Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HUMAN SPARC-HOMOLOGOUS (HSPARC-H1) GENE AND METHODS AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2002/002771
Kind Code:
A2
Abstract:
The present invention relates to at least one novel hSPARC-h1 polypeptide, isolated nucleic acids that encode such hSPARC-h1 polypeptides, as well as vectors, host cells, transgenics, chimerics, methods of making and using thereof, and hSPARC-h1 antibodies and uses thereof.

Inventors:
MITCHELL PAMELA JEAN (US)
SMITH ROSAMUND CAROL (US)
SU ERIC WEN (US)
Application Number:
PCT/US2001/014859
Publication Date:
January 10, 2002
Filing Date:
June 15, 2001
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
LILLY CO ELI (US)
MITCHELL PAMELA JEAN (US)
SMITH ROSAMUND CAROL (US)
SU ERIC WEN (US)
International Classes:
A61P17/02; A61P19/00; A61P21/00; A61P25/00; A61P35/00; C07K14/78; C12N15/12; A61K38/00; (IPC1-7): C12N15/12; A01K67/027; A61K38/39; C07K14/78; C07K16/18; C12N5/10; G01N33/50; G01N33/68
Domestic Patent References:
WO1999058675A21999-11-18
WO2000061755A22000-10-19
WO2002002603A22002-01-10
WO2001079454A12001-10-25
WO2001060850A12001-08-23
WO2001053455A22001-07-26
Other References:
DATABASE EMBL [Online] Mus musculus SPARC-related protein (SRG), 1 July 1999 (1999-07-01) Database accession no. AF070470 XP002205641 cited in the application
MOTAMED K: "SPARC (osteonectin/BM-40)." THE INTERNATIONAL JOURNAL OF BIOCHEMISTRY & CELL BIOLOGY, vol. 31, no. 12, December 1999 (1999-12), pages 1363-1366, XP002205640 ISSN: 1357-2725
Attorney, Agent or Firm:
Sharp, Robert L. (IN, US)
Davis, Paula K. (IN, US)
Download PDF:
Claims:
What is claimed :
1. An isolated nucleic acid comprising an hSPARChl polynucleotide encoding a polypeptide having a amino acid sequence as shown in SEQ ID NO : 4,5, or 6.
2. The isolated nucleic acid of claim 1 wherein said polynucleotide further comprises at least one mutation corresponding to at least one substitution, insertion or deletion selected from the group consisting of D6,7V, 25G, 42P, 45P, 48P, 78A, 80A, 161P, 218V, 225H, 239R, 300S, 301V, 302E, 364A, 377D, 432G, 433S, and 434L of SEQ ID NO : 4 or the corresponding amino acid of SEQ ID NO : 5.
3. An isolated nucleic acid complementary to the nucleic acid according to claim 1 or 2.
4. A composition comprising at least one isolated nucleic acid according to any of claims 13 and a carrier or diluent.
5. The composition according to claim 4 wherein said carrier or diluent is pharmaceutically acceptable.
6. A recombinant vector comprising at least one nucleic acid according to any of claims 13.
7. A host cell comprising the vector of claim 6.
8. A method for producing a hSPARChl polypeptide comprising culturing a host cell according to claim 7 under conditions such that said hSPARChl polypeptide is expressed in detectable or recoverable amounts.
9. A transgenic or chimeric nonhuman animal comprising the nucleic acid of claim 1,2, or 3.
10. An isolated polypeptide comprising an hSPARChl polypeptide having an amino acid sequence as shown in SEQ ID NO : 4,5, or 6.
11. The polypeptide of claim 10 further comprising at least one mutation corresponding to at least one substitution, insertion, or deletion selected from the group consisting of D6,7V, 25G, 42P, 45P, 48P, 78A, 80A, 161P, 218V, 225H, 239R, 300S, 301V, 302E, 364A, 377D, 432G, 433S, and 434L of SEQ ID NO : 4 or the corresponding amino acid of SEQ ID NO : 5.
12. A composition comprising a carrier or diluent and at least one isolated polypeptide according to claim 10 or 11.
13. The composition of claim 11 wherein said carrier or diluent is pharmaceutically acceptable.
14. An antibody that binds an epitope specific to at least one hSPARChl polypeptide according to claim 10 or 11.
15. A host cell expressing the antibody of claim 14.
16. A method for identifying compounds that bind at least one isolated hSPARChl polypeptide according to any of claims 10 or 11 comprising (a) admixing said polypeptide with at least one test compound or composition; and (b) detecting at least one binding interaction between said polypeptide and the test compound or composition.
17. A compound or composition detected by the method according to claim 16.
18. Use of a hSPARChl agonist for the manufacture of a medicament to treat or prevent a disease, condition, or disorder in a mammal selected from the group consisting of: a. Alzheimer's disease; b. GullainBarre syndrome; c. Parkinson's disease; d. multiple sclerosis; e. epilepsy; f. schizophrenia; g. sarcopenia; h. periodontal disease; i. cartilage degradation or trauma; j. wounds; k. tissue atrophy; 1. neuronal degeneration; and m. neuronal trauma.
19. Use of a hSPARChl antagonist for the manufacture of a medicament to treat or prevent a disease, condition, or disorder in a mammal selected from the group consisting of: a. Alzheimer's disease; b. GullainBarre syndrome; c. Parkinson's disease; d. multiple sclerosis; e. epilepsy; f. schizophrenia; and g. tumor.
20. The use of a hSPARChl polypeptide according to claim 10 or 11 for the manufacture of a medicament to treat or prevent a disease, condition, or disorder in a mammal selected from the group consisting of: a. Alzheimer's disease; b. GullainBarre syndrome; c. Parkinson's disease; d. multiple sclerosis; e. epilepsy; f. schizophrenia; g. sarcopenia; h. periodontal disease; i. cartilage degradation or trauma; j. wounds; k. tissue atrophy; 1. neuronal degeneration; and m. neuronal trauma.
21. Use of a SPARChl antibody for the manufacture of a medicament to treat or prevent a disease, condition, or disorder in a mammal selected from the group consisting of: a. Alzheimer's disease; b. GullainBarre syndrome; c. Parkinson's disease; d. multiple sclerosis; e. epilepsy; f. schizophrenia; and g. tumor.
Description:
NOVEL hSPARC-hl GENE AND METHODS AND USES THEREOF BACKGROUND OF THE INVENTION FIELD OF THE INVENTION The present invention relates to compounds and compositions comprising novel human SPARC-homologous (hSPARC-h) polypeptides, nucleic acids, host cells, transgenics, chimerics, antibodies, compositions, and methods of making and using thereof.

RELATED ART Matricellular proteins comprise a non-homologous group of extra-cellular regulatory macromolecules that mediate cell matrix interactions but may not contribute significantly to extra-cellular matrix structure (Bornstein, P., J. Cell Biol., 130: 503-506 (1995)). The matricellular class of secreted glycoproteins includes SPARC, thrombospondins 1 and 2, tenascins C and X, and osteopontin.

Although structurally unrelated, these molecules appear to perform related functions, e. g., they exhibit counter- adhesive effects that lead to cell rounding and changes in cell shape that result in the disruption of cell-matrix interactions.

Events that are characterized by changes in cell shape and motility, e. g., tissue renewal, tissue remodeling, and embryonic development often require expression of these proteins. However, the matricellular proteins are different from traditional extra-cellular matrix proteins such as fibronectin, laminin, fibrillar collages, and vitronectin, all of which are adhesive proteins and contribute to the structural stability of the extra-cellular matrix.

SPARC is the prototype of the matricellular proteins.

While SPARC is expressed at high levels in bone tissue, it

is also distributed widely in other tissues and cell types (Maillard, C., et al., Bone, 13: 257-264 (1992)). SPARC is associated generally with remodeling tissues, e. g., tissues undergoing morphogenesis, mineralization, angiogenesis, tumorigenesis, and pathological responses to injury.

Experiments in vitro have also identified SPARC in tumors (Schulz, A., et al., Am. J. Pathol., 132: 233-238 (1988); Porter, P. L., et al., J. Histochem. Cytochem., 43: 791-800 (1995)) and in tissues involved in repair and turnover. The high levels of SPARC expressed in the adult eye relative to the immature eye (Yan, Q., et al., J. Histo. Cytochem., 46: 3-10 (1998)) indicate that the protein might play an important role in the maintenance of ocular physiological functions. Interestingly, SPARC-null mice of three genetic backgrounds all exhibit a predominant phenotype: opacity of the lens, or cataract (Gilmour, D. T., et al., EMBO J., 17: 1860-1870 (1998); Norose, K., et al., Invest. Ophthalmolb Vis. Sci. 39: 2674-2680 (1998)).

Additionally, SPARC may have neuronal activity including at least one activity such as, but not limited to, promoting or inhibiting neurite outgrowth and/or neurite adhesion (Kolodkin, A., et al., Neuron, 21: 1079-1092, (1998); Kolodkin, et al., (1997); Wilson et al., J. Cell Sci. 109: 3129-3138 (1996); Pimenta et al., Neuron, 15: 287- 297 (1995)), inducing neural regeneration, inhibiting neural degeneration, preventing seizures, reducing frequency and/or severity of seizures, promoting or inhibiting primary or secondary sexual development, and altering behavioral patterns including, but not limited to, sleep and eating disorders.

In view of the overall importance of extracellular matrix proteins in tissue remodeling, tissue repair, general modulation of various growth factor activities, and neuronal activity, there is a need to provide novel human SPARC-like

polypeptides, nucleic acids, host cells, transgenics, chimerics, as well as methods of making and using such.

Accordingly, we provide here hSPARC-hl, a novel human SPARC- like protein.

SUMMARY OF THE INVENTION The present invention provides isolated nucleic acids and hSPARC-hl polypeptides encoded thereby, including specified fragments and variants thereof, as well as hSPARC- hl compositions, probes, primers, vectors, host cells, antibodies, transgenics, chimerics and methods of making and using thereof, as described and enabled herein.

Having the cloned hSPARC-hl gene enables the production of recombinant hSPARC-hl proteins, the isolation of homologous genes from other organisms, and/or related genes from the same organism, chromosome mapping studies, and the implementation of large scale screens to identify compounds that bind said protein and modulate the activity thereof.

The proteins disclosed herein are also useful, among other things, to treat cellular proliferation and/or neurological disorders.

The present invention provides, in one aspect, isolated nucleic acid molecules comprising or complementary to a polynucleotide encoding specific hSPARC-hl polypeptides, as well as fragments or specified variants comprising at least one domain thereof.

Such polypeptides are provided as non-limiting examples by the corresponding domains, specified fragments, and/or specified variants of hSPARC-hl polypeptides corresponding to at least 90-100% of the contiguous amino acids of at least one of SEQ ID NO : 4 or 5.

The present invention further provides recombinant vectors, comprising 1-40 of said isolated hSPARC-hl nucleic

acid molecules of the present invention, host cells containing'such nucleic acids and/or recombinant vectors, as well as methods of making and/or using such nucleic acid, vectors and/or host cells.

The present invention also provides methods of making or using such nucleic acids, vectors and/or host cells, such as, but not limited to, using them for the production of hSPARC-hl nucleic acids and/or polypeptides by known recombinant, synthetic and/or purification techniques, based on the teaching and guidance presented herein in combination with what is known in the art.

The present invention also provides an isolated hSPARC- hl polypeptide, comprising at least one fragment, domain, or specified variant of at least 90-100% of the contiguous amino acids of at least one portion of at least one of SEQ ID NO : 4 or 5.

In another embodiment the present invention relates to an isolated protein molecule, or functional fragment thereof, wherein said protein molecule comprises the sequence identified as SEQ ID NO : 4 or 5. Examples of functional fragments of preference include polypeptides comprising SEQ ID NO : 4 wherein said polypeptide lacks from 1 to 26 amino acid residues from the amino terminus of SEQ ID NO : 4 or from 1 to 50 amino acid residues from the carboxy- terminus of SEQ ID NO : 4. More preferable functional fragments are polypeptides comprising SEQ ID NO: 4 wherein said polypeptide lacks from 1 to 26 amino acid residues from the amino-terminus of SEQ ID NO : 4 and from 1 to 90 amino acid residues from the carboxy-terminus of SEQ ID NO : 4. A most preferred functional fragment is a polypeptide comprising amino acid residues 26-446 of SEQ ID NO : 4.

The present invention also provides an isolated hSPARC- hl polypeptide as described herein, wherein the polypeptide further comprises at least one specified substitution,

insertion or deletion corresponding to portions or residues of at least one of SEQ ID NO : 4 or 5.

The present invention also provides an isolated hSPARC- hl polypeptide as described herein, wherein the polypeptide has at least one activity, such as, but not limited to, promoting or inhibiting angiogenesis, neovascularization, tumorigenesis, cataractogenesis, wound healing, growth- factor mediated chemotaxis, neurite outgrowth and/or neurite adhesion.

The present invention also provides a composition comprising an isolated hSPARC-hl nucleic acid, polypeptide, and/or a antibody of the present invention as described herein and a carrier or diluent. The carrier or diluent can optionally be pharmaceutically acceptable, according to known methods.

The present invention also provides an isolated nucleic acid probe, primer or fragment, as described herein, wherein the nucleic acid comprises a polynucleotide of at least 10 nucleotides, corresponding or complementary to at least 10 nucleotides of at least one of SEQ ID NOS: 1, 2, or 3.

The present invention also provides a recombinant vector comprising an isolated hSPARC-hl nucleic acid as described herein.

The present invention also provides a host cell, comprising an isolated hSPARC-hl nucleic acid as described herein.

The present invention also provides a method for constructing a recombinant host cell that expresses an hSPARC-hl polypeptide, comprising introducing into the host cell an hSPARC-hl nucleic acid in replicatable form as described herein to provide the recombinant host cell. The present invention also provides a recombinant host cell provided by a method as described herein.

The present invention also provides a method for expressing at least one hSPARC-hl polypeptide in a recombinant host cell, comprising culturing a recombinant host cell as described herein under conditions wherein at least one hSPARC-hl polypeptide is expressed in detectable or recoverable amounts.

The present invention also provides an isolated hSPARC- hl polypeptide produced by a recombinant, synthetic, and/or any suitable purification method as described herein and/or as known in the art.

The present invention also provides an hSPARC-hl antibody or fragment, comprising a polyclonal and/or monoclonal antibody or fragment that specifically binds at least one epitope specific to at least one hSPARC-hl polypeptide as described herein.

The present invention also provides a method for producing an hSPARC-hl antibody or antibody fragment, comprising generating the antibody or fragment that binds at least one epitope that is specific to an isolated hSPARC-hl polypeptide as described herein, the generating done by known recombinant, synthetic and/or hybridoma methods.

The present invention also provides an hSPARC-hl antibody or fragment produced by a method as described herein or as known in the art.

The invention also encompasses nucleotide sequences that can be used to inhibit hSPARC-hl gene expression (e. g., antisense and ribozyme molecules, and gene or regulatory sequence replacement constructs) or to enhance hSPARC-hl gene expression (e. g., expression constructs that place the hSPARC-hl gene under the control of a strong promoter system), and transgenic animals that express hSPARC-hl transgene or"knock-outs"that do not express hSPARC-hl.

The present invention provides compounds and pharmaceutical compositions comprising hSPARC-hl nucleic

acids, polypeptides, any fragments or variants thereof, and/or anti-hSPARC-hl antibodies, for use in methods for treating or preventing tumorigenicity and/or other disorders associated with aberrant hSPARC-hl activity in mammals in need thereof.

The present invention also provides a method of inducing or inhibiting angiogenesis, neovascularization, tumorigenesis, cataractogenesis, wound healing, growth- factor mediated chemotaxis, neurite outgrowth, and/or neurite adhesion in a patient in need thereof wherein said method comprises administering to said patient a therapeutically effective amount of a composition comprising an isolated hSPARC-hl nucleic acid, polypeptide, and/or anti-hSPARC-hl antibody as described herein and a carrier or diluent. The carrier or diluent can optionally be pharmaceutically acceptable, according to known methods.

The present invention also provides a method for identifying compounds that bind an hSPARC-hl polypeptide, comprising a) admixing at least one isolated hSPARC-hl polypeptide as described herein with a test compound or composition; and b) detecting at least one binding interaction between the polypeptide and the compound or composition, optionally further comprising detecting a change in biological activity, such as a reduction or increase.

In still a further embodiment, the invention relates to hSPARC-hl antagonists and/or agonist molecules. In one aspect, the invention provides a method of screening compounds that mimic the activity of hSPARC-hl polypeptides (agonists) or diminish the effect of hSPARC-hl activity (antagonists).

DESCRIPTION OF THE INVENTION Citations All publications or patents cited herein are entirely incorporated herein by reference as they show the state of the art at the time of the present invention to provide description and enablement of the present invention.

Publications refer to scientific, patent publication, or any other information available in any media format, including all recorded, electronic or printed formats. The following citations are also entirely incorporated by reference: Ausubel, et al., eds., Current Protocols in Molecular Biology, John Wiley & Sons, N. Y., N. Y. (1987-1998); Coligan et al., eds., Current Protocols in Protein Science, John Wiley & Sons, Inc., N. Y., N. Y. (1995-1999); Sambrook, et al., Molecular Cloning: A Laboratory Manual, 2nd Edition, Cold Spring Harbor, N. Y. (1989); Harlow and Lane, Antibodies, a Laboratory Manual, Cold Spring Harbor, N. Y.

(1989); Coligan, et al., eds., Current Protocols in Immunology, John Wiley & Sons, N. Y., N. Y. (1992-1999).

Definitions The following definitions of terms are intended to correspond to those known in the art. The following terms are therefore not limited to the definitions given, but are used according to the state of the art, as demonstrated by cited and/or contemporary publications or patents.

"Active"or"activity"for the purposes herein refers to form (s) of hSPARC-hl which retain the biologic and/or immunologic activities of native or naturally-occurring hSPARC-hl polypeptide. Elaborating further,"activity"or "biological activity"in regards to hSPARC-hl refers to a biological function (either inhibitory or stimulatory) caused by a native or naturally-occurring hSPARC-hl other

than the ability to induce the production of an antibody against an antigenic epitope possessed by a native or naturally-occurring hSPARC-hl. An"immunological"activity refers only to the ability to induce the production of an antibody against an antigenic epitope possessed by a native or naturally occurring hSPARC-hl. The term"activity"or the phrase"biological activity"in reference to an hSPARC- hl agonist or antagonist relates to the capacity of the particular agent to induce or inhibit, respectively, in vivo and/or in vitro, the biological consequences associated with hSPARC-hl by the present disclosure. Preferred biological activities of hSPARC-hl agonists or antagonists as disclosed herein include, but are not limited to, the induction and/or inhibition of angiogenesis, neovascularization, tumor- igenesis, cataractogenesis, rate of wound healing, growth- factor mediated chemotaxis, neurite outgrowth, and/or neurite adhesion. Accordingly, such activities can be assessed by one or more of the in vitro or in vivo assays disclosed herein or otherwise known in the art.

The term"amino acid"is used herein in its broadest sense, and includes naturally occurring amino acids as well as non-naturally occurring amino acids, including amino acid analogs and derivatives. The latter includes molecules containing an amino acid moiety. One skilled in the art will recognize, in view of this broad definition, that reference herein to an amino acid includes, for example, naturally occurring proteogenic L-amino acids; D-amino acids; chemically modified amino acids such as amino acid analogs and derivatives; naturally occurring non-proteogenic amino acids such as norleucine, 0-alanine, ornithine, and chemically synthesized compounds having properties known in the art to be characteristic of amino acids.

The incorporation of non-natural amino acids, including

synthetic non-native amino acids, substituted amino acids, or one or more D-amino acids into the hSPARC-hl analogs of the present invention is advantageous in a number of different ways. D-amino acid-containing polypeptides exhibit increased stability in vitro or in vivo compared to L-amino acid-containing counterparts. Thus, the construction of polypeptides incorporating D-amino acids can be particularly useful when greater stability is desired or required in vivo. More specifically, D-peptides are resistant to endogenous peptidases and proteases, thereby providing improved bioavailability of the molecule, and prolonged lifetimes in vivo when such properties are desirable. When it is desirable to allow the peptide to remain active for only a short period of time, the use of L- amino acids therein will permit endogenous peptidases, proteases to digest the molecule, thereby limiting the cell's exposure to the molecule. Additionally, D-peptides cannot be processed efficienty for major histocompatibility complex class II-restricted presentation to T helper cells, and are therefore less likely to induce humoral immune responses in the whole organism.

In addition to using D-amino acids, those of ordinary skill in the art are aware that modifications in the amino acid sequence of a peptide, polypeptide, or protein can result in equivalent, or possibly improved, second generation peptides that display equivalent or superior functional characteristics when compared to the original amino acid sequences. Alterations in the hSPARC-hl analogs of the present invention can include one or more amino acid insertions, deletions, substitutions, truncations, fusions, shuffling of subunit sequences, and the like, either from natural mutations or human manipulation, provided that the sequences produced by such modifications have substantially

the same (or improved or reduced, as may be desirable) activity (ies) as the hSPARC-hl analog sequences disclosed herein. The term"hSPARC-hl analog"refers to any modified form of a hSPARC-hl polypeptide that exhibits substantially the same or enhanced biological activity in vivo and/or in vitro as compared to the corresponding unmodified form and is pharmaceutically more desirable, in at least one aspect, as compared to the corresponding unmodified hSPARC-hl polypeptide. As used herein, the term"hSPARC-hl analog"is intended to encompass hSPARC-hl polypeptides as defined herein wherein the hSPARC-hl polypeptide further comprises at least one modification not normally native to hSPARC-hl polypeptides. The term"modification"includes any change in structure (ie., a qualititive change) of a protein. Such modifications can include, but are not limited to, changes in the amino acid sequence, transcriptional or translational splice variation, pre-or post-translational modifications to the DNA or RNA sequence, addition of macromolecules or small molecules to the DNA, RNA or protein, such as peptides, ions, vitamins, atoms, sugar-containing molecules, lipid-containing molecules, small molecules and the like, as well-known in the art. One type of protein modification according to the present invention is by one or more changes in the amino acid sequence (substitution, deltion or insertion). Such changes could include, at one or more amino acids, a change from a charged amino acid to a different charged amino acid, a non-charged to a charged amino acid, a charged amino acid to a non-charged amino acid as discussed, infra. or supra. Any other change in amino acid sequence is also included in the invention. Another type of protein modification is by changes in processing of the protein in the cell. A non-limiting example is where some proteins have an"address label"specifying where in

(or outside of) the cell they should be used. Such a label or tag can be in the form of a peptide, a sugar or a lipid, which when added or removed from the protein, determines where the protein is located in the cell. A further type of protein modification is due to the attachment of other macromolecules to a protein. This group can include, but is not limited to, any addition/removal of such a macromolecule. These molecules can be of many types and can be either permanent or temporary. Examples include: (i) polyribosylation, (ii) DNA/RNA (single or double stranded); (iii) lipids and phosphlipids (e. g., for membrane attachment); (iv) saccharides/polysaccharides; and (v) glycosylation (addition of different types of sugar and sialic acids--in a variety of single and branched structures). Another type of protein modification is due to the attachment of other small molecules to proteins.

Examples can include, but are not limited to: (i) phosphorylation ; (ii) acetylation ; (iii) uridylation; (iv) adenylation ; (v) methylation, and (vi) capping (diverse complex modification of the N-terminus of the protein for assorted reasons). Most of these changes are often used to regulate a protein's activity. (v) and (vi) are also. used to change the half-life of the protein itself. These protein changes can be detected on 2 dimensional gel electrophoresis incorporating several methods, such as labeling, changes in pI, antibodies or other specific techniques directed to the molecules themselves, as known in the art. Molecular weight changes can be, but may not usually be detectable by 2DGE.

MALD (matrix assisted laser desorption of flight mass spectrometry) is preferred to detect and characterize these modifications. Such modifications are generally directed at improving upon the poor therapeutic character of the native hSPARC-hl polypeptide by increasing that molecule's target specificity, solubility, stability, serum half-life,

affinity for targeted receptors, susceptibility to proteolysis, resistance to clearing in vivo, ease of purification, and/or decreasing the antigenicity and/or required frequency of administration.

The term"antagonist"is used in the broadest sense, and includes any molecule that partially or fully blocks, inhibits, or neutralizes a biological activity of a native hSPARC-hl polypeptide disclosed herein.

The term"agonist"is used similarly in the broadest sense and includes any molecule that mimics a biological activity of a native hSPARC-hl polypeptide disclosed herein.

Suitable agonists and antagonists specifically include agonistic and antagonistic antibodies, respectively, or antibody fragments thereof. Human SPARC-hl agonists also specifically include hSPARC-hl polypeptides of the present invention as well as fragments and/or polypeptide variants thereof, and small organic molecules. Methods for identifying agonists or antagonists of an hSPARC-hl polypeptide may comprise contacting an hSPARC-hl polypeptide with a candidate agonist or antagonist molecule and measuring a detectable change in one or more biological activities normally associated with the hSPARC-hl polypeptide.

The term"antibody"is also meant to include both intact molecules as well as fragments thereof, such as, for example, Fab and Fab', which are capable of binding antigen.

Fab and Fab', fragments lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody (Wahl et al., J. Nucl. Med., 24: 316-325 (1983)).

It will be appreciated that Fab and F (ab') 2 and other fragments of the antibodies useful in the present invention may be used for the detection and quantitation of a hSPARC-h protein or glycoprotein according to methods disclosed

herein for intact antibody molecules. Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F (ab') 2 fragments).

The terms"complementary"or"complementarity"as used herein refer to the capacity of purine, pyrimidine, synthetic or modified nucleotides to associate by partial or complete complementarity through hydrogen or other bonding to form partial or complete double-or triple-stranded nucleic acid molecules. The following base pairs occur by complete complementarity: (i) guanine (G) and cytosine (C); (ii) adenine (A) and thymine (T); and adenine (A) and uracil (U)."Partial complementarity"refers to association of two or more bases by one or more hydrogen bonds or attraction that is less than the complete complementarity as described above. Partial or complete complementarity can occur between any two nucleotides, including naturally occurring or modified bases, e. g., as listed in 37 CFR § 1.822. All such nucleotides are included in polynucleotides of the invention as described herein.

"Conservative substitution"or"conservative amino acid substitution"refers to a replacement of one or more amino acid residue (s) in a protein or peptide as stipulated in Table 1.

The term"deletion"refers to the omission of one or more amino acid residue (s) in a protein or peptide, herein represented by a"D"preceding the number of the amino acid which is being deleted from the protein or peptide.

"Functional fragment"or"functionally equivalent fragment", as used herein, refers to a region, or fragment of a full length protein, or sequence of amino acids that, for example, comprises an active site, or any other conserved motif, relating to biological function.

Functional fragments are capable of providing a biological

activity substantially similar to a full-length protein disclosed herein. Functional fragments may be produced by cloning technology, or as the natural products of alternative splicing mechanisms.

The term"fusion protein"denotes a hybrid protein molecule not found in nature comprising a translational fusion or enzymatic fusion in which two or more different proteins or fragments thereof are covalently linked on a single polypeptide chain. The term"polypeptide"also includes such fusion proteins.

As used herein"half-life"refers to the time required for approximately half of the molecules making up a population of said molecules to be cleaved in vitro or in vivo. More specifically,"plasma half-life"refers to the time required for approximately half of the molecules making up a population of said molecules to be removed from circulation or be, otherwise, rendered inactive in vivo.

The term"homolog"or"homologous"describes the relationship between different nucleic acid molecules or amino acid sequences such that said sequences or molecules are related by partial identity or similarity at one or more regions within said molecules or sequences.

The term"hSPARC-hl"refers to a polynucleotide, or amino acid sequence encoded thereby, encoding a novel secreted matricellular proteins as disclosed herein that is related to mouse SPARC-related gene (mSRG ; AF07047). SPARC is a prototypical matricellular protein known to be associated with various physiological processes such as the regulation of cell adhesion, cell migration, proliferation, counter adhesion, and growth factor expression and/or growth factor activity in numerous tissues.

More specifically, the term"hSPARC-hl polypeptide" when used herein encompass native sequence hSPARC-hl

polypeptides and polypeptide fragments and/or variants thereof (which are further defined herein).

"Host cell"refers to any eucaryotic, procaryotic, or fusion or other cell or pseudo cell or membrane-containing construct that is suitable for propagating and/or expressing an isolated nucleic acid that is introduced into a host cell by any suitable means known in the art (e. g., but not limited to, transformation or transfection, or the like), or induced to express an endogenous nucleic acid encoding an hSPARC-hl polypeptide according to the present invention.

The cell can be part of a tissue or organism, isolated in culture or in any other suitable form.

The term"hybridization"as used herein refers to a process in which a partially or completely single-stranded nucleic acid molecule joins with a complementary strand through nucleotide base pairing. Hybridization can occur under conditions of low, moderate or high stringency, with high stringency preferred. The degree of hybridization depends upon, for example, the degree of homology, the stringency conditions, and the length of hybridizing strands as known in the art.

In the present disclosure,"isolated"refers to material removed from its original environment (e. g., the natural environment if it is naturally occurring), and thus is altered"by the hand of man"from its natural state. For example, the term"isolated"in reference to a polypeptide refers to a polypeptide that has been identified and separated and/or recovered from at least one contaminant from which it has been produced. Contaminants may include cellular components, such as enzymes, hormones, and other proteinaceous or non-proteinaceous solutes. Ordinarily, however, isolated polypeptides will be prepared by at least one purification step.

The term"isolated"in reference to a nucleic acid

compound refers to any specific RNA or DNA molecule, however constructed or synthesized or isolated, which is locationally distinct from its natural location. For example, an isolated polynucleotide could be part of a vector or a composition of matter, or could be contained within a cell, and still be"isolated"because that vector, composition of matter, or particular cell is not the original environment of the polynucleotide.

An"isolated"antibody is one that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or non-, proteinaceous solutes. Ordinarily, an isolated antibody is prepared by at least one purification step. In preferred embodiments, the antibody will be purified (1) to greater than 95% by weight of antibody as determined by the Lowry method, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue, or preferably, silver stain.

An"isolated antibody"is also intended to mean an antibody that is substantially purified from other antibodies having different antigenic specificities. An isolated antibody that specifically binds hSPARC-hl epitopes may bind hSPARC-hl homologous molecules from other species.

The term"isolated"may be used interchangeably with the phrases"substantially pure"or"substantially purified"in reference to a macromolecule that is separated from other cellular and non-cellular molecules, including other proteins, lipids, carbohydrates or other materials with which it is

naturally associated when produced recombinantly or synthesized without any general purifying steps. A "substantially pure"or"isolated"protein as described herein could be prepared by a variety of techniques well known to the skilled artisan. In preferred embodiments, a polypeptide will be isolated or substantialy purified upon purification (1) to greater than 85% by weight of polypeptide to the weight of total protein as determined by the Lowry method, and most preferably to more than 95% by weight of polypeptide to the weight of total protein, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to apparent homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie Blue, or preferably, silver stain, such that the major band constitutes at least 85%, and, more preferably 95%, of stained protein observed on the gel.

The term"mature protein"or"mature polypeptiden as used herein refers to the form (s) of the protein produced by expression in a mammalian cell. It is generally hypothesized that once export of a growing protein chain across the rough endoplasmic reticulum has been initiated, proteins secreted by mammalian cells have a signal peptide (SP) sequence which is cleaved from the complete polypeptide to produce a"mature"form of the protein. Oftentimes, cleavage of a secreted protein is not uniform and may result in more than one species of mature protein. The cleavage site of a secreted protein is determined by the primary amino acid sequence of the complete protein and generally can not be predicted with complete accuracy.

Methods for predicting whether a protein has a SP sequence, as well as the cleavage point for that sequence, are available. The analysis of the amino acid sequence of the proteins described herein indicated the cleavage point is after amino acid 20-30, preferably between 26-27, as

presented in SEQ ID NO : 4. The mature protein can be represented by, but not limited to, SEQ ID NO: 5. As one of ordinary skill would appreciate, however, cleavage sites sometimes vary from organism to organism and cannot be predicted with absolute certainty. Accordingly, the present invention provides polypeptides having a sequence of 90-100% of the contiguous sequence shown in SEQ ID NO : 4 which have an N-terminus beginning within 10 residues (i. e., + or-10 residues) of the predicted cleavage point between amino acid 26 and 27 of SEQ ID NO : 4. However, cleavage sites for a secreted protein may be determined experimentally by amino- terminal sequencing of the one or more species of mature proteins found within a purified preparation of the protein.

A"nucleic acid probe,""oligonucleotide probe,"or "probe"as used herein comprises at least one detectably labeled or unlabeled nucleic acid which hybridizes under specified hybridization conditions with at least one other nucleic acid. This term also refers to a single-or partially double-stranded nucleic acid, oligonucleotide or polynucleotide that will associate with a complementary or partially complementary target nucleic acid to form at least a partially double-stranded nucleic acid molecule. A nucleic acid probe may be an oligonucleotide or a nucleotide polymer. A probe can optionally contain a detectable moiety which may be attached to the end (s) of the probe or be internal to the sequence of the probe, termed a"detectable probe"or"detectable nucleic acid probe." The term"plasmid"refers to an extrachromosomal genetic element. The plasmids disclosed herein are commercially available, publicly available on an unrestricted basis, or can be constructed from readily available plasmids in accordance with published procedures.

A"polynucleotide"comprises at least 10-20 nucleotides of a nucleic acid (RNA, DNA or combination thereof), provided

by any means, such as synthetic, recombinant isolation or purification method steps.

A"primer"is a nucleic acid fragment or oligonucleotide which functions as an initiating substrate for enzymatic or synthetic elongation of, for example, a nucleic acid molecule, e. g., using an amplification reaction, such as, but not limited to, a polymerase chain reaction (PCR), as known in the art.

The term"promoter"refers to a nucleic acid sequence that directs transcription, for example, of DNA to RNA. An inducible promoter is one that is regulatable by environmental signals, such as carbon source, heat, or metal ions, for example. A constitutive promoter generally operates at a constant level and is not regulatable.

"Recombinant DNA cloning vector"as used herein refers to any autonomously replicating agent, including, but not limited to, plasmids and phages, comprising a DNA molecule to which one or more additional DNA segments can or have been incorporated.

The term"recombinant DNA expression vector"or "expression vector"as used herein refers to any recombinant DNA cloning vector, for example a plasmid or phage, in which a promoter and other regulatory elements are present thereby enabling transcription of an inserted DNA, which may encode a protein.

The term"resistant"or more specifically"protease- resistant"or"glycosylation resistant"refers to a hSPARC- hl analog that is more resistant to proteolysis or glycosylation relative to native hSPARC-hl as shown in SEQ ID NO : 2. Protease or glycosylation resistant analogs may differ from hSPARC-hl by one or more amino acid substitutions, deletions, inversions, additions, and/or other changes at any site susceptible to proteolysis or glycosylation. The term"resistant"contemplates degrees of

resistance to at each of the different susceptible sites from complete resistance to partial resistance. Thus, a "substantially resistant"analog shows a degree of resistance a particular susceptible position such that the number of analogs cleaved or glycosylated at any particular position is at least about 25% fewer than the number of native hSPARC-hl molecules cleaved or glycosylated when similarly treated. Preferably a substantially protease resistant hSPARC-hl analog possesses a half-life that is at least about 2-fold greater than the corresponding native hSPARC-hl polypeptide. Similarly, a glycosylation resistant hSPARC-hl analog exhibits a clearance rate that is at least about 2-fold slower than the clearance rate of the corresponding native hSPARC-hl polypeptide Susceptibility to proteolysis will depend on such factors as the amino acid sequence at or near the recognition site of the particular proteolytic enzyme involved, and on the physical and chemical environment in which a sample protein is located. Factors such as these can affect the KM and/or rate of proteolysis by a proteolytic enzyme. The charge density and steric properties operative at the enzymes active site will also determine the degree to which proteolysis occurs. Susceptibility to glycosylation will depend on the amino acid sequence and the presence or absence of glycosylation motifs that are known in the art.

The term"stability"in reference to a hSPARC-hl polypeptide and/or hSPARC-hl analog may refer to its half- life in vivo, in serum, and/or in solution.

The term"stringency"refers to hybridization conditions for nucleic acids in solution. High stringency conditions disfavor non-homologous base pairing. Low stringency conditions have much less of this effect.

Stringency may be altered, for example, by changes in

temperature and/or salt concentration, or other conditions, as well known in the art.

A non-limiting example of"high stringency"conditions includes, for example, (a) a temperature of about 42 °C, a formamide concentration of about 20%, and a low salt (SSC) concentration, or, alternatively, a temperature of about 65 °C, or less, and a low salt (SSPE) concentration; (b) hybridization in 0.5 M NaHP04, 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 65 °C (see, e. g., Ausubel, et al., ed., Current Protocols in Molecular Biology, 1987-1998, Wiley Interscience, New York, at § 2.10.3)."SSC"comprises a hybridization and wash solution. A stock 20X SSC solution contains 3M sodium chloride, 0.3 M sodium citrate, pH 7.0.

"SSPE"comprises a hybridization and wash solution. A 1X SSPE solution contains 180 mM NaCl, 9 mM Na2HP04, 0.9 mM NaH2PO4 and 1 mM EDTA, pH 7.4.

The term"variant"in reference to a hSPARC-hl polypeptide means an hSPARC-hl polypeptide having at least about 80% amino acid sequence identity to a full-length or mature native sequence of hSPARC-hl polypeptide as shown in SEQ ID NO : 4 or 5, respectively and further having at least one of the activities associated with said full-length or mature native hSPARC-hl polypeptide.

The term"variant"in reference to a hSPARC-hl polynucleotide means an active hSPARC-hl polypeptide- encoding nucleic acid molecule as defined below having at least about 65% nucleic acid sequence identity with at least one of the hSPARC-hl-encoding nucleotide sequences shown in SEQ ID NOS: 1,2, or 3. Variants specifically exclude or do not encompass the native nucleotide sequence, as well as those prior art sequences that share 100% identity with the nucleotide sequences of the invention.

"Percent (%) nucleic acid sequence identity"with respect to the hSPARC-hl sequences identified herein is defined as the percentage of nucleotides in a candidate sequence that are identical with the nucleotides in the hSPARC-hl sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent nucleic acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as ALIGN, Align-2, Megalign (DNASTAR), or BLAST (e. g., Blast, Blast-2) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For. purposes herein, however, % nucleic acid identity values are generated using the WU-BLAST-2 (BlastN module) computer program (Altschul et al., Methods in Enzymology m: 460-480 (1996). Most of the WU-BLAST-2 search parameters are set to the default values. Those not set default values, i. e., the adjustable parameters, are set with the following values: overlap span = 1, overlap fraction = 0.125, word threshold (T) = 11 and scoring matrix = BLOSUM62. For purposes herein, a % nucleic acid sequence identity value is determined by dividing (a) the number of matching identical nucleotides between the nucleic acid sequence of the PRO polypeptide-encoding nucleic acid molecule of interest and the comparison nucleic acid molecule of interest (i. e., the sequence against which the PRO polypeptide-encoding nucleic acid molecule of interest is being compared) as determined by WU-BLAST-2 by (b) the total number of nucleotides of the hSPARC-hl polypeptide- encoding nucleic acid molecule of interest.

"Percent (%) amino acid sequence identity"with respect to the hSPARC-hl amino acid sequences identified herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in an hSPARC-hl polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as ALIGN, ALIGN-2, Megalign (DNASTAR) or BLAST (e. g., Blast, Blast-2, WU-Blast-2) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For example, the % identity values used herein are generated using WU-BLAST-2 (Altschul et al., Methods in Enzymology 266: 460-480 (1996)). Most of the WU-BLAST-2 search parameters are set to the default values. Those not set to default values, i. e., the adjustable parameters, are set with the following values: overlap span = 1. overlap fraction = 0.125: word threshold (T) = 11, and scoring matrix = BLOSUM 62. For purposes herein, a % amino acid sequence identity value is determined by divided (a) the number of matching identical ammo acid residues between the amino acid sequence of the hSPARC-hl polypeptide of interest and the comparison amino acid sequence of interest (i. e., the sequence against which the hSPARC-hl polypeptide of interest is being compared) as determined by WU-BLAST-2 by (b) the total number of amino acid residues of the hSPARC-hl polypeptide of interest, respectively.

The term"vector"as used herein refers to a nucleic acid compound used for introducing exogenous or endogenous nucleic acid into host cells. A vector comprises a nucleotide sequence which may encode one or more polypeptide molecules. Plasmids, cosmids, viruses and bacteriophages, in a natural state or which have undergone recombinant engineering, are non-limiting examples of commonly used vectors to provide recombinant vectors comprising at least one desired isolated nucleic acid molecule.

Pharmaceutical terms The term"administer"or"administering"means to introduce by any means a therapeutic agent into the body of a mammal in order to prevent or treat a disease or condition.

"Chronic"administration refers to administration of the agent (s) in a continuous mode as opposed to an acute mode, so as to maintain the initial therapeutic effect (activity) for an extended period of time."Intermittent" administration is treatment that is not consecutively done without interruption, but rather is cyclic in nature.

Administration"in combination with"one or more further therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order.

A"biologically-effective amount"is the minimal amount of a compound or agent that is necessary to impart a biological consequence to the extent that the biological consequence is measurable either directly or indirectly.

Such determinations are routine and within the skill of an ordinarily skilled artisan.

A"therapeutically-effective amount"is the minimal amount of a compound or agent that is necessary to impart therapeutic benefit to a mammal. By administering graduated levels of a hSPARC-hl polypeptide or hSPARC-hl analog to a

mammal in need thereof, a clinician skilled in the art can determine the therapeutically effective amount of the hSPARC-hl polypeptide or hSPARC-hl analog required for administration in order to treat or prevent the diseases, condition, disorders, and/or at least one symptom thereof, discussed herein. Such determinations are routine in the art and within the skill of an ordinarily skilled clinician.

"Carriers"as used herein include pharmaceutically acceptable carriers, excipients, or stabilizers which are nontoxic to the cell or mammal being exposed thereto at the dosages and concentrations employed. Often the physio- logically acceptable carrier is an aqueous pH buffered solution. Examples of physiologically acceptable carriers include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecule weight (less than about 10 residues) polypeptides ; proteins, such as serum albumin, gelatin, or immuno- globulins; hydrophilic polymers such as polyvinyl- pyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEEN@, polyethylene glycol (PEG), and PLURONICSTM.

"Pharmaceutically acceptable salt"includes, but is not limited to, salts prepared with inorganic acids, such as chloride, sulfate, phosphate, diphosphate, hydrobromide, and nitrate salts, or salts prepared with an organic acid, such as malate, maleate, fumarate, tartrate, succinate, ethylsuccinate, citrate, acetate, lactate, methanesulfonate, benzoate, ascorbate, para-toluenesulfonate, palmoate, salicylate and stearate, as well as estolate, gluceptate and

lactobionate salts. Similarly, salts containing pharmaceutically acceptable cations include, but are not limited to, sodium, potassium, calcium, aluminum, lithium, and ammonium (including substituted ammonium).

The term"mammal"as used herein refers to any mammal, including humans, domestic and farm animals, and zoo, sports or pet animals, such as cattle (e. g. cows), horses, dogs, sheep, pigs, rabbits, goats, cats, and non-domesticated animals like mice and rats. In a preferred embodiment of the present invention, the mammal being treated or administered to is a human or mouse.

A"small molecule"is defined herein to have a molecular weight below about 500 daltons.

The terms"treating","treatment"and"therapy"as used herein refer to curative therapy, prophylactic therapy, and preventative therapy. An example of"preventative therapy" is the prevention or lessening of a targeted disease or related condition thereto. Those in need of treatment include those already with the disease or condition as well as those prone to have the disease or condition to be prevented. The terms"treating","treatment", and"therapy" as used herein also describe the management and care of a mammal for the purpose of combating a disease, or related condition, and includes the administration of hSPARC-hl polypeptides or hSPARC-hl analogs to alleviate the symptoms or complications of said disease, condition. Treating as used herein also includes the administration of the protein for cosmetic purposes.

A"therapeutically-effective amount"is the minimal amount of a compound or agent that is necessary to impart therapeutic benefit to a mammal. By administering graduated levels of a hSPARC-hl polypeptide or hSPARC-hl analog to a mammal in need thereof, a clinician skilled in the art can determine the therapeutically effective amount of the

hSPARC-hl polypeptide or hSPARC-hl analog in order to treat or prevent a particular disease condition, or disorder when it is administered, such as intravenously, subcutaneously, intraperitoneally, orally, or through inhalation. The precise amount of the compound required to be therapeutically effective will depend upon numerous factors, e. g., such as the specific binding activity of the compound, the delivery device employed, physical'characteristics of the compound, purpose for the administration, in addition to patient specific considerations. The amount of a compound that must be administered to be therapeutically effective are routine in the art and within the skill of an ordinarily skilled clinician.

All references herein to a disease, condition, or disorder are contemplated to encompass other diseases, conditions, disorders, and/or symptoms that are generally associated with that particular disease, condition, or disorder by the medical community.

The various restriction enzymes disclosed and described herein are commercially available and the manner of use of said enzymes including reaction conditions, cofactors, and other requirements for activity are well known to one of ordinary skill in the art. Reaction conditions for particular enzymes were carried out according to the manufacturer's recommendation.

Nucleic Acid Molecules The present invention provides isolated, recombinant and/or synthetic nucleic acid molecules comprising at least one polynucleotide encoding at least one hSPARC-hl polypeptide comprising specific full length sequences, fragments and specified variants thereof, such polypeptides, and methods of making and using said nucleic acids and

polypeptides thereof. An hSPARC-hl polypeptide of the invention comprises at least one fragment, domain, and/or specified variant of any portion or fragment of any hSPARC- hl protein as described herein.

The present invention also provides at least one utility by providing isolated nucleic, acids comprising polynucleotides of sufficient length and complementarity to an hSPARC-hl nucleic acid for use as probes or amplification primers in the detection, quantitation, or isolation of gene sequences or transcripts. For example, isolated nucleic acids of the present invention can be used as probes for detecting deficiencies in the level of mRNA, in screens for detection of mutations in at least one hSPARC-hl gene (e. g., substitutions, deletions, or additions), or for monitoring upregulation of expression of said gene, or changes in biological activity as described herein in screening assays of compounds, and/or for detection of any number of allelic variants (polymorphisms or isoforms) of the gene.

The isolated nucleic acids of the present invention can also be used for recombinant expression of hSPARC-hl polypeptides for use as immunogens in the preparation and/or screening of antibodies. The isolated nucleic acids of the present invention can also be employed for use in sense or antisense suppression of one or more hSPARC-hl genes or nucleic acids, in a host cell, or tissue in vivo or in vitro.

Attachment of chemical agents which bind, intercalate, cleave and/or crosslink to the isolated nucleic acids of the present invention can also be used. to modulate transcription or translation of at least one nucleic acid disclosed herein.

Using the information provided herein, such as the nucleotide sequences encoding at least 80-100% of the contiguous amino acids of at least one of SEQ ID NOS: 4 or 5, specified fragments or variants thereof, or a deposited vector comprising at least one of these sequences, a nucleic

acid molecule of the present invention encoding an hSPARC-hl polypeptide can be obtained using well-known methods.

Nucleic acid molecules of the present invention can be in the form of RNA, such as mRNA, hnRNA, tRNA or any other form, or in the form of DNA, including, but not limited to, cDNA and genomic DNA obtained by cloning or produced synthetically, or any combination thereof. The DNA can be triple-stranded, double-stranded or single-stranded, or any combination thereof. Any portion of at least one strand of the DNA or RNA can be the coding strand, also known as the sense strand, or it can be the non-coding strand, also referred to as the anti-sense strand.

Isolated nucleic acid molecules of the present invention include nucleic acid molecules comprising an open reading frame (ORF),. shown in at least one of SEQ ID NOS: 1, 2,3; nucleic acid molecules comprising the coding sequence for an hSPARC-hl polypeptide; and nucleic acid molecules which comprise a nucleotide sequence substantially different from those described above but which, due to the degeneracy of the genetic code, still encode at least one hSPARC-hl polypeptide as described herein. Of course, the genetic code is well known in the art. Thus, it would be routine for one skilled in the art to generate such degenerate nucleic acid variants that code for specific hSPARC-hl polypeptides of the present invention. See, e. g., Ausubel, et al., supra, and such nucleic acid variants are included in the present invention.

In one embodiment, the isolated nucleic acid comprises DNA having at least about 80% sequence identity, preferably at least about 81% sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at

least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity. yet more preferably at least about 91% sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity. yet more preferably at least about 97% sequence identity, yet more preferably at least about 98% sequence identity, yet more preferably at least about 99% sequence identity to (a) a DNA molecule encoding an hSPARC-hl polypeptide comprising the sequence of amino acid residues from 1 or about to 26 through amino acid residue 446, inclusive, of SEQ ID NO: 4 or (b) the complement of the DNA molecule of (a). Alternatively, the isolated nucleic acid comprises DNA encoding a hSPARC-hl polypeptide having the sequence of amino acid residues from about 1 to about 446, inclusive, of SEQ ID NO: 4.

In another aspect, the invention concerns an isolated nucleic acid molecule encoding an hSPARC-hl polypeptide comprising DNA hybridizing to the complement of the nucleic acid between about residues: (a) 1 to about 1338, inclusive, of SEQ ID NO : 2 and (b) 1 to about 1275, inclusive, of SEQ ID NO : 3.

In another aspect, the invention concerns an isolated nucleic acid molecule encoding an active hSPARC-h polypeptide comprising a nucleotide sequence that hybridizes to the complement of a nucleic acid sequence that encodes amino acids (a) 1 or about 10 to about 26, inclusive, SEQ ID NO : 4 or (b) 1 or about 27 to about 446, inclusive, of SEQ ID

NO : 4. Preferably, hybridization occurs under stringent hybridization and wash conditions.

In a further aspect, the invention concerns an isolated nucleic acid molecule comprising DNA having at least about 80% sequence identity, preferably at least about 81% sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98% sequence identity, yet more preferably at least about 99% sequence identity to a DNA molecule encoding the same mature polypeptide encoded by the polynucleotide sequence as shown in SEQ ID NO : 1.

In a further aspect, the invention concerns an isolated nucleic acid molecule produced by hybridizing a test DNA molecule under stringent conditions with: (a) a DNA molecule encoding (i) an hSPARC-hl polypeptide having the sequence of amino acid residues from about 1 or about 27 to about 446, inclusive, of SEQ ID NO : 4, or (b) the complement of the DNA molecule of (a), and if the DNA molecule has at least about an 80% sequence identity, preferably at least about an 81%

sequence identity, more preferably at least about a 82% sequence identity, yet more preferably at least about a 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91% sequence identity. yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98% sequence identity, yet more preferably at least about 99% sequence identity to (a) or (b), and isolating the test DNA molecule.

In yet a further aspect, the invention concerns an isolated nucleic acid molecule comprising: (a) DNA encoding a polypeptide scoring at least about 80% positives, preferably at least about 81% positives, more preferably at least about 82% positives, yet more preferably at least about 83% positives, yet more preferably at least about 84% positives, yet more preferably at least about 85% positives, yet more preferably at least about 86% positives, yet more preferably at least about 87% positives, yet more preferably at least about 88% positives, yet more preferably at least about 89% positives, yet more preferably at least about 90% positives, yet more preferably at least about 91% positives, yet more preferably at least about 92% positives, yet more preferably at least about 93% positives, yet more preferably

at least about 94% positives, yet more preferably at least about 95% positives, yet more preferably at least about 96% positives, yet more preferably at least about 97% positives, yet more preferably at least about 98% positives, yet more preferably at least about 99% positives, when compared with the amino acid sequence of residues about 1 to about 446, inclusive, of SEQ ID NO : 4, or (b) the complement of the DNA of (a). In a specific aspect, the invention provides an isolated nucleic acid molecule comprising DNA encoding an hSPARC-hl polypeptide without the N-terminal signal sequence and/or initiating methionine, or is complementary to such encoding nucleic acid molecule. The signal peptide has been tentatively identified as extending from about amino acid residue (a) 1 to about amino acid residue 26, inclusive, in the sequence of SEQ ID NO : 4.

The invention also provides an isolated nucleic acid molecule having the nucleotide sequence shown in at least one of SEQ ID NOS : 1, 2,3, or the nucleotide sequence of the hSPARC-hl cDNA contained in at least one of the above- referenced deposited clones, or a nucleic acid molecule having a sequence complementary thereto. Such isolated molecules, particularly nucleic acid molecules, are useful as probes for gene mapping by in situ hybridization with chromosomes, and for detecting transcription, translation and/or expression of the hSPARC-hl gene in human tissue, for instance, by Northern blot analysis for mRNA detection.

Unless otherwise indicated, all nucleotide sequences identified by sequencing a nucleic acid molecule herein can be or were identified using an automated nucleic acid sequencer, and all amino acid sequences of polypeptides encoded by nucleic acid molecules identified herein can be or were identified by codon correspondence or by translation of a nucleic acid sequence identified using method steps as described herein or as known in the art. Therefore, as is

well known in the art that for any nucleic acid sequence identified by this automated approach, any nucleotide sequence identified herein may contain some errors which are reproducibly correctable by resequencing based upon an available or a deposited vector or host cell containing the nucleic acid molecule using well-known methods.

Nucleotide sequences identified by automation are typically at least about 95% to at least about 99.999% identical to the actual nucleotide sequence of the sequenced nucleic acid molecule. The actual sequence can be more precisely identified by other approaches including manual nucleic acid sequencing methods well known in the art. As is also known in the art, a single insertion or deletion in an identified nucleotide sequence compared to the actual sequence will cause a frame shift in translation of the nucleotide sequence such that the identified amino acid sequence encoded by an identified nucleotide sequence will be completely different from the amino acid sequence actually encoded by the sequenced nucleic acid molecule, beginning at the point of such an insertion or deletion.

Nucleic Acid Fragments The present invention is further directed to fragments of a hSPARC-hl-encoding polynucleotide sequence that may find use as, for example, hybridization probes or for encoding fragments of an hSPARC-hl polypeptide that may optionally encode a polypeptide comprising a binding site for an anti-hSPARC-hl antibody using the methods disclosed herein.

Other useful fragments of the hSPARC-hl nucleic acids include antisense or sense oligonucleotides comprising a single-stranded nucleic acid sequence (either RNA or DNA) capable of binding to target hSPARC-hl mRNA (sense) of hSPARC-

hi DNA (anti-sense) sequences. Antisense or sense oligo- nucleotides, according to the present invention, comprise a fragment of the coding region of hSPARC-hl DNA. Such a fragment generally comprises at least about 14 nucleotides, preferably from about 14 to 30 nucleotides. The ability to derive an antisense or a sense oligonucleotide, based upon a cDNA sequence encoding a given protein is described in, for example. Stein and Cohen, Cancer Res., 48 (10): 2659-2668 (1988) and van der Krol et al., Bio/Techniques, 6 (10): 958-976 (1988).

Binding of antisense or sense oligonucleotides to target nucleic acid sequences results in the formation of duplexes that block transcription or translation of the target sequence by one of several means, including enhanced degradation of the duplexes, premature termination of transcription or translation, or by other means. The antisense oligonucleotides thus may be used to block expression of hSPARC-hl proteins. Antisense or sense oligonucleotides further comprise oligonucleotides having modified sugar- phosphodiester backbones (or other sugar linkages, such as those described in WO 91/06629) and wherein such sugar linkages are resistant to endogenous nucleases. Such oligonucleotides with resistant sugar linkages are stable in vivo (i. e., capable of resisting enzymatic degradation) but retain sequence specificity to be able to bind to target nucleotide sequences.

Other examples of sense or antisense oligonucleotides include those oligonucleotides which are covalently linked to organic moieties, such as those described in WO 90/10448, and other moieties that increase affinity of the oligonucleotide for a target nucleic acid sequence, such poly-L-lysine.

Further still, intercalating agents, such as ellipticine, and alkylating agents or metal complexes may be attached to sense or antisense oligonucleotides to modify binding specificities

of the antisense or sense oligonucleotide for the target nucleotide sequence. Antisense or sense oligonucleotides may be introduced into a cell containing the target nucleic acid sequence by any gene transfer method, including, for example, CaP04-mediated DNA transfection, electroporation, or by using gene transfer vectors such as Epstein-Barr virus. In a preferred procedure, an antisense or sense oligonucleotide is inserted into a suitable retroviral vector. A cell containing the target nucleic acid sequence is contacted with the recombinant retroviral vector, either in vivo or ex vivo.

Suitable retroviral vectors include, but are not limited to, those derived from the murine retrovirus M-MuLV, N2 (a retrovirus derived from M-MuLV), or the double copy vectors designated DCT5A, DCT5B and DCT5C (see WO 90/13641).

Sense or antisense oligonucleotides also may be introduced into a cell containing the target nucleotide sequence by formation of a conjugate with a ligand binding molecule, as described in WO 91/04753. Suitable ligand binding molecules include, but are not limited to, cell surface receptors, growth factors, other cytokines, or other ligands that bind to cell surface receptors. Preferably, conjugation of the ligand binding molecule does not substantially interfere with the ability of the ligand binding molecule to bind to its corresponding molecule or receptor, or block entry of the sense or antisense oligonucleotide or its conjugated version into the cell. Alternatively, a sense or an antisense oligonucleotide may be introduced into a cell containing the target nucleic acid sequence by formation of an oligonucleotide-lipid complex, as described in WO 90/10448.

The sense or antisense oligonucleotide-lipid complex is preferably dissociated within the cell by an endogenous lipase.

By a fragment of an isolated nucleic acid molecule is meant a molecule having at least 10 nucleotides of a nucleotide sequence of a deposited cDNA or a nucleotide sequence shown in at least one of SEQ ID NOS: 1, 2,3, and is intended to mean fragments at least about 10 nucleotides, which are useful, inter alia as diagnostic probes and primers as described herein. By a fragment at least 10 nucleotides in length, for example, is intended fragments which include 10 or more contiguous nucleotides from the nucleotide sequence of the deposited cDNA or the nucleotide sequence as shown in SEQ ID NOS: 1, 2,3, as determined by methods known in the art (See e. g., Ausubel, supra, Chapter 7). Of course, larger fragments such as at least about 50, 100,120,200,500,1000,1500,2000,2500,3000,3500, and/or 4000 or more nucleotides in length, are also useful according to the present invention as are fragments corresponding to most, if not all, of the nucleotide sequence as shown at least one of SEQ ID NOS: 1, 2,3, or any previously referenced deposited cDNA. However, such nucleic acids fragments are usually at least about 20 nucleotides in length, preferably at least about 30 nucleotides in length, more preferable at least about 40 nucleotides in length, yet more preferably at least about 50 nucleotides in length, yet more preferably at least about 60 nucleotides in length, yet more preferably at least about 70 nucleotides in length, yet more preferably at least about 50 nucleotides in length, yet more preferably at least about 90 nucleotides in length, yet more preferably at least about 100 nucleotides in length, yet more preferably at least about 110 nucleotides in length, yet more preferably at least about 120 nucleotides in length, yet more preferably at least about 130 nucleotides in length, yet more preferably at least about 140 nucleotides in length, yet

more preferably at least about 150 nucleotides in length, yet more preferably at least about 160 nucleotides in length, yet more preferably at least about 170 nucleotides in length, yet more preferably at least about 180 nucleotides in length, yet more preferably at least about 190 nucleotides in length, yet more preferably at least about 200 nucleotides in length, yet more preferably at least about 250 nucleotides in length, yet more preferably at least about 300 nucleotides in length, yet more preferably at least about 350 nucleotides in length, yet more preferably at least about 400 nucleotides in length, yet more preferably at least about 450 nucleotides in length, yet more preferably at least about 500 nucleotides in length, yet more preferably at least about 600 nucleotides in length, yet more preferably at least about 700 nucleotides in length, yet more preferably at least about 800 nucleotides in length, yet more preferably at least about 900 nucleotides in length, yet more preferably at least about 1000 nucleotides in length, wherein in this context"about"means the referenced nucleotide sequence length plus or minus 10% of that referenced length.

Such nucleotide fragments are also useful according to the present invention for screening DNA, cDNA, or mRNA sequences that code for one or more fragments of an hSPARC- hl polypeptide as described herein. Such screening, as a non-limiting example can include the use of so-called"DNA chips"for screening DNA, cDNA, or mRNA sequences of the present invention, as described, e. g., in U. S. Patent Nos.

5,631,734,5,624,711,5,744,305,5,770,456,5,770,722, 5,675,443,5,695,940,5,710,000,5,733,729, which are entirely incorporated herein by reference.

Of course, a polynucleotide which hybridizes only to a poly A sequence (such as the 3'terminal poly (A) of an hSPARC-hl cDNA shown in at least one of SEQ ID NOS: 1, 2,3,

or to a complementary stretch of T (or U) resides, would not be included in a probe of the invention, since such a polynucleotide would hybridize to any nucleic acid molecule containing a poly (A) stretch or the complement thereof (e. g., practically any double-stranded cDNA clone).

The present invention also provides subsequences of full-length nucleic acids. Any number of subsequences can be obtained by reference to at least one of SEQ ID NOS: 1, 2,3, or a complementary sequence, and using primers which selectively amplify, under stringent conditions to: at least two sites to the polynucleotides of the present invention, or to two sites within the nucleic acid which flank and comprise a polynucleotide of the present invention, or to a site within a polynucleotide of the present invention and a site within the nucleic acid which comprises it. A variety of methods for obtaining 5'and/or 3'ends is well known in the art. See, e. g., RACE (Rapid Amplification of Complementary Ends) as described in M. A. Frohman, PCR Protocols: A Guide to Methods and Applications, M. A. Innis, et al, Eds., Academic Press, Inc., San Diego, CA, pp. 28-38 (1990); see also, U. S. Patent No. 5,470,722, and Ausubel, et al., Current Protocols in Molecular Biology, Chapter 15, Eds., John Wiley & Sons, N. Y. (1989-1999). Thus, the present invention provides hSPARC-hl polynucleotides having the sequence of the hSPARC-hl gene, nuclear transcript, cDNA, or complementary sequences and/or subsequences thereof.

Primer sequences can be obtained by reference to a contiguous subsequence of a polynucleotide of the present invention. Primers are chosen to selectively hybridize, under PCR amplification conditions, to a polynucleotide of the present invention in an amplification mixture comprising a genomic and/or cDNA library from the same species.

Generally, the primers are complementary to a subsequence of the amplified nucleic acid. In some embodiments, the primers

will be constructed to anneal at their 5'terminal ends to the codon encoding the carboxy or amino terminal amino acid residue (or the complements thereof) of the polynucleotides of the present invention. The primer length in nucleotides is selected from the group of integers consisting of from at least 15 to 50. Thus, the primers can be at least 15,18, 20,25,30,40, or 50 nucleotides in length or any range or value therein. A non-annealing sequence at the 5'end of the primer (a"tail") can be added, for example, to introduce a cloning site at the terminal ends of the amplified DNA.

The amplification primers may optionally be elongated in the 3'direction with additional contiguous or complementary nucleotides from the polynucleotide sequences, such as shown in at least one of SEQ ID NOS: 1, 2,3, from which they are derived. The number of nucleotides by which the primers can be elongated is selected from the group of integers consisting of from at least 1 to at least 25. Thus, for example, the primers can be elongated with an additional 1, 5,10, or 15 nucleotides or any range or value therein.

Those of skill will recognize that a lengthened primer sequence can be employed to increase specificity of binding (i. e., annealing) to a target sequence, or to add useful sequences, such as links or restriction sites (See e. g., Ausubel, supra, Chapter 15).

The amplification products can be translated using expression systems well known to those of skill in the art and as discussed, infra. The resulting translation products can be confirmed as polypeptides of the present invention by, for example, assaying for the appropriate catalytic activity (e. g., specific activity and/or substrate specificity), or verifying the presence of one or more linear epitopes which are specific to a polypeptide of the present invention.

Methods for protein synthesis from PCR derived templates are

known in the art (See e. g., Ausubel, supra, Chapters 9,10, 15; Coligan, Current Protocols in Protein Science, supra, Chapter 5) and available commercially. See, e. g., Amersham Life Sciences, Inc., Catalog'97, p. 354.

Polynucleotides which Selectively Hybridize to a Polynucleotide as Described Herein The present invention provides isolated nucleic acids that hybridize under selective hybridization conditions to a polynucleotide disclosed herein, e. g., SEQ ID NO : 1. Thus, the polynucleotides of this embodiment can be used for isolating, detecting, and/or quantifying nucleic acids comprising such polynucleotides. For example, polynucleotides of the present invention can be used to identify, isolate, or amplify partial or full-length clones in a deposited library. In some embodiments, the polynucleotides are genomic or cDNA sequences isolated, or otherwise complementary to, a cDNA from a human or mammalian nucleic acid library.

Preferably, the cDNA library comprises at least 80% full-length sequences, preferably at least 85% or 90% full- length sequences, and more preferably at least 95% full- length sequences. The cDNA libraries can be normalized to increase the representation of rare sequences. Low stringency hybridization conditions are typically, but not exclusively, employed with sequences having a reduced sequence identity relative to complementary sequences.

Moderate and high stringency conditions can optionally be employed for sequences of greater identity. Low stringency conditions allow selective hybridization of sequences having about 70% sequence identity and can be employed to identify orthologous or paralogous sequences.

Optionally, polynucleotides of this invention will encode an epitope of a polypeptide described herein. The polynucleotides of this invention embrace nucleic acid sequences that can be employed for selective hybridization to a polynucleotide encoding a polypeptide of the present invention.

Screening polypeptides for specific binding to antibodies or fragments can be conveniently achieved using peptide display libraries. This method involves the screening of large collections of peptides for individual members having the desired function or structure. Antibody screening of peptide display libraries is well known in the art. The displayed peptide sequences can be from 3 to 5000 or more amino acids in length, frequently from 5-100 amino acids long, and often from about 8 to 15 amino acids long.

In addition to direct chemical synthetic methods for generating peptide libraries, several recombinant DNA methods have been described. One type involves the display of a peptide sequence on the surface of a bacteriophage or cell.

Each bacteriophage or cell contains the nucleotide sequence encoding the particular displayed peptide sequence. Such methods are described in PCT Patent Publication Nos.

91/17271,91/18980,91/19818, and 93/08278. Other systems for generating libraries of peptides have aspects of both in vitro chemical synthesis and recombinant methods. See, PCT Patent Publication Nos. 92/05258,92/14843, and 96/19256.

See also, U. S. Patent Nos. 5,658,754; and 5,643,768. Peptide display libraries, vector, and screening kits are commercially available from such suppliers as Invitrogen (Carlsbad, CA).

Polynucleotides Complementary to the Polynucleotides

As indicated above, the present invention provides isolated nucleic acids comprising hSPARC-hl polynucleotides, wherein the polynucleotides are complementary to the polynucleotides described herein. As those of skill in the art will recognize, complementary sequences base pair throughout the entirety of their length with such polynucleotides (i. e., have 100% sequence identity over their entire length). Complementary bases associate through hydrogen bonding in double-stranded nucleic acids. For example, the following base pairs are complementary: guanine and cytosine; adenine and thymine; and adenine and uracil (See, e. g., Ausubel, supra, Chapter 67; or Sambrook, supra).

Construction of Nucleic Acids The isolated nucleic acids of the present invention can be made using (a) standard recombinant methods, (b) synthetic techniques, (c) purification techniques, or combinations thereof, as well known in the art.

The nucleic acids may conveniently comprise sequences in addition to a polynucleotide of the present invention. For example, a multi-cloning site comprising one or more endonuclease restriction sites may be inserted into the nucleic acid to aid in isolation of the polynucleotide.

Also, translatable sequences may be inserted to aid in the isolation of the translated polynucleotide of the present invention. For example, a hexa-histidine marker sequence provides a convenient means to purify the proteins of the present invention. The nucleic acid of the present invention -excluding the polynucleotide sequence-is optionally a vector, adapter, or linker for cloning and/or expression of a polynucleotide of the present invention.

Additional sequences may be added to such cloning and/or expression sequences to optimize their function in cloning

and/or expression, to aid in isolation of the polynucleotide, or to improve the introduction of the polynucleotide into a cell. Typically, the length of a nucleic acid of the present invention less the length of its polynucleotide of the present invention is less than 20 kilobase pairs, often less than 15 kb, and frequently less than 10 kb. Use of cloning vectors, expression vectors, adapters, and linkers is well known in the art. (See, e. g., Ausubel, supra, Chapters 1-5; or Sambrook, supra) Recombinant Methods for Constructing Nucleic Acids The isolated nucleic acid compositions of this invention, such as RNA, cDNA, genomic DNA, or a hybrid thereof, can be obtained from biological sources using any number of cloning methodologies known to those of skill in the art. In some embodiments, oligonucleotide probes that selectively hybridize, under stringent conditions, to the polynucleotides of the present invention are used to identify the desired sequence in a cDNA or genomic DNA library. While isolation of RNA, and construction of cDNA and genomic libraries is well known to those of ordinary skill in the art. (See, e. g., Ausubel, supra, Chapters 1-7; or Sambrook, supra) Nucleic Acid Screening and Isolation Methods A cDNA or genomic library can be screened using a probe based upon the sequence of a polynucleotide of the present invention, such as those disclosed herein. Probes may be used to hybridize with genomic DNA or cDNA sequences to isolate homologous genes in the same or different organisms.

Those of skill in the art will appreciate that various degrees of stringency of hybridization can be employed in the assay; and either the hybridization or the wash medium can be

stringent. As the conditions for hybridization become more stringent, there must be a greater degree of complementarity between the probe and the target for duplex formation to occur. Temperature, ionic strength, pH and the presence of a partially denaturing solvent such as formamide can control the degree of stringency. Changing the polarity of the reactant solution through, for example, manipulation of the concentration of formamide within the range of 0% to 50%- conveniently varies the stringency of hybridization. The degree of complementarity (sequence identity) required for detectable binding will vary in accordance with the stringency of the hybridization medium and/or wash medium.

The degree of complementarity will optimally be 100%; however, it should be understood that minor sequence variations in the probes and primers may be compensated for by reducing the stringency of the hybridization and/or wash medium.

Methods of amplification of RNA or DNA are well known in the art and can be used according to the present invention without undue experimentation, based on the teaching and guidance presented herein.

Known methods of DNA or RNA amplification include, but are not limited to, polymerase chain reaction (PCR) and related amplification processes (see, e. g., U. S. Patent Nos.

4,683,195,4,683,202,4,800,159,4,965,188, to Mullis, et al.; 4,795,699 and 4,921,794 to Tabor, et al; 5,142,033 to Innis; 5,122,464 to Wilson, et al.; 5,091,310 to Innis; 5,066,584 to Gyllensten, et al; 4,889,818 to Gelfand, et al; 4,994,370 to Silver, et al; 4,766,067 to Biswas; 4,656,134 to Ringold) and RNA mediated amplification which uses anti- sense RNA to the target sequence as a template for double- stranded DNA synthesis (U. S. Patent No. 5,130,238 to Malek, et al, with the tradename NASBA), the entire contents of

which are herein incorporated by reference. (See, e. g., Ausubel, supra, Chapter 15; or Sambrook, supra) For instance, polymerase chain reaction (PCR) technology can be used to amplify the sequences of polynucleotides of the present invention and related genes directly from genomic DNA or cDNA libraries. PCR and other in vitro amplification methods may also be useful, for example, to clone nucleic acid sequences that code for proteins to be expressed, to make nucleic acids to use as probes for detecting the presence of the desired mRNA in samples, for nucleic acid sequencing, or for other purposes. Examples of techniques sufficient to direct persons of skill through in vitro amplification methods are found in Berger, Sambrook, and Ausubel (e. g., Chapter 15) supra, as well as Mullis, et al., U. S. Patent No. 4,683,202 (1987); and Innis, et al., PCR Protocols A Guide to Methods and Applications, Eds., Academic Press Inc., San Diego, CA (1990). Commercially available kits for genomic PCR amplification are known in the art. See, e. g., Advantage-GC Genomic PCR Kit (Clontech). The T4 gene 32 protein (Boehringer Mannheim) can be used to improve yield of long PCR products.

Synthetic Methods for Constructing Nucleic Acids The isolated nucleic acids of the present invention can also be prepared by direct chemical synthesis by methods such as the phosphotriester method of Narang, et al., Meth.

Enzymol., 68: 90-98 (1979); the phosphodiester method of Brown, et al., Meth. Enzymol., 68: 109-151 (1979); the diethylphosphoramidite method of Beaucage, et al., Tetra.

Letts., 22: 1859-1862 (1981) ; the solid phase phosphoramidite triester method described by Beaucage and Caruthers, Tetra.

Letts., 22 (20): 1859-1862 (1981), e. g., using an automated synthesizer, e. g., as described in Needham-VanDevanter, et

al., Nucleic Acids Res., 12: 6159-6168 (1984); and the solid support method of U. S. Patent No. 4,458,066. Chemical synthesis generally produces a single-stranded oligonucleotide, which may be converted into double-stranded DNA by hybridization with a complementary sequence, or by polymerization with a DNA polymerase using the single strand as a template. One of skill in the art will recognize that while chemical synthesis of DNA can be limited to sequences of about 100 or more bases, longer sequences may be obtained by the ligation of shorter sequences.

Recombinant Expression Cassettes The present invention further provides recombinant expression cassettes comprising a nucleic acid of the present invention. A nucleic acid sequence of the present invention, for example, a cDNA or a genomic sequence encoding a full- length polypeptide of the present invention, can be used to construct a recombinant expression cassette which can be introduced into at least one desired host cell. A recombinant expression cassette will typically comprise a polynucleotide of the present invention operably linked to transcriptional initiation regulatory sequences that will direct the transcription of the polynucleotide in the intended host cell.

Both heterologous and non-heterologous (i. e., endogenous) promoters can be employed to direct expression of the nucleic acids of the present invention. These promoters can also be used, for example, in recombinant expression cassettes to drive expression of antisense nucleic acids to reduce, increase, or alter hSPARC-hl content and/or composition in a desired tissue.

In some embodiments, isolated nucleic acids which serve as promoter or enhancer elements can be introduced in the

appropriate position (generally upstream) of a non- heterologous form of a polynucleotide of the present invention so as to up or down regulate expression of a polynucleotide of the present invention. For example, endogenous promoters can be altered in vivo or in vitro by mutation, deletion and/or substitution.

A polynucleotide of the present invention can be expressed in either sense or anti-sense orientation as desired. It will be appreciated that control of gene expression in either sense or anti-sense orientation can have a direct impact on the observable characteristics.

Another method of suppression is sense suppression.

Introduction of nucleic acid configured in the sense orientation has been shown to be an effective means by which to block the transcription of target genes.

A variety of cross-linking agents, alkylating agents and radical generating species as pendant groups on polynucleotides of the present invention can be used to bind, label, detect and/or cleave nucleic acids. Knorre, et al., Biochimie, 67: 785-789 (1985); Vlassov, et al., Nucleic Acids Res., 14: 4065-4076 (1986); Iverson and Dervan, J. Am. Chem.

Soc., 109: 1241-1243 (1987); Meyer, et al., J. Am. Chem. Soc., 111: 8517-8519 (1989); Lee, et al., Biochemistry, 27: 3197-3203 (1988); Home, et al., J. Am. Chem. Soc., 112: 2435-2437 (1990); Webb and Matteucci, J. Am. Chem. Soc., 108: 2764-2765 (1986); Nucleic Acids Res., 14: 7661-7674 (1986); Feteritz, et al., J. Am. Chem. Soc., 113: 4000 (1991). Various compounds to bind, detect, label, and/or cleave nucleic acids are known in the art. See, for example, U. S. Patent Nos. 5,543,507; 5,672,593 ; 5,484,908; 5,256,648; and 5,681941, each entirely incorporated herein by reference.

Vectors and Host Cells The present invention also relates to vectors that include isolated nucleic acid molecules of the present invention, host cells that are genetically engineered with the recombinant vectors, and the production of hSPARC-hl polypeptides or fragments thereof by recombinant techniques, as is well known in the art. See, e. g., Sambrook, et al., supra; Ausubel, supra, Chapters 1-9, each entirely incorporated herein by reference.

The polynucleotides can optionally be joined to a vector containing a selectable marker for propagation in a host. Generally, a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it can be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.

The DNA insert should be operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the E. coli lac, trp and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, or any other suitable promoter. The skilled artisan will know other suitable promoters. The expression constructs will further contain sites for transcription initiation, termination and, in the transcribed region, a ribosome- binding site for translation. The coding portion of the mature transcripts expressed by the constructs will preferably include a translation initiating at the beginning and a termination codon (e. g., UAA, UGA or UAG) appropriately positioned at the end of the mRNA to be translated, with VAA and VAG preferred for mammalian or eukaryotic cell expression.

Expression vectors will preferably include at least one selectable marker. Such markers include, e. g.,

dihydrofolate reductase, ampicillin (G418), hygromycin or neomycin resistance for eukaryotic cell culture, and tetracycline or ampicillin resistance genes for culturing in E. coli and other bacteria or prokaryotics. Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells ; fungal cells, such as yeast cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS and Bowes melanoma cells; and plant cells. Appropriate culture mediums and conditions for the above-described host cells are known in the art. Vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from Qiagen; pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A, pNH16a, pNH18A, pNH46A, available from Stratagene; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia.

Preferred eucaryotic vectors include pWLNEO, pSV2CAT, pOG44, pXT1 and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia. Other suitable vectors will be readily apparent to the skilled artisan. See, e. g., Ausubel, supra, Chapter 1; Coligan, Current Protocols in Protein Science, supra, Chapter 5.

Introduction of a vector construct into a host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection or other methods.

Such methods are described in many standard laboratory manuals, such as Sambrook, supra, Chapters 1-4 and 16-18; Ausubel, supra, Chapters 1,9,13,15,16.

As indicated, nucleic acid molecules of the present invention which comprise a nucleic acid encoding an hSPARC- hl polypeptide can include, but are not limited to, those encoding the amino acid sequence of the mature polypeptide;

the coding sequence for the mature polypeptide and additional sequences, such as the coding sequence of at least one signal leader or fusion peptide; the mature polypeptide, with at least one intron, together with additional, non-coding sequences such as, but not limited to, introns and non-coding 5'and 3'sequences (i. e., the transcribed, non-translated sequences that play a role in transcription, ribosome binding, stability, or mRNA processing, including splicing and polyadenylation signals).

Further, any of the above. described nucleic acids can further comprise additional coding sequences which encodes additional amino acids which provide additional functionalities known to one skilled in the art to be useful additions to such nucleic acids. Thus, the sequence encoding a polypeptide can be fused to a marker sequence, such as a sequence encoding a peptide that facilitates purification of the fused polypeptide.

Preferred nucleic acid fragments of the present invention also include nucleic acid molecules encoding epitope-bearing portions of an hSPARC-hl polypeptide.

Polypeptide (s) of the present invention can be expressed in a modified form, such as a fusion protein, and can include not only secretion signals, but also additional heterologous functional regions. For instance, a region of additional amino acids, particularly charged amino acids, can be added to the N-terminus of a polypeptide to improve stability and persistence in the host cell, during purification, or during subsequent handling. and storage.

Also, peptide moieties can be added to a polypeptide to facilitate purification. Such regions can be removed prior to final preparation of a polypeptide. Such methods are described in many standard laboratory manuals, such as Sambrook, supra, Chapters 17 and 18; Ausubel, supra, Chapters 16,17 and 18.

Expression of Proteins in Host Cells Using nucleic acids of the present invention, one may express a protein of the present invention in a recombinantly engineered cell, such as bacteria, yeast, insect, or mammalian cells. The cells produce the protein in a non- natural condition (e. g., in quantity, composition, location, and/or time), because they have been genetically altered through human intervention to do so.

It is expected that those of skill in the art are knowledgeable in the numerous expression systems available for expression of a nucleic acid encoding a protein of the present invention. No attempt to describe in detail the various methods known for the expression of proteins in prokaryotes or eukaryotes will be made.

In brief summary, the expression of isolated nucleic acids encoding a protein of the present invention will typically be achieved by operably linking, for example, the DNA or CDNA to a promoter (which is either constitutive or inducible) followed by incorporation into an expression vector. The vectors can be suitable for replication and integration in either prokaryotes or eukaryotes. Typical expression vectors contain transcription and translation terminators, initiation sequences and promoters useful for regulation of the expression of the DNA encoding a protein of the present invention. To obtain high level expression of a cloned gene, it is desirable to construct expression vectors which contain, at the minimum, a strong promoter to direct transcription, a ribosome binding site for translational initiation, and a transcription/translation terminator. One of skill would recognize that modifications can be made to a protein of the present invention without diminishing its biological activity. Some modifications may be made to

facilitate the cloning, expression, or incorporation of the targeting molecule into a fusion protein. Such modifications are well known to those of skill in the art and include, for example, a methionine added at the amino terminus to provide an initiation site, or additional amino acids (e. g., poly His) placed on either terminus to create conveniently located restriction sites or termination codons or purification sequences.

Alternatively, nucleic acids of the present invention can be expressed in a host cell by turning on (by manipulation) in a host cell that contains endogenous DNA encoding a polypeptide of the present invention. Such methods are well known in the art, e. g., as described in US patent Nos. 5,580,734,5,641,670,5,733,746, and 5,733,761, entirely incorporated herein by reference.

Expression in Prokaryotes Prokaryotic cells may be used as hosts for expression.

Prokaryotes most frequently are represented by various strains of E. coli; however, other microbial strains may also be used. Commonly used prokaryotic control sequences which are defined herein to include promoters for transcription initiation, optionally with an operator, along with ribosome binding site sequences, include such commonly used promoters as the beta lactamase (penicillinase) and lactose (lac) promoter systems (Chang, et al., Nature, 198: 1056 (1977)), the tryptophan (trp) promoter system (Goeddel, et al., Nucleic Acids Res., 8: 4057-4074 (1980)) and the lambda derived P L promoter and N-gene ribosome binding. site (Simatake, et al., Nature, 292: 128-132 (1981)). The inclusion of selection markers in DNA. vectors transfected in E. coli is also useful. Examples of such markers include

genes specifying resistance to ampicillin, tetracycline, or chloramphenicol.

The vector is selected to allow introduction into the appropriate host cell. Bacterial vectors are typically of plasmid or phage origin. Appropriate bacterial cells are infected with phage vector particles or transfected with naked phage vector DNA. If a plasmid vector is used, the bacterial cells are transformed with the plasmid vector DNA.

Expression systems for expressing a protein of the present invention are available using Bacillus sp. and Salmonella (Palva, et al., Gene, 22: 229-235 (1983); Mosbach, et al., Nature, 302: 543-545 (1983)). See, e. g., Ausubel, supra, Chapters 1-3,16 (Sec. 1); and Coligan, supra, Current Protocols in Protein Science, Units 5.1,6.1-6.7.

Expression in Eukaryotes A variety of eukaryotic expression systems such as yeast, insect cell lines, plant and mammalian cells, are known to those of skill in the art. As explained briefly below, a nucleic acid of the present invention can be expressed in these eukaryotic systems.

Synthesis of heterologous proteins in yeast is well known. F. Sherman, et al., Methods in Yeast Genetics, Cold Spring Harbor Laboratory (1982) is a well-recognized work describing the various methods available to produce the protein in yeast. Two widely utilized yeast for production of eukaryotic proteins are Saccharomyces cerevisiae and Pichia pastoris. Vectors, strains, and protocols for expression in Saccharomyces and Pichia are known in the art and available from commercial suppliers (e. g., Invitrogen).

Suitable vectors usually have expression control sequences, such as promoters, including 3-phosphoglycerate kinase or

alcohol oxidase, and an origin of replication, termination sequences and the like as desired.

A protein of the present invention, once expressed, can be isolated from yeast by lysing the cells and applying standard protein isolation techniques to the lysates. The monitoring of the purification process can be accomplished by using Western blot techniques or radioimmunoassay of other standard immunoassay techniques.

The sequences encoding proteins of the present invention can also be ligated to various expression vectors for use in transfecting cell cultures of, for instance, mammalian, insect, or plant origin. Illustrative of cell cultures useful for the production of the peptides are mammalian cells. Mammalian cell systems often will be in the form of monolayers of cells although mammalian cell suspensions may also be used. A number of suitable host cell lines capable of expressing intact proteins have been developed in the art, and include the HEK293, BHK21, and CHO cell lines.

Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter (e. g., the CMV promoter, a HSV tk promoter or pgk (phosphoglycerate kinase) promoter), an enhancer (Queen, et al., Immunol. Rev., 89: 49-68 (1986)), and processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites (e. g., an SV40 large T Ag poly A addition site), and transcriptional terminator sequences.

Other animal cells useful for production of proteins of the present invention are available, for instance, from the American Type Culture Collection Catalogue of Cell Lines and Hybridomas (7th edition, 1992).

Appropriate vectors for expressing proteins of the present invention in insect cells are usually derived from the SF9 baculovirus. Suitable insect cell lines include mosquito larvae, silkworm, armyworm, moth and Drosophila cell

lines such as a Schneider cell line (See Schneider, J.

Embryol. Exp. Morphol., 27: 353-365 (1972).

As with yeast, when higher animal or plant host cells are employed, polyadenlyation or transcription terminator sequences are typically incorporated into the vector. An example of a terminator sequence is the polyadenlyation sequence from the bovine growth hormone gene. Sequences for accurate splicing of the transcript may also be included. An example of a splicing sequence is the VP1 intron from SV40 (Sprague, et al., J. Virol., 45: 773-781 (1983)).

Additionally, gene sequences to control replication in the host cell may be incorporated into the vector such as those found in bovine papilloma virus type-vectors. M. Saveria- Campo, bovine Papilloma Virus DNA, a Eukaryotic Cloning Vector in DNA Cloning Vol. II, a Practical Approach, D. M.

Glover, Ed., IRL Press, Arlington, VA, pp. 213-238 (1985).

Protein Purification An hSPARC-hl polypeptide can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxyapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification. Polypeptides of the present invention include naturally purified products, products of chemical synthetic procedures, and products produced by recombinant techniques from a prokaryotic or eucaryotic host, including, for example, bacterial, yeast, higher plant, insect and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the

present invention can be glycosylated or can be non- glycosylated. In addition, polypeptides of the invention can also include an initial modified methionine residue, in some cases as a result of host-mediated processes. Such methods are described in many standard laboratory manuals, such as Sambrook, supra, Chapters 17.37-17.42; Ausubel, supra, Chapters 10,12,13,16,18 and 20. hSPARC-hl Polypeptides and Fragments and Variants Thereof The invention further provides an isolated hSPARC-hl polypeptides having fragments or specified variants of the amino acid sequence encoded by the deposited cDNAs, or the amino acid sequence in SEQ ID NO : 4 or 5.

The isolated proteins of the present invention comprise a polypeptide encoded by any one of the polynucleotides of the present invention as discussed herein, or polypeptides which are specified fragments or variants thereof.

Exemplary polypeptide sequences are provided in SEQ ID NO : 4 or 5. The proteins of the present invention or variants thereof can comprise any number of contiguous amino acid residues from a polypeptide of the present invention.

Optionally, the subsequence of contiguous amino acids is at least 20,30,40,50,60,70,80, or 90 amino acids in length. Further, the number of contiguous amino acid residues in such subsequences can be any integer selected from the group consisting of from 1 to 20, such as 2,3,4, or 5.

In a further aspect, the invention concerns an isolated hSPARC-hl polypeptide comprising an amino acid sequence having at least about 80% sequence identity, preferably at least about 81% sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86%

sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity. yet more preferably at least about 98% sequence identity, yet more preferably at least about 99% sequence identity to a hSPARC-hl polypeptide of the present invention.

In a further aspect, the invention concerns an isolated hSPARC-hl polypeptide comprising an amino acid sequence scoring at least about 80% positives, preferably at least about 81% positives, more preferably at least about 82% positives, yet more preferably at least about 83% positive, yet more preferably at least about 84% positives, yet more preferably at least about 85% positives, yet more preferably at least about 86% positives, yet more preferably at least about 87% positives, yet more preferably at least about 88% positives, yet more preferably at least about 89% positives, yet more preferably at least about 90% positives, yet more preferably at least about 91% positives, yet more preferably at least about 92% positives, yet more preferably at least about 93% positives, yet more preferably at least about 94% positives, yet more preferably at least about 95% positives, yet more preferably at least about 96% positives, yet more preferably at least about 97% positives, yet more preferably at least about 98% positives, yet more preferably at least about 99% positives, when compared with the amino acid

sequence of residues from about (1) 1 or about 27 to about 446, inclusive, of SEQ ID NO : 4.

In a specific aspect, the invention provides an isolated hSPARC-hl polypeptide without the N-terminal signal sequence and/or initiating methionine and is encoded by a nucleotide sequence that encodes such an amino acid sequence as hereinbefore described. Processes for producing the same are also herein described, wherein those processes comprise culturing a host cell comprising a vector which comprises the appropriate encoding nucleic acid molecule under conditions suitable for expression of hSPARC-hl polypeptide and recovering the hSPARC-hl polypeptide, respectively, from the cell culture.

In still a further aspect, the invention provides a polypeptide produced by: (1) hybridizing a test DNA molecule under stringent conditions with (a) a DNA molecule encoding a (i) hSPARC-hl polypeptide having the sequence of amino acid residues from about 27 to about 446, inclusive, of SEQ ID NO : 4, or (b) the complement of the DNA molecule of (a); and if the test DNA molecule has at least about an 80% sequence identity, preferably at least about an 81% sequence identity, more preferably at least about an 82% sequence identity, yet more preferably at least about an 83% sequence identity, yet more preferably at least about an 84% sequence identity, yet more preferably at least about an 85% sequence identity, yet more preferably at least about an 86% sequence identity, yet more preferably at least about an 87% sequence identity, yet more preferably at least about an 88% sequence identity, yet more preferably at least about an 89% sequence identity, yet more preferably at least about a 90% sequence identity, yet more preferably at least about a 91% sequence identity, yet more preferably at least about a 92% sequence identity, yet more preferably at least about a 93% sequence identity, yet more preferably at least about a 94% sequence identity, yet

more preferably at least about a 95% sequence identity, yet more preferably at least about a 96% sequence identity, yet more preferably at least about a 97% sequence identity, yet more preferably at least about 98% sequence identity, yet more preferably at least about a 99% sequence identity to (a) or (b); (2) culturing a host cell comprising the test DNA molecule under conditions suitable for expression of the polypeptide, and (3) recovering the polypeptide from the cell culture.

In yet another aspect, the invention concerns an isolated hSPARC-hl polypeptide comprising the sequence of amino acid residues from about 1 or about 27 to about 446, inclusive, of SEQ ID NO: 4, or a fragment or variant thereof which is biologically active or sufficient to provide a binding site for an anti-hSPARC-hl antibody, wherein the identification of hSPARC-hl polypeptide or fragments thereof that possess biological activity or provide a binding site for an anti-hSPARC-hl antibody may be accomplished in a routine manner using techniques which are well known in the art.

As those of skill will appreciate, the present invention includes biologically active polypeptides of the present invention (i. e., enzymes). Biologically active polypeptides have a specific activity at least 20%, 30%, or 40%, and preferably at least 50%, 60%, or 70%, and most preferably at least 80%, 90%, or 95%-100% of that of the native (non-synthetic), endogenous polypeptide. Further, the substrate specificity (e. g., kcat/Km) is optionally substantially similar to the native (non-synthetic), endogenous polypeptide. Typically, the Km will be at least 30%, 40%, or 50%, that of the native (non-synthetic), endogenous polypeptide; and more preferably at least 60%, 70%, 80%, or 90%-1000%. Methods of assaying and quantifying

measures of enzymatic activity and substrate specificity, are well known to those of skill in the art.

Generally, the polypeptides of the present invention will, when presented as an immunogen, elicit production of an antibody specifically reactive to a polypeptide of the present invention encoded by a polynucleotide of the present invention as described, supra. Exemplary polypeptides include those which are full-length, such as those disclosed herein. Further, the proteins of the present invention will not bind to antisera raised against a polypeptide of the present invention which has been fully immunosorbed with the same polypeptide. Immunoassays for determining binding are well known to those of skill'in the art. A preferred immunoassay is a competitive immunoassay as discussed, infra.

Thus, the proteins of the present invention can be employed as immunogens for constructing antibodies immunoreactive to a protein of the present invention for such exemplary utilities as immunoassays or protein purification techniques.

An hSPARC-hl polypeptide of the present invention can include one or more amino acid substitutions, deletions or additions, either from natural mutations or human manipulation, as specified herein. Functional analogs of hSPARC-hl polypeptide (s) are typically generated by deletion, insertion, or substitution of a single (or few) amino acid residues. Functional analogs of the polypeptide of SEQ ID NO : 4 or 5, may be those in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code.

Substitution modifications can generally be made in accordance with the following Table.

Table 1 ORIGINAL RESIDUE EXEMPLARY SUBSTITUTIONS ALA SER ARG LYS ASN GLN, HIS ASPGLU CYS SER GLN ASN GLU ASP GLY PRO HIS ASN, GLN ILE LEU, VAL LEU ILE, VAL LYS ARG, GLN, GLU MET LEU, ILE PHE MET, LEU, TYR SER THR THR SER TRP TYR TYR TRP, PHE VAL ILE, LEU

of course, the number of amino acid substitutions a skilled artisan would make depends on many factors, including those described above. Generally speaking, the number of amino acid substitutions, insertions or deletions for any given hSPARC-hl polypeptide will not be more than 40,30,20,10,5, or 3, such as 1-30 or any range or value therein, as specified herein.

Amino acids in an hSPARC-hl polypeptide of the present invention that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (Cunningham and Wells, Science 244: 1081-1085 (1989)). The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity. Sites that are critical for ligand-protein binding can also be identified by structural analysis such as crystallization, nuclear magnetic resonance or photoaffinity labeling (Smith, et al., J. Mol. Biol.

224: 899-904 (1992) and de Vos, et al., Science 255: 306-312 (1992)).

Non-limiting mutants that can enhance or maintain at least one of the listed activities include any of the polypeptides described herein further comprising at least one mutation corresponding to at least one substitution, insertion or deletion selected from the group consisting of D6,7V, 25G, 42P, 45P, 48P, 78A, 80A, 161P, 218V, 225H, 239R, 300S, 301V, 302E, 364A, 377D, 432G, 433S, and 434L of SEQ ID NO : 4 or the corresponding amino acids of SEQ ID NO: 5.

Covalent modifications of hSPARC-hl polypeptides are also included within the scope of this invention. One type of covalent modification includes reacting targeted amino acid residues of an hSPARC-hl polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or the N-or C-terminal residues of an hSPARC-hl polypeptide. Derivatization with bifunctional agents is useful, for instance, for crosslinking hSPARC-hl to a water- insoluble support matrix or surface for use in the method for purifying anti-hSPARC-hl antibodies, and vice-versa. Commonly used crosslinking agents include. e. g., 1. l-bis (diazo- acetyl)-2-phenylethane, glutaraldehyde, N-hydroxy-succinimide

esters. for example, esters with 4-azidosalicylic acid, homobifunctional imidoesters including disuccinimidyl esters such as 3,3'-dithiobis- (succinimidyhydroprionate). bifunctional maleimides such as bis-N-maleimidol, 8-octane and agents such as methyl-3-[(p-azidophenyI)-dithioproprioimidate.

Other modifications include deamidation of glutaminyl and asparaginyl residues to the corresponding glutamyl and aspartyl residues, respectively, hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the a-amino groups of lysine, arginine, and histidine side chains [T. E. Creighton.

Proteins: Structure and Molecular Properties. W. H. Freeman & Co.. San Francisco, pp. 79-86 (1983)], acetylation of the N- terminal amine. and amidation of any C-terminal carboxyl group.

Another type of covalent modification of the hSPARC-hl polypeptide included within the scope of this invention comprises altering the native glycosylation pattern of the polypeptide."Altering the native glycosylation patterns intended for purposes herein to mean deleting one or more carbohydrate moieties found in native sequence hSPARC-hl polypeptide. and/or adding one or more glycosylation sites that are not present in the native sequence hSPARC-hl polypeptide. Additionally, the phrase includes qualitative changes in the glycosylation of the native proteins as well as changes in the nature and proportions of the various carbohydrate moieties present.

Another class of hSPARC-hl variant that would be useful in the methods of the present invention is a hSPARC-hl polypeptide as defined herein further comprising one or more amino acid substitutions that result in an altered glycosylation pattern as compared to the corresponding unsubstituted hSPARC-hl polypeptide.

The term"N-glycosyled polypeptide"refers to

polypeptides having one or more NXS/T motifs in which the nitrogen atom in the side chain amide of the asparagine is covalently bonded to a glycosyl group."X"refers to any naturally occurring amino acid residue except proline. The "naturally occurring amino acids"are glycine, alanine, valine, leucine, isoleucine, proline, serine, threonine, cysteine, methionine, lysine, arganine, glutamic acid, asparatic acid, glutamine, asparagine, phenylalanine, histidine, tyrosine and tryptophan. N-glycosylated proteins are optionally 0-glycosylation.

The term"O-glycosylated polypeptide"refers to polypeptides having one or more serines and/or threonine in which the oxygen atom in the side chain is covalently bonded to a glycosyl group. O-Glycosylated proteins are optionally N-glycosylation. Glycosylated polypeptides can be prepared recombinantly by expressing a gene encoding a polypeptide in a suitable mammalian host cell, resulting in glycosylation of side chain amides found in accessible NXT/S motifs on the polypeptide surface and/or of side chain alcohols of surface accessible serines and threonines. Specific procedures for recombinantly expressing genes in mammalian cells are provided hereinbelow. Other procedures for preparing glycosylated proteins are disclosed in EP 640,619 to Elliot and Burn, the entire teachings of which are incorporated herein by reference. Unglycosylated polypeptides can be prepared recombinantly by expressing a gene encoding a polypeptide in a suitable prokaryotic host cell. The hSPARC-hl polypeptides and hSPARC-hl fragments and/or hSPARC-hl variants of the present invention can also be glycosylated or unglycosylated. A glycosylated polypeptide is modified with one or more monosaccharides or oligosaccharides. A monosaccharide is a chiral polyhydroxyalkanol or polyhydroxyalkanone which typically exists in hemiacetal

form. An"oligosaccharide"is a polymer of from about 2 to about 18 monosaccharides which are generally linked by acetal bonds. One type of glycosyl group commonly found in glycosylated proteins is N-acetylneuraminic acid. A glycosylated polypeptide can be N-glycosylated and/or O- glycosylated, preferably N-glycosylated.

Addition of glycosylation sites to hSPARC-hl polypeptides may be accomplished by altering the amino acid sequence thereof. The alteration may be made, for example, by the addition of, or substitution by, one or more serine or threonine residues to the native sequence hSPARC-hl polypeptide (for 0-linked glycosylation sites). The hSPARC-hl amino acid sequence may optionally be altered through changes at the DNA level, particularly by mutating the DNA encoding the hSPARC-hl polypeptide at preselected bases such that codons are generated that will translate into the desired amino acids.

Another means of increasing the number of carbohydrate moieties on the hSPARC-hl polypeptide is by chemical or enzymatic coupling of glycosides to the polypeptide. Such methods are described in the art, e. g., in WO 87105330 published 11 September 1987, and in Aplin and Wriston, CRC Crit. Rev. Biochem., pp. 259-306 (1981).

Removal of carbohydrate moieties present on the hSPARC-hl polypeptide may be accomplished chemically or enzymatically or by mutational substitution of codons encoding for amino acid residues that serve as targets for glycosylation. Chemical deglycosylation techniques are known in the art and described, for instance, by Hakimuddin, et al., Arch. Biochem. Biophys., 259: 52 (1987) and by Edge et al., Anal. Biochem.) m: 131 (1981). Enzymatic cleavage of carbohydrate moieties on polypeptides can be achieved by the use of a variety of. endo- and exo-glycosidases as described by Thotakura et al., Meth.

Enzymof., 138: 350 (1987).

Another type of covalent modification of hSPARC-hl comprises linking the hSPARC-hl polypeptide to one of a variety of nonproteinaceous polymers, e. g., polyethylene glycol, polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U. S. Patent Nos. 4640,835; 4,496,689: 4,301,144; 4.670,417; 4.791,192 or 4,179,337, Mumtaz and Bachhawat, Indian Journal of Biochemistry and Biophysics 28 : 346 (1991) and Franciset al., International Journal of Hematology 68: 1 (1998), the entire teachings of which are incorporated herein by reference. Therefore, the hSPARC-hl variants useful in the methods of the present invention also include hSPARC-hl polypeptides as defined herein further comprising one or more polyethylene glycol groups (hereinafter "PEG"groups). Suitable PEG groups generally have a molecular weight between about 5000 and 40,000 atomic'mass units.

Human SPARC-hl polypeptides of the present invention may also be modified in a way to form chimeric molecules comprising an hSPARC-hl polypeptide fused to another heterologous polypeptide or amino acid sequence. hSPARC-hl fusion proteins represent a hybrid protein molecule not found in nature comprising a translational fusion or enzymatic fusion in which two or more different proteins, fragments, or variants thereof are covalently linked on a single polypeptide chain. In one embodiment, such a chimeric molecule comprises a fusion of an hSPARC-hl polypeptide with a tag polypeptide that provides an epitope to which an anti-tag antibody can selectively bind. The epitope tag is generally placed at the amino-or carboxyl-terminus of the hSPARC-hl polypeptide.

The presence of such epitope-tagged forms of an hSPARC-hl polypeptide can be detected using an antibody against the tag polypeptide. Also, another provision of the epitope tag

enables the hSPARC-hl polypeptide to be readily purified by affinity purification using an anti-tag antibody or another type of affinity matrix that binds to the epitope tag. Human serum albumin, the C-terminal domain of thrombopoietin, the C- terminal extension peptide of hCG, and/or a Fc fragment are examples of proteins which could be fused with hSPARC-hl polypeptides, hSPARC-hl fragments and/or hSPARC-hl variants for use in the present invention. As used herein,"Fc fragment"of an antibody has the meaning commonly given to the term in the field of immunology. Specifically, this term refers to an antibody fragment which binds complement and is obtained by removing the two antigen binding regions (the Fab Fragments) from the antibody. Thus, the Fc fragment is formed from approximately equal sized fragments from both heavy chains, which associate through non-covalent interactions and disulfide bonds. The Fc Fragment includes the hinge regions and extends through the CH2 and CH3 domains to the C-terminus of the antibody. Procedures for preparing fusion proteins are disclosed in EP394, 827, Tranecker et al., Nature 331 : 84 (1988) and Fares, et al., Proc. Natl. Acad. Sci. USA 89 : 4304 (1992), the entire teachings of which are incorporated herein by reference.

Many fusion proteins can be secreted by virtue of heterologous secretion signals in regions that can be removed prior to final preparation of the polypeptide. Such methodologies are described in many standard laboratory manuals, such as Sambrook, supra, Chapters 17.29-17.42 and 18.1-18.74; Ausubel, supra, Chapters 16,17 and 18, the entire relevant teachings of which are incorporated herein by reference.

In a preferred process for protein expression and subsequent purification, the hSPARC-hl gene can be modified at the 5'end to incorporate several histidine residues at

the amino terminus of the hSPARC-hl protein resulting from its expression. This"histidine tag"enables a single-step protein purification method referred to as"immobilized metal ion affinity chromatography" (IMAC), essentially as described in U. S. Patent 4,569,794, which hereby is incorporated by reference. The IMAC method enables rapid isolation of substantially pure recombinant hSPARC-hl protein starting from a crude extract of cells that express a modified recombinant protein, as described above.

Antigenic/Epitope Comprising hSPARC-hl Peptide and Polypeptides In another aspect, the invention provides a peptide or polypeptide comprising an epitope-bearing portion of a polypeptide of the invention according to methods well known in the art (See, e. g., Coligan, et al,. ed., Current Protocols in Immunology, Greene Publishing, NY (1993-1998), Ausubel, supra, each entirely incorporated herein by reference).

The epitope of this polypeptide portion is an immunogenic or antigenic epitope of a polypeptide described herein. An"immunogenic epitope"can be defined as a part of a polypeptide that elicits an antibody response when the whole polypeptide is the immunogen. On the other hand, a region of a polypeptide molecule to which an antibody can bind is defined as an"antigenic epitope."The number of immunogenic epitopes of a polypeptide generally is less than the number of antigenic epitopes. See, for instance, Geysen, et al., Proc. Natl. Acad. Sci. USA, 81: 3998-4002 (1984).

As to the selection of peptides or polypeptides bearing an antigenic epitope (i. e., that contain at least a portion of a region of a polypeptide molecule to which an antibody

can bind), it is well known in the art that relatively short synthetic peptides that mimic part of a polypeptide sequence are routinely capable of eliciting an antiserum that reacts with the partially mimicked polypeptide. See, for instance, J. G. Sutcliffe, et al.,"Antibodies that react with pre- identified sites on polypeptides,"Science, 219: 660-666 (1983).

Antigenic epitope-bearing peptides and polypeptides of the invention are useful to raise antibodies, including monoclonal antibodies, or screen antibodies, including fragments or single chain antibodies, that bind specifically to a polypeptide of the invention. See, for instance, Wilson, et al., Cell, 37: 767-778 (1984) at 777. Antigenic epitope-bearing peptides and polypeptides of the invention preferably contain a sequence of at least five, more preferably at least nine, and most preferably between at least about 15 to about 30 amino acids contained within the amino acid sequence of a polypeptide of the invention.

The epitope-bearing peptides and polypeptides of the invention can be produced by any conventional means. R. A.

Houghten,"General method for the rapid solid-phase synthesis of large numbers of peptides: specificity of antigen-antibody interaction at the level of individual amino acids,"Proc. Natl. Acad. Sci. USA, 82: 5131-5135 (1985). This"Simultaneous Multiple Peptide Synthesis (SMPS)"process is further described in U. S. Patent No.

4,631,211 to Houghten, et al. (1986).

As one of skill in the art will appreciate, hSPARC-hl polypeptides of the present invention and the epitope- bearing fragments thereof described above can be combined with parts of the constant domain of immunoglobulins (IgG), resulting in chimeric polypeptides. These fusion proteins facilitate purification and show an increased half-life in vivo. This has been shown, e. g., for chimeric proteins

consisting of the first two domains of the human CD4- polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins (EPO 394,827 ; Traunecker, et al., Nature, 331: 84-86 (1988)).

Fusion proteins that have a disulfide-linked dimeric structure due to the IgG part can also be more efficient in binding and neutralizing other molecules than the monomeric hSPARC-hl polypeptide or polypeptide fragment alone (Fountoulakis, et al., J. Biol. Chem. 270: 3958-3964 (1995)).

Production of Antibodies The polypeptides of this invention and fragments thereof may be used in the production of antibodies. The term"antibody"as used herein describes antibodies, fragments of antibodies (such as, but not limited, to Fab, Fab', Fab2', and Fv fragments), and modified versions thereof, as well known in the art (e. g., chimeric, humanized, recombinant, veneered, resurfaced or CDR- grafted). The term"antibody"is meant to include polyclonal antibodies, monoclonal antibodies (MAbs), chimeric antibodies, single-chain polypeptide binding molecules, and anti-idiotypic (anti-id) antibodies.

Polyclonal antibodies are heterogeneous populations of antibody molecules derived from the sera of animals immu- nised with an antigen while monoclonal antibodies (MAbs) are a substantially homogeneous population of antibodies to specific antigens. Polyclonal and MAbs may be obtained by methods known to those skilled in the art (for MAbs, see, for example, Kohler et al., Nature 256 : 495-497 (1975), Colligan, supra., and U. S. Pat. No. 4,376,110).

Single chain antibodies and libraries thereof are yet another variety of genetically engineered antibody technology that is well known in the art. (See, e. g., R. E.

Bird, et al., Science 242: 423-426 (1988) ; PCT Publication Nos. WO 88/01649, WO 90/14430, and WO 91/10737. Single chain antibody technology involves covalently joining the binding regions of heavy and light chains to generate a single polypeptide chain. The binding specificity of the. intact antibody molecule is thereby reproduced on a single polypeptide chain.

MAbs may be of any immuno-globulin class including IgG, IgM, IgE, IgA, IgD and any subclass thereof. The hybridoma producing the MAbs of this invention may be cultivated in vitro or in vivo. Production of high titers of MAbs in vivo makes this the presently preferred method of production.

Briefly, cells from the individual hybridomas are injected intraperitoneally into pristane-primed BALB/C mice to produce ascites fluid containing high concentrations of the desired MAbs. MAbs of isotype IgM or IgG may be purified from such ascites fluids, or from culture supernatants, using column chromatography methods well known to those of skill in the art.

Chimeric antibodies are molecules in which different portions are derived from different animal species, such as those having variable region derived from a murine MAb and a human immunoglobulin constant region. Chimeric antibodies and methods for their production are known in the art (Cabilly et al., Proc. Natl. Acad. Sci. (USA) 71-3273-3277 (1984); Morrison et al., Proc. Natl. Acad. Sci. (USA) 81: 6851-6855 (1984); Boulianne et al., Nature 312: 643646 (1984); Cabilly et al., European Patent Application 125023 (published Nov. 14,1984); Neuberger et al., Nature 314: 268- 270 (1985); Taniguchi et al., European Patent Application 171496 (published Feb. 19,1985) ; Morrison et al., European Patent Application 173494 (published Mar. 5,1986); Neuberger et al., PCT Application WO 86/01533 (published

Mar. 13,1986); Kudo et al., European Patent Application 184187 (published Jun. 11,1986); Sahagan et al., J.

Immunol. 137: 1066-1074 (1986); Robinson et al., International Patent Publication #PCT/US86/02269 (published May 7,1987); Liu et al., Proc. Natl. Acad. Sci. (USA) 84: 3439-3443 (1987); Sun et al., Proc. Natl. Acad. Sci.

(USA) 84: 214-218 (1987); Better et al., Science 140: 1041- 1043 (1988)). These documents are hereby incorporated by reference.

The most preferred method of generating MAbs to the polypeptides and glycopeptides of the present invention comprises producing said MAbs in a transgenic mammal modified in such a way that they are capable of producing fully humanized MAbs upon antigenic challenge. Fully humanized MAbs and methods for their production are generally known in the art (PCT/W09634096, PCT/W09633735, and PCT/W09824893). These documents are hereby incorporated by reference.

An anti-idiotypic (anti-Id) antibody is an antibody which recognizes unique determinants generally associated with the antigen-binding site of an antibody. An anti-Id antibody can be prepared by immunizing an animal of the same species and genetic type (e. g. mouse strain) as the source of the MAb with the MAb to which an anti-Id is being prepared. The immunized animal will recognize and respond to the idiotypic determinants of the immunizing antibody by producing an antibody to these idiotypic determinants (the anti-Id antibody). The anti-Id antibody may also be used as an"immunogen"to induce an immune response in yet another animal, producing a so-called anti-anti-Id antibody. The anti-anti-Id may be epitopically identical to the original MAb which induced the anti-Id. Thus, by using antibodies to the idiotypic determinants of a MAb, it is possible to

identify other clones expressing antibodies of identical specificity. Accordingly, MAbs generated against a hSPARC- hl protein or glycoprotein of the present invention may be used to induce anti-Id antibodies in suitable animals, such as BALB/C mice and/or any transgenically altered mouse capable of producing fully humanized MAbs. Spleen cells from such immunized mice are used produce anti-Id hybridomas secreting anti-Id MAbs. Further, the anti-Id MAbs can be coupled to a carrier such as keyhole limpet hemocyanin (KLH) and used to immunize additional similar mice. Sera from these mice will contain anti-anti-Id antibodies that have the binding properties of the original MAb specific for a hSPARC-hl epitope. The anti-Id MAbs thus have their own idiotypic epitopes, or"idiotopes"structurally similar to the epitope being evaluated, such as a hSPARC-hl protein or glycoprotein.

The term"antibody"is also meant to include both intact molecules as well as fragments thereof, such as, for example, Fab and F (ab'),, which are capable of binding antigen. Fab and F (ab'), fragments lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody (Wahl et al., J. Nucl. Med. 24: 316-325 (1983)).

It will be appreciated that Fab and F (ab') a and other fragments of the antibodies useful in the present invention may be used for the detection and quantitation of a hSPARC- hl protein or glycoprotein according to methods disclosed herein for intact antibody molecules. Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F (ab') 2 fragments).

A polypeptide used as an immunogen may be modified or administered in an adjuvant, by subcutaneous or intraperitoneal injection into, for example, a mouse or a

rabbit. For the production of monoclonal antibodies, spleen cells from immunized animals are removed, fused with myeloma or other suitable known cells, and allowed to become monoclonal antibody producing hybridoma cells in the manner known to the skilled artisan. Hybridomas that secrete a desired antibody molecule can be screened by a variety of well known methods, for example ELISA assay, Western blot analysis, or radioimmunoassay (Lutz, et al. Exp. Cell Res.

175: 109-124 (1988); Monoclonal Antibodies: Principles & Applications, Ed. J. R. Birch & E. S. Lennox, Wiley-Liss (1995); Colligan, supra).

Antibodies included in this invention are useful in diagnostics, therapeutics or in diagnostic/therapeutic combinations. Thus, polypeptides of this invention or suitable fragments thereof can be used to generate polyclonal or monoclonal antibodies, and various inter- species hybrids, or humanized antibodies, or antibody fragments, or single-chain antibodies.

In one aspect, the present invention relates to a method for detecting the presence of or measuring the quantity of a hSPARC-hl protein or glycoprotein in a cell, comprising : (a) contacting said cell or an extract thereof with an antibody specific for an epitope of a hSPARC-hl protein or glycoprotein; and (b) detecting the binding of said antibody to said cell or extract thereof, or measuring the quantity of antibody bound, thereby determining the presence of or measuring the quantity of said hSPARC-hl protein or glycoprotein.

For some applications labeled antibodies are desirable.

Procedures for labeling antibody molecules are widely known, including for example, the use of radioisotopes, affinity labels, such as biotin or avidin, enzymatic labels, for example horseradish peroxidase, and fluorescent labels, such

as FITC or rhodamine (see, e. g., Colligan, supra). Labeled antibodies are useful for a variety of diagnostic applications. In one embodiment the present invention relates to the use of labeled antibodies to detect the presence of an hSPARC-hl polypeptide. Alternatively, the antibodies could be used in a screen to identify potential modulators of an hSPARC-hl polypeptide. For example, in a competitive displacement assay, the antibody or compound to be tested is labeled by any suitable method. Competitive displacement of an antibody from an antibody-antigen complex by a test compound such that a test compound-antigen complex is formed provides a method for identifying compounds that bind hSPARC-hl.

Transgenics and Chimeric Non-Human Mammals Another embodiment of the present invention provides transgenic non-human mammals carrying a recombinant hSPARC- hl gene construct in its somatic and germ cells. The recombinant gene construct may be composed of regulatory DNA sequences that belong to the native hSPARC-hl gene or those which are derived from an alternative source. These regulatory sequences are functionally linked to the hSPARC- hl coding region, resulting in the constitutive and/or regulatable expression of hSPARC-hl in the body of the transgenic non-human mammal. The most important of such regulatory sequences is the promoter. Promoters are defined in this context as any and all DNA elements necessary for the functional expression of a gene. Promoters drive the expression of structural genes and may be modulated by inducers and repressors. Numerous promoters have been described in the literature and are easily within the grasp of the ordinarily skilled artisan. Viral promoters, such as the SV40 early promoter, are consistent with the invention

though mammalian promoters are preferred. The promoter is chosen such that the level of expression is sufficient to promote physiological consequences in the transgenic non-. human mammal, or ancestor of said mammal. Preferably, the genome of the transgenic mammal contains at least 30 copies of a transgene. More preferably, the genome of the transgenic mammal contains at least 50 copies, and may contain 100-200 or more copies of the transgene. Generally, said nucleic acid is introduced into said mammal at an embryonic stage, preferably the 1-1000 cell or oocyte stage, and, most preferably not later than about the 64-cell stage.

Most preferably the transgenic mammal is homozygous for the transgene.

The techniques described in Leder, U. S. Patent No.

4,736,866 (hereby entirely incorporated by reference) for producing transgenic non-human mammals may be used for the production of a transgenic non-human mammal of the present invention. The various techniques described in U. S. patent Nos. 5,454,807,5,073,490,5,347,075,4870,009, and 4,736,866, the entire contents of which are hereby incorporated by reference, may also be used. Such methods are also described in Methods in Molecular Biology, Vol. 18, 1993, Transgenesis Techniques, Principles and Protocols, (Murphy, D., and Carter, D. A.) as well as in U. S. Patents #5, 174,986, #5, 175,383, #5, 175,384, and #5, 175,385, all of which are herein incorporated by reference.

Also intended to be within the scope of the present invention are chimeric non-human mammals in which fewer than all of the somatic and germ cells contain a DNA construct comprising a nucleic acid encoding a hSPARC-hl polypeptide of the present invention. Contemplated chimeric non-human mammals include animals produced when fewer than all of the

cells of the morula are transfected in the process of producing the transgenic animal.

Transgenic and chimeric non-human mammals having human cells or tissue engrafted therein are also encompassed by the present invention. Methods for providing chimeric non-human mammals are provided, e. g., in U. S. Serial Nos. 07/508,225, 07/518,748,07/529,217,07/562,746,07/596,518,07/574,748, 07/575,962,07/207,273,07/241,590 and 07/137,173, which are entirely incorporated herein by reference, for their description of how to engraft human cells or tissue into non- human mammals.

Alternatively, genetic constructs comprising at least one of the hSPARC-hl nucleic acid sequences as defined herein may be used to create transgenic"knockouts"of the hSPARC-hl gene. Accordingly, the present invention also provides a transgenic animal which has been engineered by homologous recombination to be deficient in the expression of the endogenous SPARC gene. Further, the invention provides a method of producing an heterozygous or homozygous transgenic animal deficient in or lacking functional SPARC proteins, respectfully, said method comprising: a) obtaining a DNA construct comprising a disrupted hSPARC-hl gene, wherein said disruption is by the insertion of an heterologous marker sequence; b) introducing said DNA construct into an ES cell of said animal such that the endogenous SPARC gene is disrupted by homologous recombination; c) selecting ES cells comprising said disrupted allele; d) incorporating the ES cells of step c) into a mouse embryo; e) transferring said embryo into a pseudopregnant animal of the said species; f) developing said embryo into a viable offspring;

g) screening offspring to identify heterozygous animal comprising said disrupted SPARC gene; and h) if desired, breeding said heterozygous animal to produce homozygous transgenic animals of said species, wherein the said homozygous animal does not express functional SPARC proteins.

Transgenic and chimeric non-human mammals of the present invention may be used for analyzing the consequences of over- expression of at least one hSPARC-hl polypeptide in vivo.

Such animals are also useful for testing the effectiveness of therapeutic and/or diagnostic agents, either associated or unassociated with delivery vectors or vehicles, which preferentially bind to an hSPARC-hl polypeptide of the present invention or act to indirectly modulate hSPARC-hl activity.

SPARC-hl transgenic non-human mammals are useful as an animal models in both basic research and drug development endeavors. Transgenic animals carrying at least one SPARC-hl polypeptide or nucleic acid can be used to test compounds or other treatment modalities which may prevent, suppress, or cure a pathology or disease associated with at least one of the above mentioned SPARC-hl activities. Such transgenic animals can also serve as a model for the testing of diagnostic methods for those same diseases. Furthermore, tissues derived from SPARC-hl transgenic non-human mammals are useful as a source of cells for cell culture in efforts to develop in vitro bioassays to identify compounds that modulate SPARC-hl activity or SPARC-hl dependent signaling.

Accordingly, another aspect of the present invention contemplates a method of identifying compounds efficacious in the treatment of at least one previously described . disease or pathology associated with SPARC activity. A non- limiting example of such a method comprises:

a) generating an SPARC-hl transgenic non-human animal which is, as compared to a wild-type animal, pathologically distinct in some detectable or measurable manner from wild- type version of said non-human mammal; b) exposing said transgenic animal to a compound, and; c) determining the progression of the pathology in the treated transgenic animal, wherein an arrest, delay, or reversal in disease progression in transgenic animal treated with-said compound as compared to the progression of the pathology in an untreated control animals is indicative that the compound is useful for the treatment of said pathology.

Another embodiment of the present invention provides a method of identifying compounds capable of inhibiting hSPARC-hl activity in vivo and/or in vitro wherein said method comprises: a) administering an experimental compound to an hSPARC-hl transgenic non-human animal, or tissues derived therefrom, exhibiting one or more physiological or pathological conditions attributable to the overexpression of an hSPARC-hl transgene; and b) observing or assaying said animal and/or animal tissues to detect changes in said physiological or pathological condition or conditions.

Another embodiment of the invention provides a method for identifying compounds capable of overcoming deficiencies in hSPARC-hl activity in vivo or in vitro wherein said method comprises: a) administering an experimental compound to an hSPARC- hl transgenic non-human animal, or tissues derived therefrom, exhibiting one or more physiological or pathological conditions attributable to the disruption of the endogenous SPARC gene; and b) observing or assaying said animal and/or animal

tissues to detect changes in said physiological or pathological condition or conditions.

Various means for determining a compound's ability to modulate hSPARC-hl in the body of the transgenic animal are consistent with the invention. Observing the reversal of a pathological condition in the transgenic animal after administering a compound is one such means. Another more preferred means is to assay for markers of hSPARC-hl activity in the blood of a transgenic animal before and after administering an experimental compound to the animal.

The level of skill of an artisan in the relevant arts readily provides the practitioner with numerous methods for assaying physiological changes related to therapeutic modulation of hSPARC-hl activity.

In all previously described in vitro and in vivo assays, the experimental compound may be administered when applicable, either superficially, orally, parenterally (e. g. by intravenous infusion or injection) or a combination of injection and infusion (iv), intramuscularly (im), or subcutaneously (sc). A preferred route of compound administration to an animal is iv, while oral administration is most preferred.

Biological and Therapeutic Uses Northern analysis of hSPARC-hl indicates expression of a 3.5 Kb band and a slightly fainter 2. 0 Kb band in RNA isolated from normal brain tissue and skeletal tissue. Data from tissue scan analysis indicates binding of hSPARC-hl protein to sections of the cerebrum and cerebellum. Clones of hSPARC-hl were identified in multiple normal types of neural tissue like brain, spinal cord and dorsal root ganglia, as well as from developing brain and diseased brain with pathologies like Alzheimer's disease, Huntington's

disease, multiple sclerosis, epilepsy, and schizophrenia. hSPARC-hl contains a signal peptide for secretion and there is a region of hSPARC-hl that fits the consensus pattern for predicting kazal type domains. Kazal domains are commonly found in extracellular matrix proteins which are often associated with neuromuscular junctions and the synaptic extracellular matrix. They are associated with the development or regeneration of synaptic junctions. There are also two EF hand Ca-binding domains and two thyroglobulin domains within hSPARC-hl. These motifs are often associated with cell motility and adhesion, extracellular matrix interactions and are thought to play a role in the control of proteolytic degradation and correlates with hSPARC-hl activities disclosed herein such as modulation of axon outgrowth during neural development and regeneration, wound healing, in particular tissue remodeling during repair of tissue and damaged neuromuscular junctions or in wound repair where re-innervation of regenerating muscle is necessary. Similarly, hSPARC-hl is useful in treatment of muscle degenerative disorders and pathologies characterized by reduced muscle function including muscular dystrophy, rhabdosarcomas, and muscle atrophy.

Northern analysis of hSPARC-hl also shows high expression in the spleen. hSPARC-hl polypeptides bound to the red pulp of the spleen and clones of hSPARC-hl were isolated from the spleen. The kazal domain of hSPARC-hl has two potential functions relating to this data. The ECM protein properties are suspected to have involvement in the creation of signaling pathways in both the nervous and immune system. These combined properties of hSPARC-hl may be used in the regulation of the immune response.

Accordingly, the present invention provides hSPARC-hl

agonists and antagonists as described herein, wherein the molecule has at least one activity, such as, but not limited to, promoting or inhibiting angiogenesis, neo- vascularization, tumorigenesis, cataractogenesis, wound healing, growth-factor mediated chemotaxis, promoting or inhibiting neurite outgrowth and/or neurite adhesion, inducing neural regeneration, inhibiting neural degeneration, preventing seizures, reducing frequency and/or severity of seizures, promoting or inhibiting primary or secondary sexual development, and altering behavioral patterns including, but not limited to, sleep and eating disorders. An hSPARC-hl agonist or antagonist can thus be screened for a corresponding activity according to methods known in the art (Sage, E. H., and Vernon, R. B., J.

Hypertension 12: S145-152 (1994)); Sage, E. H., Adv. Oncol.

12: 17-29 (1996); Ledda F., et al., Nature Med., 3: 171-176 (1997); Sage E. H., et al., Nature Med., 3: 144-146 (1997); Gilmour, D. T., et al., EMBO J., 17: 1860-1870 (1998); Kolodkin, A., et al., Neuron, 21: 1079-1092, (1998); Kolodkin, et al., (1997); Pimenta et al., Neuron, 15: 287-297 (1995); Norose, K., et al., Invest. Ophthalmol. Vis. Sci., 39: 2674-2680t (1998); Reed, M. J., et al., Current Topics in Microbiology and Immunology, 213 : 81-94 (1996); Mendis, D. B., et al., Brain Research 713: 53-63 (1996)); Wilson et al., J. Cell Sci. 109: 3129-3138 (1996); or methods specified herein (see Examples 7-9). Further obvious modifications of screening assays disclosed herein or screening assays otherwise known in the art enable a skilled artisan to identify substantially similar polypeptides having substantially similar activity as the hSPARC-hl polypeptides disclosed herein. Similarly, obvious modifications of screening assays disclosed herein (see Examples 7-9) or screening assays otherwise known in the art enable a skilled artisan to identify compounds which act to modulate the

activity of an hSPARC-hl polypeptide (e. g., anti-hSPARC-hl antibodies) are within the contemplation of an ordinarily skilled artisan.

The invention also provides methods for treating conditions associated with altered expression of hSPARC-hl polynucleotides and/or polypeptides, for example, cell proliferative disorders (e. g., cell proliferative disorders of the central or peripheral nervous systems, for example, conditions affecting neural tissue, testes, heart tissue, and cells of the eye). Treatment of an hSPARC-hl-associated cell proliferative disorder can be carried out, for example, by modulating hSPARC-hl gene expression or hSPARC-hl activity in a cell. The term"modulate"includes, for example, suppressing expression of an hSPARC-hl when it is over-expressed, and augmenting expression of an hSPARC-hl when it is under-expressed. In cases where a disorder is associated with over-expression of an hSPARC-hl including, but not limited to, Alzheimer's disease, Gullain-Barre syndrome, Parkinson's disease, multiple sclerosis, epilepsy, schizophrenia, and cancer, antagonists that interfere with hSPARC-hl expression, at transcriptional or translational levels, or endogenous hSPARC-hl activity can be used to treat the disorder. This approach may employ, for example, antisense nucleic acids (i. e., nucleic acids that are complementary to, or capable of hybridizing with, a target . nucleic acid, e. g., a nucleic acid encoding an hSPARC-hl polypeptide), ribozymes, or triplex agents. The antisense and triplex approaches function by masking the nucleic acid, while the ribozyme strategy functions by cleaving the nucleic acid. In addition, antibodies that bind to hSPARC- hl polypeptides can be used in methods to block the activity of an hSPARC-hl.

The use of antisense methods to inhibit the in vitro translation of genes is well-known in the art (see, e. g.,

Marcus-Sakura, Anal. Biochem., 172: 289,1988). Antisense nucleic acids are nucleic acid molecules (e. g., molecules containing DNA nucleotides, RNA nucleotides, or modifications (e. g., modification that increase the stability of the molecule, such as 2'-0-alkyl (e. g., methyl) substituted nucleotides) or combinations thereof) that are complementary to, or that hybridize to, at least a portion of a specific nucleic acid molecule, such as an RNA molecule (e. g., an mRNA molecule) (see, e. g., Weintraub, Scientific American, 262: 40,1990). The antisense nucleic acids hybridize to corresponding nucleic acids, such as mRNAs, to form a double-stranded molecule, which interferes with translation of the mRNA, as the cell will not translate an double-stranded mRNA. Antisense nucleic acids used in the invention are typically at least 10-12 nucleotides in length, for example, at least 15,20,25,50,75, or 100 nucleotides in length. The antisense nucleic acid can also be. as long as the target nucleic acid with which it is intended that it form an inhibitory duplex. As is described further below, the antisense nucleic acids can be introduced into cells as antisense oligonucleotides, or can be produced in a cell in which a nucleic acid encoding the antisense nucleic acid has been introduced by, for example, using gene therapy methods.

Introduction of hSPARC-hl antisense or ribozyme nucleic acids into cells affected by a proliferative disorder, for the purpose of gene therapy, can be achieved using a recombinant expression vector, such as a chimeric virus or a colloidal dispersion system, such as a targeted liposome.

Those of skill in this art know or can easily ascertain the appropriate route and means for introduction of sense or antisense hSPARC-hl nucleic acids, without resort to undue experimentation.

Ribozymes are RNA molecules possessing the ability to

specifically cleave other single stranded RNA molecules in a manner somewhat analogous to DNA restriction endonucleases.

Ribozymes were discovered from the observation that certain mRNAs have the ability to excise their own introns. By modifying the nucleotide sequence of these RNAs, researchers have been able to engineer molecules that recognize specific nucleotide sequences in an RNA molecule and cleave it (Cech, 1988.). Because they are sequence-specific, only mRNAs with particular sequences are inactivated. Six basic varieties of naturally-occurring enzymatic RNAs are known presently.

Each can catalyze the hydrolysis of RNA phosphodiester bonds in trans (and thus can cleave other RNA molecules) under physiological conditions. Tetrahymena-type ribozymes recognize four-base sequences, while"hammerhead"-type recognize eleven-to eighteen-base sequences (Hasselhoff and Gerlach, 1988). The longer the recognition sequence, the more likely it is to occur exclusively in the target mRNA species. Therefore, hammerhead-type ribozymes are preferable to Tetrahymena-type ribozymes for inactivating a specific mRNA species, and eighteen base recognition sequences are preferable to shorter recognition sequences.

The DNA sequences described herein may thus be used to prepare antisense molecules against, and ribozymes that cleave mRNAs encoding hhSPARC-hl polypeptides and their ligands directed at decreasing hhSPARC-hl activity in a mammal. These specific motifs are not limiting in the invention and those skilled in the art will recognize that all that is important in an enzymatic RNA molecule of this invention is that it has a specific substrate binding site which is complementary to one or more of the target gene RNA regions, and that it have nucleotide sequences within or surrounding that substrate binding site which impart an RNA cleaving activity to the molecule (see, for instance, United States Patents 6022962,5977343,5891684,5837855,5831071,

5631359, the entirety of these patents are hereby incorporated by reference herein).

In addition to blocking mRNA translation, oligonucleotides, such as antisense oligonucleotides, can be used in methods to stall transcription, such as the triplex method. In this method, an oligonucleotide winds around double-helical DNA in a sequence-specific manner, forming a three-stranded helix, which blocks transcription from the targeted gene. These triplex compounds can be designed to recognize a unique site on a chosen gene (Maher, et al., Antisense Res. and Dev., 1 (3): 227,1991; Helene, Anticancer Drug Design, 6 (6): 569,1991).

The present invention also provides a method to induce or inhibit a neurite outgrowth, induce or inhibit neurite adhesion, induce neuronal regeneration, inhibit neuronal degeneration, prevent or reduce frequency and/or severity of seizures, induce or inhibit growth-factor mediated chemotaxis, prevent or treat cell-proliferative disorders, induce or inhibit primary or secondary sexual development, or alter behavioral patterns including, but not limited to, sleep or eating disorders in a mammal wherein said method comprises administering to said mammal a biologically active and pharmaceutically acceptable composition of the present invention. Preferably, the condition to be treated involves one or more of the following disorders: nervous system nerve damage, neurodegeneration, trigeminal neuralgia, glossopharyngeal neuralgia, Bell's Palsy, myasthenia gravis, muscular dystrophy, progressive muscular atrophy, progressive bulbar inherited muscular atrophy, herniated, ruptured or prolapsed invertebrae disk, cervical spondylosis, plexus disorders, thoracic outlet destruction, peripheral neuropathy, such as those caused by lead, dapsone, ticks or porphyria, peripheral myelin disorders, Alzheimer's disease, Gullain-Barre syndrome, Parkinson's

disease, Parkinsonian disorders, ALS, multiple sclerosis, other central myelin disorders, stroke, ischemia associated with stroke, neural paropathy, other neural degenerative diseases, motor. neuron. diseases, sciatic crush, neuropathy associated with diabetes, spinal cord injuries, facial nerve crush, chemotherapy-or pharmacotherapy-induced neuropathy, Huntington's disease, cancer, retinal degenerative diseases, such as retinitis pigmentosa and macular degeneration, and peripheral neuropathies, abnormal primary or secondary sexual development, impotence, infertility, reduced libido, and behavioral disorders such as sleeping or eating disorders.

Even more preferred embodiments of the present methods comprise administering to a patient a biologically active and pharmaceutically acceptable composition of the present invention which further comprises at least one other anti- tumorigenic, neurotrophic, neuroprotective, thrombolytic, anti-proliferative, and/or anti-thrombotic agent known to be biologically active.

The methods of the present invention may be particularly useful for treating or preventing neuronal damage resulting from NO mediated toxicity and/or enhancing recovery of neuronal function.

The invention further provides for the use of a hSPARC- hl agonist, hSPARC-hl antagonist, hSPARC-hl polypeptide, hSPARC-hl nucleic acid, or hSPARC-hl antibody in the manufacture of a medicament for the treatment or prevention of, tissue damage, cell proliferation or tissue growth disorders such as nerve damage, neurodegeneration, trigeminal neuralgia, glossopharyngeal neuralgia, Bell's Palsy, myasthenia gravis, muscular dystrophy, progressive muscular atrophy, progressive bulbar inherited muscular atrophy, herniated, ruptured or prolapsed invertebrae disk, cervical. spondylosis, plexus disorders, thoracic outlet

destruction, peripheral neuropathy, such as those caused by lead, dapsone, ticks or porphyria, peripheral myelin disorders, Alzheimer's disease, Gullain-Barre syndrome, Parkinson's disease, Parkinsonian disorders, ALS, multiple sclerosis, other central myelin disorders, stroke, ischemia associated with stroke, neural paropathy, other neural degenerative diseases, motor neuron diseases, sciatic crush, neuropathy associated with diabetes, spinal cord injuries, facial nerve crush, chemotherapy-or pharmacotherapy-induced neuropathy, Huntington's disease, cancer, retinal degenerative diseases, such as retinitis pigmentosa and macular degeneration, and abnormal primary or secondary sexual development.

The present invention contemplates the use of hSPARC-hl agonists, hSPARC-hl antagonists, hSPARC-hl polynucleotides, hSPARC-hl polypeptides, hSPARC-hl variants, and hSPARC-hl antibodies to treat metabolic diseases such as obesity, cachexia, bulimia, anorexia, and/or disorders commonly associated with these conditions such as dyslipidemia and diabetes, especially non-insulin dependent diabetes. Those skilled in the art are able to recognize the presence of such diseases in a subject (e. g, a mammalian subject, such as a human, a domesticated animal, or an animal used in agriculture). Accordingly, the present invention includes the administration of hSPARC-hl agonists, hSPARC-hl antagonists, hSPARC-hl polynucleotides, hSPARC-hl polypeptides, hSPARC-hl variants, and/or hSPARC-hl antibodies for veterinary or human therapeutic uses According to another embodiment, the invention provides methods to induce or inhibit angiogenesis, neovascular- ization, tumorigenesis, cataractogenesis, wound healing, growth-factor mediated chemotaxis, neurite outgrowth and/or neurite adhesion comprising administering to patient a pharmaceutically acceptable composition comprising a hSPARC-

hl nucleic acid, polypeptide, cells, and/or antibody described herein and a pharmaceutically acceptable carrier.

For therapeutic utility, an effective amount of hSPARC-hi polypeptide, hSPARC-hl variant, and/or hSPARC-hl antibody is administered to a mammal in need thereof in a dose between about 0.1 and 1000 pg/kg body weight. In practicing the methods contemplated by this invention, the hSPARC-hl polypeptides, hSPARC-hl variants, and/or hSPARC-hl antibodies as defined herein can be administered in multiple doses per day, in single daily doses, in weekly doses, or at any other regular interval. The amount per administration and frequency of administration will be determined by a physician and depend on such factors as the nature and severity of the disease, and the age and general health of the patient.

The present invention also provides a pharmaceutical composition comprising as the active agent an hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody, and/or a pharmaceutically acceptable non-toxic salt thereof, and a pharmaceutically acceptable solid or liquid carrier. For example, compounds comprising at least one hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody can be admixed with conventional pharmaceutical carriers and excipients, and used in the form of tablets, capsules, elixirs, suspensions, syrups, wafers, parenteral formulations, and the like. The compositions comprising at least one hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody, will contain from about 0.1% to 90% by

weight of the active compound, and more generally from about 10% to 30%. The compositions may contain common carriers and excipients such as corn-starch or gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride, and alginic acid.

As a general proposition, the total pharmaceutically effective amount of a hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody administered parenterally per dose will be in the range of about 1 tg/kg/day to 10 mg/kg/day of patient body weight, particularly 2 mg/kg/day to 8 mg/kg/day, more particularly 2 mg/kg/day to 4 mg/kg/day, even more particularly 2.2 mg/kg/day to 3.3 mg/kg/day, and finally 2.5 mg/kg/day, although, as noted above, this will be subject to therapeutic discretion. More preferably, this dose is at least 0.01 mg/kg/day. If given continuously, a hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody is typically administered at a dose rate of about 1 pg/kg/hour to about 50 llg/kg/hour, either by 1-4 injections per day or by continuous subcutaneous infusions, for example, using a mini-pump. An intravenous bag solution may also be employed. The length of treatment needed to observe changes and the interval following treatment for responses to occur appears to vary depending on the desired effect.

Pharmaceutical compositions containing a hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody may be administered orally, rectally, intracranially, parenterally, intracisternally, intravaginally, intraperitoneally,

topically (as by powders, ointments, drops or transdermal patch), transdermally, intrathecally, bucally, or as an oral or nasal spray. By"pharmaceutically acceptable carrier"is meant a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. The term"parenteral"as used herein includes, but is not limited to, modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intra-articular injection, infusion and implants comprising a hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody.

The compounds can be formulated for oral or parenteral administration. A preferred parenteral formulation for subcutaneous administration. would comprise a buffer (phosphate, citrate, acetate, borate, TRIS), salt (NaCl, KCl), divalent metal (Zn, Ca), and isotonicty agent (glycerol, mannitol), detergent (Polyoxyethylene sorbitan fatyy acid esters, poloxamer, ddicusate sodium, sodium lauryl sulfate), antioxidants (ascorbic acid), and antimicrobial agent (phenol, m-cresol, alcohol, benzyl alcohol, butylparben, methylparaben, ethylparaben, chlorocresol, phenoxyethanol, phenylethyl alcohol, propylparaben.

For intravenous (IV) use, a hSPARC-hl agonist, hSPARC- hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody is administered in commonly used intravenous fluid (s) and administered by infusion. Such fluids, for example, physiological saline, Ringer's solution or 5% dextrose solution can be used.

For intramuscular preparations, a sterile formulation,

preferably a suitable soluble salt form of a hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody such as the hydrochloride salt, can be dissolved and administered in a pharmaceutical diluent such as pyrogen-free water (distilled), physiological saline or 5% glucose solution. A suitable insoluble form of the compound may be prepared and administered as a suspension in an aqueous base or a pharmaceutically acceptable oil base, e. g. an ester of a long chain fatty acid such as ethyl oleate.

A hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody is also suitably administered by sustained-release systems.

Suitable examples of sustained-release compositions include semi-permeable polymer matrices in the form of shaped articles, e. g., films, or microcapsules. Sustained-release matrices include polylactides (U. S. Pat. No. 3,773.919, EP 58, 481), copolymers of L-glutamic acid and gamma-ethyl-L- glutamate (Sidman, U. et al., Biopolymers 22: 547-556 (1983)), poly (2-hydroxyethyl methacrylate) (R. Langer et al., J. Biomed. Mater. Res. 15: 167-277 (1981), and R.

Langer, Chem. Tech. 12: 98-105 (1982)), ethylene vinyl acetate (R. Langer et al., Id.) or poly-D- (-)-3- hydroxybutyric acid (EP 133,988). Other sustained-release compositions also include liposomally entrapped modified hSPARC-hl polypeptides and/or fragments thereof and/or variants thereof. Such liposomes are prepared by methods known per se: DE 3,218,121; Epstein et al., Proc. Natl.

Acad. Sci. (USA) 82: 3688-3692 (1985); Hwang et al.,. Proc.

Natl. Acad. Sci. (USA) 77 : 4030-4034 (1980); EP 52,322; EP 36,676; EP 88,046; EDP 143,949; EP 142,641; Japanese Pat.

Appl. 83-118008; U. S. Pat. Nos. 4,485,045 and 4,544,545; and EP 102,324. Ordinarily, the liposomes are of the small (about 200-800 Angstroms) unilamellar type in which the lipid content is greater than about 30 mol. percent cholesterol, the selected proportion being adjusted for the optimal therapy.

For parenteral administration, in one embodiment, the hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody is formulated generally by mixing it at the desired degree of purity, in a unit dosage injectable form (solution, suspension, or emulsion), with a pharmaceutically acceptable carrier, i. e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation. For example, the formulation preferably does not include oxidizing agents and other compounds that are known to be deleterious to polypeptides.

Generally, the formulations are prepared by contacting hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody uniformly and intimately with liquid carriers or finely divided solid carriers or both. Then, if necessary, the product is shaped into the desired formulation. Preferably the carrier is a parenteral carrier, more preferably a solution that is isotonic with the blood of the recipient. Examples of such carrier vehicles include water, saline, Ringer's solution, and dextrose solution. Non-aqueous vehicles such as fixed oils and ethyl oleate are also useful herein, as well as liposomes.

The carrier suitably contains minor amounts of additives such as substances that enhance isotonicity and

chemical stability. Such materials are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, succinate, acetic acid, and other organic acids or their salts; antioxidants such as ascorbic acid; low molecular weight (less than about ten residues) polypeptides, e. g., polyarginine or tripeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids, such as glycine, glutamic acid, aspartic acid, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, manose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; counterions such as sodium; and/or nonionic surfactants such as polysorbates, poloxamers, or PEG.

A hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody is typically formulated in such vehicles at a concentration of about 0.1 mg/ml to 100 mg/ml, preferably 1-10 mg/ml, at a pH of about 3 to 8. It will be understood that the use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of salts of the particular active ingredient (s).

Compositions to be used for therapeutic administration must be sterile. Sterility is readily accomplished by filtration through sterile filtration membranes (e. g., 0.2 micron membranes). Therapeutic compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.

Compositions comprising a hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl

antibody ordinarily will be stored in unit or multi-dose containers, for example, sealed ampoules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution. As an example of a lyophilized formulation, 10-ml vials are filled with 5 ml of sterile-filtered 1% (w/v) aqueous solution of one of the hSPARC-hl polypeptides and/or fragments and/or hSPARC-hl variants of the present invention, and the resulting mixture is lyophilized. The infusion solution is prepared by reconstituting the lyophilized polypeptide using bacteriostatic Water-for- Injection.

The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Associated with such container (s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. In addition, a hSPARC-hl agonist, hSPARC-hl antagonist, hSPARC-hl polynucleotide, hSPARC-hl polypeptide, hSPARC-hl fragment, hSPARC-hl variant, and/or hSPARC-hl antibody may be administered in the methods of the present invention in combination with other therapeutic compounds.

Administration in combination with one or more further therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order.

Gene Therapy Nucleic acids encoding the hSPARC-hl polypeptides may also be used in gene therapy. In gene therapy applications, genes are introduced into cells in order to achieve in vivo synthesis of a therapeutically effective genetic product, for

example, for replacement of a defective gene or to treat conditions associated with insufficient hSPARC-hl expression.

"Gene therapy"includes both conventional gene therapy where a lasting effect is achieved by a single treatment, and the administration of gene therapeutic agents, which involves the one time or repeated administration of a therapeutically effective DNA or mRNA. Antisense RNAs and DNAs can be used as therapeutic agents for blocking the expression of certain genes in vivo. It has already been shown that short antisense oligo-nucleotides can be imported into cells where act as inhibitors, despite their low intracellular concentrations caused by their restricted uptake by the cell membrane (Zamecnik et al., Proc. Natl. Acad Sci. USA, 83: 4143-4146 (1986)). The oligonucleotides can be modified to enhance their uptake, e. g., by substituting their negatively charged phosphodiester groups by uncharged groups. There are a variety of techniques available for introducing nucleic acids into viable cells. The techniques vary depending upon whether the nucleic acid is transferred into cultured cell in vitro, or in vivo in the cells of the intended host. Techniques suitable for the transfer of nucleic acid into mammalian cells in vitro include the use of liposomes, electroporation, microinjection, cell fusion, DEAE-dextran, the calcium phosphate precipitation method, etc. The currently preferred in vivo gene transfer techniques include transfection with viral (typically retroviral) vectors and viral coat protein-liposome mediated transfection (Dzau, et al., Trends in Biotechnology, 11: 205- 210 (1993). In some situations it is desirable to provide the nucleic acid source with an agent that targets the target cells, such as an antibody specific for a cell surface membrane protein or the target cell, a ligand for a receptor on the target cells, etc. Where liposomes are employed, proteins which bind to a cell surface membrane protein associated with endocytosis may by used for targeting and/or

to facilitate uptake, e. g., capsid proteins or fragments thereof tropic for a particular cell type, antibodies for proteins which undergo internalization in cycling, protein that target intracellular localization and enhance intracellular half-life. The technique of receptor-mediated endocytosis is described, for example by Wu et al., J. Biol.

Chem., 262: 4429-4432 (1987). For a review of gene marking and gene therapy protocols see Anderson, Science, 256: 808-813 (1992).

The principles, preferred embodiments, and modes of operation of the present invention have been described in the foregoing specification. The invention which is intended to be protected herein, however, is not to be construed as limited to the particular forms disclosed, since they are to be regarded as illustrative rather than restrictive. Variations and changes may be made by those skilled in the art without departing from the spirit of the invention.

Having generally described the invention, the same will be more readily understood by reference to the following examples, which are provided by way of illustration and are not intended as limiting.

EXAMPLES Example 1: Expression and Purification of an Human SPARC-hl Polypeptide in E. coli The bacterial expression vector pQE60 is used for bacterial expression in this example. (QIAGEN, Inc., Chatsworth, CA). pQE60 encodes ampicillin antibiotic resistance ("Ampr") and contains a bacterial origin of replication ("ori"), an IPTG inducible promoter, a ribosome binding site ("RBS"), six codons encoding histidine residues

that allow affinity purification using nickel-nitrilo-tri- acetic acid ("Ni-NTA") affinity resin sold by QIAGEN, Inc., and suitable single restriction enzyme cleavage sites.

These elements are arranged such that a DNA fragment encoding a polypeptide can be inserted in such a way as to produce that polypeptide with the six His residues (i. e., a "6 X His tag") covalently linked to the carboxyl terminus of that polypeptide. However, a polypeptide coding sequence can optionally be inserted such that translation of the six His codons is prevented and, therefore, a polypeptide is produced with no 6 X His tag.

The nucleic acid sequence encoding the desired portion of an hSPARC-hl polypeptide lacking the hydrophobic leader sequence is amplified from the deposited cDNA clone using PCR oligonucleotide primers (based on the sequences presented, e. g., as presented in at least one of SEQ ID NOS: 1, 2,3), which anneal to the amino terminal encoding DNA sequences of the desired portion of an hSPARC-hl polypeptide and to sequences in the deposited construct 3' to the cDNA coding sequence. Additional nucleotides containing restriction sites to facilitate cloning in the pQE60 vector are added to the 5'and 3'sequences, respectively.

For cloning an hSPARC-hl polypeptide, the 5'and 3' primers have nucleotides corresponding or complementary to a portion of the coding sequence of an hSPARC-hl, e. g., as presented in at least one of SEQ ID NOS: 1, 2,3, according to known method steps. One of ordinary skill in the art would appreciate, of course, that the point in a polypeptide coding sequence where the 5'primer begins can be varied to amplify a desired portion of the complete polypeptide shorter or longer than the mature form.

The amplified hSPARC-hl nucleic acid fragments and the vector pQE60 are digested with appropriate restriction

enzymes and the digested DNAs are then ligated together.

Insertion of the hSPARC-hl DNA into the restricted pQE60 vector places an hSPARC-hl polypeptide coding region including its associated stop codon downstream from the IPTG-inducible promoter and in-frame with an initiating AUG codon. The associated stop codon prevents translation of the six histidine codons downstream of the insertion point.

The ligation mixture is transformed into competent E. coli cells using standard procedures such as those described in Sambrook, et al., 1989; Ausubel, 1987-1998. E. coli strain Ml5/rep4, containing multiple copies of the plasmid pREP4, which expresses the lac repressor and confers kanamycin resistance ("Kanr"), is used in carrying out the illustrative example described herein. This strain, which is only one of many that are suitable for expressing hSPARC- hl polypeptide, is available commercially from QIAGEN, Inc.

Transformants are identified by their ability to grow on LB plates in the presence of ampicillin and kanamycin. Plasmid DNA is isolated from resistant colonies and the identity of the cloned DNA confirmed by restriction analysis, PCR and DNA sequencing.

Clones containing the desired constructs are grown overnight ("0/N") in liquid culture in LB media supplemented with both ampicillin (100 pg/ml) and kanamycin (25, ug/ml).

The O/N culture is used to inoculate a large culture, at a dilution of approximately 1: 25 to 1: 250. The cells are grown to an optical density at 600 nm ("OD600") of between 0.4 and 0.6. Isopropyl-b-D-thiogalactopyranoside ("IPTG") is then added to a final concentration of 1 mM to induce transcription from the lac repressor sensitive promoter, by inactivating the laci repressor. Cells subsequently are incubated further for 3 to 4 hours. Cells then are harvested by centrifugation.

The cells are then stirred for 3-4 hours at 4°C in 6M guanidine-HCl, pH8. The cell debris is removed by centrifugation, and the supernatant containing the hSPARC-hl is dialyzed against 50 mM Na-acetate buffer pH6, supplemented with 200 mM NaCl. Alternatively, a polypeptide can be successfully refolded by dialyzing it against 500 mM NaCl, 20% glycerol, 25 mM Tris/HCl pH7.4, containing protease inhibitors.

If insoluble protein is generated, the protein is made soluble according to known method steps. After renaturation the polypeptide is purified by ion exchange, hydrophobic interaction and size exclusion chromatography.

Alternatively, an affinity chromatography step such as an antibody column is used to obtain pure hSPARC-hl polypeptide. The purified polypeptide is stored at 4°C or frozen at-40°C to-120°C.

Example 2: Cloning and Expression of an hSPARC-hl Polypeptide in a Baculovirus Expression System In this illustrative example, the plasmid shuttle vector pA2 GP is used to insert the cloned DNA encoding the mature polypeptide into a baculovirus to express an hSPARC- hl polypeptide, using a baculovirus leader and standard methods as described in Summers, et al., A Manual of Methods for Baculovirus Vectors and Insect Cell Culture Procedures, Texas Agricultural Experimental Station Bulletin No. 1555 (1987). This expression vector contains the strong polyhedrin promoter of the Autographa californica nuclear polyhedrosis virus (AcMNPV) followed by the secretory signal peptide (leader) of the baculovirus gp67 polypeptide and convenient restriction sites such as BamHI, Xba I and Asp718. The polyadenylation site of the simian virus 40 ("SV40") is used for efficient polyadenylation. For easy

selection of recombinant virus, the plasmid contains the beta-galactosidase gene from E. coli under control of a weak Drosophila promoter in the same orientation, followed by the polyadenylation signal of the polyhedrin gene. The inserted genes are flanked on both sides by viral sequences for cell- mediated homologous recombination with wild-type viral DNA to generate viable virus that expresses the cloned polynucleotide.

Other baculovirus vectors are used in place of the vector above, such as pAc373, pVL941 and pAcIMl, as one skilled in the art would readily appreciate, as long as the construct provides appropriately located signals for transcription, translation, secretion and the like, including a signal peptide and an in-frame AUG as required.

Such vectors are described, for instance, in Luckow, et al., Virology, 170: 31-39 (1989).

The cDNA sequence encoding the mature hSPARC-hl polypeptide in the deposited or other clone, lacking the AUG initiation codon and the naturally associated nucleotide binding site, is amplified using PCR oligonucleotide primers corresponding to the 5'and 3'sequences of the gene. Non- limiting examples include 5'and 3'primers having nucleotides corresponding or complementary to a portion of the coding sequence of an hSPARC-hl polypeptide, e. g., as presented in at least one of SEQ ID NOS: 1, 2,3, according to known method steps.

The amplified fragment is isolated from a 1% agarose gel using a commercially available kit (e. g.,"Geneclean," BIO 101 Inc., La Jolla, CA). The fragment then is then digested with the appropriate restriction enzyme and again is purified on a 1% agarose gel. This fragment is designated herein"F1".

The plasmid is digested with the corresponding restriction enzymes and optionally, can be dephosphorylated

using calf intestinal phosphatase, using routine procedures known in the art. The DNA is then isolated from a 1% agarose gel using a commercially available kit ("Geneclean" BIO 101 Inc., La Jolla, CA). This vector DNA is designated herein"V1".

Fragment F1 and the dephosphorylated plasmid V1 are ligated together with T4 DNA ligase. E. coli HB101 or other suitable E. coli hosts such as XL-1 Blue (Stratagene Cloning Systems, La Jolla, CA) cells are transformed with the ligation mixture and spread on culture plates. Bacteria are identified that contain the plasmid with the hSPARC-hl gene using the PCR method, in which one of the primers that is used to amplify the gene and the second primer is from well within the vector so that only those bacterial colonies containing the hSPARC-hl gene fragment will show amplification of the DNA. The sequence of the cloned fragment is confirmed by DNA sequencing. This plasmid is designated herein pBac hSPARC-hl.

Five jug of the plasmid pBachSPARC-hl is co-transfected with 1.0 J. g of a commercially available linearized baculovirus DNA ("BaculoGoldTM baculovirus DNA", Pharmingen, San Diego, CA), using the lipofection method described by Felgner, et al., Proc. Natl. Acad. Sci. USA, 84: 7413-7417 (1987). 1 jug of BaculoGold virus DNA and 5 g of the plasmid pBac hSPARC-hl are mixed in a sterile well of a microtiter plate containing 50 ttl of serum-free Grace's medium (Life Technologies, Inc., Rockville, MD).

Afterwards, 10, ul Lipofectin plus 90 1 Grace's medium are added, mixed and incubated for 15 minutes at room temperature. Then the transfection mixture is added drop- wise to Sf9 insect cells (ATCC CRL 1711) seeded in a 35 mm tissue culture plate with 1 ml Grace's medium without serum.

The plate is rocked back and forth to mix the newly added

solution. The plate is then incubated for 5 hours at 27°C.

After 5 hours the transfection solution is removed from the plate and 1 ml of Grace's insect medium supplemented with 10% fetal calf serum is added. The plate is put back into an incubator and cultivation is continued at 27°C for four days.

After four days the supernatant is collected and a plaque assay is performed, according to known methods. An agarose gel with"Blue Gal" (Life Technologies, Inc., Rockville, MD) is used to allow easy identification and isolation of gal-expressing clones, which produce blue- stained plaques. (A detailed description of a"plaque assay"of this type can also be found in the user's guide for insect cell culture and baculovirology distributed by Life Technologies, Inc., Rockville, MD, page 9-10). After appropriate incubation, blue stained plaques are picked with a micropipettor tip (e. g., Eppendorf). The agar containing the recombinant viruses is then resuspended in a microcentrifuge tube containing 200 1ll of Grace's medium and the suspension containing the recombinant baculovirus is used to infect Sf9 cells seeded in 35 mm dishes. Four days later the supernatants of these culture dishes are harvested and then they are stored at 4°C. The recombinant virus is called V-hSPARC-hl.

To verify the expression of the hSPARC-hl gene, Sf9 cells are grown in Grace's medium supplemented with 10% heat-inactivated FBS. The cells are infected with the recombinant baculovirus V-hSPARC-hl at a multiplicity of infection ("MOI") of about 2. Six hours later the medium is removed and is replaced with SF900 II medium minus methionine and cysteine (available, e. g., from Life Technologies, Inc., Rockville, MD). If radiolabeled polypeptides are desired, 42 hours later, 5 mCi of 35S-

methionine and 5 mCi 35S-cysteine (available from Amersham) are added. The cells are further incubated for 16 hours and then they are harvested by centrifugation. The polypeptides in the supernatant as well as the intracellular polypeptides are analyzed by SDS-PAGE followed by autoradiography (if radiolabeled). Microsequencing of the amino acid sequence of the amino terminus of purified polypeptide can be used to determine the amino terminal sequence of the mature polypeptide and thus the cleavage point and length of the secretory signal peptide.

Example 3: Cloning and Expression of hSPARC-hl in Mammalian Cells A typical mammalian expression vector contains at least one promoter element, which mediates the initiation of transcription of mRNA, the polypeptide coding sequence, and signals required for the termination of transcription and polyadenylation of the transcript. Additional elements include enhancers, Kozak sequences and intervening sequences flanked by donor and acceptor sites for RNA splicing.

Highly efficient transcription can be achieved with the early and late promoters from SV40, the long terminal repeats (LTRS) from Retroviruses, e. g., RSV, HTLVI, HIVI and the early promoter of the cytomegalovirus (CMV). However, cellular elements can also be used (e. g., the human actin promoter). Suitable expression vectors for use in practicing the present invention include, for example, vectors such as pIRESlneo, pRetro-Off, pRetro-On, PLXSN, or pLNCX (Clontech Labs, Palo Alto, CA), pcDNA3. 1 (+/-), pcDNA/Zeo (+/-) or pcDNA3. 1/Hygro (+/-) (Invitrogen), PSVL and PMSG (Pharmacia, Uppsala, Sweden), pRSVcat (ATCC 37152), pSV2dhfr (ATCC 37146) and pBC12MI (ATCC 67109). Mammalian host cells that could be used include human Hela 293, H9 and

Jurkat cells, mouse NIH3T3 and C127 cells, Cos 1, Cos 7 and CV 1, quail QC1-3 cells, mouse L cells and Chinese hamster ovary (CHO) cells.

Alternatively, the gene can be expressed in stable cell lines that contain the gene integrated into a chromosome.

The co-transfection with a selectable marker such as dhfr, gpt, neomycin, or hygromycin allows the identification and isolation of the transfected cells.

The transfected gene can also be amplified to express large amounts of the encoded polypeptide. The DHFR (dihydrofolate reductase) marker is useful to develop cell lines that carry several hundred or even several thousand copies of the gene of interest. Another useful selection marker is the enzyme glutamine synthase (GS) (Murphy, et al., Biochem. J., 277: 277-279 (1991); Bebbington, et al., Bio/Technology, 10: 169-175 (1992)). Using these markers, the mammalian cells are grown in selective medium and the cells with the highest resistance are selected. These cell lines contain the amplified gene (s) integrated into a chromosome. Chinese hamster ovary (CHO) and NSO cells are often used for the production of polypeptides.

The expression vectors pC1 and pC4 contain the strong promoter (LTR) of the Rous Sarcoma Virus (Cullen, et al., Mol. Cell. Biol., 5: 438-447 (1985)) plus a fragment of the CMV-enhancer (Boshart, et al., Cell, 41: 521-530 (1985)).

Multiple cloning sites, e. g., with the restriction enzyme cleavage sites BamHI, XbaI and Asp718, facilitate the cloning of the gene of interest. The vectors contain in addition the 3'intron, the polyadenylation and termination signal of the rat preproinsulin gene.

Example 3 (a): Cloning and Expression in COS Cells

The expression plasmid, phSPARC-hl HA, is made by cloning a cDNA encoding hSPARC-hl into the expression vector pcDNAI/Amp or pcDNAIII (which can be obtained from Invitrogen, Inc.).

The expression vector pcDNAI/amp contains: (1) an E. coli origin of replication effective for propagation in E. coli and other prokaryotic cells; (2) an ampicillin resistance gene for selection of plasmid-containing prokaryotic cells; (3) an SV40 origin of replication for propagation in eucaryotic cells; (4) a CMV promoter, a polylinker, an SV40 intron; (5) several codons encoding a hemagglutinin fragment (i. e., an"HA"tag to facilitate purification) or HIS tag (see, e. g, Ausubel, supra) followed by a termination codon and polyadenylation signal arranged so that a cDNA can be conveniently placed under expression control of the CMV promoter and operably linked to the SV40 intron and the polyadenylation signal by means of restriction sites in the polylinker. The HA tag corresponds to an epitope derived from. the influenza hemagglutinin polypeptide described by Wilson, et al., Cell, 37: 767-778 (1984). The fusion of the HA tag to the target polypeptide allows easy detection and recovery of the recombinant polypeptide with an antibody that recognizes the HA epitope. pcDNAIII contains, in addition, the selectable neomycin marker.

A DNA fragment encoding the hSPARC-hl is cloned into the polylinker region of the vector so that recombinant polypeptide expression is directed by the CMV promoter. The plasmid construction strategy is as follows. The hSPARC-hl cDNA of the deposited clone is amplified using primers that contain convenient restriction sites, much as described above for construction of vectors for expression of hSPARC- hl in E. coli. Non-limiting examples of suitable primers include those based on the coding sequences presented in at

least one of SEQ ID NOS: 1, 2, 3, as they encode hSPARC-hl polypeptides as described herein.

The PCR amplified DNA fragment and the vector, pcDNAI/Amp, are digested with suitable restriction enzyme (s) and then ligated. The ligation mixture is transformed into E. coli strain SURE (available from Stratagene Cloning Systems, 11099 North Torrey Pines Road, La Jolla, CA 92037), and the transformed culture is plated on ampicillin media plates which then are incubated to allow growth of ampicillin resistant colonies. Plasmid DNA is isolated from resistant colonies and examined by restriction analysis or other means for the presence of the hSPARC-hl-encoding fragment.

For expression of recombinant hSPARC-hl, COS cells are transfected with an expression vector, as described above, using DEAE-DEXTRAN, as described, for instance, in Sambrook, et al., Molecular Cloning: a Laboratory Manual, Cold Spring Laboratory Press, Cold Spring Harbor, New York (1989).

Cells are incubated under conditions for expression of hSPARC-hl by the vector.

Expression of the hSPARC-hl-HA fusion polypeptide is detected by radiolabeling and immunoprecipitation, using methods described in, for example Harlow, et al., Antibodies: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1988). To this end, two days after transfection, the cells are labeled by incubation in media containing 35S-cysteine for 8 hours.

The cells and the media are collected, and the cells are washed and lysed with detergent-containing RIPA buffer: 150 mM NaCl, 1% NP-40,0.1% SDS, 0.5% DOC, 50 mM TRIS, pH 7.5, as described by Wilson, et al. cited above. Proteins are precipitated from the cell lysate and from the culture media using an HA-specific monoclonal antibody. The precipitated polypeptides then are analyzed by SDS-PAGE and

autoradiography. An expression product of the expected size is seen in the cell lysate, which is not seen in negative controls.

Example 3 (b): Cloning and Expression in CHO Cells The vector pC4 is used for the expression of hSPARC-hl polypeptide. Plasmid pC4 is a derivative of the plasmid pSV2-dhfr (ATCC Accession No. 37146). The plasmid contains the mouse DHFR gene under control of the SV40 early promoter. Chinese hamster ovary-or other cells lacking dihydrofolate activity that are transfected with these plasmids can be selected by growing the cells in a selective medium (alpha minus MEM, Life Technologies) supplemented with the chemotherapeutic agent methotrexate. The amplification of the DHFR genes in cells resistant to methotrexate (MTX) has been well documented (see, e. g., F.

W. Alt, et al., J. Biol. Chem., 253: 1357-1370 (1978); J. L.

Hamlin and C. Ma, Biochem. et Biophys. Acta, 1087: 107-125 (1990) ; and M. J. Page and M. A. Sydenham, Bio/Technology, 9: 64-68 (1991)). Cells grown in increasing concentrations of MTX develop resistance to the drug by overproducing the target enzyme, DHFR, as a result of amplification of the DHFR gene. If a second gene is linked to the DHFR gene, it is usually co-amplified and over-expressed. It is known in the art that this approach can be used to develop cell lines carrying more than 1,000 copies of the amplified gene (s).

Subsequently, when the methotrexate is withdrawn, cell lines are obtained which contain the amplified gene integrated into one or more chromosome (s) of the host cell.

Plasmid pC4 contains for expressing the gene of interest the strong promoter of the long terminal repeat (LTR) of the Rous Sarcoma Virus (Cullen, et al., Mol. Cell.

Biol., 5 : 438-447 (1985)) plus a fragment isolated from the enhancer of the immediate early gene of human

cytomegalovirus (CMV) (Boshart, et al., Cell, 41: 521-530 (1985)). Downstream of the promoter are BamHI, XbaI, and Asp718 restriction enzyme cleavage sites that allow integration of the genes. Behind these cloning sites the plasmid contains the 3'intron and polyadenylation site of the rat preproinsulin gene. Other high efficiency promoters can also be used for the expression, e. g., the human b-actin promoter, the SV40 early or late promoters or the long terminal repeats from other retroviruses., e. g., HIV and HTLVI. Clontech's Tet-Off and Tet-On gene expression systems and similar systems can be used to express the hSPARC-hl in a regulated way in mammalian cells (M. Gossen, and H. Bujard, Proc. Natl. Acad. Sci. USA, 89: 5547-5551 (1992)). For the polyadenylation of the mRNA other signals, e. g., from the human growth hormone or globin genes can be used as well. Stable cell lines carrying a gene of interest integrated into the chromosomes can also be selected upon co-transfection with a selectable marker such as gpt, G418 or hygromycin. It is advantageous to use more than one selectable marker in the beginning, e. g., G418 plus methotrexate.

The plasmid pC4 is digested with restriction enzymes and then dephosphorylated using calf intestinal phosphatase by procedures known in the art. The vector is then isolated from a 1% agarose gel.

The DNA sequence encoding the complete hSPARC-hl polypeptide is amplified using PCR oligonucleotide primers corresponding to the 5'and 3'sequences of the gene. Non- limiting examples include 5'and 3'primers having nucleotides corresponding or complementary to a portion of the coding sequence of an hSPARC-hl, e. g., as presented in at least one of SEQ ID NOS: 1, 2,3, according to known method steps.

The amplified fragment is digested with suitable endonucleases and then purified again on a 1% agarose gel.

The isolated fragment and the dephosphorylated vector are then ligated with T4 DNA ligase. E. coli HB101 or XL-1 Blue cells are then transformed and bacteria are identified that contain the fragment inserted into plasmid pC4 using, for instance, restriction enzyme analysis.

Chinese hamster ovary (CHO) cells lacking an active DHFR gene are used for transfection. 5 g of the expression plasmid pC4 is cotransfected with 0.5 g of the plasmid pSV2-neo using lipofectin. The plasmid pSV2neo contains a dominant selectable marker, the neo gene from Tn5 encoding an enzyme that confers resistance to a group of antibiotics including G418. The cells are seeded in alpha minus MEM supplemented with 1 pg/ml G418. After 2 days, the cells are trypsinized and seeded in hybridoma cloning plates (Greiner, Germany) in alpha minus MEM supplemented with 10,25, or 50 ng/ml of methotrexate plus 1 ug/ml G418. After about 10-14 days single clones are trypsinized and then seeded in 6-well petri dishes or 10 ml flasks using different concentrations of methotrexate (50 nM, 100 nM, 200 nM, 400 nM, 800 nM).

Clones growing at the highest concentrations of methotrexate are then transferred to new 6-well plates containing even higher concentrations of methotrexate (1 mM, 2 mM, 5 mM, 10 mM, 20 mM). The same procedure is repeated until clones are obtained which grow at a concentration of 100-200 mM.

Expression of the desired gene product is analyzed, for instance, by SDS-PAGE and Western blot or by reverse phase HPLC analysis.

Example 4: Tissue Distribution of hSPARC-hl mRNA Expression Northern blot analysis is carried out to examine hSPARC-hl gene expression in human tissues, using

essentially methods described by, among others, Sambrook, et al., cited above. Briefly, a cDNA probe containing the entire nucleotide sequence of an hSPARC-hl polypeptide (SEQ ID NO : 1) is labeled with 32p using the Rediprime DNA labeling system (Amersham Life Science), according to the manufacturer's instructions. After labeling, the probe is purified using a CHROMA SPIN-100 column (Clontech Laboratories, Inc.), according to the manufacturer's protocol number PT1200-1. The purified and labeled probe is used to examine various human tissues for hSPARC-hl mRNA.

Multiple Tissue Northern (MTN) blots containing various human tissues (H) or human immune system tissues (IM) are obtained from Clontech and are examined with the labeled probe using ExpressHyb hybridization solution (Clontech) according to manufacturer's protocol number PT1190-1.

Following hybridization and washing, the blots are mounted and exposed to film at-70°C overnight, and films developed according to standard procedures.

Northern analysis of hSPARC-hl expression indicated hSPARC-hl sequences were present in the molecules comprising two bands at 3.5 Kb and 2.0 Kb. The 3.5 Kb band was present in liver with strong intensity, the spleen with moderate intensity, brain with moderate intensity, skeletal muscle with moderate intensity, and placenta with moderate intensity. The same band appears upon longer exposure in the heart, thymus, kidney, small intestine and lung. The smaller 2.0 Kb band is present in the same tissues excluding lung.

Example 5: Determination of hSPARC-hl Protein Binding in Human Tissue Binding of hSPARC-hl proteins to human tissues was determined by protein staining with fluorescent dye. All

tissues were fixed with 3% paraformaldehyde and embedded in paraffin.

Tissues were prepared for analysis by removing the paraffin with xylene then gradually rehydrating the tissue with graded solutions of ethanol and water. Antigen retrieval was performed to unmask antigenic sites so that antibodies can recognize the antigen. This was accomplished by soaking the tissue in citrate buffer (Dako, Carpinteria, CA) for twenty minutes at 80 to 90°C followed 10 minutes at ambient temperature. The tissue was then washed in tris- buffered saline (TBS) containing 0.05% TWEEN 20 and 0.01% thimerosol. To minimize non-specific background staining, the tissue was soaked in non-serum protein block (Dako) for 45 minutes, after which the protein block was removed by blowing air over the tissue.

The tissue was exposed for 2 hours to the FLAG-HIS tagged hSPARC-hl protein at 10 g/mL. Following exposure, the tissue was washed twice with tris-buffered saline (TBS) containing 0.05% TWEEN 20 and 0.01% thimerosol. The tissue sample was then incubated for one hour with mouse anti-FLAG antibody at 10 Mg/mL. Subsequently, the tissue was washed twice with tris-buffered saline (TBS) containing 0.05% TWEEN 20 and 0.01% thimerosol. Next, the tissue was exposed to rabbit anti-mouse Ig with Alexa 568, a fluorescent dye, at 10 ug/mL for one hour, followed again by two washes with tris-buffered saline (TBS) containing 0.05% TWEEN 20 and 0.01% thimerosol. Finally, the tissue was coverslipped with fluorescence mounting media, and the fluorescence was measured. A positive fluorescence reading indicates that the protein binds with antigens on the tissue, suggesting that the protein was expressed in that tissue.

Human SPARC-hl polypeptides bound to the following tissues: the pancreas in the islet and acinar cells with

moderate intensity; the gut in villous epithelial cells, Brunner's gland, and muscularis with moderate intensity ; the adrenal cortex with moderate intensity; the cerebellum and cerebrum with moderate intensity; the ductal epithelial cells of the breast with moderate intensity; the red pulp of the spleen with moderate intensity; and the tubular epithelial cells of the kidney with weak/moderate intensity. hSPARC-hl polypeptide binding was also observed in breast, colon, ovarian, and prostate cancer tissue. Binding of hSPARC-hl was localized to both cytoplasmic and nuclear regions within the tissues.

EXAMPLE 6: Directed Mutagenesis of hSPARC-hl polypeptides to provide DNA encoding specified substitutions, insertions or deletions of SEQ ID NO : 1 Using the Polymerase Chain Reaction The polymerase chain reaction (PCR) can be used for the enzymatic amplification and direct sequencing of small quantities of nucleic acids (see, e. g., Ausubel, supra, section 15) to provide specified substitutions, insertions or deletions in DNA encoding an hSPARC-hl polypeptide of the present inventions, e. g., SEQ ID NO : 1, 2,3, or any sequence described herein, as presented herein, to provide an hSPARC- hl polypeptide sequence of interest including at least one substitution, insertion or deletion selected from the group consisting of D6,7V, 25G, 42P, 45P, 48P, 78A, 80A, 161P, 218V, 225H, 239R, 300S, 301V, 302E, 364A, 377D, 432G, 433S, and 434L of SEQ ID NO : 4, or the corresponding amino acid of SEQ ID NO : 5. This technology can be used as a quick and efficient method for introducing any desired sequence change into the DNA of interest.

Unit 8.5 of Ausubel, supra, contains two basic protocols for introducing base changes into specific DNA sequences. Basic Protocol 1, as presented in the first section 8.5 of Ausubel, supra (entirely incorporated. herein

by reference), describes the incorporation of a restriction site and Basic Protocol 2, as presented below and in the second section of Unit 8.5 of Ausubel, supra, details the generation of specific point mutations (all of the following references in this example are to sections of Ausubel et al., eds., Current Protocols in Molecular Biology, Wiley Interscience, New York (1987-1999)). An alternate protocol describes generating point mutations by sequential PCR steps. Although the general procedure is the same in all three protocols, there are differences in the design of the synthetic oligonucleotide primers and in the subsequent cloning and analyses of the amplified fragments.

The PCR procedure described here can rapidly, efficiently, and/or reproducibly introduce any desired change into a DNA fragment. It is similar to the oligonucleotide-directed mutagenesis method described in UNIT 8. 1, but does not require the preparation of a uracil- substituted DNA template.

The main disadvantage of PCR-generated mutagenesis is related to the fidelity of the Taq DNA polymerase. The mutation frequency for Taq DNA polymerase was initially estimated to be as high as 1/5000 per cycle (Saiki et al., 1988). This means that the entire amplified fragment must be sequenced to be sure that there are no Taq-derived mutations. To reduce the amount of sequencing required, it is best to introduce the mutation by amplifying as small a fragment as possible. With rapid and reproducible methods of double-stranded DNA sequencing (UNIT 7.4), the entire amplified fragment can usually be sequenced from a single primer. If the fragment is somewhat longer, it is best to subclone the fragment into an M13-derived vector, so that both forward and reverse primers can be used to sequence the amplified fragment.

If there are no convenient restriction sites flanking the fragment of interest, the utility of this method is somewhat reduced. Many researchers prefer the mutagenesis procedure in UNIT 8.1 to avoid excessive sequencing.

A full discussion of critical parameters for PCR amplification can be found in UNIT 15.1.

Anticipated Results Each of the procedures presented here has a 100% efficiency rate. All or substantially all of the cloned, amplified fragments will contain the mutation corresponding to the synthesized oligonucleotide.

Literature Cited Saiki, R. K., Gelfand, D. H., Stoffel, S., Scharf, S. J., Higuchi, R., Horn, G. T., Mullis, K. B., and Erlich, H. A.

1988. Primer-directed enzymatic amplification of DNA with a thermostable DNA polymerase. Science 239: 487-491.

BASIC PROTOCOL (2): INTRODUCTION OF POINT MUTATIONS BY PCR In this protocol, synthetic oligonucleotides are designed to incorporate a point mutation at one end of an amplified fragment. Following PCR, the amplified fragments are made blunt-ended by treatment with Klenow fragment.

These fragments are then ligated and subcloned into a vector to facilitate sequence analysis. This procedure is summarized in Figure 8.5.2 of Ausubel, supra.

Materials DNA sample to be mutagenized Klenow fragment of E. coli DNA polymerase I (UNIT 3.5 of Ausubel, supra).

Appropriate restriction endonuclease (Table 8.5.1)

Additional reagents and equipment for synthesis and purification of oligonucleotides (UNITS 2.11 & 2.12), phosphorylation of oligonucleotides (UNIT 3.10), electrophoresis of DNA on nondenaturing agarose and low gelling/melting agarose gels (UNITS 2.5A & 2.6), restriction endonuclease digestion (UNIT 3.1), ligation of DNA fragments (UNIT 3.16), transformation of E. coli (UNIT 1.8), plasmid DNA miniprep (UNIT 1.6), and DNA sequence analysis (UNIT 7.4) Prepare the template DNA and oligonucleotide primers Prepare template DNA (see Basic Protocol 1, steps 1 and 2).

Synthesize (UNIT 2.11) and purify (UNIT 2.12) the oligonucleotide primers (primers 3 and 4 in Fig. 8.5.2B).

The oligonucleotide primers must be homologous to the template DNA for more than 15 bases. No four-base"clamp" sequence is added to these primers. The primer sequences are based on a DNA encoding the hSPARC-hl polypeptide sequence of interest including at least one substitution, insertion or deletion selected from the group consisting of D6,7V, 25G, 42P, 45P, 48P, 78A, 80A, 161P, 218V, 225H, 239R, 300S, 301V, 302E, 364A, 377D, 432G, 433S, and 434L of SEQ ID NO: 4, or the corresponding amino acid of SEQ ID NO : 5. Phosphorylate the 5'end of the oligonucleotides (UNIT 3.10). This step is necessary because the 5'end of the oligonucleotide will be used directly in cloning.

Amplify DNA and prepare blunt-end fragments Amplify the template DNA (see Basic Protocol 1, steps 5 and 6). After the final extension step, add 5 U Klenow fragment to the reaction mix and incubate 15 min at 30°C.

During PCR, the Taq polymerase adds an extra nontemplated nucleotide to the 3'end of the fragment. The 3'-5' exonuclease activity of the Klenow fragment is required to

make the ends flush and suitable for blunt-end cloning (UNIT 3.5). Analyze and process the reaction mix (see Basic Protocol 1, steps 7 and 8). Digest half the amplified fragments with the restriction endonucleases for the flanking sequences (UNIT 3.1). Purify digested fragments on a low gelling/melting agarose gel (UNIT 2.6).

Subclone the two amplified fragments into an appropriately digested vector by blunt-end ligation (UNIT 3.16). Transform recombinant plasmid into E. coli (UNIT 1.8). Prepare DNA by plasmid miniprep (UNIT 1.6). Analyze the amplified fragment portion of the plasmid DNA by DNA sequencing to confirm the point mutation (UNIT 7.4). This is critical because the Taq DNA polymerase can introduce additional mutations into the fragment (see Critical Parameters).

ALTERNATE PROTOCOL: INTRODUCTION OF A POINT MUTATION BY SEQUENTIAL PCR STEPS In this procedure, the two fragments encompassing the mutation are annealed with each other and extended by mutually primed synthesis; this fragment is then amplified by a second PCR step, thereby avoiding the blunt-end ligation required in Basic Protocol 2. This strategy is outlined in Figure 8.5.3. For materials, see Basic Protocols 1 and 2 of Ausubel, supra.

Prepare template DNA (see Basic Protocol 1, steps 1 and 2). Synthesize (UNIT 2.11) and purify (UNIT 2.12) the oligonucleotide primers (primers 5 and 6 in Fig. 8.5.3B) to generate an hSPARC-hl polypeptide sequence of interest including at least one substitution, insertion or deletion selected from the group consisting of D6,7V, 25G, 42P, 45P, 48P, 78A, 80A, 161P, 218V, 225H, 239R, 300S, 301V, 302E, 364A, 377D, 432G, 433S, and 434L of SEQ ID NO : 4, or the corresponding amino acid of SEQ ID NO : 5. The oligonucleotides must be homologous to the template for 15

to 20 bases and must overlap with one another by at least 10 bases. The 5'end does not have a"clamp"sequence.

Amplify the template DNA and generate blunt-end fragments (see Basic Protocol 2, steps 4 and 5). Purify the fragments by nondenaturing agarose gel electrophoresis (UNIT 2.5A). Resuspend in TE buffer at 1 ng/pL.

Carry out second PCR amplification. Combine the following in a 500-lull microcentrifuge tube: 10 J. L (10 ng) each amplified fragment 1 p. L (500 ng) each flanking sequence primer (each 1 g final) 10 tL lOx amplification buffer 10 uL 2 mM 4dNTP mix H20 to 99.5 J. L 0. 5 uL Taq DNA polymerase (5 U/L).

Overlay with 100 L mineral oil. Carry out PCR for 20 to 25 cycles, using the conditions for introduction of restriction endonuclease sites by PCR (see Basic Protocol 1, step 6). Analyze and process the reaction mix (see Basic Protocol 1, Ausubel, supra, steps 7 and 8).

Digest the DNA fragment with the appropriate restriction endonuclease for the flanking sites (UNIT 3.1).

Purify the digested fragment on a low gelling/melting agarose gel (UNIT 2.6). Subclone into an appropriately digested vector. Transform recombinant plasmid into E. coli (UNIT 1.8). Prepare DNA by plasmid miniprep (UNIT 1.6).

Analyze the amplified fragment portion of the plasmid DNA by DNA sequencing (UNIT 7.4) to confirm the point mutation.

This is critical because the Taq DNA polymerase can introduce additional mutations into the fragment (see Critical Parameters).

Example 7: Serine Proteinase Inhibitor Function of hSPARC-hl The ability of hSPARC-hl polypeptides to inhibit proteinase activity of trypsin and chymotrypsin in a dose response screen can be determined using an assay essentially as described by Araujo M. S., et al (Araujo M. S., et al, Immunopharmacology, Preliminary characterization of a Kazal- type serine protease inhibitor from Caiman Crocodilus yacare plasma, Vol. 45, pg 179-183 (1999)). The residual activity of the proteinases may be measured as a function of the hydrolysis of Bz-Arg-pNan or Suc-Phe-pNan. Activity on other serine proteases and peptides can be tested similarly as well. Results can be measured by various methods known to those skilled in the art, including affinity chromatography, SDS-PAGE, and reverse substrate zymography.

Example 8: Growth Factor Binding Assay The extracellular glycoprotein SPARC interacts with platelet-derived growth factor (PDGF)-AB and-BB and inhibits the binding of PDGF to its receptors (Raines, E. W. et al, PNAS, Vol. 89: 1281-1285 (1992)). Using essentially the method of Raines, et al. (1992), one skilled in the art can measure the ability of hSPARC-hl to interact with various growth factors. Binding can be compared to known positive and negative controls for growth factor binding.

Example 9: Cell Migration Assays Glioma cells are cultured in a monolayer and transfered to tissue culture plates coated with vitronectin. The cells are seeded into the middle of each well using sedimentation manifolds and then incubated overnight. The manifold is

removed and the medium is changed to include hSPARC-hl at increasing doses. Migration is allowed to proceed and measured essentially as described by Menon, et al. (Menon P. M. et al, Int. J. of Onc., A study of SPARC and vitronectin localization and expression in pediatric and adult gliomas: high SPARC secretion correlates with decreased migration on vitronectin, Vol. 17: 683-693 (2000)).