, CA (1990), the vector further includes an ADH2 transcription terminator downstream of the cloning site, the yeast "2-micron" replication origin, the yeast leu-2d gene, the yeast R~Pl and REP2 genes, the E. coli beta-1~ct~m~e gene, and an E. coli origin of replication. The beta-lactamase and leu-2d genes provide for selection in bacteria and yeast, respectively. The leu-2d gene also facilitates increased copy number of the plasmid in yeast to induce higher levels of expression.
The REP1 and R~iP2 genes encode proteins involved in regulation of the plasmid copy number.
The DNA construct described in the prece ling paragraph is transformed into yeast cells using a known method, e.g., lithium acetate treatment [Stearns et al., Met~z. Enz., supra, pp. 280-297]. The ADH2 promoter is in~ ced upon exhaustion of glucose in the growth media [Price e~ al., Gene, 55:287 (1987)]. The pre-pro-alpha sequence effects secretion 15 of the fusion protein from the cells. Concomitantly, the yeast KEX2 protein cleaves the pre-pro sequence from the mature Exodus chemokine [Bitter et. al., Proc. Natl. Acad. Sci. USA, 81:5330-5334 (1984)1.
Alternative1y, Exodus is recombinantly expressed in yeast using a commercially available expression system, e.g., the Pichia 20 Expression System (Invitrogen, San Diego, CA), following the manufacturer's instructions. This system also relies on the pre-pro-alpha sequence to direct secretion, but transcription of the insert is driven by the alcohol oxidase (AOX1) promoter upon induction by methanol.
--- The secreted recombinant ~xodus is purified from the yeast 25 growth medium by, ~.g., the methods used to purify Exodus from bacterial ~ and m~m m~ n cell supernatants (see Examples 4 and 5 above).
Alternatively, the cDNA encoding Exodus is cloned into the baculovirus expression vector pVL1393 (PharMingen, San Diego, CA).
This Exodus-cont~ining vector is then used according to the m~nnf~ctllrer~s 30 directions (PharMingen) to infect Spodoptera frugiperda cells in sF9 W O 98/21330 PCT~US9 m O662 __ protein-free media and to produce recombinant protein. The protein is purified and concentrated from the media using a heparin-Sepharose column (Pharmacia, Piscataway, NJ) and sequential molecular sizing columns (Amicon, Beverly, MA), and resuspended in PBS. SDS-PAGE analysis 5 shows a single band and confirms the size of the protein, and Edman sequencing on a Porton 2090 Peptide Sequencer confirms its N-terminal sequence.

Production of Exodus Analogs Recombinant techniques such as those described in the preceding examples may be used to prepare Exodus polypeptide analogs.
Morc particularly, polynucleotides encoding Exodus are modified to encode polypeptide analogs of interest using well-known techniques, e.g., site-directed mutagenesis and polymerase chain reaction. See generally Sambrook et al., s~pra, Chapter 15. The modified polynucleotides are expressed recombinantly, and the recombinant polypeptide analogs are purified as described in the preceding examples.
Residues critical for Exodus activity are identified, e.g., by homology to oth~r C-C chemokines and by substituting alanines for the native Bxodus amino acid residues. Cysteines are often critical for the functional integrity of proteins because of their capacity to forrn disulfide bonds. To determine whether any of the four cysteines in Exodus is critical for enzyme activity, each cysteine is mutated individually to a serine.
Other exemplary analogs include substitutions in the ~xodus amino acid sequencc designed to effect greater homology with the chemokines to which it is most closely related. Substitutions designed to effect greater homology with the C-C chemokine family include replacing the alanine at position 31 in the mature protein sequence with a threonine, WO 98/21330 PCTrUS97/20662 _ or replacing the phenyl~l~ninP ~t position 26 with a tyrosine. Other substitutions that would effect greater homology with MIP~ , MIP-l~ and RAN~ includc rep1acing resicEues 1-8 of Exodus with residues 1-10 of MIP-lo~ or residues 1-9 of RANTES, replacing the leucine at position 11 S with a phenyl~ nine, replacing the glycine at position 12 with a serine, replacing the glycine at position 25 with a glut~mic acid, replacing the --glutamic acid at position 36 with a serine, replacing the serine at position 46 with a glutamine, replacing the isoleucine at position 60 with a tyrosine, and replacing the serine at position 67 with an aspartic acid. These 10 substitutions may be made singly or in all combinations, and are expected to have a potential for enhancing the activity of Exodus in myelosuppression or inhibition of HIV production.
Other substitutions designed to enhance the properties of an amino acid at a given position (e.g., if an amino acid is hydrophobic, the 15 replacement is to be more hydrophobic) may also enhance the activities of Exodus: replacing the asparagine at position 6 with an aspartic acid, replacing the leucine at position 18 with an isoleucine, replacing the glut:lmine at position 29 with a glutamic acid, replacing the asparagine at position 38 with aspartic acid, replacing the valine at position 50 with 20 isoleucine, and replacing the glllt~mine at position 56 with glllt~mic acid.
These substitutions may be made singly or in all combinations.
C-terminal deletions are prepared, e.g., by digesting the 3' end of the Exodus coding sequence with exonuclease m for various amounts of time and then ligating the shortened coding sequence to plasmid 25 DNA encoding stop codons in all three reading frames. N-te.~ al deletions are prepared in a similar manner by digesting the 5' end of the coding sequence and then ligating tlle digested fragments into a plasmid cont~ining a promoter sequence and an initi~tin~g methionine immediately upstream of the promoter site. These N-terminal deletion analogs may also --~ 30 be expressed as fusion proteins.

W O 98/21330 PCTAUS97/tO662 _ Alternatively, Exodus polypeptide analogs may also be prepared by chemical peptide synthesis using techniques that have been used successfully for the production of other chemokines such as IL-8 ~CIar~-Lewis et al., J. Biol Chem., 266:23128-34 (l99l)] and MCP-l.
5 Such methods are advantageous because they are rapid, reliable for short sequences such as chemokines, and allow the selective introduction of novel, unnatural amino acids and other chemical modifications.
The properties of Exodus analogs on one or more types of cells involved in tl~e infl~mm~tory process, (e.g., T lymphocytes, lO monocytes, macrophages, basophils, eosinophils, neutrophils, mast cells, endothelial cells, epithelial cells or others) are assayed by art-recognized techniques that have been used for assaying such properties of numerous other chemokines. The properties of Exodus analogs on inhibiting myeloproliferation and HIV production are also assayed according to lS Examples lO and l l below.

Preparation of Antibodies to Exodus Exodus chemokine was chemically synthesi7ed e~enti~11y as described in Example 7. For storagc, Exodus was diluted in RPMI
20 medium cont~inin~ 1% bovine serum albumin (Sigma, St. Louis, MO).
Exodus was subsequently purified from the medium by passage over a Heparin Sepharose CL-6B column (Pharmacia, Piscataway, NJ). The column was washed with a solution of 0.2 M NaC1 and 20 mM Tris, pH
7.5, and the chemokine was eluted with 0.6 M NaCl and 20 mM Tris, pH
25 7.5.
To generate polyclonal antisera, 50 ,ug of Exodus were emulsified in Freund's Complete Adjuvant for immunization of rabbits. At intervals of 21 days, 50 ,ug of Exodus were emulsified in Freund's Incomplete Adjuvant for boosts. These antisera recognized the chemically .

W O 98/21330 PCTrUS97/20662 _ synthes~zcd Exodus and the CHO celi-derived E~xodus (prepared as described in Example 4) on Western blot.
To generate monoclonal antibodies to Exodus, a mouse is injected periodically with recombinant Exodus (e.g., 10-20 ~g emulsified in 5 Preund's Complete Adjuvant) obtained as described in any of Examples 3 through 7. The mouse is given a final pre-fusion boost of Exodus in PBS, and four days later the mouse is sacrificed and its spleen removed. The spleen is placed in 10 ml serum-free RPMI 1640, and a single cell suspension is forrned by grinding the spleen between the frosted ends of 10 two glass microscopc slides submerged ;n serum-free RPMI 1640, supplemented with 2 mM L-gll~f~FninP, 1 mM sodium pyn~vate, 100 units/ml penicillin, and 100 ~bg/ml streptomycin (RPMI) (Gibco, Canada).
The cell suspension is filtered through sterile 70-mesh Nitex cell strainer (Becton Dickinson,,P~l~i~any, New Jersey), and is washed twice by centrifuging at 200 g for 5 minutes and resuspending the pellet in 20 ml serum-free RPMI. Splenocytes taken from three naive Balb/c mice are prepared in a similar manner and used as a control. NS-1 myeloma cells, kept in log phase in RPMI with 11% fetal bovine serum (FBS) (Hyclone Laboratories, Inc., Logan, Utah) for three days prior to fusion, are 20 centrifilged at 200 g for 5 minutes, and the pellet is washed twice as described in the foregoing paragraph.
One x 108 spleen cells are combined with 2.0 x 107 NS-1 cells and c~..tliruged, and the supernatant is aspirated. The cell pellet is dislodged by tapping the tube, and 1 ml of 37~~ PEG 1500 (50% in 75mM
25 ~epes, pH 8.0) (Boehringer Mannheim~ is added with stirring over the - course of I minute, followed by the addition oiC 7 ml of serum-free RPMI
over 7 minutes. An additional 8 ml RPMI is added and the cells are centrifuged at 200 g for 10 minutes. After discarding the supernatant, the pellet is resuspended in 200 ml RPMI Cont~ining 15% FBS, 100 ~M
30 sodium hypo~nthine, 0.4',uM aminopterin, 16 ~uM thymidine (HAT) W O 98/21330 PCT~US97/20662 _ (Gibco), 25 units/ml IL-6 (Boehringer Mannheim) and 1.5 x 106 splenocytes/ml and plated into 10 Corning flat-bottom 96-well tissue culture plates (~orning, Corning New York).
On days 2, 4, and 6, after the fusion, 100 ,ul of medium is 5 removed from the wells of the fusion plates and replaced with fresh mediull]. On day 8, the fusion is screened by ~T TSA, testing for the presence of mouse IgG binding to Exodus as follows. Immulon 4 plates (Dynatech, Cambridge, MA) are coated for 2 hours at 37~C with 100 ng/well of Exodus diluted in 25mM Tris, pH 7.5. The coating solution is aspirated and 200 ul/well of blocking solution [0.5 % ~lsh skin gelatin (Sigma) diluted in CMF-PBS]is added and incubated for 30 min. at 37~C.
Plates are washed three times witll PBS with 0.05% Tween 20 (PBST) and 50 ,ul culture supernatant is added. After incubation at 37~C for 3û
minutes, and washing as above, 50 ,ul of horseradish peroxidase conjugated goat anti-mouse IgG(fc) (Jackson ImmunoResearch, West Grove, Pennsy}vania) diluted 1:3500 in PBSTis added. Plates are incub~t~cl as above, washed four times with PBST, and 100 ~I substrate, Con~ tin~ of 1 mg/ml o-phenylene diamine (Sigma) and 0.1 f~l/ml 30% H2~2 in 100 mM
Citrate, pH 4.5, are added. The color reaction is stopped after 5 minutes with the addition of 50 ,ul of 15% H2SO4. A490 is read on a plate reader (Dynatech) .
Selected fusion wells are cloned twice by dilution into 96-well plàtes and visual scoring of the number of colonies/well after 5 days.
The monoclonal antibodies produced by hybridomas are isotyped using the Isostrip system (Boehringer Mannheim, Indianapolis, IN).

Effect of Exodus on Monocyte Chemotaxis The activity of Exodus was evaluated in chemotaxis assays perfonned as previously described in Martinet et al., J. Immunol. Meth., CA 02242384 l998-07-09 W O 98/21330 PCTrUS97/20662 _ 174:209, 1994 and Keller et al., J. Imntunol. Meth., 1:165, 1972. Twent~
ml of peripheral blood was collected from healthy volunteers in 10 ml heparinized tubes. Blood was diluted 1:1 with PBS and then underlaid with 10 ml of Histopaque (Sigma). ~fter centrifugation at 400 g for 25 mins, 5 cells at the interface were collected and washed twice in P~S. Cells were resuspended in DM~M (Life Technologies, Gaithersburg, MD) with 100 U/ml penicillin and 100 ,ug/ml streptomycin (tissue culture antibiotics, LiiFe Technologies) at 106fml. Sterile bovine serum albumin (Sigma) was added to final concentration of 0.2 mg/ml.
100 ,ul of this cell suspension was added to each transwell insert (Costar). DMEM with antibiotics and 0.2% BSA with or without pure synthetic Exodus was added to the lower wells in the 24 well plate. All Exodus concentrations were done in triplicate. Transwell inserts were placed into the lower walls, and incubated at 37~ C for 90 mins. At the completion of the incubation period inserts were removed and the top of the filter scraped with a rubber policeman to remove adherent cells. The entire insert was then stained with Wright-Giemsa. Cells adherent to the lower surface of the insert and those that migrated to the lower well were counted under 3 high power fields, and added together to obtain a total number of migrating cells.~~
Purified synthetic Exodus was tested at concentrations of 5, 50 and 500 ng/ml. For comparison, MIP-l~ was tested at a concentration of 833 ng/ml. The control contained no chemo~ine. Results are shown in Figure 1. The values represent the average of two experiments performed in triplicate, plus or minus the standard error. The stars represent statistically significant diff~l~nces from the control at p<0.05 using the unpaired Student's t-test.
These results show that Exodus stimulated chemotactic activity of normal human peripheral blood mononuclear cells, as measured by transwell migration. The highest concentrations of Exodus stimulated ---W O 98/21330 PCTrUS97/20662_ chemotaxis more efficiently than maximally effective concentrations of MIP-l ~Y.
Similar results were obtained with an Exodus protein product, which was Exodus with an additional alanine after residue 4.

Effect of Exodus on Proliferation of Myeloid Cells A. Effect on Myeloid Pro~enitor Cells The effect of Exodus protein products on hematopoietic colony formation were assayed essentially as previously described in, e.g., Broxmeyer et al., Blood, 76:1110 (1990). Bone marrow cells were collccted from human donors after obtaining informed consent. Low density human bone marrow cells at 5 x 104/ml were plated in 1%
methylcellulose in Iscove's Modified Essential Medium (Biowhitaker, Walkersville, MD) supplemented with 30% FCS (Hyclone~, recombinant human erythropoietin (EPO, 1 U/ml, Amgen, Thousand Oalcs, CA), recombinant human interleukin-3 (IL-3, 100 U/ml, Immunex, Seattle, WA), and recombinant human stem cell factor (SCF, 50 ng/ml, Amgen) for colony forming unit granulocyte/macrophage (CFU-GM), colony fonning unit granulocyte/erythrocyte/macrophage/megakalyocyte (CFU-GEMM) or blast forming unit-erythrocyte (BFU-E) analysis. Cultures were incubated at 5% C02 and low oxygen tension (5~) for 14 days, and then scored for colony formation using an inverted m;croscope in a blinded fashion. Experiments were perfolmed at least twice in triplicate.
Varying amounts of COS cell supelnal~lll cont~ining Exodus, prepared as described in Example 3 above, were tested in this assay, as was MIP-l(x (R&D Systems, Minneapolis, MN) at 50 ng/ml. Results are shown in Table 1 below, which displays the mean count of hematopoietic progenitor colonies per plate, plus or minus the standard deviation.

W O 9~/2133~ PCTrUS97/20662 _ Chemokine in ~ m CFU-GM BFU-E CFU-GEMM
Control (no chemokine)53 + 8 56 + 8 24 + 5 MIP-lo~ 50 ng/ml 23 + 4* 37 + 3* 12 + 1*
~xodus COS cell 21 + 3 29 + 2* 11 + 1*
supernatant (0.2 ml in 2 ml tota} medium) Exodus COS cell 24 + 4 27 + S 11 + 2 supernatant (0.1 ml in 2 ml total medium) Exodus COS cell 23 + 5 36 + 3 14 + 1 supernatant (0.05 ml in 2 ml total medium) Exodus COS cell 47 + 8 58 + 6 24 + 1 lS supernatant (0.025 ml in 2 ml total me~ m) pECE-only COS cell 50 + 6 60 + 7 23 + 3 supernatant (0.2 ml in 2 ml total medium~
20 Ik p<0.005 (the other values are not significantly different from control or pECE at p < 0. 05) The Exodus in the COS cell supernatant inhibited hematopoietic progenitor colony forrnation in a dose-dependent manner, slightly more efficiently than a maximal dose of MIP-l~. There was no 25 statistical difference between COS cell medium alone and medium from COS cells that had been tranfected with the empty pECE expression vector.
At 50 ng/ml of recombinant human MIP-l~x, a dose at which the biological effect plateaus, there was stz~ti.ctic~lly significant reduction of both CFU-GM (43% of medium control), BFU-E (66% of control), and CFU-GEMM
30 (50% of control). At the highest concentrations of recombinant Exodus used in these experiments there was also a statistically significant decrease in both CFU-GM (42% of control), BFU-E (48% of control), and CFU-GEMM (48 % of control) .

W O 98/21330 PCT~US97/20662_ This inhibition by Exodus was dose-dependent, in that the three highest levels of Exodus showed inhibition of the proliferation of hematopoietic progellilol~ as measured by colony formation assays.
However, the lowest concentration of Exodus used did not show such an S inhibition. Like ~[P~ , Exodus inhibited progenitors in a multi-lineage fashion.
Purified synthetic Exodus was also tested in this assay.
Results are shown in Table 2 below, which displays the mean count of hematopoietic progenitor colonies per plate, plus or minus the standard 10 deviation.
TA~LE~ 2 Conce.,lralion of CFU-GM BFU-~: CFU-GEl\~M
Chemokine in Medium Control (no chemokine) 85 + 3 97 + 4 39 + 3 Exodus (200 ng/ml) 43 + 11 43 + 2 19 + 3 Exodus (100 ng/ml) 39 + 3 41 + 2 20 + 2 Exodus (50 ng/ml) 42 + 10 42 + 3 17 i 2 Exodus (25 ng/ml) 41 + 2 50 + 2 20 + 4 Exodus (12.5 ng/ml) 51 + 3 70 + 10 27 + 1 Exodus (6.25 ng/ml) 64 + 8 87 + 3 32 + 2 MIP-l~x (100 ng/ml) 41 + 2 44 + 3 19 + 2 IL-8 (100 ng/ml) 42 + 2 44 + 2 19 + 2 PF-4 (100 ng/ml) 42 + 4 44 + 5 19 + 1 RANTES (100 ng/ml) 81 + 7 99 + 4 37 + 1 NAP-2 (100 ng/ml) 83 + 2 93 + 3 39 + 4 There was a statistically significant (Student's T test) reduction in colony formation of all three types at concentrations of Exodus down to 25 ng/ml (p ~ 0.005). The purified synthetic Exodus behaves identically to the Exodus in the COS cell supell-al~nls. Both sources of CA 02242384 l998-07-09 W O 98/21330 PCTrUS97/20662 _ Exodus are effective at inhibiting hematopoietic marrow progenitor proliferation, at least as good if not better than -MIP~
Similar results showing inhibition of the proliferation of hematopoietic progenitors, as measured by colony formation assays, were 5 obtained with a purified synthetic Exodus protein product, which was Exodus with an additional alanine after residue 4.
These results show that Exodus protein products inhibited proliferation of hematopoietic progenitors. Indeed, Exodus protein products were as effective as MIP-l~x. This indicates that Exodus protein 10 products will be useful as cycle-specific chemoprotective agents.
Further experiments confirmed the effect of Exodus protein product in vivo on hematopoiesis in mice. Experiments were carried out essentially as described in Broxmeyer et al., Ann. Hematol., 71:235-246 (1995), using untreated pure synthetic Exodus and Exodus treated with a 15 30% acetonitrile/1% trifluoroacetic acid (ACN) solution as described in Mantel et al., Proc. Natl. Acad, Sci, USA, 90:2232-2236 (1993). For chemokines which exist in solution as multimers, treatment with ACN
stimulates formation of monomers, which are the active forrn in vivo, and thus e~h~nces chemokine activity. ACN-treated chemokines can be active 20 at concentrations 200-fold lower than untreated chemokines.
Briefly, solutions of untreated Exodus or ACN-treated Exodus were prepared at concentrations varying from 0.00l to 50 ng/ml Exodus. ACN-treated or untreated diluent served as a control to show that the ACN was not toxic. Normall C3HtHeJ mice were in}ected 25 intravenously with a 0.2 ml dose of each solution and were sacrificed after 24 hours. Unseparated bone marrow cells were obtained from their femurs and were plated in either agar with 10% v/v pokeweed mitogen mouse spleen cell-conditioned medium (PWMSCM), for CFU-GM assessment, or in methylcellulose with human erythropoietin ~Epogen~, Amgen, Thousand 30 Oaks, CA), PWMSCM and hemin (F~tm;ln Kodak Co., Rochester, NY), W O 98/21330 PCTrUS97120662 _ for BFU-E/CFU-GE;MM assessment. Colony counts were determined after seven days of incubation in a humidified environment in an ESPEC N2-O2-CO2 incubatorBNP-210 (Taboi ESPEC Corp., South Plainffeld, NJ) in 5%
C~2 and 5% ~2 Results are displayed in Figure 2.
The results showed that untreated Exodus reduced CFU-GM
colony formation to an average of 56% of control when a single dose of 0.2 ml of 25 ng/ml Exodus was injected. ACN-treated Exodus reduced CFU-GM colony formation to an average of 53 % of control when a single dose of 0.2 ml of 0.1 ng/ml Exodus was injected. The inhibition of in 1nvo CF~-GM formation in~ cecl by both the ACN-treated and the untreated Exodus was statistically significant at p<0.05.
In another experiment, the effect of untreated Exodus on the in vivo cycling of hematopoietic progenitors was evaluated with a tritiated thymidine kill assay essentially as described in Broxmeyer et al., Ann.
Hematol., supra and in Mantel et al., Proc. Natl. Acad. Sci., supra.
Briefly, mice were treated with varying concentrations of untreated Exodus (0.5, 1 or 10 ng/ml) alone or in combination with other untreated chemokines (Exodus at 0.01 or O. l ng/ml with either MCP-1 or MIG at 0.01 or 0.1 ng/ml). The ~nim:~l.c were then sacrificed after 24 hours and bone marrow was collected for a tritiated thymidine kill assay. Rer~ll,se cells in the S-phase of the cell cycle pler~;renLially incorporate thymidine, they are killed by the incorporation of tritiated thymidine whereas cells that - are not in S-phase are unharmed. The number of cells in- S-phase is estim~tPcl by calculating the number of cells killed, based on control colony numbers of cells that were not treated with tritiated thymidine. The values are reported as a percentage of progenitors in S-phase and are norm~li7erl for the total number of progenitors per femur. Results for CFU-~;M are shown in Figure 3.
The results showed that a single injection of 10 ng/mouse of Exodus significantly reduced the percentage of CFU-GM progenitors in S-W O 98/21330 PCTAUS97/20662_ phase of the cell cycle to an average of 4% (p~0.001), as compared to an average control value of 56% of CFU-GM in S-phase. In addition, Exodus-induced inhibition of cell cycle progression for CFU-GM was synergistic with MCP-l and MIG treatment. When very low concentrations of Exodus was injected with MIG or MCP-l, there was an even greater reduction of CFU-GM in S-phase. Thus, a combination of chemokines may produce a more potent inhibition of hematopoiesis.
These results indicate that Exodus can temporarily arrest cell cycle progression of bone marrow progenitor cells, and thus can be used to protect no~nal bone marrow against S-phase cytotoxic chemotherapy.

B. Effect on Myeloid Cell Lines The effect of Exodus protein products on the proliferation of cytokine dependent myeloid cell lines was also tested. The human myeloid cell lines TP-l and MO7E [Avanzi et al., B~t. J. Haematol., 69:359 (1988)~ require GM-CSF and SCF for maximal proliferation. The cytokine-dependent primitive acute myeloid leukemia cell lines TF-l and MO7E (both gifts from Dr. Hal Broxmeyer, Tn(li:~n~ University, Tn~i~n~) were cultured in l?PMI 1640 (Life Technologies, G~i~he~ rg, MD) plus 10% FCS ~Hyclone) and 100 U/ml penicillin and 100 ,ug/ml streptomycin (tissue culture antibiotics, Life Technologies, Gaithersburg, MD). This media was supplemented with granulocyte-macrophage colony stimulating factor (GM-CSF, 100 U/ml, Immunex, Seattle, WA) and stem cell factor - - (SCP, 50 ng/ml, Amgen, Thousand Oaks, CA) for normal log phase growth.
Exodus in COS cell supernatants prepared as described in Example 3 was tested at a final dilution of 1/10. Results are shown in Figure 4 (for MO7E cells) and Figure 5 (for TF-l cells). When COS cell supelllala--L was added to log phase MO7E cells, in the presence of GM-CSF and SCF, proliferation over the next 72 hours was reduced to 10.4%

- -W O 98/21330 PCTnUS97/20662 of control. When COS cell supernatant was added to log phase TF-l cells that were also continuously exposed to GM-CSF and SCF, proliferation was completely inhibited. Exodus was not exerting a cytotoxic effect, as the Exodus-treated cells had greater than 95 % viability at every time point, as ~ccces~ed by trypan blue exclusion, which was identical to that of the control cells.
Purified synthetic Exodus also completely inhibited proliferation of the MO7E cells, as shown in Figure 6. Each data point is the mean of four separate experiments. Stars denote st~ti~ticzll significance 10 at p<0.05 using a paired t-test. Viability also did not change with the addition of Exodus.
These results indicate that Exodus will also be useful in treating myeloproliferative disorders such as chronic myelogenous leukemia.

15 C. ~ffect on Chronic Myelogenous Leukemia Progenitors The effect of Exodus (also called "Exodus-l "~ on progenitor proliferation in chronic myelogenous leukemia (CML) was evaluated using colony formation assays as described in Hromas et al., Blood~ 89:3315- _ 3322 (1997). Briefly, bone marrow cells were collected from six CML
20 patients in chronic phase. Low density marrow cells at 5 x 104 cells/mL
were plated in l ~ methylcellulose in Iscove's modified Dulbecco's medium supplemented with 30% fetal calf serum, 1 U/mL human erythropoietin (Epogen~D, Amgen~, lO0 U/mL human interleukin-3 (Genetics Tn~tit~lt~) and 50 ng/mL human stem cell factor (Arngen), in the presence or absence of 25 100 ng/ml Exodus and in the presence or absence of 100 ng/ml MIP-l~.
Cultures were incubated at 5% CO2 and low (5%) oxygen tension for 14 days, and then scored using an inverted n1icroscope for CFU-GM, C~U-GEMM and BFU-P. Colony counts for cultures treated with Exodus or MIP-lo~ were compared to colony counts of the control cultures and were CA 02242384 l998-07-09 W O 98/21330 PCTrUS97/20662 _ expressed as a percentage of control CFU or BFU. Data is displayed in Table 3 below.
Table 3 Treatment % of Contr~l % of Control % of Control CFU-GM BFU-E CFU-GEMM
100 ng/ml 52 + 5* 44 ~ 19* ~7 + 6~ --Exodus 100 nglml 9 ~ 3 1 + 8 3 ~ 6 MIP- 1 ~
Statistically signif cant (p<0.05) using unpaired Studen 's t-test.
These data demonstrate that in these six patients with CML
in chronic phase, Exodus markedly inhibited progenitor colony formation.
Exodus was much more effective than MIP-l~ in suppressing proliferation, suggesting that the effects of Exodus are mediated by a receptor that MIP-1~ does not activate CML progenitors overexpress the BCR-A13L fusion onco~ teill, a constitutively activated cytoplasmic tyrosine kinase that stimulates proliferation. In fact, forced over-expression of BCR-ABL in cell culture is transforming in many cell types, including NIH 3T3 cells.
The effect of Exodus on the cell cycle progression of progenitors from 20 three chronic phase CML patients was further explored using a tritiated thymidine kill assay as describecl above. Exodus treatment of the CML
progenitors arrested cell cycle progression in an average of 55 + 5 % of CFU-GM[, 45 ~ 13% of BFU-E, and 50 ~ 10% of CFU-GEMM. Thus, the Exodus inhibitory signal was able to overcome the aggressive 25 proliferative signal of BCR-ABL in CML progel.ilol~.
These results in~lic~te that Exodus suppresses hematopoiesis and may be effective for treating CML in chronic phase.

W O 98/21330 PCT~US97/20662_ Effect of Exodus on HIV Proliferation The ability of Exodus protein products to irhibit HIV
proliferation, as measured by HIV production of p24 protein, was tested 5 using a standard p24 T ~SA assay as previously described in Cocchi et al., Science, 270:1811 (1996). Normal volunteer human peripheral blood mononuclear cells were isolated on a Ficoll gradient. These cells were activated with 1 ng/ml PHA (Sigma, St. Louis, MO~ in RPMI 1640 (Life Technologies, Gaithersburg, MD) plus 10% FCS (Hyclone) and 100 U/ml 10 penicillin and 100 ~g/ml streptomycin (tissue culture antibiotics, Life Technologies, Gaithersburg, MD) for 48 hours at 37~ C, washed in complete media, then infected witll TCID50=5000 of the HIV strains BAL
(from ATCC) or A018-H112-2 (from ATCC) for 1 hour in complete media at 37~ C. Cells were then washed three times in media to remove excess virus, and resuspended at 5 x 105 cells/0.3 ml per test in complete media plus recombinant human IL-2 (10 ng/ml, Boehringer-~nnheim, Tn~ n~rolis, IN) plus recombinant Exodus or pECE-transfected COS cell supell.ala~ as controls. After six days of culture, cell-free supt;lndlallls were assessed for their content of HIV p24 using an enzyme-linked immunoabsorbent assay (ELISA, Abbott Laboratories, Chicago, IL).
Experiments were perfonned in triplicate.
Exodus-cont~ining COS cell supernatant at a final dilution of 1:2 (i.e., 0.15 ml of COS cell supernatant in 0.3 ml total in each well), Exodus-cont~inin~ COS cell supernatant at a final dilution of 1:4, MlP-1a~
at 625 ng/ml and 1250 ng/ml (bar 1 and bar 2, respectively, in Figure 7) and pECE COS cell supernatant (without Exodus) were measured at 6 days after infection. Results are shown in Figure 7. When normal human peripheral blood mononuclear cells stimulated by PMA were infected at a high multiplicity with two strains of HIV, Exodus was able to significantly inhibit HIV proliferation in both strains. At the highest concentration of W O 98/21330 PCT~US97/20662 _ recombinant Exodus used, proliferation of the ~IIV strain BAL was decreased to 39% of control, while proliferation of the HIV strain A018 was reduced to 27% of control. This inhibition was less marked when the concentration of Exodus was recl~lce~l In addition, this inhibition was S consistent with that seen with MIP-l(x, which was used as a positive control in these experiments. The inhibition by Exodus was not due to cytotoxicity, as there was no difference in the viability of cells treated with Exodus as with control cells.
SimiIar results for a purified synthetic Exodus protein product (Exodus with an additional alanine after residue 4) at a concentration of 1 ~g/ml and MIP-l(x at a concentration of 1 ,ug/ml are shown in Figure 8. Results are shown at 3, 6, and 9 days after infection.
At 9 days after infection, the inhibitory effect of Exodus was similar to that seen with MIP-lcY at the same concentration. In this preliminary experiment, no effects were seen with this Exodus protein product at concentrations less than 1 ,ug/mL.
These results infli(~.~te that Exodus protein products inhibit the proliferation of HIV, and will therefore be useful in methods of increasing re~i~t:~n~ e to HIV infection and methods of treating HIV
20 infection.

l~XAMPLE 12 Assay of ChPmo~ttractant and Cell-Activation Properties of Exodus on ~~ n Mo~lo~les/Macrophalges and ~llman N~ullu~llils The effects of Exodus upon human monocytestmacrophages or human neutrophils is evaluated, e.g., by methods described by Devi et al., J. Immunol., 153:5376-5383 (1995) for evaluating murine TCA3-ind~lced activation of neutrophils and macrophages. Indices of activation measured in such studies include increased adhesion to fibrinogen due to integrin activation, chemotaxis, induction of reactive nitrogen CA 02242384 Isss-07-os w o 98/21330 PCT~US97/Z0662 _ intermediates, respiratory burst (superoxide and hydrogen peroxide production), and exocytosis of lysozyme and elastase in the presence of cytochalasin B. As discussed by Devi et al., these activities correlate to several stages of the leukocyte response to infl~mm:~til n. This leukocyte response, reviewed by Springer, Cell, 76:301-314 (1994), involves adherence of leukocytes to endothelial cells of blood vessels, migration through the endothelial layer, chemotaxis toward a source of chemokines, and site-specific release of infl~mm~tory mediators. The involvement of Exodus at any one of these stages provides an important target for clinical intervention by mod~ tin~ the inflammatory response.

EX~PLE 13 ~xodus In Vivo Tunnor Growth Inhibition ~say Tumor growth-inhibition properties of Exodus are assayed, e.g., by modifying the protocol described by Laning et al., J. Immunol., 153:4625-4635 (1994) for assaying the tumor growth-inhibitory properties of murine TCA3. An Exodus-encoding cDNA is transfected by electroporation into the myeloma-derived cell line J558 (American Type Culhlre Collection, Rockville, MD). Transfectants are screened for Exodus production by standard techniques such as EI~SA (enzyme-linked immunoadsorbant assay) using a monoclonal antibody generated against Exodus as det~ilerl in Example 8. A bolus of 10 million cells from an Exodus-producing clone is injected subcutaneously into the lower right quadrant of BALB/c mice. For comparison, lQ million non-transfected cells are injected into control mice. The rate and frequency of tumor formation in the two groups is compared to determine efficacy of Exodus in inhibiting tumor growth. The nature of the cellular infiltrate subseqllently associated with the tumor cells is identified by histologic means. In addition, recombinant Lxodus (20 ng) is mixed with non-transfected J558 cells and injected ~20 ng/day) into tumors derived from W O 98/21330 PCTrUS97/20662._ such cells, to assay the effect of Exodus ~(lmini~tered exogenously to tumor cells.
-Intraperitoneal Injection Assay S The cells which respond to Exodus in vivo are determined through injection of 1-100 ng of purified Exodus into the intraperitoneal cavity ol~ mice, as described by ~uo et al., J. Immunol., 153:4616-4624 (1994). Following injection, leukocytes are isolated from peripheral blood and from the peritoneal cavity and identified by st~ining with the Diff 10 Quick kit (Baxter, McGraw, IL). The profile of leukocytes is measured at various times to assess the kinetics of appearance of different cell types. In separate experiments, neutralizing antibodies directed against Exodus (Example 8) are injected along with Exodus to confirrn that the infiltration of leukocytes is due to the activity of Exodus.

In vivo Activity Assay - Sub~--t~neous Injection The chemoattractant properties of Exodus are assayed in vivo by adapting the protocol described by Meurer et al., J. Exp. Med., 178:1913-1921 (1993). Recombinant Exodus (10-500 pmol/site) is injected 20 intradennally into a suitable m~mm~l, e.g., dogs or rabbits. At times of 4 to 24 hours, cell infiltration at the site of injection is ~ses~ed by histologicmethods. The presence of Exodus is confirrned by immunocytochemistry using antibodies directed against Exodus. The nature of the cellular infiltrate is identified by staining with Baxter's Diff Quick kit.

W O 98/21330 PCT~US97/20662 _ Cloning of an E~odus Receptor DNA encoding an Exodus receptor is cloned by adapting procedures previously described for isolation of the IL-8 receptor gene in 5 Holn1es et al., supra, and isolation of the MCP-l receptor gene in Charo et al., supra.
A cDNA library is ~ d, preferably from cells that respond to Exodus by chemotaxis and activation. Radiolabelled E~odus can also be used to identify cell types which express high levels of receptor 10 for Exodus. Cells which do not respond to MIP-l~Y or RANTES, or cells which show a different pattern of receptor desen~iti7~tion in response to these ligands are of particular interest. Pools of transfected clones in the cDNA library are screened for binding of radiolabelled Exodus by autoradiography. Positive pools are successively subfractionated and 15 rescreened until individual positive clones are obtained.
Alternatively, a degenerate PCR strategy may be used in which the sequences of the P~R primers are based on conserved regions of the sequences of known chemokine receptors. To increase the chance of isolating an Exodus receptor, the template DNA used in the reaction may 20 be cDNA derived from a cell typc rcsponsive to Exodus.

While the present invention has been described in terms of specific embodiments, it is understood that variations and modi~lcations will occur to those skilled in the art. Accordingly, only such limitations as appear in the appended claims should be placed on the invention.

CA 02242384 l998-07-09 WO 98/21330 PCTrUS97/20662 _ SEQUENCE LISTING

(1) GENER~L INFORMATION:
~i) APPLICANT: Indiana Univer6ity Foundation ~ii) TITLE OF lNvhNllON: EXODUS CHEMOKINE ~ATERIALS AND METHODS
(iii) NUMBER OF SEQUENCES: 4 (iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Mar6hall, O'Toole, Gerstein, Murray & Borun (B) STREET: 233 South Wacker Drive/ 6300 Sear6 Tower (C) CITY: Chlcago (D) STATE: Illinois (E) COUNTRY: United States o~ America (F) ZIP: 60606-6402 (v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy di6k (B) COMPUTER: IBM PC compatible (C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Relea6e #1.0, Version #1.30 (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/749,513 ~B) FILING DATE: 15-NOV-1996 (viii) Allo~N~y/AGENT INFORMATION:
(A) NAME: Rin-Laure6, Li-H6ien (B) REGISTRATION NUMBER: 33,547 (C) REh~N~/DOCKET NUMBER: 27866/34328 PCT
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (312) 474-6300 (B) TELEFAX: (312) 474-0448 (2) lN~o~vLATIoN FOR SEQ ID NO:1:
_ (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 821 ba6e pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: 8ingle (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (ix)--FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 43..327 ~ix) FEATURE:
(A) NAME/KEY: mat peptide (B) LOCATION: 109..327 W O 98/21330 PCTrUS97/20662 _ (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:
GGTACTCAAC ACTGAGCAGA lCl~ll~ll"l' GAGCTAAAAA CC ATG TGC TGT ACC 54 Met Cy~ Cys Thr Lys Ser Leu Leu Leu Ala Ala Leu Met Ser Val Leu Leu Leu His Leu Cy8 Gly Glu Ser Glu Ala Ser Asn Phe Asp Cy~ Cys Leu Gly Tyr Thr Asp Arg Ile Leu His Pro Lys Phe Ile Val Gly Phe Thr Arg Gln Leu GCC AAT GAA GGC TGT GAC ATC AAT GCT ATC ATC TTT CAC ACA AAG A~A 246 Ala A8n Glu Gly Cys Asp Ile Asn Ala Ile Ile Phe His Thr Lys Lys A~G TTG TCT GTG TGC GCA AAT CCA A~A CAG ACT TGG GTG A~A TAT ATT 294 Lys Leu Ser Val Cy~ Ala Asn Pro Lys Gln Thr Trp Val Lys Tyr Ile GTG CGT CTC CTC AGT A~A A~A GTC AAG AAC ATG TA~AAACTGT GC~lll~l~l~ 347 Val Arg Leu Leu Ser Lys Lys Val Ly8 Asn Met AATGAAGTTG ATTcATATTG CATCATAGTT TG~ll~ AAGC~TCACA TTAAAGTTAA 527 ACTGTATTTT ATGTTATTTA TAGCTGTAGG 'l"l"l"l'~l'G'l'~l~ TTAGCTATTT AATACTAATT 587 TTCCATAAGC TATTTTGGTT TAGTGCA~AG TATA~AATTA TATTTGGGGG GGAATA~GAT 647 TATATGGACT TTCTTGCAAG CAACAAGCTA ~ A~AA A~AACTATTT AACAll~ ll 707 TGTTTATATT ~~ ACT CCTAAATTGT TGTA~TTGCA TTATA~AATA AGA~AAATAT 767 TAATAAGACA AATATTGA~A ATA~AGA~AC A~AAAGTTCT T~l~G~ ~AAA A~AA 821 (2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 95 amino acid~
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECU~E TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
Met Cyg Cys Thr Ly~ Ser Leu Leu Leu Ala Ala Leu Met Ser Val Leu Leu Leu His Leu Cys Gly Glu Ser Glu Ala Ser A6n Phe A8p Cy~ Cys -5 l 5 10 W O 98/21330 PCT~US97/20662 Leu Gly Tyr Thr Asp Arg Ile Leu His Pro Lys Phe Ile Val Gly Phe -- Thr Arg Gln Leu Ala A~n Glu Gly Cys Asp Ile Asn Ala Ile Ile Phe Hi~ Thr Lys Lys Lys Leu Ser Val Cy~ Ala Asn Pro Ly~ Gln Thr Trp Val Ly8 Tyr Ile ~al Arg Leu Leu Ser Lys Lys Val Lys Asn Met (2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 26 ba~e pair~
(B) TYPE: nucleic acid (C) STRANDEDNESS: single - (D) TOPOLOGY: linear (ii) MO~ECuLE TYPE: nuclelc acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
GGCGAAGCTT TGAGCTA~AA ACCATG _ 26 (2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 26 base pairs (B) TYPE: nucleic acid (C) STR~NDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: nucleic acid (Xi ) ~U~N~ DESCRIPTION: SEQ ID NO:4:

Claims (26)

- 58 -What is claimed is:
1. A purified polynucleotide encoding the Exodus amino acid sequence of SEQ ID NO: 2.
2. The polynucleotide of claim 1 which is a DNA.
3. The DNA of claim 2 comprising a nucleotide sequence consisting of nucleotides 43 to 327 of SEQ ID NO:1.
4. A purified polynucleotide encoding amino acids 1 to 73 of SEQ ID NO:2.
5. The polynucleotide of claim 4 which is a DNA.
6. The DNA of claim 5 comprising a nucleotide sequence consisting of nucleotides 109 to 327 of SEQ ID NO: 1.
7. A vector comprising the DNA of claim 2, 3, 5 or 6.
8. The vector of claim 7 that is an expression vector, wherein the DNA is operatively linked to an expression control DNA
sequence.
9. A host cell stably transformed or transfected with the DNA of claim 2, 3, 5 or 6 in a manner allowing the expression in said host cell of Exodus.
10. A method for producing Exodus comprising culturing the host cell of claim 9 in a nutrient medium and isolating Exodus from said host cell or said nutrient medium.
11. A purified polypeptide produced by the method of claim 10.
12. A purified polypeptide comprising the Exodus amino acid sequence of SEQ ID NO: 2.
13. A purified polypeptide comprising Exodus amino acids 1 to 73 of SEQ ID NO: 2.
14. A hybridoma cell line producing a monoclonal antibody that is specifically reactive with the polypeptide of claim 15.
15. The monoclonal antibody produced by the hybridoma of claim 14.
16. A method of increasing resistance to human immunodeficiency virus (HIV) infection comprising administering to a subject an amount of Exodus protein product effective to inhibit HIV
proliferation.
17. The method of claim 16 wherein the subject is at risk of exposure to HIV, has been exposed to HIV, or has been infected with HIV.
18. A method of treating human immunodeficiency virus (HIV) infection comprising administering to a subject infected with HIV an amount of Exodus protein product effective to inhibit HIV proliferation.
19. Use of Exodus protein product in the preparation of a medicament for the prophylactic or therapeutic treatment of HIV infection.
20. The use according to claim 19 wherein the medicament inhibits HIV proliferation.
21. A method of protecting bone marrow progenitor cells from cytotoxic effects comprising administering to a subject an amount of Exodus protein product effective to suppress bone marrow progenitor cell proliferation.
22. The method of claim 21 wherein the subject is undergoing chemotherapy or radiotherapy.
23. Use of Exodus protein product in the preparation of a medicament for suppression of bone marrow progenitor cell proliferation.
24. The method of claim 23 wherein the medicament is administered to a subject undergoing chemotherapy or radiotherapy.

25. A method of treating myeloproliferative diseases comprising administering to a subject suffering from a myeloproliferative disease an amount of Exodus protein product effective to suppress malignant bone marrow progenitor cell proliferation.

26. The method of claim 25 wherein the myeloproliferative disease is chronic myelogenous leukemia, essential thrombocytosis, myelofibrosis, or polycythemia vera.
25. Use of Exodus protein product in the preparation of a medicament for treatment of myeloproliferative diseases.
26. The usc according to claim 25 wherein the myeloproliferative disease is chronic myelogenous leukemia, essential thrombocytosis, myelofibrosis, or polycythemia vera.
CA002242384A 1996-11-15 1997-11-14 Exodus chemokine materials and methods Abandoned CA2242384A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US74951396A 1996-11-15 1996-11-15
US08/749,513 1996-11-15

Publications (1)

Publication Number Publication Date
CA2242384A1 true CA2242384A1 (en) 1998-05-22

Family

ID=25014053

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002242384A Abandoned CA2242384A1 (en) 1996-11-15 1997-11-14 Exodus chemokine materials and methods

Country Status (14)

Country Link
US (1) US6096300A (en)
EP (1) EP0894134A1 (en)
JP (1) JP2001516203A (en)
CN (1) CN1213401A (en)
AU (1) AU5255398A (en)
BR (1) BR9707142A (en)
CA (1) CA2242384A1 (en)
CZ (1) CZ220298A3 (en)
HU (1) HUP0000316A2 (en)
IL (1) IL125351A0 (en)
NO (1) NO983238L (en)
PL (1) PL327853A1 (en)
SK (1) SK96398A3 (en)
WO (1) WO1998021330A1 (en)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6458349B1 (en) 1995-06-02 2002-10-01 Human Genome Sciences, Inc. Chemokine β-4 polypeptides
US6174995B1 (en) 1994-08-23 2001-01-16 Haodong Li Human chemokines, CKβ4 and CKβ10/MCP-4
US6391589B1 (en) 1994-08-23 2002-05-21 Human Genome Sciences, Inc. Human chemokine beta-10 mutant polypeptides
EP0979282A1 (en) * 1997-04-30 2000-02-16 F. Hoffmann-La Roche Ag Rat st38.2 chemokine
US20030144225A1 (en) * 2001-12-28 2003-07-31 Karras James G. Antisense modulation of macrophage inflammatory protein 3-alpha expression
AU6497799A (en) * 1998-10-15 2000-05-01 Canji, Inc. Methods and compositions to induce antitumor response
US6649158B1 (en) 1998-10-15 2003-11-18 Canji, Inc. Methods and compositions to induce antitumor response
AU2002228742A1 (en) * 2000-10-19 2002-04-29 Genaissance Pharmaceuticals, Inc. Haplotypes of the scya20 gene
ATE518884T1 (en) 2002-05-01 2011-08-15 Human Genome Sciences Inc ANTIBODIES SPECIFICALLY BINDING TO CHEMOKIN BETA-4
US7563810B2 (en) 2002-11-06 2009-07-21 Celgene Corporation Methods of using 3-(4-amino-1-oxo-1,3-dihydroisoindol-2-yl)-piperidine-2,6-dione for the treatment and management of myeloproliferative diseases
US8034831B2 (en) 2002-11-06 2011-10-11 Celgene Corporation Methods for the treatment and management of myeloproliferative diseases using 4-(amino)-2-(2,6-Dioxo(3-piperidyl)-isoindoline-1,3-dione in combination with other therapies
SG10201509499RA (en) 2010-11-19 2015-12-30 Eisai R&D Man Co Ltd Neutralizing anti-ccl20 antibodies

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5037630A (en) * 1985-01-14 1991-08-06 Neorx Corporation Metal radionuclide labeled proteins for diagnosis and therapy
US4965392A (en) * 1987-03-26 1990-10-23 Neorx Corporation Chelating compounds for metal-radionuclide labeled proteins
US5179078A (en) * 1989-05-12 1993-01-12 Dana Farber Cancer Institute Method of suppressing tumor formation in vivo
US5413778A (en) * 1992-10-05 1995-05-09 The Regents Of The University Of Michigan Labelled monocyte chemoattractant protein material and medical uses thereof
KR970705405A (en) * 1994-08-23 1997-10-09 벤슨 로버트 에이취 Human Chemokine Polypeptides &lt; RTI ID = 0.0 &gt;
US5602008A (en) * 1994-11-29 1997-02-11 Incyte Pharmaceuticals, Inc. DNA encoding a liver expressed chemokine
AU3574997A (en) * 1996-07-05 1998-02-02 Schering Corporation Mammalian chemokine reagents

Also Published As

Publication number Publication date
US6096300A (en) 2000-08-01
CZ220298A3 (en) 1999-01-13
PL327853A1 (en) 1999-01-04
HUP0000316A2 (en) 2000-07-28
IL125351A0 (en) 1999-03-12
CN1213401A (en) 1999-04-07
SK96398A3 (en) 1999-04-13
NO983238D0 (en) 1998-07-14
WO1998021330A1 (en) 1998-05-22
EP0894134A1 (en) 1999-02-03
BR9707142A (en) 1999-12-28
NO983238L (en) 1998-09-15
JP2001516203A (en) 2001-09-25
AU5255398A (en) 1998-06-03

Similar Documents

Publication Publication Date Title
Bozic et al. Expression and biologic characterization of the murine chemokine KC.
JP4502408B2 (en) CXCR3 chemokine receptor, antibody, nucleic acid and method of use
US5935567A (en) Cytokine IP-10 as an anti-tumor agent
AU708743B2 (en) Macrophage derived chemokine and chemokine analogs
US6498015B1 (en) Methods of identifying agents that modulate the binding between MDC and an MDC receptor
JP2009278990A (en) Cxcr3 chemokine receptor, antibody, nucleic acid and method of use
JP2002514887A (en) Method of secreting thrombopoietin polypeptide
EP0740701A1 (en) Mammalian monocyte chemoattractant protein receptors
US6096300A (en) Treatment of myeloproliferative disease with exodus chemokine
US5965697A (en) Disaggregated mutant human RANTES
US5688927A (en) Macrophage derived chemokine
AU733925B2 (en) Monocyte chemotactic protein-5 materials and methods
AU705064B2 (en) Novel polypeptides
US7018627B1 (en) Macrophage derived chemokine (MDC), MDC analogs, MDC inhibitor substances, and uses thereof
US6737513B1 (en) Macrophage derived chemokine (MDC) and chemokine analogs and assay to identify modulators of MDC activity, and therapeutic uses for same
US6737252B2 (en) 7 transmembrane receptor family member BLRX
AU1552602A (en) Exodus chemokine materials and methods
WO1998024907A1 (en) Novel protein with chemokine activity
US6320023B1 (en) Macrophage derived chemokine
KR100367213B1 (en) A cDNA ENCODING C6 β-CHEMOKINE LEUKOTACTIN-1(LKN-1) ISOLATED FROM HUMAN
MXPA98005598A (en) Exodus chemokine materials and methods
AU760296B2 (en) Macrophage derived chemokine (MDC), MDC analogs, MDC inhibitor sustances, and uses thereof
US6790947B1 (en) Polynucleotides encoding macrophage derived chemokine
AU779916B2 (en) CXCR3 chemokine receptor, antibodies, nucleic acids, and methods of use
MXPA98002380A (en) Short shapes of bet chemiscino

Legal Events

Date Code Title Description
FZDE Discontinued