Language selection

Search

Patent 2384672 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2384672
(54) English Title: STEROID HORMONE RECEPTOR POLYNUCLEOTIDES, POLYPEPTIDES, AND ANTIBODIES
(54) French Title: POLYNECLUOTIDES, POLYPEPTIDES ET ANTICORPS DE RECEPTEURS A HORMONES STEROIDES
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12N 15/12 (2006.01)
  • C07K 14/47 (2006.01)
  • C07K 14/705 (2006.01)
  • C07K 14/72 (2006.01)
  • C12N 5/10 (2006.01)
  • C12N 15/63 (2006.01)
  • C12N 15/64 (2006.01)
(72) Inventors :
  • NI, JIAN (United States of America)
  • SHI, YANGGU (United States of America)
  • RUBEN, STEVEN M. (United States of America)
(73) Owners :
  • HUMAN GENOME SCIENCES, INC. (United States of America)
(71) Applicants :
  • HUMAN GENOME SCIENCES, INC. (United States of America)
(74) Agent: MBM INTELLECTUAL PROPERTY LAW LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2000-09-07
(87) Open to Public Inspection: 2001-03-15
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2000/024517
(87) International Publication Number: WO2001/018049
(85) National Entry: 2002-03-07

(30) Application Priority Data:
Application No. Country/Territory Date
60/152,932 United States of America 1999-09-09
60/189,032 United States of America 2000-03-14

Abstracts

English Abstract




The present invention relates to novel human steroid hormone receptor
polypeptides and isolated nucleic acids containing the coding regions of the
genes encoding such polypeptides. Also provided are vectors, host cells,
antibodies, and recombinant methods for producing human steroid hormone
receptor polypeptides. The invention further relates to diagnostic and
therapeutic methods useful for diagnosing and treating disorders related to
these novel human steroid hormone receptor polypeptides.


French Abstract

La présente invention concerne des polypeptides de récepteurs à hormones stéroïdes humains et des acides nucléiques isolés contenant les zones de codage des gènes codant pour ces polypeptides. Cette invention concerne également des vecteurs, des cellules hôtes, des anticorps, et des procédés de recombinaison permettant de produire des polypeptides de récepteurs à hormones stéroïdes humains. L'invention concerne également des procédés diagnostiques et thérapeutiques permettant le diagnostique et le traitement des troubles liés à ces polypeptides de récepteurs à hormones stéroïdes humains.

Claims

Note: Claims are shown in the official language in which they were submitted.





227

What Is Claimed Is:

1. An isolated nucleic acid molecule comprising a polynucleotide
selected from the group consisting of:
(a) the polynucleotide shown as SEQ ID NO:X or the polynucleotide encoded
by a cDNA included in ATCC Deposit No:Z;
(b) a polynucleotide encoding a biologically active polypeptide fragment of
SEQ ID NO:Y or a biologically active polypeptide fragment encoded by the cDNA
sequence included in ATCC Deposit No:Z;
(c) a polynucleotide encoding a polypeptide epitope of SEQ ID NO:Y or a
polypeptide epitope encoded by the cDNA sequence included in ATCC Deposit
No:Z;
(d) a polynucleotide capable of hybridizing under stringent conditions to any
one of the polynucleotides specified in (a)-(c), wherein said polynucleotide
does not
hybridize under stringent conditions to a nucleic acid molecule having a
nucleotide
sequence of only A residues or of only T residues.

2. The isolated nucleic acid molecule of claim 1, wherein the
polynucleotide comprises a nucleotide sequence encoding a soluble polypeptide.

3. The isolated nucleic acid molecule of claim 1, wherein the
polynucleotide comprises a nucleotide sequence encoding the sequence
identified as
SEQ ID NO:Y or the polypeptide encoded by the cDNA sequence included in ATCC
Deposit No:Z.

4. The isolated nucleic acid molecule of claim 1, wherein the
polynucleotide comprises the entire nucleotide sequence of SEQ ID NO:X or a
cDNA
included in ATCC Deposit No:Z.

5. The isolated nucleic acid molecule of claim 2, wherein the
polynucleotide is DNA.




228

6. The isolated nucleic acid molecule of claim 3, wherein the
polynucleotide is RNA.

7. A vector comprising the isolated nucleic acid molecule of claim 1.

8. A host cell comprising the vector of claim 7.

9. A recombinant host cell comprising the nucleic acid molecule of claim
1 operably limited to a heterologous regulating element which controls gene
expression.

10. A method of producing a polypeptide comprising expressing the
encoded polypeptide from the host cell of claim 9 and recovering said
polypeptide.

11. An isolated polypeptide comprising an amino acid sequence at least
95% identical to a sequence selected from the group consisting of
(a) the polypeptide shown as SEQ ID NO:Y or the polypeptide encoded by
the cDNA;
(b) a polypeptide fragment of SEQ ID NO:Y or the polypeptide encoded by
the cDNA;
(c) a polypeptide epitope of SEQ ID NO:Y or the polypeptide encoded by the
cDNA; and
(d) a variant of SEQ ID NO:Y.

12. The isolated polypeptide of claim 11, comprising a polypeptide having
SEQ ID NO:Y.

13. An isolated antibody that binds specifically to the isolated polypeptide
of claim 11.

14. A recombinant host cell that expresses the isolated polypeptide of
claim 11.





229

15. A method of making an isolated polypeptide comprising:
(a) culturing the recombinant host cell of claim 14 under conditions such that
said polypeptide is expressed; and
(b) recovering said polypeptide.

16. The polypeptide produced by claim 15.

17. A method for preventing, treating, or ameliorating a medical condition,
comprising administering to a mammalian subject a therapeutically effective
amount
of the polypeptide of claim 11 or the polynucleotide of claim 1.

18. A method of diagnosing a pathological condition or a susceptibility to
a pathological condition in a subject comprising:
(a) determining the presence or absence of a mutation in the polynucleotide of
claim 1; and
(b) diagnosing a pathological condition or a susceptibility to a pathological
condition based on the presence or absence of said mutation.

19. A method of diagnosing a pathological condition or a susceptibility to
a pathological condition in a subject comprising:
(a) determining the presence or amount of expression of the polypeptide of
claim 11 in a biological sample; and
(b) diagnosing a pathological condition or a susceptibility to a pathological
condition based on the presence or amount of expression of the polypeptide.

20. A method for identifying a binding partner to the polypeptide of claim
11 comprising:
(a) contacting the polypeptide of claim 11 with a binding partner; and
(b) determining whether the binding partner effects an activity of the
polypeptide.




230

21. A method of screening for molecules which modify activities of the
polypeptide of claim 11 comprising:
(a) contacting said polypeptide with a compound suspected of having agonist or
antagonist activity; and
(a) assaying for activity of said polypeptide.

Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
Steroid Hormone Receptor Polynucleotides, Polypeptides, and
Antibodies
Field of the Invention
The present invention relates to novel steroid hormone receptor proteins. More
specifically, isolated nucleic acid molecules are provided encoding novel
steroid hormone
receptor polypeptides. Novel steroid hormone receptor polypeptides and
antibodies that bind
to these polypeptides are provided. Also provided are vectors, host cells, and
recombinant
and synthetic methods for producing human steroid hormone receptor
polynucleotides and/or
polypeptides. The invention further relates to diagnostic and therapeutic
methods useful for
diagnosing, treating, preventing and/or prognosing disorders related to these
novel steroid
hormone receptor polypeptides. The invention further relates to screening
methods for
identifying agonists and antagonists of polynucleotides and polypeptides of
the invention.
The present invention further relates to methods and/or compositions for
inhibiting the
production and fiinction of the polypeptides of the present invention.
Background of the Invention
Physiological and therapeutic activities of steroid hormones are mediated by
the
family of Steroid Hormone Receptor (SHR) proteins, which are members of the
nuclear
receptor family of proteins. These nuclear receptors include the receptors for
steroids, thyroid
hormone, vitamins A and D-derived hormones, and certain fatty acids (Kumar,
R., and E.B.
Thompson, Steroids, 64:310-19 (1999)).
Several common structural features are shared by members of the nuclear
receptor
family of proteins. All members of this family of proteins contain a variable
N-terminal
domain that is thought to be involved in the transactivation of transcription.
Further, all
members of this family of proteins contain a well-conserved DNA-binding
domain, which is
thought to be important in DNA binding and protein-protein interactions, as
well as a ligand-
binding domain at the C-terminal end of the protein which is thought to be
involved in


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
2
hormone binding, protein-protein interactions, and transactivation activity
(Kumar, R.,
( 1999)).
Steroid hormone receptors constitute a family of inducible transcription
factors that
transduce the effects of small, lipophilic hormones into transcriptional
responses. Activation
of these receptors by binding of the cognate ligand induces the receptors to
bind with high
affinity to specific response elements in the promoter regions of target
genes, known as
hormone responsive elements (HREs), inducing transcription of certain genes
(Klein-Hitpass,
L., et al., J. Mol Med, 76:490-6 (1998)).
In addition to promoting transcription of particular genes, Steroid Hormone
Receptor
Proteins also function as transcriptional repressors. For example, elevated
expression of AP-1
subunits, or their activation by growth factors or phorbol esters, results in
inhibition of
hormone responsive element (HRE) promoters and interference with DNA binding
(Beato,
M., et al., Cell, 83:851-7 (1995)).
Thus there exists a clear need for identifying and exploiting novel members of
the
nuclear hormone receptor family of proteins. Although structurally related,
such proteins may
possess diverse and multifaceted functions in a variety of cell and tissue
types. Receptor type
molecules should prove useful in target based screens for small molecules and
other such
pharmacologically valuable factors. The purified steroid hormone receptor
proteins of the
invention are research tools useful for the identification, characterization
and purification of
additional interacting proteins or receptor proteins, or other signal
transduction pathway
proteins that interact with steroid hormone receptor proteins. Additionally,
assays designed to
monitor the expression of steroid hormone receptor proteins may be useful as
diagnostic tools
to monitor the presence or progression of certain types of cancers, such as
breast cancer.
Summary of the Invention
The present invention includes isolated nucleic acid molecules comprising, or
alternatively, consisting of a polynucleotide sequence disclosed in the
sequence listing and/or
contained in a human cDNA plasmid described in Table 1 and deposited with the
American
Type Culture Collection (ATCC). Fragments, variants, and derivatives of these
nucleic acid
molecules are also encompassed by the invention. The present invention also
includes
isolated nucleic acid molecules comprising, or alternatively, consisting of, a
polynucleotide


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
3
encoding steroid hormone receptor polypeptides. The present invention further
includes
steroid hormone receptor polypeptides encoded by these polynucleotides.
Further provided
for are amino acid sequences comprising, or alternatively, consisting of,
steroid hormone
receptor polypeptides as disclosed in the sequence listing and/or encoded by
the human
cDNA plasmids described in Table 1 and deposited with the ATCC. Antibodies
that bind
these polypeptides are also encompassed by the invention. Polypeptide
fragments, variants,
and derivatives of these amino acid sequences are also encompassed by the
invention, as are
polynucleotides encoding these polypeptides and antibodies that bind these
polypeptides.
l0 Detailed Description
Tables
Table 1 summarizes ATCC Deposits, Deposit dates, and ATCC designation numbers
of deposits made with the ATCC in connection with the present application.
Table 1 further
summarizes the information pertaining to each "Gene No." described below,
including cDNA
clone identifier, the type of vector contained in the cDNA clone identifier,
the nucleotide
sequence identifier number, nucleotides contained in the disclosed sequence,
the location of
the 5' nucleotide of the start codon of the disclosed sequence, the amino acid
sequence
identifier number, and the last amino acid of the ORF encoded by the disclosed
sequence.
Table 2 indicates public ESTs, of which at least one, two, three, four, five,
ten, or
more of any one or more of these public EST sequences are optionally excluded
from certain
embodiments of the invention.
Table 3 summarizes the expression profile of polynucleotides corresponding to
the
clones disclosed in Table 1. The first column provides a unique clone
identifier, "Clone ID
NO:Z", for a cDNA clone related to each contig sequence disclosed in Table 1.
Column 2,
"Library Code" shows the expression profile of tissue and/or cell line
libraries which express
the polynucleotides of the invention. Each Library Code in column 2 represents
a tissue/cell
source identifier code corresponding to the Library Code and Library
description provided in
Table 4. Expression of these polynucleotides was not observed in the other
tissues and/or cell
libraries tested. One of skill in the art could routinely use this information
to identify tissues
which show a predominant expression pattern of the corresponding
polynucleotide of the


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
4
invention or to identify polynucleotides which show predominant and/or
specific tissue
expression.
Table 4, column 1, provides the Library Code disclosed in Table 3, column 2.
Column
2 provides a description of the tissue or cell source from which the
corresponding library was
derived.
Definitions
The following definitions are provided to facilitate understanding of certain
terms
used throughout this specification.
In the present invention, "isolated" refers to material removed from its
original
environment (e.g., the natural environment if it is naturally occurnng), and
thus is altered "by
the hand of man" from its natural state. 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. The term "isolated" does not refer to
genomic or cDNA
libraries, whole cell total or mRNA preparations, genomic DNA preparations
(including
those separated by electrophoresis and transferred onto blots), sheared whole
cell genomic
DNA preparations or other compositions where the art demonstrates no
distinguishing
features of the polynucleotide/sequences of the present invention.
As used herein, a "polynucleotide" refers to a molecule having a nucleic acid
sequence contained in SEQ ID NO:X (as described in column 5 of Table 1 ), or
cDNA
plasmid:Z (as described in column 3 of Table 1 and contained within a pool of
plasmids
deposited with the ATCC). For example, the polynucleotide can contain the
nucleotide
sequence of the full length cDNA sequence, including the 5' and 3'
untranslated sequences,
the coding region, with or without a natural or artificial signal sequence,
the protein coding
region, as well as fragments, epitopes, domains, and variants of the nucleic
acid sequence.
Moreover, as used herein, a "polypeptide" refers to a molecule having an amino
acid
sequence encoded by a polynucleotide of the invention as broadly defined
(obviously
excluding poly-Phenylalanine or poly-Lysine peptide sequences which result
from translation
of a polyA tail of a sequence corresponding to a cDNA).
In the present invention, a representative plasmid containing the sequence of
SEQ )D
NO:X was deposited with the American Type Culture Collection ("ATCC") and/or
described


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
in Table 1. As shown in Table 1, each plasmid is identified by a cDNA Clone ID
(Identifier)
and the ATCC Deposit Number (ATCC Deposit No:Z). Plasmids that were pooled and
deposited as a single deposit have the same ATCC Deposit Number. The ATCC is
located at
10801 University Boulevard, Manassas, Virginia 20110-2209, USA. The ATCC
deposit was
5 made pursuant to the terms of the Budapest Treaty on the international
recognition of the
deposit of microorganisms for purposes of patent procedure.
A "polynucleotide" of the present invention also includes those
polynucleotides
capable of hybridizing, under stringent hybridization conditions, to sequences
contained in
SEQ ID NO:X, or the complement thereof (e.g., the complement of any one, two,
three, four,
or more of the polynucleotide fragments described herein) and/or sequences
contained in
cDNA plasmid:Z (e.g., the complement of any one, two, three, four, or more of
the
polynucleotide fragments described herein). "Stringent hybridization
conditions" refers to an
overnight incubation at 42 degree C in a solution comprising 50% formamide, 5x
SSC (750
mM NaCI, 75 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5x
Denhardt's
solution, 10% dextran sulfate, and 20 pg/ml denatured, sheared salmon sperm
DNA,
followed by washing the filters in O.lx SSC at about 65 degree C.
Also included within "polynucleotides" of the present invention are nucleic
acid
molecules that hybridize to the polynucleotides of the present invention at
lower stringency
hybridization conditions. Changes in the stringency of hybridization and
signal detection are
primarily accomplished through the manipulation of formamide concentration
(lower
percentages of formamide result in lowered stringency); salt conditions, or
temperature. For
example, lower stringency conditions include an overnight incubation at 37
degree C in a
solution comprising 6X SSPE (20X SSPE = 3M NaCI; 0.2M NaHZP04; 0.02M EDTA, pH
7.4), 0.5% SDS, 30% formamide, 100 ug/ml salmon sperm blocking DNA; followed
by
washes at 50 degree C with 1XSSPE, 0.1% SDS. In addition, to achieve even
lower
stringency, washes performed following stringent hybridization can be done at
higher salt
concentrations (e.g. 5X SSC).
Note that variations in the above conditions may be accomplished through the
inclusion and/or substitution of alternate blocking reagents used to suppress
background in
hybridization experiments. Typical blocking reagents include Denhardt's
reagent, BLOTTO,
heparin, denatured salmon sperm DNA, and commercially available proprietary
formulations.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
6
The inclusion of specific blocking reagents may require modification of the
hybridization
conditions described above, due to problems with compatibility.
Of course, a polynucleotide which hybridizes only to polyA+ sequences (such as
any
3' terminal polyA+ tract of a cDNA shown in the sequence listing), or to a
complementary
stretch of T (or U) residues, would not be included in the definition of
"polynucleotide," 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
generated using oligo dT as a primer).
The polynucleotides of the present invention can be composed of any
polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or
DNA or
modified RNA or DNA. For example, polynucleotides can be composed of single-
and
double-stranded DNA, DNA that is a mixture of single- and double-stranded
regions, single-
and double-stranded RNA, and RNA that is mixture of single- and double-
stranded regions,
hybrid molecules comprising DNA and RNA that may be single-stranded or, more
typically,
double-stranded or a mixture of single- and double-stranded regions. In
addition, the
polynucleotide can be composed of triple-stranded regions comprising RNA or
DNA or both
RNA and DNA. A polynucleotide may also contain one or more modified bases or
DNA or
RNA backbones modified for stability or for other reasons. "Modified" bases
include, for
example, tritylated bases and unusual bases such as inosine. A variety of
modifications can
be made to DNA and RNA; thus, "polynucleotide" embraces chemically,
enzymatically, or
metabolically modified forms.
In specific embodiments, the polynucleotides of the invention are at least 15,
at least
30, at least 50, at least 100, at least 125, at least 500, or at least 1000
continuous nucleotides
but are less than or equal to 300 kb, 200 kb, 100 kb, SO kb, 15 kb, 10 kb,
7.Skb, S kb, 2.5 kb,
2.0 kb, or 1 kb, in length. In a further embodiment, polynucleotides of the
invention
comprise a portion of the coding sequences, as disclosed herein, but do not
comprise all or a
portion of any intron. In another embodiment, the polynucleotides comprising
coding
sequences do not contain coding sequences of a genomic flanking gene (i.e., 5'
or 3' to the
gene of interest in the genome). In other embodiments, the polynucleotides of
the invention
do not contain the coding sequence of more than 1000, 500, 250, 100, 50, 25,
20, 15, 10, 5, 4,
3, 2, or 1 genomic flanking gene(s).


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
7
"SEQ ID NO:X" refers to a polynucleotide sequence described in column 5 of
Table
1, while "SEQ ID NO:Y" refers to a polypeptide sequence described in column 10
of Table 1.
SEQ 117 NO:X is identified by an integer specified in column 6 of Table 1. The
polypeptide
sequence SEQ ID NO:Y is a translated open reading frame (ORF) encoded by
polynucleotide
SEQ ID NO:X. The polynucleotide sequences are shown in the sequence listing
immediately
followed by all of the polypeptide sequences. Thus, a polypeptide sequence
corresponding to
polynucleotide sequence SEQ ID N0:2 is the first polypeptide sequence shown in
the
sequence listing. The second polypeptide sequence corresponds to the
polynucleotide
sequence shown as SEQ ID N0:3, and so on.
The polypeptides of the present invention can be composed of amino acids
joined to
each other by peptide bonds or modified peptide bonds, i.e., peptide
isosteres, and may
contain amino acids other than the 20 gene-encoded amino acids. The
polypeptides may be
modified by either natural processes, such as posttranslational processing, or
by chemical
modification techniques which are well known in the art. Such modifications
are well
described in basic texts and in more detailed monographs, as well as in a
voluminous research
literature. Modifications can occur anywhere in a polypeptide, including the
peptide
backbone, the amino acid side-chains and the amino or carboxyl termini. It
will be
appreciated that the same type of modification may be present in the same or
varying degrees
at several sites in a given polypeptide. Also, a given polypeptide may contain
many types of
modifications. Polypeptides may be branched, for example, as a result of
ubiquitination, and
' they may be cyclic, with or without branching. Cyclic, branched, and
branched cyclic
polypeptides may result from posttranslation natural processes or may be made
by synthetic
methods. Modifications include acetylation, acylation, ADP-ribosylation,
amidation,
covalent attachment of flavin, covalent attachment of a heme moiety, covalent
attachment of
a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid
derivative,
covalent attachment of phosphotidylinositol, cross-linking, cyclization,
disulfide bond
formation, demethylation, formation of covalent cross-links, formation of
cysteine, formation
of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor
formation,
hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation,
proteolytic
processing, phosphorylation, prenylation, racemization, selenoylation,
sulfation, transfer-
RNA mediated addition of amino acids to proteins such as arginylation, and
ubiquitination.
(See, for instance, PROTEINS - STRUCTURE AND MOLECULAR PROPERTIES, 2nd


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
8
Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993);
POSTTRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson,
Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et al., Meth Enzymol
182:626-646
(1990); Rattan et al., Ann NY Acad Sci 663:48-62 (1992)).
The polypeptides of the invention can be prepared in any suitable manner. Such
polypeptides include isolated naturally occurring polypeptides, recombinantly
produced
polypeptides, synthetically produced polypeptides, or polypeptides produced by
a
combination of these methods. Means for preparing such polypeptides are well
understood in
the art.
The polypeptides may be in the form of the secreted protein, including the
mature
form, or may be a part of a larger protein, such as a fusion protein (see
below). It is often
advantageous to include an additional amino acid sequence which contains
secretory or
leader sequences, pro-sequences, sequences which aid in purification, such as
multiple
histidine residues, or an additional sequence for stability during recombinant
production.
The polypeptides of the present invention are preferably provided in an
isolated form,
and preferably are substantially purified. A recombinantly produced version of
a
polypeptide, including the secreted polypeptide, can be substantially purified
using
techniques described herein or otherwise known in the art, such as, for
example, by the one-
step method described in Smith and Johnson, Gene 67:31-40 (1988). Polypeptides
of the
invention also can be purified from natural, synthetic or recombinant sources
using
techniques described herein or otherwise known in the art, such as, for
example, antibodies of
the invention raised against the polypeptides of the present invention in
methods which are
well known in the art.
By a polypeptide demonstrating a "functional activity" is meant, a polypeptide
capable of displaying one or more known functional activities associated with
a full-length
(complete) protein of the invention. Such functional activities include, but
are not limited to,
biological activity, antigenicity [ability to bind (or compete with a
polypeptide for binding)
to an anti-polypeptide antibody], immunogenicity (ability to generate antibody
which binds to
a specific polypeptide of the invention), ability to form multimers with
polypeptides of the
invention, and ability to bind to a receptor or ligand for a polypeptide.
"A polypeptide having functional activity" refers to polypeptides exhibiting
activity
similar, but not necessarily identical to, an activity of a polypeptide of the
present invention,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
9
including mature forms, as measured in a particular assay, such as, for
example, a biological
assay, with or without dose dependency. In the case where dose dependency does
exist, it
need not be identical to that of the polypeptide, but rather substantially
similar to the dose-
dependence in a given activity as compared to the polypeptide of the present
invention (i.e.,
the candidate polypeptide will exhibit greater activity or not more than about
25-fold less
and, preferably, not more than about tenfold less activity, and most
preferably, not more than
about three-fold less activity relative to the polypeptide of the present
invention).
The functional activity of the polypeptides, and fragments, variants
derivatives, and
analogs thereof, can be assayed by various methods.
For example, in one embodiment where one is assaying for the ability to bind
or
compete with full-length polypeptide of the present invention for binding to
an antibody to
the full length polypeptide, various immunoassays known in the art can be
used, including
but not limited to, competitive and non-competitive assay systems using
techniques such as
radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich"
immunoassays, immunoradiometric assays, gel diffusion precipitation reactions,
immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or
radioisotope
labels, for example), western blots, precipitation reactions, agglutination
assays (e.g., gel
agglutination assays, hemagglutination assays), complement fixation assays,
immunofluorescence assays, protein A assays, and immunoelectrophoresis assays,
etc. In one
embodiment, antibody binding is detected by detecting a label on the primary
antibody. In
another embodiment, the primary antibody is detected by detecting binding of a
secondary
antibody or reagent to the primary antibody. In a further embodiment, the
secondary
antibody is labeled. Many means are known in the art for detecting binding in
an
immunoassay and are within the scope of the present invention.
In another embodiment, where a ligand is identified, or the ability of a
polypeptide
fragment, variant or derivative of the invention to multimerize is being
evaluated, binding can
be assayed, e.g., by means well-known in the art, such as, for example,
reducing and non-
reducing gel chromatography, protein affinity chromatography, and affinity
blotting. See
generally, Phizicky, E., et al., Microbiol. Rev. 59:94-123 (1995). In another
embodiment,
physiological correlates polypeptide of the present invention binding to its
substrates (signal
transduction) can be assayed.
In addition, assays described herein (see Examples) and otherwise known in the
art


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
may routinely be applied to measure the ability of polypeptides of the present
invention and
fragments, variants derivatives and analogs thereof to elicit polypeptide
related biological
activity (either in vitro or in vivo). Other methods will be known to the
skilled artisan and
are within the scope of the invention.
Polynucleotides and Polypeptides of the Invention
FEATURES OF PROTEIN ENCODED BY GENE NO: 1
In specific embodiments, polypeptides of the invention comprise, or
alternatively
10 consist of, the following amino acid sequence: TSGEKHHQRLQTGGTMNLMFPA
PSLQMPGLWPVNEPRGAGTVGFSWNGHGVAGSLSPRGLGRSRPLCGVLPTGVRWW
LAGTPHRNYVSAVERSCPRARKAGSAFPHVPRVKENEKETHKCCYDLGCWLANTAL
IETFFHPRIPSTICFGQGAARGFKSNKKKTKQC (SEQ ID NO: 12). Fragments and/or
variants of these polypeptides, such as, for example, fragments and/or
variants as described
herein, are encompassed by the invention. Polynucleotides encoding these
polypeptides
(including fragments and/or variants) are also encompassed by the invention,
as are
antibodies that bind these polypeptides.
Preferred polypeptides of the present invention comprise, or alternatively
consist of,
the immunogenic epitope shown in SEQ ID NO: 7 as residues: Thr-1 to Ser-6.
Fragments
and/or variants of these polypeptides, such as, for example, fragments and/or
variants as
described herein, are encompassed by the invention. Polynucleotides encoding
these
polypeptides (including fragments and/or variants) are also encompassed by the
invention, as
are antibodies that bind these polypeptides.
This gene is expressed in a number of fetal tissues, such as fetal lung and
fetal heart
tissues, as well as in placental tissue.
Therefore, polynucleotides and polypeptides of the invention, including
antibodies,
are useful as reagents for differential identification of the tissues) or cell
types) present in a
biological sample and for diagnosis of diseases and conditions which include
but are not
limited to: diseases and/or disorders of the placenta and developing fetus.
Similarly,
polypeptides and antibodies directed to these polypeptides are useful in
providing
immunological probes for differential identification of the tissues) or cell
type(s). For a
number of disorders of the above tissues or cells, particularly of the fetus
or the placenta,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
11
expression of this gene at significantly higher or lower levels may be
routinely detected in
certain tissues or cell types (e.g., fetal, placental, cancerous and wounded
tissues) or bodily
fluids (e.g., lymph, serum, plasma, urine, synovial fluid and spinal fluid) or
another tissue or
sample taken from an individual having such a disorder, relative to the
standard gene
expression level, i.e., the expression level in healthy tissue or bodily fluid
from an individual
not having the disorder.
The tissue distribution in fetal and placental tissues indicates that
polynucleotides,
translation products and antibodies corresponding to this gene are useful for
the diagnosis,
detection and/or treatment of diseases and/or disorders of the developing
fetus and of the
placenta.
This gene shares sequence homology to other steroid hormone receptors, and
accordingly it is thought that polynucleotides and translation products
corresponding to this
gene may share some of the biological activities of other steroid hormone
receptors. Thus,
translation products corresponding to this gene may be involved in transducing
the signals of
hormones that specifically bind, resulting in transcriptional activation of
target genes.
Therefore, translation products corresponding to this gene may be expressed in
other specific
tissues or organs where it may play related functional roles in other
processes, such as
hematopoiesis, inflammation, bone formation, and kidney function, to name a
few possible
target indications. Given the homology, translation products corresponding to
this gene are
also useful in identifying modulators for control of reproduction andlor
social behavior, to
increase fertility, or control hormone secretion.
The tissue distribution suggests that polynucleotides, translation products
and
antibodies corresponding to this gene are useful for the diagnosis and/or
treatment of
disorders of the placenta. Specific expression within the placenta suggests
that this gene
product may play a role in the proper establishment and maintenance of
placental function.
Alternatively, this gene product may be produced by the placenta and then
transported to the
embryo, where it may play a crucial role in the development and/or survival of
the
developing embryo or fetus. Expression of this gene product in a vascular-rich
tissue such as
the placenta also suggests that this gene product may be produced more
generally in
endothelial cells or within the circulation. In such instances, it may play
more generalized
roles in vascular function, such as in angiogenesis. It may also be produced
in the vasculature
and have effects on other cells within the circulation, such as hematopoietic
cells. It may


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
12
serve to promote the proliferation, survival, activation, and/or
differentiation of
hematopoietic cells, as well as other cells throughout the body. Protein, as
well as, antibodies
directed against the protein may show utility as a tumor marker and/or
immunotherapy
targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 2
Translation products corresponding to this gene share sequence homology with a
pheromone receptor isolated from Rattus norwegicus (See International
Publication No.
W097/14790), which is thought to be involved in binding to pheromones. Based
upon the
homology, it is thought that these proteins will share at least some
biological activities.
In specific embodiments, polypeptides of the invention comprise, or
alternatively
consist of, an amino acid sequence selected from the group consisting of:
WVINSLIYIKLVPFVDTIKYGSVTKNLSIKMCLATPHMGNTIAVSHTSVITFQDLIF
LVLMS (SEQ ID NO: 13) and SGYLVIFLHRHQKNNPHLHNNSCSSIASHETGTITTAL
LLMICFVVFNVSNSCHSIYLSTVKKRDQLWTISDLISS CYPIFWSIFAH (SEQ >D NO:
14). Fragments and/or variants of these polypeptides, such as, for example,
fragments and/or
variants as described herein, are encompassed by the invention.
Polynucleotides encoding
these polypeptides (including fragments and/or variants) are also encompassed
by the
invention, as are antibodies that bind these polypeptides.
Preferred polypeptides of the present invention comprise, or alternatively
consist of,
one, two, or both of the immunogenic epitopes shown in SEQ ID NO: 8 as
residues: His-1 to
Leu-11 and Ser-46 to Arg-51. Fragments and/or variants of these polypeptides,
such as, for
example, fragments and/or variants as described herein, are encompassed by the
invention.
Polynucleotides encoding these polypeptides (including fragments andlor
variants) are also
encompassed by the invention, as are antibodies that bind these polypeptides.
This gene is expressed in hippocampal and testes tumor tissues.
Therefore, polynucleotides and polypeptides of the invention, including
antibodies,
are useful as reagents for differential identification of the tissues) or cell
types) present in a
biological sample and for diagnosis of diseases and conditions which include
but are not
limited to: neural and/or reproductive diseases and/or disorders. Similarly,
polypeptides and
antibodies directed to these polypeptides are useful in providing
immunological probes for
differential identification of the tissues) or cell type(s). For a number of
disorders of the


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
13
above tissues or cells, particularly of the neural and reproductive systems,
expression of this
gene at significantly higher or lower levels may be routinely detected in
certain tissues or cell
types (e.g., neural, reproductive, cancerous and wounded tissues) or bodily
fluids (e.g.,
lymph, serum, plasma, urine, synovial fluid and spinal fluid) or another
tissue or sample
S taken from an individual having such a disorder, relative to the standard
gene expression
level, i.e., the expression level in healthy tissue or bodily fluid from an
individual not having
the disorder.
The tissue distribution in hippocampus tissue and testes tumor tissue, and the
homology to pheromone receptors, indicates that polynucleotides, translation
products and
antibodies corresponding to this gene are useful for the diagnosis, detection
and/or treatment
of diseases and/or disorders of the neural and/or immune systems.
The tissue distribution in hippocampus tissue suggests that polynucleotides,
translation products and antibodies corresponding to this gene are useful for
the detection
and/or treatment of neurodegenerative disease states and behavioural disorders
such as
Alzheimer's Disease, Parkinson's Disease, Huntington's Disease, Tourette
Syndrome,
schizophrenia, mania, dementia, paranoia, obsessive compulsive disorder, panic
disorder,
learning disabilities, ALS, psychoses, autism, and altered behaviors,
including disorders in
feeding, sleep patterns, balance, and perception. In addition, the gene or
gene product may
also play a role in the treatment and/or detection of developmental disorders
associated with
the developing embryo, or sexually-linked disorders. This protein may play a
role in
transducing the signals of hormones, thus resulting in the activation of
particular genes that
may play relevant roles in normal neural activity or development.
Alternatively, the tissue distribution in testes tissue indicates that the
protein product
of this clone is useful for the treatment and diagnosis of conditions
concerning proper
testicular function (e.g., endocrine function, sperm maturation), as well as
cancer. Therefore,
this gene product is useful in the treatment of male infertility and/or
impotence. This gene
product is also useful in assays designed to identify binding agents, as such
agents
(antagonists) are useful as male contraceptive agents. Similarly, the protein
is believed to be
useful in the treatment and/or diagnosis of testicular cancer. The testes are
also a site of active
gene expression of transcripts that may be expressed, particularly at low
levels, in other
tissues of the body. Thus, again this protein may be involved in transducing
the signals of
hormones that specifically bind it, resulting in transcriptional activation of
target genes.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
14
Therefore, this gene product may be expressed in other specific tissues or
organs where it
may play related functional roles in other processes, such as hematopoiesis,
inflammation,
bone formation, and kidney function, to name a few possible target
indications.
Given the homology, the translation product of this gene is also useful in
identifying
modulators for control of reproduction and/or social behavior, to increase
fertility, or control
hormone secretion. Furthermore, the translation product of this gene may be
useful for
recreational uses, such as, for example, cosmetics or aphrodesiacs. Protein,
as well as,
antibodies directed against the protein may show utility as a tumor marker
and/or
immunotherapy targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 3
Preferred polypeptides of the present invention comprise, or alternatively
consist of,
one, two, three, or all three of the immunogenic epitopes shown in SEQ >D NO:
9 as residues:
Asp-65 to Gln-72, Arg-78 to Thr-92 and Lys-106 to Lys-112. Fragments and/or
variants of
these polypeptides, such as, for example, fragments and/or variants as
described herein, are
encompassed by the invention. Polynucleotides encoding these polypeptides
(including
fragments and/or variants) are also encompassed by the invention, as are
antibodies that bind
these polypeptides.
This gene is expressed in pregnant uterus and total fetus tissues, as well as
in
senescent fibroblasts and immune system tissues, such as activated T cells, T
cell lymphoma,
and primary dendritic cells.
Therefore, polynucleotides and polypeptides of the invention, including
antibodies,
are useful as reagents for differential identification of the tissues) or cell
types) present in a
biological sample and for diagnosis of diseases and conditions which include
but are not
limited to: diseases and/or disorders of the fetus and the musculo-skeletal
system. Similarly,
polypeptides and antibodies directed to these polypeptides are useful in
providing
immunological probes for differential identification of the tissues) or cell
type(s). For a
number of disorders of the above tissues or cells, particularly of the fetus,
immune system,
and the musculo-skeletal system, expression of this gene at significantly
higher or lower
levels may be routinely detected in certain tissues or cell types (e.g.,
musculo-skeletal, fetal,
immune, cancerous and wounded tissues) or bodily fluids (e.g., lymph, serum,
plasma, urine,
synovial fluid and spinal fluid) or another tissue or sample taken from an
individual having


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
such a disorder, relative to the standard gene expression level, i.e., the
expression level in
healthy tissue or bodily fluid from an individual not having the disorder.
The tissue distribution in pregnant uterus and total fetus tissues, as well as
in
senescent fibroblasts and immune system tissues, indicates that
polynucleotides, translation
5 products and antibodies corresponding to this gene are useful for the
diagnosis, detection
and/or treatment of diseases and/or disorders of the developing fetus, the
immune system, and
of the musculo-skeletal system.
Expression within embryonic tissue and other cellular sources marked by
proliferating
cells suggests that translation products corresponding to this gene may play a
role in the
10 regulation of cellular division, and may show utility in the diagnosis and
treatment of cancer
and other proliferative disorders. Similarly, embryonic development also
involves decisions
involving cell differentiation and/or apoptosis in pattern formation. Thus,
polynucleotides and
translation products corresponding to this gene may also be involved in
apoptosis or tissue
differentiation and could again be useful in cancer therapy.
15 Alternatively, the tissue distribution in senescent fibroblasts indicates
that
polynucleotides, translation products and antibodies corresponding to this
gene are useful for
the diagnosis, detection and/or treatment of diseases and/or disorders of the
musculo-skeletal
system, such as heart disease, restenosis, atherosclerosis, stoke, angina,
thrombosis, and
wound healing, as well as disorders afflicting connective tissues (e.g.
arthritis, trauma,
tendonitis, chrondomalacia and inflammation), such as in the diagnosis or
treatment of
various autoimmune disorders such as rheumatoid arthritis, lupus, scleroderma,
and
dermatomyositis as well as dwarfism, spinal deformation, and specific joint
abnormalities as
well as chondrodysplasias (ie. spondyloepiphyseal dysplasia congenita,
familial arthritis,
Atelosteogenesis type II, metaphyseal chondrodysplasia type Schmid).
Also, the tissue distribution in immune system tissues, such as T cells and
primary
dendritic cells, indicates that polynucleotides, translation products and
antibodies
corresponding to this gene are useful for the diagnosis, detection and/or
treatment of immune
system diseases and/or disorders. This gene product may be involved in the
regulation of
cytokine production, antigen presentation, or other processes that may also
suggest a
usefulness in the treatment of cancer (e.g. by boosting immune responses).
Since the gene is
expressed in cells of lymphoid origin, the gene or protein, as well as,
antibodies directed
against the protein may show utility as a tumor marker and/or immunotherapy
targets for the


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
16
above listed tissues. Therefore it may be also used as an agent for
immunological disorders
including arthritis, asthma, immune deficiency diseases such as AIDS,
leukemia, rheumatoid
arthritis, inflammatory bowel disease, sepsis, acne, and psoriasis. In
addition, this gene
product may have commercial utility in the expansion of stem cells and
committed
progenitors of various blood lineages, and in the differentiation and/or
proliferation of
various cell types.
This gene shares sequence homology to other steroid hormone receptors, and
accordingly it is thought that polynucleotides and translation products
corresponding to this
gene may share some of the biological activities of other steroid hormone
receptors. Thus,
translation products corresponding to this gene may be involved in transducing
the signals of
hormones that specifically bind, resulting in transcriptional activation of
target genes.
Therefore, translation products corresponding to this gene may be expressed in
other specific
tissues or organs where it may play related functional roles in other
processes, such as
hematopoiesis, inflammation, bone formation, and kidney function, to name a
few possible
target indications. Given the homology, translation products corresponding to
this gene are
also useful in identifying modulators for control of reproduction and/or
social behavior, to
increase fertility, or control hormone secretion. Protein, as well as,
antibodies directed against
the protein may show utility as a tumor marker and/or immunotherapy targets
for the above
listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 4
In specific embodiments, polypeptides of the invention comprise, or
alternatively
consist of, the following amino acid sequence: HEPPSWKTKVTVFSFLKVLLRSVND
SQRNSTWVYLKLVFIHLC (SEQ ID NO: 15). Fragments and/or variants of these
polypeptides, such as, for example, fragments and/or variants as described
herein, are
encompassed by the invention. Polynucleotides encoding these polypeptides
(including
fragments and/or variants) are also encompassed by the invention, as are
antibodies that bind
these polypeptides.
Preferred polypeptides of the present invention comprise, or alternatively
consist of,
the immunogenic epitope shown in SEQ ID NO: 10 as residues: Lys-43 to Arg-48.
Fragments
and/or variants of these polypeptides, such as, for example, fragments and/or
variants as
described herein, are encompassed by the invention. Polynucleotides encoding
these


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
17
polypeptides (including fragments and/or variants) are also encompassed by the
invention, as
are antibodies that bind these polypeptides.
This gene is expressed in 9-week old human tissues, fetal brain tissue, and
placental
tissue.
Therefore, polynucleotides and polypeptides of the invention, including
antibodies,
are useful as reagents for differential identification of the tissues) or cell
types) present in a
biological sample and for diagnosis of diseases and conditions which include
but are not
limited to: developmental and placental diseases and/or disorders. Similarly,
polypeptides and
antibodies directed to these polypeptides are useful in providing
immunological probes for
differential identification of the tissues) or cell type(s). For a number of
disorders of the
above tissues or cells, particularly of the fetus and placenta, expression of
this gene at
significantly higher or lower levels may be routinely detected in certain
tissues or cell types
(e.g., fetal, developmental, placental, cancerous and wounded tissues) or
bodily fluids (e.g.,
lymph, serum, plasma, urine, synovial fluid and spinal fluid) or another
tissue or sample
taken from an individual having such a disorder, relative to the standard gene
expression
level, i.e., the expression level in healthy tissue or bodily fluid from an
individual not having
the disorder.
The tissue distribution in 9-week old tissues, fetal brain tissue, and
placental tissue,
indicates that polynucleotides, translation products and antibodies
corresponding to this gene
are useful for the diagnosis, detection and/or treatment of diseases and/or
disorders of the
fetus, placenta, and developing systems.
This gene shares sequence homology to other steroid hormone receptors, and
accordingly it is thought that polynucleotides and translation products
corresponding to this
gene may share some of the biological activities of other steroid hormone
receptors. Thus,
translation products corresponding to this gene may be involved in transducing
the signals of
hormones that specifically bind, resulting in transcriptional activation of
target genes.
Therefore, translation products corresponding to this gene may be expressed in
other specific
tissues or organs where it may play related functional roles in other
processes, such as
hematopoiesis, inflammation, bone formation, and kidney function, to name a
few possible
target indications. Given the homology, translation products corresponding to
this gene are
also useful in identifying modulators for control of reproduction and/or
social behavior, to
increase fertility, or control hormone secretion.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
18
The tissue distribution suggests that the protein product of this clone is
useful for the
diagnosis and/or treatment of disorders of the placenta. Specific expression
within the
placenta suggests that this gene product may play a role in the proper
establishment and
maintenance of placental function. Alternately, this gene product may be
produced by the
placenta and then transported to the embryo, where it may play a crucial role
in the
development and/or survival of the developing embryo or fetus. Expression of
this gene
product in a vascular-rich tissue such as the placenta also suggests that this
gene product may
be produced more generally in endothelial cells or within the circulation. In
such instances, it
may play more generalized roles in vascular function, such as in angiogenesis.
It may also be
produced in the vasculature and have effects on other cells within the
circulation, such as
hematopoietic cells. It may serve to promote the proliferation, survival,
activation, and/or
differentiation of hematopoietic cells, as well as other cells throughout the
body. Protein, as
well as, antibodies directed against the protein may show utility as a tumor
marker and/or
immunotherapy targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 5
In specific embodiments, polypeptides of the invention comprise, or
alternatively
consist of, the following amino acid sequence: ARAKIEENQNLYFRKYPIEIFRFIPNSK
(SEQ ID NO: 16). Fragments and/or variants of these polypeptides, such as, for
example,
fragments andlor variants as described herein, are encompassed by the
invention.
Polynucleotides encoding these polypeptides (including fragments and/or
variants) are also
encompassed by the invention, as are antibodies that bind these polypeptides.
This gene is expressed in neutrophils.
Therefore, polynucleotides and polypeptides of the invention, including
antibodies,
are useful as reagents for differential identification of the tissues) or cell
types) present in a
biological sample and for diagnosis of diseases and conditions which include
but are not
limited to: diseases and/or disorders of the immune system. Similarly,
polypeptides and
antibodies directed to these polypeptides are useful in providing
immunological probes for
differential identification of the tissues) or cell type(s). For a number of
disorders of the
above tissues or cells, particularly of the immune system, expression of this
gene at
significantly higher or lower levels may be routinely detected in certain
tissues or cell types
(e.g., immune, cancerous and wounded tissues) or bodily fluids (e.g., lymph,
serum, plasma,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
19
urine, synovial fluid and spinal fluid) or another tissue or sample taken from
an individual
having such a disorder, relative to the standard gene expression level, i.e.,
the expression
level in healthy tissue or bodily fluid from an individual not having the
disorder.
The tissue distribution in neutrophils indicates that polynucleotides,
translation
products and antibodies corresponding to this gene are useful for the
diagnosis, detection,
andlor treatment of diseases and/or disorders of the immune system.
This gene shares sequence homology to other steroid hormone receptors, and
accordingly it is thought that polynucleotides and translation products
corresponding to this
gene may share some of the biological activities of other steroid hormone
receptors. Thus,
translation products corresponding to this gene may be involved in transducing
the signals of
hormones that specifically bind, resulting in transcriptional activation of
target genes.
Therefore, translation products corresponding to this gene may be expressed in
other specific
tissues or organs where it may play related functional roles in other
processes, such as
hematopoiesis, inflammation, bone formation, and kidney function, to name a
few possible
1 S target indications. Given the homology, translation products corresponding
to this gene are
also useful in identifying modulators for control of reproduction and/or
social behavior, to
increase fertility, or control hormone secretion.
This gene product may be involved in the regulation of cytokine production,
antigen
presentation, or other processes that may also suggest a usefulness in the
treatment of cancer
(e.g. by boosting immune responses). Expression of this gene product in
neutrophils also
strongly suggests a role for this protein in immune function and immune
surveillance. This
gene product may be involved in the regulation of cytokine production, antigen
presentation,
or other processes that may also suggest a usefulness in the treatment of
cancer (e.g. by
boosting immune responses). Since the gene is expressed in cells of immune
origin, the gene
or protein, as well as, antibodies directed against the protein may show
utility as a tumor
marker and/or immunotherapy targets for the above listed tissues. Therefore it
may be also
used as an agent for immunological disorders including arthritis, asthma,
immune deficiency
diseases such as AIDS, leukemia, rheumatoid arthritis, inflammatory bowel
disease, sepsis,
acne, and psoriasis. In addition, this gene product may have commercial
utility in the
expansion of stem cells and committed progenitors of various blood lineages,
and in the
differentiation and/or proliferation of various cell types. Protein, as well
as, antibodies


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
directed against the protein may show utility as a tumor marker and/or
immunotherapy
targets for the above listed tissues.
TABLE 1
5
NT 5' 3' AA
NT NT


ATCC SEQ of of 5' SEQ Last
NT


Deposit ID TotalCloneCloneof ID AA


Gene cDNA No:Z NO: NT Seq.Seq. StartNO: of


No. Clone and Vector X Se CodonY ORF
ID Date .


1 HWHQR64 PTA629 pCMVSport 2 1079 1 1079 7 76


09/09/993.0


2 HHPFB87 PTA1477Uni-ZAP 3 1734 1 1734 8 98
XR


03/13/00


3 HOUFT74 PTA629 Uni-ZAP 4 642 1 642 9 120
XR


09/09/99


4 HPWSB15 PTA629 pBluescript5 1697 1 1697 538 10 81


09/09/99


5 HNGAY56 PTA629 Uni-ZAP 6 1446 1 1446 160 11 51
XR


09/09/99


Table 1 summarizes the information corresponding to each "Gene No:" described
above. The nucleotide sequence identified as "NT SEQ ID NO:X" was assembled
from
10 partially homologous ("overlapping") sequences obtained from the "cDNA
clone ID"
identified in Table 1 and, in some cases, from additional related DNA clones.
The
overlapping sequences were assembled into a single contiguous sequence of high
redundancy
(usually three to five overlapping sequences at each nucleotide position),
resulting in a final
sequence identified as SEQ ID NO:X.
15 The cDNA Clone ID was deposited on the date and given the corresponding
deposit
number listed in "ATCC Deposit No:Z and Date." Some of the deposits contain
multiple
different clones corresponding to the same gene. "Vector" refers to the type
of vector
contained in the cDNA Clone ID.
"Total NT Seq." refers to the total number of nucleotides in the contig
identified by
20 "Gene No:" The deposited plasmid contains all of these sequences, reflected
by the
nucleotide position indicated as "5' NT of Clone Seq." and the "3' NT of Clone
Seq." of SEQ
ID NO:X. The nucleotide position of SEQ ID NO:X of the putative methionine
start codon


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
21
(if present) is identified as "5' NT of Start Codon." Similarly , the
nucleotide position of SEQ
ID NO:X of the predicted signal sequence (if present) is identified as "5' NT
of First AA of
Signal Pep."
The translated amino acid sequence, beginning with the first translated codon
of the
polynucleotide sequence, is identified as "AA SEQ ID NO:Y," although other
reading frames
can also be easily translated using known molecular biology techniques. The
polypeptides
produced by these alternative open reading frames are specifically
contemplated by the
present invention.
SEQ ID NO:X (where X may be any of the polynucleotide sequences disclosed in
the
sequence listing) and the translated SEQ ID NO:Y (where Y may be any of the
polypeptide
sequences disclosed in the sequence listing) are sufficiently accurate and
otherwise suitable
for a variety of uses well known in the art and described further below. For
instance, SEQ 1D
NO:X has uses including, but not limited to, in designing nucleic acid
hybridization probes
that will detect nucleic acid sequences contained in SEQ ID NO:X or the cDNA
contained in
a deposited plasmid. These probes will also hybridize to nucleic acid
molecules in biological
samples, thereby enabling a variety of forensic and diagnostic methods of the
invention.
Similarly, polypeptides identified from SEQ ID NO:Y have uses that include,
but are not
limited to generating antibodies, which bind specifically to the secreted
proteins encoded by
the cDNA clones identified in Table 1.
Nevertheless, DNA sequences generated by sequencing reactions can contain
sequencing errors. The errors exist as misidentified nucleotides, or as
insertions or deletions
of nucleotides in the generated DNA sequence. The erroneously inserted or
deleted
nucleotides cause frame shifts in the reading frames of the predicted amino
acid, sequence. In
these cases, the predicted amino acid sequence diverges from the actual amino
acid sequence,
even though the generated DNA sequence may be greater than 99.9% identical to
the actual
DNA sequence (for example, one base insertion or deletion in an open reading
frame of over
1000 bases).
Accordingly, for those applications requiring precision in the nucleotide
sequence or
the amino acid sequence, the present invention provides not only the generated
nucleotide
sequence identified as SEQ ID NO:X, and the predicted translated amino acid
sequence
identified as SEQ 117 NO:Y, but also a sample of plasmid DNA containing a
human cDNA of
the invention deposited with the ATCC, as set forth in Table 1. The nucleotide
sequence of


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
22
each deposited plasmid can readily be determined by sequencing the deposited
plasmid in
accordance with known methods.
The predicted amino acid sequence can then be verified from such deposits.
Moreover, the amino acid sequence of the protein encoded by a particular
plasmid can also be
directly determined by peptide sequencing or by expressing the protein in a
suitable host cell
containing the deposited human cDNA, collecting the protein, and determining
its sequence.
Also provided in Table 1 is the name of the vector which contains the cDNA
plasmid.
Each vector is routinely used in the art. The following additional information
is provided for
convenience.
Vectors Lambda Zap (U.5. Patent Nos. 5,128,256 and 5,286,636), Uni-Zap XR
(U.5.
Patent Nos. 5,128, 256 and 5,286,636), Zap Express (U.S. Patent Nos. 5,128,256
and
5,286,636), pBluescript (pBS) (Short, J. M. et al., Nucleic Acids Res. 16:7583-
7600 (1988);
Aping-Mees, M. A. and Short, J. M., Nucleic Acids Res. 17:9494 (1989)) and pBK
(Alting-
Mees, M. A. et al., Strategies 5:58-61 (1992)) are commercially available from
Stratagene
Cloning Systems, Inc., 11011 N. Torrey Pines Road, La Jolla, CA, 92037. pBS
contains an
ampicillin resistance gene and pBK contains a neomycin resistance gene.
Phagemid pBS
may be excised from the Lambda Zap and Uni-Zap XR vectors, and phagemid pBK
may be
excised from the Zap Express vector. Both phagemids may be transformed into E.
coli strain
XL-1 Blue, also available from Stratagene.
Vectors pSportl, pCMVSport 1.0, pCMVSport 2.0 and pCMVSport 3.0, were
obtained from Life Technologies, Inc., P. O. Box 6009, Gaithersburg, MD 20897.
All Sport
vectors contain an ampicillin resistance gene and may be transformed into E.
coli strain
DH10B, also available from Life Technologies. See, for instance, Gruber, C.
E., et al., Focus
15:59 (1993). Vector lafmid BA (Bento Soares, Columbia University, New York,
NY)
contains an ampicillin resistance gene and can be transformed into E. coli
strain XL-1 Blue.
Vector pCR~2.1, which is available from Invitrogen, 1600 Faraday Avenue,
Carlsbad, CA
92008, contains an ampicillin resistance gene and may be transformed into E.
coli strain
DH10B, available from Life Technologies. See, for instance, Clark, J. M., Nuc.
Acids Res.
16:9677-9686 (1988) and Mead, D. et al., BiolTechnology 9: (1991).
The present invention also relates to the genes corresponding to SEQ ID NO:X,
SEQ
ID NO:Y, and/or a deposited plasmid (cDNA plasmid:Z). The corresponding gene
can be
isolated in accordance with known methods using the sequence information
disclosed herein.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
23
Such methods include, but are not limited to, preparing probes or primers from
the disclosed
sequence and identifying or amplifying the corresponding gene from appropriate
sources of
genomic material.
Also provided in the present invention are allelic variants, orthologs, and/or
species
homologs. Procedures known in the art can be used to obtain full-length genes,
allelic
variants, splice variants, full-length coding portions, orthologs, and/or
species homologs of
genes corresponding to SEQ ID NO:X, SEQ ID NO:Y, and/or cDNA plasmid:Z, using
information from the sequences disclosed herein or the clones deposited with
the ATCC. For
example, allelic variants and/or species homologs may be isolated and
identified by making
suitable probes or primers from the sequences provided herein and screening a
suitable
nucleic acid source for allelic variants and/or the desired homologue.
The present invention provides a polynucleotide comprising, or alternatively
consisting of, the nucleic acid sequence of SEQ ID NO:X and/or cDNA plasmid:Z.
The
present invention also provides a polypeptide comprising, or alternatively,
consisting of, the
polypeptide sequence of SEQ ID NO:Y, a polypeptide encoded by SEQ 117 NO:X,
and/or a
polypeptide encoded by the cDNA in cDNA plasmid:Z. Polynucleotides encoding a
polypeptide comprising, or alternatively consisting of the polypeptide
sequence of SEQ ID
NO:Y, a polypeptide encoded by SEQ ID NO:X and/or a polypeptide encoded by the
cDNA
in cDNA plasmid:Z, are also encompassed by the invention. The present
invention further
encompasses a polynucleotide comprising, or alternatively consisting of the
complement of
the nucleic acid sequence of SEQ ID NO:X, and/or the complement of the coding
strand of
the cDNA in cDNA plasmid:Z.
Many polynucleotide sequences, such as EST sequences, are publicly available
and
accessible through sequence databases and may have been publicly available
prior to
conception of the present invention. Preferably, such related polynucleotides
are specifically
excluded from the scope of the present invention. To list every related
sequence would
unduly burden the disclosure of this application. Accordingly, preferably
excluded from SEQ
B7 NO:X are one or more polynucleotides comprising a nucleotide sequence
described by the
general formula of a-b, where a is any integer between l and the final
nucleotide minus 15 of
SEQ ID NO:X, b is an integer of 15 to the final nucleotide of SEQ ID NO:X,
where both a
and b correspond to the positions of nucleotide residues shown in SEQ ID NO:X,
and where
b is greater than or equal to a + 14.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
24
RACE Protocol For Recovery of Full-Length Genes
Partial cDNA clones can be made full-length by utilizing the rapid
amplification of
cDNA ends (RACE) procedure described in Frohman, M.A., et al., Proc. Nat'1.
Acad. Sci.
S USA, 85:8998-9002 (1988). A cDNA clone missing either the S' or 3' end can
be
reconstructed to include the absent base pairs extending to the translational
start or stop
codon, respectively. In some cases, cDNAs are missing the start of
translation, therefor. The
following briefly describes a modification of this original 5' RACE procedure.
Poly A+ or
total RNA is reverse transcribed with Superscript II (Gibco/BRL) and an
antisense or
complementary primer specific to the cDNA sequence. The primer is removed from
the
reaction with a Microcon Concentrator (Amicon). The first-strand cDNA is then
tailed with
dATP and terminal deoxynucleotide transferase (GibcolBRL). Thus, an anchor
sequence is
produced which is needed for PCR amplification. The second strand is
synthesized from the
dA-tail in PCR buffer, Taq DNA polymerase (Perkin-Elmer Cetus), an oligo-dT
primer
containing three adjacent restriction sites (XhoI, SaII and CIaI) at the 5'
end and a primer
containing just these restriction sites. This double-stranded cDNA is PCR
amplified for 40
cycles with the same primers as well as a nested cDNA-specific antisense
primer. The PCR
products are size-separated on an ethidium bromide-agarose gel and the region
of gel
containing cDNA products the predicted size of missing protein-coding DNA is
removed.
cDNA is purified from the agarose with the Magic PCR Prep kit (Promega),
restriction
digested with XhoI or SaII, and ligated to a plasmid such as pBluescript SKII
(Stratagene) at
XhoI and EcoRV sites. This DNA is transformed into bacteria and the plasmid
clones
sequenced to identify the correct protein-coding inserts. Correct 5' ends are
confirmed by
comparing this sequence with the putatively identified homologue and overlap
with the
partial cDNA clone. Similar methods known in the art and/or commercial kits
are used to
amplify and recover 3' ends.
Several quality-controlled kits are commercially available for purchase.
Similar
reagents and methods to those above are supplied in kit form from Gibco/BRL
for both S' and
3' RACE for recovery of full length genes. A second kit is available from
Clontech which is
a modification of a related technique, SLIC (single-stranded ligation to
single-stranded
cDNA), developed by Dumas et al., Nucleic Acids Res., 19:5227-32 (1991). The
major
differences in procedure are that the RNA is alkaline hydrolyzed after reverse
transcription


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
and RNA ligase is used to join a restriction site-containing anchor primer to
the first-strand
cDNA. This obviates the necessity for the dA-tailing reaction which results in
a polyT
stretch that is difficult to sequence past.
An alternative to generating 5' or 3' cDNA from RNA is to use cDNA library
double
s stranded DNA. An asymmetric PCR-amplified antisense cDNA strand is
synthesized with an
antisense cDNA-specific primer and a plasmid-anchored primer. These primers
are removed
and a symmetric PCR reaction is performed with a nested cDNA-specific
antisense primer
and the plasmid-anchored primer.
10 RNA Ligase Protocol For Generating The 5' or 3' End Sequences To Obtain
Full Length
Genes
Once a gene of interest is identified, several methods are available for the
identification of the 5' or 3' portions of the gene which may not be present
in the original
cDNA plasmid. These methods include, but are not limited to, filter probing,
clone
15 enrichment using specific probes and protocols similar and identical to 5'
and 3'RACE.
While the full length gene may be present in the library and can be identified
by probing, a
useful method for generating the 5' or 3' end is to use the existing sequence
information from
the original cDNA to generate the missing information. A method similar to
5'RACE is
available for generating the missing 5' end of a desired full-length gene.
(This method was
20 published by Fromont-Racine et al., Nucleic Acids Res., 21(7):1683-1684
(1993)). Briefly, a
specific RNA oligonucleotide is ligated to the 5' ends of a population of RNA
presumably
containing full-length gene RNA transcript and a primer set containing a
primer specific to
the ligated RNA oligonucleotide and a primer specific to a known sequence of
the gene of
interest, is used to PCR amplify the 5' portion of the desired full length
gene which may then
25 be sequenced and used to generate the full length gene. This method starts
with total RNA
isolated from the desired source, poly A RNA may be used but is not a
prerequisite for this
procedure. The RNA preparation may then be treated with phosphatase if
necessary to
eliminate 5' phosphate groups on degraded or damaged RNA which may interfere
with the
later RNA ligase step. The phosphatase if used is then inactivated and the RNA
is treated
with tobacco acid pyrophosphatase in order to remove the cap structure present
at the S' ends
of messenger RNAs. This reaction leaves a 5' phosphate group at the 5' end of
the cap
cleaved RNA which can then be ligated to an RNA oligonucleotide using T4 RNA
ligase.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
26
This modified RNA preparation can then be used as a template for first strand
cDNA
synthesis using a gene specific oligonucleotide. The first strand synthesis
reaction can then
be used as a template for PCR amplification of the desired 5' end using a
primer specific to
the ligated RNA oligonucleotide and a primer specific to the known sequence of
the steroid
hormone receptor gene of interest. The resultant product is then sequenced and
analyzed to
confirm that the 5' end sequence belongs to the relevant steroid hormone
receptor gene.
Polynucleotide and Polypeptide Fragments
The present invention is also directed to polynucleotide fragments of the
polynucleotides (nucleic acids) of the invention. In the present invention, a
"polynucleotide
fragment" refers to a polynucleotide having a nucleic acid sequence which: is
a portion of the
cDNA contained in cDNA plasmid:Z or encoding the polypeptide encoded by the
cDNA
contained in cDNA plasmid:Z; is a portion of the polynucleotide sequence in
SEQ ID NO:X
or the complementary strand thereto; is a polynucleotide sequence encoding a
portion of the
polypeptide of SEQ m NO:Y; or is a polynucleotide sequence encoding a portion
of a
polypeptide encoded by SEQ ID NO:X. The nucleotide fragments of the invention
are
preferably at least about 15 nt, and more preferably at least about 20 nt,
still more preferably
at least about 30 nt, and even more preferably, at least about 40 nt, at least
about 50 nt, at
least about 75 nt, at least about 100 nt, at least about 125 nt, or at least
about 150 nt in length.
A fragment "at least 20 nt in length," for example, is intended to include 20
or more
contiguous bases from, for example, the sequence contained in the cDNA in cDNA
plasmid:Z, or the nucleotide sequence shown in SEQ ID NO:X or the
complementary stand
thereto. In this context "about" includes the particularly recited value, or a
value larger or
smaller by several (5, 4, 3, 2, or 1 ) nucleotides. These nucleotide fragments
have uses that
include, but are not limited to, as diagnostic probes and primers as discussed
herein. Of
course, larger fragments (e.g., at least 150, 175, 200, 250, 500, 600, 1000,
or 2000
nucleotides in length ) are also encompassed by the invention.
Moreover, representative examples of polynucleotide fragments of the
invention,
include, for example, fragments comprising, or alternatively consisting of, a
sequence from
about nucleotide number 1-50, S1-100, 101-150, 151-200, 201-250, 251-300, 301-
350, 351
400, 401-450, 451-500, 501-550, 551-600, 651-700,701- 750, 751-800, 800-850,
851-900,
901-950, 951-1000, 1001-1050, 1051-1100, 1101-1150, 1151-1200, 1201-1250, 1251-
1300,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
27
1301-1350, 1351-1400, 1401-1450, 1451-1500, 1501-1550, 1551-1600, 1601-1650,
1651-
1700, and/or 1701-1734 of SEQ ID NO:X, or the complementary strand thereto. In
this
context "about" includes the particularly recited range or a range larger or
smaller by several
(5, 4, 3, 2, or 1 ) nucleotides, at either terminus or at both termini.
Preferably, these fragments
encode a polypeptide which has a functional activity (e.g. biological
activity) of the
polypeptide encoded by a polynucleotide of which the sequence is a portion.
More
preferably, these fragments can be used as probes or primers as discussed
herein.
Polynucleotides which hybridize to one or more of these fragments under
stringent
hybridization conditions or alternatively, under lower stringency conditions,
are also
encompassed by the invention, as are polypeptides encoded by these
polynucleotides or
fragments.
Moreover, representative examples of polynucleotide fragments of the
invention,
include, for example, fragments comprising, or alternatively consisting of, a
sequence from
about nucleotide number 1-50, 51-100, 101-150, 151-200, 201-250, 251-300, 301-
350, 351-
400, 401-450; 451-500, 501-550, 551-600, 651-700,701- 750, 751-800, 800-850,
851-900,
901-950, 951-1000, 1001-1050, 1051-1100, 1101-1150, 1151-1200, 1201-1250, 1251-
1300,
1301-1350, 1351-1400, 1401-1450, 1451-1500, 1501-1550, 1551-1600, 1601-1650,
1651-
1700, and/or 1701-1734 of the cDNA nucleotide sequence contained in cDNA
plasmid:Z, or
the complementary strand thereto. In this context "about" includes the
particularly recited
range or a range larger or smaller by several (5, 4, 3, 2, or 1) nucleotides,
at either terminus or
at both termini. Preferably, these fragments encode a polypeptide which has a
functional
activity (e.g. biological activity) of the polypeptide encoded by the cDNA
nucleotide
sequence contained in cDNA plasmid:Z. More preferably, these fragments can be
used as
probes or primers as discussed herein. Polynucleotides which hybridize to one
or more of
these fragments under stringent hybridization conditions, or alternatively,
under lower
stringency conditions are also encompassed by the invention, as are
polypeptides encoded by
these polynucleotides or fragments.
In the present invention, a "polypeptide fragment" refers to an amino acid
sequence
which is a portion of that contained in SEQ ID NO:Y, a portion of an amino
acid sequence
encoded by the polynucleotide sequence of SEQ ID NO:X, and/or encoded by the
cDNA in
cDNA plasmid:Z. Protein (polypeptide) fragments may be "free-standing," or
comprised
within a larger polypeptide of which the fragment forms a part or region, most
preferably as a


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
28
single continuous region. Representative examples of polypeptide fragments of
the
invention, include, for example, fragments comprising, or alternatively
consisting of, an
amino acid sequence from about amino acid number 1-20, 21-40, 41-60, 61-80, 81-
100,
and/or 101-120 of the coding region of SEQ ID NO:Y. Moreover, polypeptide
fragments of
the invention may be at least about 10, 15, 20, 25, 30, 35, 40, 45, 50, 55,
60, 65, 70, 75, 80,
85, 90, 100, or 110 amino acids in length. In this context "about" includes
the particularly
recited ranges or values, or ranges or values larger or smaller by several (S,
4, 3, 2, or 1)
amino acids, at either terminus or at both termini. Polynucleotides encoding
these
polypeptide fragments are also encompassed by the invention.
Even if deletion of one or more amino acids from the N-terminus of a protein
results
in modification of loss of one or more biological functions of the protein,
other functional
activities (e.g., biological activities, ability to multimerize, ability to
bind a ligand) may still
be retained. For example, the ability of shortened muteins to induce and/or
bind to antibodies
which recognize the complete or mature forms of the polypeptides generally
will be retained
when less than the majority of the residues of the complete or mature
polypeptide are
removed from the N-terminus. Whether a particular polypeptide lacking N-
terminal residues
of a complete polypeptide retains such immunologic activities can readily be
determined by
routine methods described herein and otherwise known in the art. It is not
unlikely that a
mutein with a large number of deleted N-terminal amino acid residues may
retain some
biological or immunogenic activities. In fact, peptides composed of as few as
six amino acid
residues may often evoke an immune response.
Accordingly, polypeptide fragments of the invention include the secreted
protein as
well as the mature form. Further preferred polypeptide fragments include the
secreted protein
or the mature form having a continuous series of deleted residues from the
amino or the
carboxy terminus, or both. For example, any number of amino acids, ranging
from 1-60, can
be deleted from the amino terminus of either the secreted polypeptide or the
mature form.
Similarly, any number of amino acids, ranging from 1-30, can be deleted from
the carboxy
terminus of the secreted protein or mature form. Furthermore, any combination
of the above
amino and carboxy terminus deletions are preferred. Similarly, polynucleotides
encoding
these polypeptide fragments are also preferred.
The present invention further provides polypeptides having one or more
residues
deleted from the amino terminus of the amino acid sequence of a polypeptide
disclosed


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
29
herein (e.g., a polypeptide of SEQ IID NO:Y, a polypeptide encoded by the
polynucleotide
sequence contained in SEQ ID NO:X, and/or a polypeptide encoded by the cDNA
contained
in cDNA plasmid:Z). In particular, N-terminal deletions may be described by
the general
formula m-q, where q is a whole integer representing the total number of amino
acid residues
in a polypeptide of the invention (e.g., the polypeptide disclosed in SEQ ID
NO:Y), and m is
defined as any integer ranging from 2 to q-6. Polynucleotides encoding these
polypeptides,
including fragments and/or variants, are also encompassed by the invention.
Also as mentioned above, even if deletion of one or more amino acids from the
C-terminus of a protein results in modification of loss of one or more
biological functions of
the protein, other functional activities (e.g., biological activities, ability
to multimerize,
ability to bind a ligand) may still be retained. For example the ability of
the shortened mutein
to induce and/or bind to antibodies which recognize the complete or mature
forms of the
polypeptide generally will be retained when less than the majority of the
residues of the
complete or mature polypeptide are removed from the C-terminus. Whether a
particular
polypeptide lacking C-terminal residues of a complete polypeptide retains such
immunologic
activities can readily be determined by routine methods described herein and
otherwise
known in the art. It is not unlikely that a mutein with a large number of
deleted C-terminal
amino acid residues may retain some biological or immunogenic activities. In
fact, peptides
composed of as few as six amino acid residues may often evoke an immune
response.
Accordingly, the present invention further provides polypeptides having one or
more
residues from the carboxy terminus of the amino acid sequence of a polypeptide
disclosed
herein (e.g., a polypeptide of SEQ ID NO:Y, a polypeptide encoded by the
polynucleotide
sequence contained in SEQ ID NO:X, and/or a polypeptide encoded by the cDNA
contained
in cDNA plasmid:Z). In particular, C-terminal deletions may be described by
the general
formula 1-n, where n is any whole integer ranging from 6 to q-1, and where n
corresponds to
the position of an amino acid residue in a polypeptide of the invention.
Polynucleotides
encoding these polypeptides, including fragments and/or variants, are also
encompassed by
the invention.
In addition, any of the above described N- or C-terminal deletions can be
combined to
produce a N- and C-terminal deleted polypeptide. The invention also provides
polypeptides
having one or more amino acids deleted from both the amino and the carboxyl
termini, which
may be described generally as having residues m-n of a polypeptide encoded by
SEQ >D


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
NO:X (e.g., including, but not limited to, the preferred polypeptide disclosed
as SEQ ID
NO:Y), and/or the cDNA in cDNA plasmid:Z, and/or the complement thereof, where
n and m
are integers as described above. Polynucleotides encoding these polypeptides,
including
fragments and/or variants, are also encompassed by the invention.
5 Any polypeptide sequence contained in the polypeptide of SEQ ID NO:Y,
encoded by
the polynucleotide sequences set forth as SEQ ID NO:X, or encoded by the cDNA
in cDNA
plasmid:Z may be analyzed to determine certain preferred regions of the
polypeptide. For
example, the amino acid sequence of a polypeptide encoded by a polynucleotide
sequence of
SEQ >D NO:X or the cDNA in cDNA plasmid:Z may be analyzed using the default
10 parameters of the DNASTAR computer algorithm (DNASTAR, Inc., 1228 S. Park
St.,
Madison, WI 53715 USA; http://www.dnastar.comn.
Polypeptide regions that may be routinely obtained using the DNASTAR computer
algorithm include, but are not limited to, Gamier-Robson alpha-regions, beta-
regions,
turn-regions, and coil-regions, Chou-Fasman alpha-regions, beta-regions, and
turn-regions,
15 Kyte-Doolittle hydrophilic regions and hydrophobic regions, Eisenberg alpha-
and
beta-amphipathic regions, Karplus-Schulz flexible regions, Emini surface-
forming regions
and Jameson-Wolf regions of high antigenic index. Among highly preferred
polynucleotides
of the invention in this regard are those that encode polypeptides comprising
regions that
combine several structural features, such as several (e.g., l, 2, 3 or 4) of
the features set out
20 above.
Additionally, Kyte-Doolittle hydrophilic regions and hydrophobic regions,
Emini
surface-forming regions, and Jameson-Wolf regions of high antigenic index
(i.e., containing
four or more contiguous amino acids having an antigenic index of greater than
or equal to
1.5, as identified using the default parameters of the Jameson-Wolf program)
can routinely be
25 used to determine polypeptide regions that exhibit a high degree of
potential for antigenicity.
Regions of high antigenicity are determined from data by DNASTAR analysis by
choosing
values which represent regions of the polypeptide which are likely to be
exposed on the
surface of the polypeptide in an environment in which antigen recognition may
occur in the
process of initiation of an immune response.
30 Preferred polypeptide fragments of the invention are fragments comprising,
or
alternatively, consisting of, an amino acid sequence that displays a
functional activity (e.g.
biological activity) of the polypeptide sequence of which the amino acid
sequence is a


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
31
fragment. By a polypeptide displaying a "functional activity" is meant a
polypeptide capable
of one or more known functional activities associated with a full-length
protein, such as, for
example, biological activity, antigenicity, immunogenicity, and/or
multimerization, as
described supra.
Other preferred polypeptide fragments are biologically active fragments.
Biologically
active fragments are those exhibiting activity similar, but not necessarily
identical, to an
activity of the polypeptide of the present invention. The biological activity
of the fragments
may include an improved desired activity, or a decreased undesirable activity.
In preferred embodiments, polypeptides of the invention comprise, or
alternatively
consist of, one, two, three, four, five or more of the antigenic fragments of
the polypeptide of
SEQ >D NO:Y, or portions thereof. Polynucleotides encoding these polypeptides,
including
fragments and/or variants, are also encompassed by the invention.
The present invention encompasses polypeptides comprising, or alternatively
consisting of, an epitope of the polypeptide sequence shown in SEQ ID NO:Y, or
an epitope
of the polypeptide sequence encoded by the cDNA in cDNA plasmid:Z, or encoded
by a
polynucleotide that hybridizes to the complement of an epitope encoding
sequence of SEQ
ID NO:X, or an epitope encoding sequence contained in cDNA plasmid:Z under
stringent
hybridization conditions, or alternatively, under lower stringency
hybridization, as defined
supra. The present invention further encompasses polynucleotide sequences
encoding an
epitope of a polypeptide sequence of the invention (such as, for example, the
sequence
disclosed in SEQ ID NO:X), polynucleotide sequences of the complementary
strand of a
polynucleotide sequence encoding an epitope of the invention, and
polynucleotide sequences
which hybridize to this complementary strand under stringent hybridization
conditions, or
alternatively, under lower stringency hybridization conditions, as defined
supra.
The term "epitopes," as used herein, refers to portions of a polypeptide
having
antigenic or immunogenic activity in an animal, preferably a mammal, and most
preferably
in a human. In a preferred embodiment, the present invention encompasses a
polypeptide
comprising an epitope, as well as the polynucleotide encoding this
polypeptide. An
"immunogenic epitope," as used herein, is defined as a portion of a protein
that elicits an
antibody response in an animal, as determined by any method known in the art,
for example,
by the methods for generating antibodies described infra. (See, for example,
Geysen et al.,
Proc. Natl. Acad. Sci. USA 81:3998- 4002 (1983)). The term "antigenic
epitope," as used


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
32
herein, is defined as a portion of a protein to which an antibody can
immunospecifically bind
its antigen as determined by any method well known in the art, for example, by
the
immunoassays described herein. Immunospecific binding excludes non-specific
binding but
does not necessarily exclude cross- reactivity with other antigens. Antigenic
epitopes need
not necessarily be immunogenic.
Fragments which function as epitopes may be produced by any conventional
means.
(See, e.g., Houghten, R. A., Proc. Natl. Acad. Sci. USA 82:5131-5135 (1985)
further
described in U.S. Patent No. 4,631,211.)
In the present invention, antigenic epitopes preferably contain a sequence of
at least 4,
at least 5, at least 6, at least 7, more preferably at least 8, at least 9, at
least 10, at least 11, at
least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at
least 30, at least 40, at
least 50, and, most preferably, between about 15 to about 30 amino acids.
Preferred
polypeptides comprising immunogenic or antigenic epitopes are at least 10, 15,
20, 25, 30,
35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues
in length.
Additional non-exclusive preferred antigenic epitopes include the antigenic
epitopes
disclosed herein, as well as portions thereof. Antigenic epitopes are useful,
for example, to
raise antibodies, including monoclonal antibodies, that specifically bind the
epitope.
Preferred antigenic epitopes include the antigenic epitopes disclosed herein,
as well as any
combination of two, three, four, five or more of these antigenic epitopes.
Antigenic epitopes
can be used as the target molecules in immunoassays. (See, for instance,
Wilson et al., Cell
37:767-778 (1984); Sutcliffe et al., Science 219:660-666 (1983)).
Similarly, immunogenic epitopes can be used, for example, to induce antibodies
according to methods well known in the art. (See, for instance, Sutcliffe et
al., supra; Wilson
et al., supra; Chow et al., Proc. Natl. Acad. Sci. USA 82:910-914; and Bittle
et al., J. Gen.
Virol. 66:2347-2354 (1985). Preferred immunogenic epitopes include the
immunogenic
epitopes disclosed herein, as well as any combination of two, three, four,
five or more of
these immunogenic epitopes. The polypeptides comprising one or more
immunogenic
epitopes may be presented for eliciting an antibody response together with a
Garner protein,
such as an albumin, to an animal system (such as rabbit or mouse), or, if the
polypeptide is of
sufficient length (at least about 25 amino acids), the polypeptide may be
presented without a
carrier. However, immunogenic epitopes comprising as few as 8 to 10 amino
acids have


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
33
been shown to be sufficient to raise antibodies capable of binding to, at the
very least, linear
epitopes in a denatured polypeptide (e.g., in Western blotting).
Epitope-bearing polypeptides of the present invention may be used to induce
antibodies according to methods well known in the art including, but not
limited to, in vivo
immunization, in vitro immunization, and phage display methods. See, e.g.,
Sutcliffe et al.,
supra; Wilson et al., supra, and Bittle et al., J. Gen. Virol., 66:2347-2354
(1985). If in vivo
immunization is used, animals may be immunized with free peptide; however,
anti-peptide
antibody titer may be boosted by coupling the peptide to a macromolecular
carrier, such as
keyhole limpet hemacyanin (KLH) or tetanus toxoid. For instance, peptides
containing
cysteine residues may be coupled to a carrier using a linker such as
maleimidobenzoyl- N-
hydroxysuccinimide ester (MBS), while other peptides may be coupled to
carriers using a
more general linking agent such as glutaraldehyde. Animals such as rabbits,
rats and mice
are immunized with either free or Garner- coupled peptides, for instance, by
intraperitoneal
and/or intradermal injection of emulsions containing about 100 pg of peptide
or Garner
protein and Freund's adjuvant or any other adjuvant known for stimulating an
immune
response. Several booster injections may be needed, for instance, at intervals
of about two
weeks, to provide a useful titer of anti-peptide antibody which can be
detected, for example,
by ELISA assay using free peptide adsorbed to a solid surface. The titer of
anti-peptide
antibodies in serum from an immunized animal may be increased by selection of
anti-peptide
antibodies, for instance, by adsorption to the peptide on a solid support and
elution of the
selected antibodies according to methods well known in the art.
As one of skill in the art will appreciate, and as discussed above, the
polypeptides of
the present invention and immunogenic and/or antigenic epitope fragments
thereof can be
fused to other polypeptide sequences. For example, the polypeptides of the
present invention
may be fused with the constant domain of immunoglobulins (IgA, IgE, IgG, IgM),
or
portions thereof (CH1, CH2, CH3, or any combination thereof and portions
thereof) resulting
in chimeric polypeptides. Such fusion proteins may facilitate purification and
may increase
half life in vivo. This has been shown 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. See, e.g., EP 394,827;
Traunecker et
al., Nature, 331:84-86 (1988). Enhanced delivery of an antigen across the
epithelial barrier to
the immune system has been demonstrated for antigens (e.g., insulin)
conjugated to an FcRn


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
34
binding partner such as IgG or Fc fragments (see, e.g., PCT Publications WO
96/22024 and
WO 99/04813). IgG Fusion proteins that have a disulfide-linked dimeric
structure due to
the IgG portion desulfide bonds have also been found to be more efficient in
binding and
neutralizing other molecules than monomeric polypeptides or fragments thereof
alone. See,
e.g., Fountoulakis et al., J. Biochem., 270:3958-3964 (1995).
Similarly, EP-A-O 464 533 (Canadian counterpart 2045869) discloses fusion
proteins
comprising various portions of constant region of immunoglobulin molecules
together with
another human protein or part thereof. In many cases, the Fc part in a fusion
protein is
beneficial in therapy and diagnosis, and thus can result in, for example,
improved
pharmacokinetic properties. (EP-A 0232 262.) Alternatively, deleting the Fc
part after the
fusion protein has been expressed, detected, and purified, may be desired. For
example, the
Fc portion may hinder therapy and diagnosis if the fusion protein is used as
an antigen for
immunizations. In drug discovery, for example, human proteins, such as hIL-5,
have been
fused with Fc portions for the purpose of high-throughput screening assays to
identify
antagonists of hIL-5. (See, D. Bennett et al., J. Molecular Recognition 8:52-
58 (1995); K.
Johanson et al., J. Biol. Chem. 270:9459-9471 (1995).)
Moreover, the polypeptides of the present invention can be fused to marker
sequences, such as a peptide which facilitates purification of the fused
polypeptide. In
preferred embodiments, the marker amino acid sequence is a hexa-histidine
peptide, such as
the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth,
CA,
91311 ), among others, many of which are commercially available. As described
in Gentz et
al., Proc. Natl. Acad. Sci. USA 86:821-824 (1989), for instance, hexa-
histidine provides for
convenient purification of the fusion protein. Another peptide tag useful for
purification, the
"HA" tag, corresponds to an epitope derived from the influenza hemagglutinin
protein.
(Wilson et al., Cell 37:767 (1984).)
Thus, any of these above fusions can be engineered using the polynucleotides
or the
polypeptides of the present invention.
Nucleic acids encoding the above epitopes can also be recombined with a gene
of
interest as an epitope tag (e.g., the hemagglutinin ("HA") tag or flag tag) to
aid in detection
and purification of the expressed polypeptide. For example, a system described
by
Janknecht et al. allows for the ready purification of non-denatured fusion
proteins expressed
in human cell lines (Janknecht et al., Proc. Natl. Acad. Sci. USA 88:8972- 897
(1991)). In


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
this system, the gene of interest is subcloned into a vaccinia recombination
plasmid such that
the open reading frame of the gene is translationally fused to an amino-
terminal tag
consisting of six histidine residues. The tag serves as a matrix binding
domain for the fusion
protein. Extracts from cells infected with the recombinant vaccinia virus are
loaded onto
5 Ni2+ nitriloacetic acid-agarose column and histidine-tagged proteins can be
selectively
eluted with imidazole-containing buffers.
Additional fusion proteins of the invention may be generated through the
techniques
of gene-shuffling, motif shuffling, exon-shuffling, and/or codon-shuffling
(collectively
referred to as "DNA shuffling"). DNA shuffling may be employed to modulate the
activities
10 of polypeptides of the invention, such methods can be used to generate
polypeptides with
altered activity, as well as agonists and antagonists of the polypeptides.
See, generally, U.S.
Patent Nos. 5,605,793; 5,811,238; 5,830,721; 5,834,252; and 5,837,458, and
Patten et al.,
Curr. Opinion Biotechnol. 8:724-33 (1997); Harayama, Trends Biotechnol.
16(2):76-82
(1998); Hansson, et al., J. Mol. Biol. 287:265-76 (1999); and Lorenzo and
Blasco,
15 Biotechniques 24(2):308- 13 (1998) (each of these patents and publications
are hereby
incorporated by reference in its entirety). In one embodiment, alteration of
polynucleotides
corresponding to SEQ ID NO:X and the polypeptides encoded by these
polynucleotides may
be achieved by DNA shuffling. DNA shuffling involves the assembly of two or
more DNA
segments by homologous or site-specific recombination to generate variation in
the
20 polynucleotide sequence. In another embodiment, polynucleotides of the
invention, or the
encoded polypeptides, may be altered by being subjected to random mutagenesis
by error-
prone PCR, random nucleotide insertion or other methods prior to
recombination. In another
embodiment, one or more components, motifs, sections, parts, domains,
fragments, etc., of a
polynucleotide encoding a polypeptide of the invention may be recombined with
one or more
25 components, motifs, sections, parts, domains, fragments, etc. of one or
more heterologous
molecules.
Polynucleotide and Polypeptide Variants
The invention also encompasses steroid hormone receptor variants. The present
30 invention is directed to variants of the polynucleotide sequence disclosed
in SEQ ID NO:X or
the complementary strand thereto, and/or the cDNA sequence contained in cDNA
plasmid:Z.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
36
The present invention also encompasses variants of the polypeptide sequence
disclosed in SEQ ID NO:Y, a polypeptide sequence encoded by the polynucleotide
sequence
in SEQ ID NO:X and/or a polypeptide sequence encoded by the cDNA in cDNA
plasmid:Z.
"Variant" refers to a polynucleotide or polypeptide differing from the
polynucleotide
or polypeptide of the present invention, but retaining properties thereof.
Generally, variants
are overall closely similar, and, in many regions, identical to the
polynucleotide or
polypeptide of the present invention.
Thus, one aspect of the invention provides an isolated nucleic acid molecule
comprising, or alternatively consisting of, a polynucleotide having a
nucleotide sequence
selected from the group consisting of : (a) a nucleotide sequence encoding a
steroid hormone
receptor polypeptide having an amino acid sequence as shown in the sequence
listing and
described in SEQ ID NO:X or the cDNA in cDNA plasmid:Z; (b) a nucleotide
sequence
encoding a mature steroid hormone receptor polypeptide having the amino acid
sequence as
shown in the sequence listing and described in SEQ ID NO:X or the cDNA in cDNA
plasmid:Z; (c) a nucleotide sequence encoding a biologically active fragment
of a steroid
hormone receptor polypeptide having an amino acid sequence shown in the
sequence listing
and described in SEQ ID NO:X or the cDNA in cDNA plasmid:Z; (d) a nucleotide
sequence
encoding an antigenic fragment of a steroid hormone receptor polypeptide
having an amino
acid sequence shown in the sequence listing and described in SEQ )D NO:X or
the cDNA in
cDNA plasmid:Z; (e) a nucleotide sequence encoding a steroid hormone receptor
polypeptide comprising the complete amino acid sequence encoded by a human
cDNA
plasmid contained in SEQ ID NO:X or the cDNA in cDNA plasmid:Z; (f) a
nucleotide
sequence encoding a mature steroid hormone receptor polypeptide having an
amino acid
sequence encoded by a human cDNA plasmid contained in SEQ ID NO:X or the cDNA
in
cDNA plasmid:Z; (g) a nucleotide sequence encoding a biologically active
fragment of a
steroid hormone receptor polypeptide having an amino acid sequence encoded by
a human
cDNA plasmid contained in SEQ ID NO:X or the cDNA in cDNA plasmid:Z; (h) a
nucleotide sequence encoding an antigenic fragment of a steroid hormone
receptor
polypeptide having an amino acid sequence encoded by a human cDNA plasmid
contained in
SEQ ID NO:X or the cDNA in cDNA plasmid:Z; (i) a nucleotide sequence
complementary to
any of the nucleotide sequences in (a), (b), (c), (d), (e), (f), (g), or (h),
above.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
37
The present invention is also directed to nucleic acid molecules which
comprise, or
alternatively consist of, a nucleotide sequence which is at least 80%, 85%,
90%, 95%, 96%,
97%, 98%, 99% or 100%, identical to, for example, any of the nucleotide
sequences in (a),
(b), (c), (d), (e), (f), (g), (h), or (i) above. Polypeptides encoded by these
nucleic acid
molecules are also encompassed by the invention. In another embodiment, the
invention
encompasses nucleic acid molecules which comprise, or alternatively, consist
of a
polynucleotide which hybridizes under stringent hybridization conditions, or
alternatively,
under lower stringency conditions, to a polynucleotide in (a), (b), (c), (d),
(e), (f), (g), (h), or
(i), above. Polynucleotides which hybridize to the complement of these nucleic
acid
molecules under stringent hybridization conditions or alternatively, under
lower stringency
conditions, are also encompassed by the invention, as are polypeptides encoded
by these
polynucleotides.
Another aspect of the invention provides an isolated nucleic acid molecule
comprising, or alternatively consisting of, a polynucleotide having a
nucleotide sequence
selected from the group consisting of : (a) a nucleotide sequence encoding a
steroid hormone
receptor polypeptide having an amino acid sequence as shown in the sequence
listing and
described in Table 1; (b) a nucleotide sequence encoding a mature steroid
hormone receptor
polypeptide having the amino acid sequence as shown in the sequence listing
and described
in Table 1; (c) a nucleotide sequence encoding a biologically active fragment
of a steroid
hormone receptor polypeptide having an amino acid sequence shown in the
sequence listing
and described in Table 1; (d) a nucleotide sequence encoding an antigenic
fragment of a
steroid hormone receptor polypeptide having an amino acid sequence shown in
the sequence
listing and described in Table 1; (e) a nucleotide sequence encoding a steroid
hormone
receptor polypeptide comprising the complete amino acid sequence encoded by a
human
cDNA in a cDNA plasmid contained in the ATCC Deposit and described in Table 1;
(f) a
nucleotide sequence encoding a mature steroid hormone receptor polypeptide
having an
amino acid sequence encoded by a human cDNA in a cDNA plasmid contained in the
ATCC
Deposit and described in Table 1; (g) a nucleotide sequence encoding a
biologically active
fragment of a steroid hormone receptor polypeptide having an amino acid
sequence encoded
by a human cDNA in a cDNA plasmid contained in the ATCC Deposit and described
in
Table 1; (h) a nucleotide sequence encoding an antigenic fragment of a steroid
hormone
receptor polypeptide having an amino acid sequence encoded by a human cDNA in
a cDNA


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
38
plasmid contained in the ATCC Deposit and described in Table 1; (i) a
nucleotide sequence
complementary to any of the nucleotide sequences in (a), (b), (c), (d), (e),
(f), (g), or (h),
above.
The present invention is also directed to nucleic acid molecules which
comprise, or
S alternatively consist of, a nucleotide sequence which is at least 80%, 85%,
90%, 95%, 96%,
97%, 98%, 99% or 100%, identical to, for example, any of the nucleotide
sequences in (a),
(b), (c), (d), (e), (f), (g), (h), or (i) above. Polypeptides encoded by these
nucleic acid
molecules are also encompassed by the invention. In another embodiment, the
invention
encompasses nucleic acid molecules which comprise, or alternatively, consist
of a
polynucleotide which hybridizes under stringent hybridization conditions, or
alternatively,
under lower stringency conditions, to a polynucleotide in (a), (b), (c), (d),
(e), (f), (g), (h), or
(i), above. Polynucleotides which hybridize to the complement of these nucleic
acid
molecules under stringent hybridization conditions or alternatively, under
lower stringency
conditions, are also encompassed by the invention, as are polypeptides encoded
by these
polynucleotides.
The present invention is also directed to polypeptides which comprise, or
alternatively
consist of, an amino acid sequence which is at least 80%, 85%, 90%, 95%, 96%,
97%, 98%,
99% or 100% identical to, for example, the polypeptide sequence shown in SEQ
ID NO:Y, a
polypeptide sequence encoded by the nucleotide sequence in SEQ ID NO:X, a
polypeptide
sequence encoded by the cDNA in cDNA plasmid:Z, and/or polypeptide fragments
of any of
these polypeptides (e.g., those fragments described herein). Polynucleotides
which hybridize
to the complement of the nucleic acid molecules encoding these polypeptides
under stringent
hybridization conditions or alternatively, under lower stringency conditions
are also
encompassed by the invention, as are polypeptides encoded by these
polynucleotides.
By a nucleic acid having a nucleotide sequence at least, for example, 95%
"identical"
to a reference nucleotide sequence of the present invention, it is intended
that the nucleotide
sequence of the nucleic acid is identical to the reference sequence except
that the nucleotide
sequence may include up to five point mutations per each 100 nucleotides of
the reference
nucleotide sequence encoding the polypeptide. In other words, to obtain a
nucleic acid
having a nucleotide sequence at least 95% identical to a reference nucleotide
sequence, up to
S% of the nucleotides in the reference sequence may be deleted or substituted
with another
nucleotide, or a number of nucleotides up to 5% of the total nucleotides in
the reference


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
39
sequence may be inserted into the reference sequence. The query sequence may
be an entire
sequence referred to in Table 1, the ORF (open reading frame), or any fragment
specified as
described herein.
As a practical matter, whether any particular nucleic acid molecule or
polypeptide is
at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide
sequence of
the present invention can be determined conventionally using known computer
programs. A
preferred method for determining the best overall match between a query
sequence (a
sequence of the present invention) and a subject sequence, also referred to as
a global
sequence alignment, can be determined using the FASTDB computer program based
on the
algorithm of Brutlag et al. (Comp. App. Biosci. 6:237-245 (1990)). In a
sequence alignment
the query and subject sequences are both DNA sequences. An RNA sequence can be
compared by converting U's to T's. The result of said global sequence
alignment is in
percent identity. Preferred parameters used in a FASTDB alignment of DNA
sequences to
calculate percent identiy are: Matrix=Unitary, k-tuple=4, Mismatch Penalty=1,
Joining
Penalty=30, Randomization Group Length=0, Cutoff Score=1, Gap Penalty=5, Gap
Size
Penalty 0.05, Window Size=S00 or the lenght of the subject nucleotide
sequence, whichever
is shorter.
If the subject sequence is shorter than the query sequence because of 5' or 3'
deletions,
not because of internal deletions, a manual correction must be made to the
results. This is
because the FASTDB program does not account for 5' and 3' truncations of the
subject
sequence when calculating percent identity. For subject sequences truncated at
the 5' or 3'
ends, relative to the query sequence, the percent identity is corrected by
calculating the
number of bases of the query sequence that are S' and 3' of the subject
sequence, which are
' not matched/aligned, as a percent of the total bases of the query sequence.
Whether a
nucleotide is matched/aligned is determined by results of the FASTDB sequence
alignment.
This percentage is then subtracted from the percent identity, calculated by
the above
FASTDB program using the specified parameters, to arrive at a final percent
identity score.
This corrected score is what is used for the purposes of the present
invention. Only bases
outside the S' and 3' bases of the subject sequence, as displayed by the
FASTDB alignment,
which are not matched/aligned with the query sequence, are calculated for the
purposes of
manually adjusting the percent identity score.
For example, a 90 base subject sequence is aligned to a 100 base query
sequence to


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
determine percent identity. The deletions occur at the 5' end of the subject
sequence and
therefore, the FASTDB alignment does not show a matched/alignment of the first
10 bases at
5' end. The 10 unpaired bases represent 10% of the sequence (number of bases
at the 5' and
3' ends not matched/total number of bases in the query sequence) so 10% is
subtracted from
S the percent identity score calculated by the FASTDB program. If the
remaining 90 bases
were perfectly matched the final percent identity would be 90%. In another
example, a 90
base subject sequence is compared with a 100 base query sequence. This time
the deletions
are internal deletions so that there are no bases on the S' or 3' of the
subject sequence which
are not matched/aligned with the query. In this case the percent identity
calculated by
10 FASTDB is not manually corrected. Once again, only bases 5' and 3' of the
subject sequence
which are not matched/aligned with the query sequence are manually corrected
for. No other
manual corrections are to made for the purposes of the present invention.
By a polypeptide having an amino acid sequence at least, for example, 95%
"identical" to a query amino acid sequence of the present invention, it is
intended that the
15 amino acid sequence of the subject polypeptide is identical to the query
sequence except that
the subject polypeptide sequence may include up to five amino acid alterations
per each 100
amino acids of the query amino acid sequence. In other words, to obtain a
polypeptide
having an amino acid sequence at least 95% identical to a query amino acid
sequence, up to
5% of the amino acid residues in the subject sequence may be inserted,
deleted, (indels) or
20 substituted with another amino acid. These alterations of the reference
sequence may occur
at the amino or carboxy terminal positions of the reference amino acid
sequence or anywhere
between those terminal positions, interspersed either individually among
residues in the
reference sequence or in one or more contiguous groups within the reference
sequence.
As a practical matter, whether any particular polypeptide is at least 80%,
85%, 90%,
25 95%, 96%, 97%, 98% or 99% identical to, for instance, the amino acid
sequence referred to
in Table 1 or a fragment thereof, the amino acid sequence encoded by the
nucleotide
sequence in SEQ ID NO:X or a fragment thereof, or to the amino acid sequence
encoded by
the cDNA in cDNA plasmid:Z, or a fragment thereof, can be determined
conventionally
using known computer programs. A preferred method for determing the best
overall match
30 between a query sequence (a sequence of the present invention) and a
subject sequence, also
referred to as a global sequence alignment, can be determined using the FASTDB
computer
program based on the algorithm of Brutlag et al. (Comp. App. Biosci.6:237-
245(1990)). In a


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
41
sequence alignment the query and subject sequences are either both nucleotide
sequences or
both amino acid sequences. The result of said global sequence alignment is in
percent
identity. Preferred parameters used in a FASTDB amino acid alignment are:
Matrix=PAM
0, k-tuple=2, Mismatch Penalty=1, Joining Penalty=20, Randomization Group
Length=0,
Cutoff Score=1, Window Size=sequence length, Gap Penalty=5, Gap Size
Penalty=0.05,
Window Size=500 or the length of the subject amino acid sequence, whichever is
shorter.
If the subject sequence is shorter than the query sequence due to N- or C-
terminal
deletions, not because of internal deletions, a manual correction must be made
to the results.
This is because the FASTDB program does not account for N- and C-terminal
truncations of
the subject sequence when calculating global percent identity. For subject
sequences
truncated at the N- and C-termini, relative to the query sequence, the percent
identity is
corrected by calculating the number of residues of the query sequence that are
N- and C-
terminal of the subject sequence, which are not matched/aligned with a
corresponding subject
residue, as a percent of the total bases of the query sequence. Whether a
residue is
matched/aligned is determined by results of the FASTDB sequence alignment.
This
percentage is then subtracted from the percent identity, calculated by the
above FASTDB
program using the specified parameters, to arrive at a final percent identity
score. This final
percent identity score is what is used for the purposes of the present
invention. Only residues
to the N- and C-termini of the subject sequence, which are not matched/aligned
with the
query sequence, are considered for the purposes of manually adjusting the
percent identity
score. That is, only query residue positions outside the farthest N- and C-
terminal residues
of the subject sequence.
For example, a 90 amino acid residue subject sequence is aligned with a 100
residue
query sequence to determine percent identity. The deletion occurs at the N-
terminus of the
subject sequence and therefore, the FASTDB alignment does not show a
matching/alignment
of the first 10 residues at the N-terminus. The 10 unpaired residues represent
10% of the
sequence (number of residues at the N- and C- termini not matched/total number
of residues
in the query sequence) so 10% is subtracted from the percent identity score
calculated by the
FASTDB program. If the remaining 90 residues were perfectly matched the final
percent
identity would be 90%. In another example, a 90 residue subject sequence is
compared with
a 100 residue query sequence. This time the deletions are internal deletions
so there are no
residues at the N- or C-termini of the subject sequence which are not
matched/aligned with


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
42
the query. In this case the percent identity calculated by FASTDB is not
manually corrected.
Once again, only residue positions outside the N- and C-terminal ends of the
subj ect
sequence, as displayed in the FASTDB alignment, which are not matched/aligned
with the
query sequnce are manually corrected for. No other manual corrections are to
made for the
purposes of the present invention.
The variants may contain alterations in the coding regions, non-coding
regions, or
both. Especially preferred are polynucleotide variants containing alterations
which produce
silent substitutions, additions, or deletions, but do not alter the properties
or activities of the
encoded polypeptide. Nucleotide variants produced by silent substitutions due
to the
degeneracy of the genetic code are preferred. Moreover, variants in which less
than 50, less
than 40, less than 30, less than 20, less than 10, or 5-50, 5-25, S-10, 1-5,
or 1-2 amino acids
are substituted, deleted, or added in any combination are also preferred.
Polynucleotide
variants can be produced for a variety of reasons, e.g., to optimize codon
expression for a
particular host (change codons in the human mRNA to those preferred by a
bacterial host
such as E. coli).
Naturally occurring variants are called "allelic variants," and refer to one
of several
alternate forms of a gene occupying a given locus on a chromosome of an
organism. (Genes
II, Lewin, B., ed., John Wiley & Sons, New York (1985).) These allelic
variants can vary at
either the polynucleotide and/or polypeptide level and are included in the
present invention.
Alternatively, non-naturally occurnng variants may be produced by mutagenesis
techniques
or by direct synthesis.
Using known methods of protein engineering and recombinant DNA technology,
variants may be generated to improve or alter the characteristics of the
polypeptides of the
present invention. For instance, as discussed herein, one or more amino acids
can be deleted
from the N-terminus or C-terminus of the polypeptide of the present invention
without
substantial loss of biological function. The authors of Ron et al., J. Biol.
Chem. 268: 2984
2988 (1993), reported variant KGF proteins having heparin binding activity
even after
deleting 3, 8, or 27 amino-terminal amino acid residues. Similarly, Interferon
gamma
exhibited up to ten times higher activity after deleting 8-10 amino acid
residues from the
carboxy terminus of this protein. (Dobeli et al., J. Biotechnology 7:199-216
(1988).)
Moreover, ample evidence demonstrates that variants often retain a biological
activity
similar to that of the naturally occurnng protein. For example, Gayle and
coworkers (J. Biol.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
43
Chem 268:22105-22111 (1993)) conducted extensive mutational analysis of human
cytokine
IL-la. They used random mutagenesis to generate over 3,500 individual IL-la
mutants that
averaged 2.5 amino acid changes per variant over the entire length of the
molecule. Multiple
mutations were examined at every possible amino acid position. The
investigators found that
"[m]ost of the molecule could be altered with little effect on either [binding
or biological
activity]." (See, Abstract.) In fact, only 23 unique amino acid sequences, out
of more than
3,500 nucleotide sequences examined, produced a protein that significantly
differed in
activity from wild-type.
Furthermore, as discussed herein, even if deleting one or more amino acids
from the
N-terminus or C-terminus of a polypeptide results in modification or loss of
one or more
biological functions, other biological activities may still be retained. For
example, the ability
of a deletion variant to induce and/or to bind antibodies which recognize the
secreted form
will likely be retained when less than the majority of the residues of the
secreted form are
removed from the N-terminus or C-terminus. Whether a particular polypeptide
lacking N- or
1 S C-terminal residues of a protein retains such immunogenic activities can
readily be
determined by routine methods described herein and otherwise known in the art.
Thus, the invention further includes polypeptide variants which show a
functional
activity (e.g. biological activity) of the polypeptide of the invention, of
which they are a
variant. Such variants include deletions, insertions, inversions, repeats, and
substitutions
selected according to general rules known in the art so as have little effect
on activity.
The present application is directed to nucleic acid molecules at least 80%,
85%, 90%,
95%, 96%, 97%, 98%, 99% or 100% identical to the nucleic acid sequences
disclosed herein,
(e.g., encoding a polypeptide having the amino acid sequence of an N and/or C
terminal
deletion), irrespective of whether they encode a polypeptide having functional
activity. This
is because even where a particular nucleic acid molecule does not encode a
polypeptide
having functional activity, one of skill in the art would still know how to
use the nucleic acid
molecule, for instance, as a hybridization probe or a polymerase chain
reaction (PCR) primer.
Uses of the nucleic acid molecules of the present invention that do not encode
a polypeptide
having functional activity include, inter alia, (1) isolating a gene or
allelic or splice variants
thereof in a cDNA library; (2) in situ hybridization (e.g., "FISH") to
metaphase chromosomal
spreads to provide precise chromosomal location of the gene, as described in
Verma et al.,
Human Chromosomes: A Manual of Basic Techniques, Pergamon Press, New York
(1988);


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
44
and (3) Northern Blot analysis for detecting mRNA expression in specific
tissues.
Preferred, however, are nucleic acid molecules having sequences at least 80%,
85%,
90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleic acid sequences
disclosed
herein, which do, in fact, encode a polypeptide having functional activity of
a polypeptide of
the invention.
Of course, due to the degeneracy of the genetic code, one of ordinary skill in
the art
will immediately recognize that a large number of the nucleic acid molecules
having a
sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical
to, for
example, the nucleic acid sequence of the cDNA in cDNA plasmid:Z, the nucleic
acid
sequence referred to in Table 1 (SEQ ID NO:X), or fragments thereof, will
encode
polypeptides "having functional activity." In fact, since degenerate variants
of any of these
nucleotide sequences all encode the same polypeptide, in many instances, this
will be clear to
the skilled artisan even without performing the above described comparison
assay. It will be
further recognized in the art that, for such nucleic acid molecules that are
not degenerate
variants, a reasonable number will also encode a polypeptide having functional
activity. This
is because the skilled artisan is fully aware of amino acid substitutions that
are either less
likely or not likely to significantly effect protein function (e.g., replacing
one aliphatic amino
acid with a second aliphatic amino acid), as further described below.
For example, guidance concerning how to make phenotypically silent amino acid
substitutions is provided in Bowie et al., "Deciphering the Message in Protein
Sequences:
Tolerance to Amino Acid Substitutions," Science 247:1306-1310 (1990), wherein
the authors
indicate that there are two main strategies for studying the tolerance of an
amino acid
sequence to change.
The first strategy exploits the tolerance of amino acid substitutions by
natural
selection during the process of evolution. By comparing amino acid sequences
in different
species, conserved amino acids can be identified. These conserved amino acids
are likely
important for protein function. In contrast, the amino acid positions where
substitutions have
been tolerated by natural selection indicates that these positions are not
critical for protein
function. Thus, positions tolerating amino acid substitution could be modified
while still
maintaining biological activity of the protein.
The second strategy uses genetic engineering to introduce amino acid changes
at
specific positions of a cloned gene to identify regions critical for protein
function. For


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
example, site directed mutagenesis or alanine-scanning mutagenesis
(introduction of single
alanine mutations at every residue in the molecule) can be used. (Cunningham
and Wells,
Science 244:1081-1085 (1989).) The resulting mutant molecules can then be
tested for
biological activity.
5 As the authors state, these two strategies have revealed that proteins are
surprisingly
tolerant of amino acid substitutions. The authors further indicate which amino
acid changes
are likely to be permissive at certain amino acid positions in the protein.
For example, most
buried (within the tertiary structure of the protein) amino acid residues
require nonpolar side
chains, whereas few features of surface side chains are generally conserved.
Moreover,
10 tolerated conservative amino acid substitutions involve replacement of the
aliphatic or
hydrophobic amino acids Ala, Val, Leu and Ile; replacement of the hydroxyl
residues Ser and
Thr; replacement of the acidic residues Asp and Glu; replacement of the amide
residues Asn
and Gln, replacement of the basic residues Lys, Arg, and His; replacement of
the aromatic
residues Phe, Tyr, and Trp, and replacement of the small-sized amino acids
Ala, Ser, Thr,
15 Met, and Gly. Besides conservative amino acid substitution, variants of the
present invention
include (i) substitutions with one or more of the non-conserved amino acid
residues, where
the substituted amino acid residues may or may not be one encoded by the
genetic code, or
(ii) substitution with one or more of amino acid residues having a substituent
group, or (iii)
fusion of the mature polypeptide with another compound, such as a compound to
increase the
20 stability and/or solubility of the polypeptide (for example, polyethylene
glycol), or (iv) fusion
of the polypeptide with additional amino acids, such as, for example, an IgG
Fc fusion region
peptide, or leader or secretory sequence, or a sequence facilitating
purification. Such variant
polypeptides are deemed to be within the scope of those skilled in the art
from the teachings
herein.
25 For example, polypeptide variants containing amino acid substitutions of
charged
amino acids with other charged or neutral amino acids may produce proteins
with improved
characteristics, such as less aggregation. Aggregation of pharmaceutical
formulations both
reduces activity and increases clearance due to the aggregate's immunogenic
activity.
(Pinckard et al., Clin. Exp. Immunol. 2:331-340 (1967); Robbins et al.,
Diabetes 36: 838-845
30 (1987); Cleland et al., Crit. Rev. Therapeutic Drug Carner Systems 10:307-
377 (1993).)
A further embodiment of the invention relates to a polypeptide which comprises
the
amino acid sequence of a polypeptide having an amino acid sequence which
contains at least


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
46
one amino acid substitution, but not more than 50 amino acid substitutions,
even more
preferably, not more than 40 amino acid substitutions, still more preferably,
not more than 30
amino acid substitutions, and still even more preferably, not more than 20
amino acid
substitutions. Of course it is highly preferable for a polypeptide to have an
amino acid
sequence which comprises the amino acid sequence of a polypeptide of SEQ ID
NO:Y, an
amino acid sequence encoded by SEQ ID NO:X, and/or the amino acid sequence
encoded by
the cDNA in cDNA plasmid:Z which contains, in order of ever-increasing
preference, at least
one, but not more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid
substitutions. In specific
embodiments, the number of additions, substitutions, and/or deletions in the
amino acid
sequence of SEQ ID NO:Y or fragments thereof (e.g., the mature form and/or
other
fragments described herein), an amino acid sequence encoded by SEQ ID NO:X or
fragments
thereof, and/or the amino acid sequence encoded by cDNA plasmid:Z or fragments
thereof, is
1-5, 5-10, 5-25, 5-50, 10-50 or SO-150, conservative amino acid substitutions
are preferable.
As discussed herein, any polypeptide of the present invention can be used to
generate fusion
1 S proteins. For example, the polypeptide of the present invention, when
fused to a second
protein, can be used as an antigenic tag. Antibodies raised against the
polypeptide of the
present invention can be used to indirectly detect the second protein by
binding to the
polypeptide. Moreover, because secreted proteins target cellular locations
based on
trafficking signals, polypeptides of the present invention which are shown to
be secreted can
be used as targeting molecules once fused to other proteins.
Examples of domains that can be fused to polypeptides of the present invention
include not only heterologous signal sequences, but also other heterologous
functional
regions. The fusion does not necessarily need to be direct, but may occur
through linker
sequences.
In certain preferred embodiments, proteins of the invention comprise fusion
proteins
wherein the polypeptides are N and/or C- terminal deletion mutants. In
preferred
embodiments, the application is directed to nucleic acid molecules at least
80%, 85%, 90%,
95%, 96%, 97%, 98% or 99% identical to the nucleic acid sequences encoding
polypeptides
having the amino acid sequence of the specific N- and C-terminal deletions
mutants.
Polynucleotides encoding these polypeptides, including fragments and/or
variants, are also
encompassed by the invention.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
47
Moreover, fusion proteins may also be engineered to improve characteristics of
the
polypeptide of the present invention. For instance, a region of additional
amino acids,
particularly charged amino acids, may be added to the N-terminus of the
polypeptide to
improve stability and persistence during purification from the host cell or
subsequent
handling and storage. Also, peptide moieties may be added to the polypeptide
to facilitate
purification. Such regions may be removed prior to final preparation of the
polypeptide. The
addition of peptide moieties to facilitate handling of polypeptides are
familiar and routine
techniques in the art.
As one of skill in the art will appreciate, polypeptides of the present
invention of the
present invention and the epitope-bearing fragments thereof described above
can be
combined with heterologous polypeptide sequences. For example, the
polypeptides of the
present invention may be fused with heterologous polypeptide sequences, for
example, the
polypeptides of the present invention may be fused with the constant domain of
immunoglobulins (IgA, IgE, IgG, IgM) or portions thereof (CH1, CH2, CH3, and
any
1 S combination thereof, including both entire domains and portions thereof),
resulting in
chimeric polypeptides. These fusion proteins facilitate purification and show
an increased
half life in vivo. One reported example describes 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. (EP A 394,827; Traunecker
et al.,
Nature 331:84-86 (1988).) Fusion proteins having disulfide-linked dimeric
structures (due to
the IgG) can also be more efficient in binding and neutralizing other
molecules, than the
monomeric protein or protein fragment alone. (Fountoulakis et al., J. Biochem.
270:3958-
3964 (1995).)
Vectors, Host Cells, and Protein Production
The present invention also relates to vectors containing the polynucleotide of
the
present invention, host cells, and the production of polypeptides by
recombinant techniques.
The vector may be, for example, a phage, plasmid, viral, or retroviral vector.
Retroviral
vectors may be replication competent or replication defective. In the latter
case, viral
propagation generally will occur only in complementing host cells.
The polynucleotides of the invention may be joined to a vector containing a
selectable
marker for propagation in a host. Generally, a plasmid vector is introduced in
a precipitate,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
48
such as a calcium phosphate precipitate, or in a complex with a charged lipid.
If the vector is
a virus, it may be packaged in vitro using an appropriate packaging cell line
and then
transduced into host cells.
The polynucleotide insert should be operatively linked to an appropriate
promoter,
such as the phage lambda PL promoter, the E. coli lac, trp, phoA and tac
promoters, the SV40
early and late promoters and promoters of retroviral LTRs, to name a few.
Other suitable
promoters will be known to the skilled artisan. 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 transcripts expressed
by the constructs
will preferably include a translation initiating codon at the beginning and a
termination codon
(UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be
translated.
As indicated, the expression vectors will preferably include at least one
selectable
marker. Such markers include dihydrofolate reductase, 6418 or neomycin
resistance for
eukaryotic cell culture and tetracycline, kanamycin or ampicillin resistance
genes for
culturing in E. coli and other bacteria. 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 (e.g., Saccharomyces
cerevisiae or Pichia
pastoris (ATCC Accession No. 201178)); insect cells such as Drosophila S2 and
Spodoptera
Sf~3 cells; animal cells such as CHO, COS, 293, and Bowes melanoma cells; and
plant cells.
Appropriate culture mediums and conditions for the above-described host cells
are known in
the art.
Among vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9,
available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pNHBA,
pNHl6a,
pNHl8A, pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a,
pKK223-
3, pKK233-3, pDR540, pRITS available from Pharmacia Biotech, Inc. Among
preferred
eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTI and pSG available from
Stratagene; and pSVK3, pBPV, ~pMSG and pSVL available from Pharmacia.
Preferred
expression vectors for use in yeast systems include, but are not limited to
pYES2, pYDI,
pTEFl/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZaIph, pPIC9, pPIC3.5, pHIL-D2, pHIL-
S1,
pPIC3.5K, pPIC9K, and PA0815 (all available from Invitrogen, Carlbad, CA).
Other suitable
vectors will be readily apparent to the skilled artisan.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
49
Introduction of the construct into the 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 Davis et al., Basic Methods In
Molecular Biology
S (1986). It is specifically contemplated that the polypeptides of the present
invention may in
fact be expressed by a host cell lacking a recombinant vector.
A polypeptide of this invention 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,
hydroxylapatite
chromatography and lectin chromatography. Most preferably, high performance
liquid
chromatography ("HPLC") is employed for purification.
Polypeptides of the present invention can also be recovered from: products
purified
from natural sources, including bodily fluids, tissues and cells, whether
directly isolated or
1 S cultured; products of chemical synthetic procedures; and products produced
by recombinant
techniques from a prokaryotic or eukaryotic 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
may be
glycosylated or may be non-glycosylated. In addition, polypeptides of the
invention may also
include an initial modified methionine residue, in some cases as a result of
host-mediated
processes. Thus, it is well known in the art that the N-terminal methionine
encoded by the
translation initiation codon generally is removed with high efficiency from
any protein after
translation in all eukaryotic cells. While the N-terminal methionine on most
proteins also is
efficiently removed in most prokaryotes, for some proteins, this prokaryotic
removal process
is inefficient, depending on the nature of the amino acid to which the N-
terminal methionine
is covalently linked.
In one embodiment, the yeast Pichia pastoris is used to express polypeptides
of the
invention in a eukaryotic system. Pichia pastoris is a methylotrophic yeast
which can
metabolize methanol as its sole carbon source. A main step in the methanol
metabolization
pathway is the oxidation of methanol to formaldehyde using O2. This reaction
is catalyzed by
the enzyme alcohol oxidase. In order to metabolize methanol as its sole carbon
source,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
Pichia pastoris must generate high levels of alcohol oxidase due, in part, to
the relatively low
affinity of alcohol oxidase for OZ. Consequently, in a growth medium depending
on
methanol as a main carbon source, the promoter region of one of the two
alcohol oxidase
genes (AOXI ) is highly active. In the presence of methanol, alcohol oxidase
produced from
the AOXI gene comprises up to approximately 30% of the total soluble protein
in Pichia
pastoris. See, Ellis, S.B., et al., Mol. Cell. Biol. 5:1111-21 (1985); Koutz,
P.J, et al., Yeast
5:167-77 (1989); Tschopp, J.F., et al., Nucl. Acids Res. 15:3859-76 (1987).
Thus, a
heterologous coding sequence, such as, for example, a polynucleotide of the
present
invention, under the transcriptional regulation of all or part of the AOXl
regulatory sequence
10 is expressed at exceptionally high levels in Pichia yeast grown in the
presence of methanol.
In one example, the plasmid vector pPIC9K is used to express DNA encoding a
polypeptide of the invention, as set forth herein, in a Pichea yeast system
essentially as
described in "Pichia Protocols: Methods in Molecular Biology," D.R. Higgins
and J. Cregg,
eds. The Humana Press, Totowa, NJ, 1998. This expression vector allows
expression and
15 secretion of a polypeptide of the invention by virtue of the strong AOXl
promoter linked to
the Pichia pastoris alkaline phosphatase (PHO) secretory signal peptide (i.e.,
leader) located
upstream of a multiple cloning site.
Many other yeast vectors could be used in place of pPIC9K, such as, pYES2,
pYDl,
pTEFI/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalpha, pPIC9, pPIC3.5, pHIL-D2, pHIL-
Sl,
20 pPIC3.5K, and PA0815, as one skilled in the art would readily appreciate,
as long as the
proposed expression construct provides appropriately located signals for
transcription,
translation, secretion (if desired), and the like, including an in-frame AUG
as required.
In another embodiment, high-level expression of a heterologous coding
sequence,
such as, for example, a polynucleotide of the present invention, may be
achieved by cloning
25 the heterologous polynucleotide of the invention into an expression vector
such as, for
example, pGAPZ or pGAPZalpha, and growing the yeast culture in the absence of
methanol.
In addition to encompassing host cells containing the vector constructs
discussed
herein, the invention also encompasses primary, secondary, and immortalized
host cells of
vertebrate origin, particularly mammalian origin, that have been engineered to
delete or
30 replace endogenous genetic material (e.g., coding sequence), and/or to
include genetic


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
51
material (e.g., heterologous polynucleotide sequences) that is operably
associated with
polynucleotides of the invention, and which activates, alters, and/or
amplifies endogenous
polynucleotides. For example, techniques known in the art may be used to
operably associate
heterologous control regions (e.g., promoter and/or enhancer) and endogenous
polynucleotide sequences via homologous recombination (see, e.g., U.S. Patent
No.
5,641,670, issued June 24, 1997; International Publication No. WO 96/29411,
published
September 26, 1996; International Publication No. WO 94/12650, published
August 4, 1994;
Koller et al., Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); and Zijlstra et
al., Nature
342:435-438 (1989), the disclosures of each of which are incorporated by
reference in their
entireties).
In addition, polypeptides of the invention can be chemically synthesized using
techniques known in the art (e.g., see Creighton, 1983, Proteins: Structures
and Molecular
Principles, W.H. Freeman & Co., N.Y., and Hunkapiller et al., Nature, 310:105-
111 (1984)).
For example, a polypeptide corresponding to a fragment of a polypeptide can be
synthesized
by use of a peptide synthesizer. Furthermore, if desired, nonclassical amino
acids or
chemical amino acid analogs can be introduced as a substitution or addition
into the
polypeptide sequence. Non-classical amino acids include, but are not limited
to, to the D-
isomers of the common amino acids, 2,4-diaminobutyric acid, a-amino isobutyric
acid, 4-
aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6-amino hexanoic
acid, Aib,
2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine,
norvaline,
hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-
butylglycine, t-
butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids,
designer
amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na-methyl
amino acids,
and amino acid analogs in general. Furthermore, the amino acid can be D
(dextrorotary) or L
(levorotary).
The invention encompasses polypeptides of the present invention which are
differentially modified during or after translation, e.g., by glycosylation,
acetylation,
phosphorylation, amidation, derivatization by known protecting/blocking
groups, proteolytic
cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any
of numerous
chemical modifications may be carried out by known techniques, including but
not limited, to
specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain,
V8
protease, NaBH4; acetylation, formylation, oxidation, reduction; metabolic
synthesis in the


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
52
presence of tunicamycin; etc.
Additional post-translational modifications encompassed by the invention
include, for
example, e.g., N-linked or O-linked carbohydrate chains, processing of N-
terminal or
C-terminal ends), attachment of chemical moieties to the amino acid backbone,
chemical
modifications of N-linked or O-linked carbohydrate chains, and addition or
deletion of an
N-terminal methionine residue as a result of procaryotic host cell expression.
The
polypeptides may also be modified with a detectable label, such as an
enzymatic, fluorescent,
isotopic or affinity label to allow for detection and isolation of the
protein.
Also provided by the invention are chemically modified derivatives of the
polypeptides of the invention which may provide additional advantages such as
increased
solubility, stability and circulating time of the polypeptide, or decreased
immunogenicity (see
U.S. Patent No. 4,179,337). The chemical moieties for derivitization may be
selected from
water soluble polymers such as polyethylene glycol, ethylene glycol/propylene
glycol
copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like.
The
polypeptides may be modified at random positions within the molecule, or at
predetermined
positions within the molecule and may include one, two, three or more attached
chemical
moieties.
The polymer may be of any molecular weight, and may be branched or unbranched.
For polyethylene glycol, the preferred molecular weight is between about 1 kDa
and about
100 kDa (the term "about" indicating that in preparations of polyethylene
glycol, some
molecules will weigh more, some less, than the stated molecular weight) for
ease in handling
and manufacturing. Other sizes may be used, depending on the desired
therapeutic profile
(e.g., the duration of sustained release desired, the effects, if any on
biological activity, the
ease in handling, the degree or lack of antigenicity and other known effects
of the
polyethylene glycol to a therapeutic protein or analog).
The polyethylene glycol molecules (or other chemical moieties) should be
attached to
the protein with consideration of effects on functional or antigenic domains
of the protein.
There are a number of attachment methods available to those skilled in the
art, e.g., EP 0 401
384, herein incorporated by reference (coupling PEG to G-CSF), see also Malik
et al., Exp.
Hematol. 20:1028-1035 (1992) (reporting pegylation of GM-CSF using tresyl
chloride). For
example, polyethylene glycol may be covalently bound through amino acid
residues via a
reactive group, such as, a free amino or carboxyl group. Reactive groups are
those to which


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
53
an activated polyethylene glycol molecule may be bound. The amino acid
residues having a
free amino group may include lysine residues and the N-terminal amino acid
residues; those
having a free carboxyl group may include aspartic acid residues glutamic acid
residues and
the C-terminal amino acid residue. Sulfhydryl groups may also be used as a
reactive group
for attaching the polyethylene glycol molecules. Preferred for therapeutic
purposes is
attachment at an amino group, such as attachment at the N-terminus or lysine
group.
One may specifically desire proteins chemically modified at the N-terminus.
Using
polyethylene glycol as an illustration of the present composition, one may
select from a
variety of polyethylene glycol molecules (by molecular weight, branching,
etc.), the
proportion of polyethylene glycol molecules to protein (polypeptide) molecules
in the
reaction mix, the type of pegylation reaction to be performed, and the method
of obtaining
the selected N-terminally pegylated protein. The method of obtaining the N-
terminally
pegylated preparation (i.e., separating this moiety from other monopegylated
moieties if
necessary) may be by purification of the N-terminally pegylated material from
a population
of pegylated protein molecules. Selective proteins chemically modified at the
N-terminus
modification may be accomplished by reductive alkylation which exploits
differential
reactivity of different types of primary amino groups (lysine versus the N-
terminal) available
for derivatization in a particular protein. Under the appropriate reaction
conditions,
substantially selective derivatization of the protein at the N-terminus with a
carbonyl group
containing polymer is achieved.
The polypeptides of the invention may be in monomers or multimers (i.e.,
dimers,
trimers, tetramers and higher multimers). Accordingly, the present invention
relates to
monomers and multimers of the polypeptides of the invention, their
preparation, and
compositions (preferably, Therapeutics) containing them. In specific
embodiments, the
polypeptides of the invention are monomers, dimers, trimers or tetramers. In
additional
embodiments, the multimers of the invention are at least dimers, at least
trimers, or at least
tetramers.
Multimers encompassed by the invention may be homomers or heteromers. As used
herein, the term homomer, refers to a multimer containing only polypeptides
corresponding
to the amino acid sequence of SEQ m NO:Y or an amino acid sequence encoded by
SEQ ID
NO:X or the complement of SEQ ID NO:X, and/or an amino acid sequence encoded
by
cDNA plasmid:Z (including fragments, variants, splice variants, and fusion
proteins,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
54
corresponding to these as described herein). These homomers may contain
polypeptides
having identical or different amino acid sequences. In a specific embodiment,
a homomer of
the invention is a multimer containing only polypeptides having an identical
amino acid
sequence. In another specific embodiment, a homomer of the invention is a
multimer
S containing polypeptides having different amino acid sequences. In specific
embodiments, the
multimer of the invention is a homodimer (e.g., containing polypeptides having
identical or
different amino acid sequences) or a homotrimer (e.g., containing polypeptides
having
identical and/or different amino acid sequences). In additional embodiments,
the homomeric
multimer of the invention is at least a homodimer, at least a homotrimer, or
at least a
homotetramer.
As used herein, the term heteromer refers to a multimer containing one or more
heterologous polypeptides (i.e., polypeptides of different proteins) in
addition to the
polypeptides of the invention. In a specific embodiment, the multimer of the
invention is a
heterodimer, a heterotrimer, or a heterotetramer. In additional embodiments,
the heteromeric
1 S multimer of the invention is at least a heterodimer, at least a
heterotrimer, or at least a
heterotetramer.
Multimers of the invention may be the result of hydrophobic, hydrophilic,
ionic
and/or covalent associations and/or may be indirectly linked, by for example,
liposome
formation. Thus, in one embodiment, multimers of the invention, such as, for
example,
homodimers or homotrimers, are formed when polypeptides of the invention
contact one
another in solution. In another embodiment, heteromultimers of the invention,
such as, for
example, heterotrimers or heterotetramers, are formed when polypeptides of the
invention
contact antibodies to the polypeptides of the invention (including antibodies
to the
heterologous polypeptide sequence in a fusion protein of the invention) in
solution. In other
embodiments, multimers of the invention are formed by covalent associations
with and/or
between the polypeptides of the invention. Such covalent associations may
involve one or
more amino acid residues contained in the polypeptide sequence (e.g., that
recited in SEQ ID
NO:Y, or contained in a polypeptide encoded by SEQ ID NO:X, and/or the cDNA
plasmid:Z). In one instance, the covalent associations are cross-linking
between cysteine
residues located within the polypeptide sequences which interact in the native
(i.e., naturally
occurnng) polypeptide. In another instance, the covalent associations are the
consequence of
chemical or recombinant manipulation. Alternatively, such covalent
associations may


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
SS
involve one or more amino acid residues contained in the heterologous
polypeptide sequence
in a fusion protein. In one example, covalent associations are between the
heterologous
sequence contained in a fusion protein of the invention (see, e.g., US Patent
Number
5,478,925). In a specific example, the covalent associations are between the
heterologous
S sequence contained in a Fc fusion protein of the invention (as described
herein). In another
specific example, covalent associations of fusion proteins of the invention
are between
heterologous polypeptide sequence from another protein that is capable of
forming covalently
associated multimers, such as for example, osteoprotegerin (see, e.g.,
International
Publication NO: WO 98/49305, the contents of which are herein incorporated by
reference in
its entirety). In another embodiment, two or more polypeptides of the
invention are joined
through peptide linkers. Examples include those peptide linkers described in
U.S. Pat. No.
5,073,627 (hereby incorporated by reference). Proteins comprising multiple
polypeptides of
the invention separated by peptide linkers may be produced using conventional
recombinant
DNA technology.
Another method for preparing multimer polypeptides of the invention involves
use of
polypeptides of the invention fused to a leucine zipper or isoleucine zipper
polypeptide
sequence. Leucine zipper and isoleucine zipper domains are polypeptides that
promote
multimerization of the proteins in which they are found. Leucine zippers were
originally
identified in several DNA-binding proteins (Landschulz et al., Science
240:1759, (1988)),
and have since been found in a variety of different proteins. Among the known
leucine
zippers are naturally occurring peptides and derivatives thereof that dimerize
or trimerize.
Examples of leucine zipper domains suitable for producing soluble multimeric
proteins of the
invention are those described in PCT application WO 94/10308, hereby
incorporated by
reference. Recombinant fusion proteins comprising a polypeptide of the
invention fused to a
polypeptide sequence that dimerizes or trimerizes in solution are expressed in
suitable host
cells, and the resulting soluble multimeric fusion protein is recovered from
the culture
supernatant using techniques known in the art.
Trimeric polypeptides of the invention may offer the advantage of enhanced
biological activity. Preferred leucine zipper moieties and isoleucine moieties
are those that
preferentially form trimers. One example is a leucine zipper derived from lung
surfactant
protein D (SPD), as described in Hoppe et al. (FEBS Letters 344:191, (1994))
and in U.S.
patent application Ser. No. 08/446,922, hereby incorporated by reference.
Other peptides


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
56
derived from naturally occurnng trimeric proteins may be employed in preparing
trimeric
polypeptides of the invention.
In another example, proteins of the invention are associated by interactions
between
Flag~ polypeptide sequence contained in fusion proteins of the invention
containing Flag~
polypeptide seuqence. In a further embodiment, associations proteins of the
invention are
associated by interactions between heterologous polypeptide sequence contained
in Flag~
fusion proteins of the invention and anti-Flag~ antibody.
The multimers of the invention may be generated using chemical techniques
known in
the art. For example, polypeptides desired to be contained in the multimers of
the invention
may be chemically cross-linked using linker molecules and linker molecule
length
optimization techniques known in the art (see, e.g., US Patent Number
5,478,925, which is
herein incorporated by reference in its entirety). Additionally, multimers of
the invention
may be generated using techniques known in the art to form one or more inter-
molecule
cross-links between the cysteine residues located within the sequence of the
polypeptides
desired to be contained in the multimer (see, e.g., US Patent Number
5,478,925, which is
herein incorporated by reference in its entirety). Further, polypeptides of
the invention may
be routinely modified by the addition of cysteine or biotin to the C-terminus
or N-terminus of
the polypeptide and techniques known in the art may be applied to generate
multimers
containing one or more of these modified polypeptides (see, e.g., US Patent
Number
5,478,925, which is herein incorporated by reference in its entirety).
Additionally, techniques
known in the art may be applied to generate liposomes containing the
polypeptide
components desired to be contained in the multimer of the invention (see,
e.g., US Patent
Number 5,478,925, which is herein incorporated by reference in its entirety).
Alternatively, multimers of the invention may be generated using genetic
engineering
techniques known in the art. In one embodiment, polypeptides contained in
multimers of the
invention are produced recombinantly using fusion protein technology described
herein or
otherwise known in the art (see, e.g., US Patent Number 5,478,925, which is
herein
incorporated by reference in its entirety). In a specific embodiment,
polynucleotides coding
for a homodimer of the invention are generated by ligating a polynucleotide
sequence
encoding a polypeptide of the invention to a sequence encoding a linker
polypeptide and then
further to a synthetic polynucleotide encoding the translated product of the
polypeptide in the
reverse orientation from the original C-terminus to the N-terminus (lacking
the leader


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
57
sequence) (see, e.g., US Patent Number 5,478,925, which is herein incorporated
by reference
in its entirety). In another embodiment, recombinant techniques described
herein or
otherwise known in the art are applied to generate recombinant polypeptides of
the invention
which contain a transmembrane domain (or hyrophobic or signal peptide) and
which can be
S incorporated by membrane reconstitution techniques into liposomes (see,
e.g., US Patent
Number 5,478,925, which is herein incorporated by reference in its entirety).
Antibodies
Further polypeptides of the invention relate to antibodies and T-cell antigen
receptors
(TCR) which immunospecifically bind a polypeptide, polypeptide fragment, or
variant of
SEQ 117 NO:Y, and/or an epitope, of the present invention (as determined by
immunoassays
well known in the art for assaying specific antibody-antigen binding).
Antibodies of the
invention include, but are not limited to, polyclonal, monoclonal,
multispecific, human,
humanized or chimeric antibodies, single chain antibodies, Fab fragments,
F(ab') fragments,
fragments produced by a Fab expression library, anti-idiotypic (anti-Id)
antibodies
(including, e.g., anti-Id antibodies to antibodies of the invention), and
epitope-binding
fragments of any of the above. The term "antibody," as used herein, refers to
immunoglobulin molecules and immunologically active portions of immunoglobulin
molecules, i.e., molecules that contain an antigen binding site that
immunospecifically binds
an antigen. The immunoglobulin molecules of the invention can be of any type
(e.g., IgG,
IgE, IgM, IgD, IgA and IgY), class (e.g., IgGI, IgG2, IgG3, IgG4, IgAl and
IgA2) or
subclass of immunoglobulin molecule.
Most preferably the antibodies are human antigen-binding antibody fragments of
the
present invention and include, but are not limited to, Fab, Fab' and F(ab')2,
Fd, single-chain
Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and fragments
comprising
either a VL or VH domain. Antigen-binding antibody fragments, including single-
chain
antibodies, may comprise the variable regions) alone or in combination with
the entirety or a
portion of the following: hinge region, CH1, CH2, and CH3 domains. Also
included in the
invention are antigen-binding fragments also comprising any combination of
variable
regions) with a hinge region, CH1, CH2, and CH3 domains. The antibodies of the
invention
may be from any animal origin including birds and mammals. Preferably, the
antibodies are
human, murine (e.g., mouse and rat), donkey, ship rabbit, goat, guinea pig,
camel, horse, or


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
58
chicken. As used herein, "human" antibodies include antibodies having the
amino acid
sequence of a human immunoglobulin and include antibodies isolated from human
immunoglobulin libraries or from animals transgenic for one or more human
immunoglobulin
and that do not express endogenous immunoglobulins, as described infra and,
for example
S in, U.S. Patent No. 5,939,598 by Kucherlapati et al.
The antibodies of the present invention may be monospecific, bispecific,
trispecific or
of greater multispecificity. Multispecific antibodies may be specific for
different epitopes of
a polypeptide of the present invention or may be specific for both a
polypeptide of the present
invention as well as for a heterologous epitope, such as a heterologous
polypeptide or solid
support material. See, e.g., PCT publications WO 93/17715; WO 92/08802; WO
91/00360;
WO 92/05793; Tutt, et al., J. Immunol. 147:60-69 (1991); U.S. Patent Nos.
4,474,893;
4,714,681; 4,925,648; 5,573,920; 5,601,819; Kostelny et al., J. Immunol.
148:1547-1553
(1992).
Antibodies of the present invention may be described or specified in terms of
the
epitope(s) or portions) of a polypeptide of the present invention which they
recognize or
specifically bind. The epitope(s) or polypeptide portions) may be specified as
described
herein, e.g., by N-terminal and C-terminal positions, or by size in contiguous
amino acid
residues. Antibodies which specifically bind any epitope or polypeptide of the
present
invention may also be excluded. Therefore, the present invention includes
antibodies that
specifically bind polypeptides of the present invention, and allows for the
exclusion of the
same.
Antibodies of the present invention may also be described or specified in
terms of
their cross-reactivity. Antibodies that do not bind any other analog,
ortholog, or homolog of
a polypeptide of the present invention are included. Antibodies that bind
polypeptides with at
least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least
70%, at least 65%, at
least 60%, at least 55%, and at least 50% identity (as calculated using
methods known in the
art and described herein) to a polypeptide of the present invention are also
included in the
present invention. In specific embodiments, antibodies of the present
invention cross-react
with murine, rat and/or rabbit homologs of human proteins and the
corresponding epitopes
thereof. Antibodies that do not bind polypeptides with less than 95%, less
than 90%, less than
85%, less than 80%, less than 75%, less than 70%, less than 65%, less than
60%, less than
SS%, and less than 50% identity (as calculated using methods known in the art
and described


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
59
herein) to a polypeptide of the present invention are also included in the
present invention.
In a specific embodiment, the above-described cross-reactivity is with respect
to any single
specific antigenic or immunogenic polypeptide, or combinations) of 2, 3, 4, 5,
or more of the
specific antigenic and/or immunogenic polypeptides disclosed herein. Further
included in the
present invention are antibodies which bind polypeptides encoded by
polynucleotides which
hybridize to a polynucleotide of the present invention under stringent
hybridization
conditions (as described herein). Antibodies of the present invention may also
be described
or specified in terms of their binding affinity to a polypeptide of the
invention. Preferred
binding affinities include those with a dissociation constant or Kd less than
5 X 10-2 M, 10-Z
M, 5 X 10-3 M, 10-3 M, 5 X 10-4 M, 10-~ M, 5 X 10-5 M, 10-5 M, 5 X 10-~ M, 10-
6M, 5 X 10-~
M, 107 M, 5 X 10-$ M, 10-g M, 5 X 10-9 M, 10-9 M, 5 X 10-' ° M, 10-'
° M, 5 X 10-" M, 10-"
M, 5 X 10-' 2 M, ' °-' 2 M, 5 X 10-' 3 M, 10-' 3 M, 5 X 10-' 4 M, 10-'
4 M, 5 X 10-' S M, or 10-' S M.
The invention also provides antibodies that competitively inhibit binding of
an
antibody to an epitope of the invention as determined by any method known in
the art for
determining competitive binding, for example, the immunoassays described
herein. In
preferred embodiments, the antibody competitively inhibits binding to the
epitope by at least
95%, at least 90%, at least 85 %, at least 80%, at least 75%, at least 70%, at
least 60%, or at
least 50%.
Antibodies of the present invention may act as agonists or antagonists of the
polypeptides of the present invention. For example, the present invention
includes antibodies
which disrupt the receptor/ligand interactions with the polypeptides of the
invention either
partially or fully. Preferrably, antibodies of the present invention bind an
antigenic epitope
disclosed herein, or a portion thereof. The invention features both receptor-
specific antibodies
and ligand-specific antibodies. The invention also features receptor-specific
antibodies
which do not prevent ligand binding but prevent receptor activation. Receptor
activation
(i.e., signaling) may be determined by techniques described herein or
otherwise known in the
art. For example, receptor activation can be determined by detecting the
phosphorylation
(e.g., tyrosine or serine/threonine) of the receptor or its substrate by
immunoprecipitation
followed by western blot analysis (for example, as described supra). In
specific
embodiments, antibodies are provided that inhibit ligand activity or receptor
activity by at
least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least
70%, at least 60%, or
at least 50% of the activity in absence of the antibody.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
The invention also features receptor-specific antibodies which both prevent
ligand
binding and receptor activation as well as antibodies that recognize the
receptor-ligand
complex, and, preferably, do not specifically recognize the unbound receptor
or the unbound
ligand. Likewise, included in the invention are neutralizing antibodies which
bind the ligand
5 and prevent binding of the ligand to the receptor, as well as antibodies
which bind the ligand,
thereby preventing receptor activation, but do not prevent the ligand from
binding the
receptor. Further included in the invention are antibodies which activate the
receptor. These
antibodies may act as receptor agonists, i.e., potentiate or activate either
all or a subset of the
biological activities of the ligand-mediated receptor activation, for example,
by inducing
10 dimerization of the receptor. The antibodies may be specified as agonists,
antagonists or
inverse agonists for biological activities comprising the specific biological
activities of the
peptides of the invention disclosed herein. The above antibody agonists can be
made using
methods known in the art. See, e.g., PCT publication WO 96/40281; U.S. Patent
No.
5,811,097; Deng et al., Blood 92(6):1981-1988 (1998); Chen et al., Cancer Res.
15 58(16):3668-3678 (1998); Harrop et al., J. Immunol. 161(4):1786-1794
(1998); Zhu et al.,
Cancer Res. 58(15):3209-3214 (1998); Yoon et al., J. Immunol. 160(7):3170-3179
(1998);
Prat et al., J. Cell. Sci. 111(Pt2):237-247 (1998); Pitard et al., J. Immunol.
Methods
205(2):177-190 (1997); Liautard et al., Cytokine 9(4):233-241 (1997); Carlson
et al., J. Biol.
Chem. 272(17):11295-11301 (1997); Taryman et al., Neuron 14(4):755-762 (1995);
Muller
20 et al., Structure 6(9):1153-1167 (1998); Bartunek et al., Cytokine 8(1):14-
20 (1996) (which
are all incorporated by reference herein in their entireties).
Antibodies of the present invention may be used, for example, but not limited
to, to
purify, detect, and target the polypeptides of the present invention,
including both in vitro and
in vivo diagnostic and therapeutic methods. For example, the antibodies have
use in
25 immunoassays for qualitatively and quantitatively measuring levels of the
polypeptides of the
present invention in biological samples. See, e.g., Harlow et al., Antibodies:
A Laboratory
Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988) (incorporated by
reference
herein in its entirety).
As discussed in more detail below, the antibodies of the present invention may
be
30 used either alone or in combination with other compositions. The antibodies
may further be
recombinantly fused to a heterologous polypeptide at the N- or C-terminus or
chemically
conjugated (including covalently and non-covalently conjugations) to
polypeptides or other


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
61
compositions. For example, antibodies of the present invention may be
recombinantly fused
or conjugated to molecules useful as labels in detection assays and effector
molecules such as
heterologous polypeptides, drugs, radionuclides, or toxins. See, e.g., PCT
publications WO
92/08495; WO 91/14438; WO 89/12624; U.S. Patent No. 5,314,995; and EP 396,387.
The antibodies of the invention include derivatives that are modified, i.e, by
the
covalent attachment of any type of molecule to the antibody such that covalent
attachment
does not prevent the antibody from generating an anti-idiotypic response. For
example, but
not by way of limitation, the antibody derivatives include antibodies that
have been modified,
e.g., by glycosylation, acetylation, pegylation, phosphylation, amidation,
derivatization by
known protecting/blocking groups, proteolytic cleavage, linkage to a cellular
ligand or other
protein, etc. Any of numerous chemical modifications may be carried out by
known
techniques, including, but not limited to specific chemical cleavage,
acetylation, formylation,
metabolic synthesis of tunicamycin, etc. Additionally, the derivative may
contain one or
more non-classical amino acids.
The antibodies of the present invention may be generated by any suitable
method
known in the art. Polyclonal antibodies to an antigen-of interest can be
produced by various
procedures well known in the art. For example, a polypeptide of the invention
can be
administered to various host animals including, but not limited to, rabbits,
mice, rats, etc. to
induce the production of sera containing polyclonal antibodies specific for
the antigen.
Various adjuvants may be used to increase the immunological response,
depending on the
host species, and include but are not limited to, Freund's (complete and
incomplete), mineral
gels such as aluminum hydroxide, surface active substances such as
lysolecithin, pluronic
polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins,
dinitrophenol, and
potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and
corynebacterium parvum. Such adjuvants are also well known in the art.
Monoclonal antibodies can be prepared using a wide variety of techniques known
in
the art including the use of hybridoma, recombinant, and phage display
technologies, or a
combination thereof. For example, monoclonal antibodies can be produced using
hybridoma
techniques including those known in the art and taught, for example, in Harlow
et al.,
Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed.
1988);
Hammerling, et al., in: Monoclonal Antibodies and T-Cell Hybridomas 563-681
(Elsevier,
N.Y., 1981) (said references incorporated by reference in their entireties).
The term


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
62
"monoclonal antibody" as used herein is not limited to antibodies produced
through
hybridoma technology. The term "monoclonal antibody" refers to an antibody
that is
derived from a single clone, including any eukaryotic, prokaryotic, or phage
clone, and not
the method by which it is produced.
Methods for producing and screening for specific antibodies using hybridoma
technology are routine and well known in the art and are discussed in detail
in the Examples.
In a non-limiting example, mice can be immunized with a polypeptide of the
invention or a
cell expressing such peptide. Once an immune response is detected, e.g.,
antibodies specific
for the antigen are detected in the mouse serum, the mouse spleen is harvested
and
splenocytes isolated. The splenocytes are then fused by well known techniques
to any
suitable myeloma cells, for example cells from cell line SP20 available from
the ATCC.
Hybridomas are selected and cloned by limited dilution. The hybridoma clones
are then
assayed by methods known in the art for cells that secrete antibodies capable
of binding a
polypeptide of the invention. Ascites fluid, which generally contains high
levels of
antibodies, can be generated by immunizing mice with positive hybridoma
clones.
Accordingly, the present invention provides methods of generating monoclonal
antibodies as well as antibodies produced by the method comprising culturing a
hybridoma
cell secreting an antibody of the invention wherein, preferably, the hybridoma
is generated by
fusing splenocytes isolated from a mouse immunized with an antigen of the
invention with
myeloma cells and then screening the hybridomas resulting from the fusion for
hybridoma
clones that secrete an antibody able to bind a polypeptide of the invention.
Antibody fragments which recognize specific epitopes may be generated by known
techniques. For example, Fab and F(ab')2 fragments of the invention may be
produced by
proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain
(to
produce Fab fragments) or pepsin (to produce F(ab')2 fragments). F(ab')2
fragments contain
the variable region, the light chain constant region and the CH1 domain of the
heavy chain.
For example, the antibodies of the present invention can also be generated
using
various phage display methods known in the art. In phage display methods,
functional
antibody domains are displayed on the surface of phage particles which carry
the
polynucleotide sequences encoding them. In a particular embodiment, such phage
can be
utilized to display antigen binding domains expressed from a repertoire or
combinatorial
antibody library (e.g., human or murine). Phage expressing an antigen binding
domain that


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
63
binds the antigen of interest can be selected or identified with antigen,
e.g., using labeled
antigen or antigen bound or captured to a solid surface or bead. Phage used in
these methods
are typically filamentous phage including fd and M13 binding domains expressed
from phage
with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused
to either the
phage gene III or gene VIII protein. Examples of phage display methods that
can be used to
make the antibodies of the present invention include those disclosed in
Brinkman et al., J.
Immunol. Methods 182:41-50 (1995); Ames et al.,' J. Immunol. Methods 184:177-
186
(1995); Kettleborough et al., Eur. J. Immunol. 24:952-958 (1994); Persic et
al., Gene 187 9-
18 (1997); Burton et al., Advances in Immunology 57:191-280 (1994); PCT
application No.
PCT/GB91/01134; PCT publications WO 90/02809; WO 91/10737; WO 92/01047; WO
92/18619; WO 93/11236; WO 95/15982; WO 95/20401; and U.S. Patent Nos.
5,698,426;
5,223,409; 5,403,484; 5,580,717; 5,427,908; 5,750,753; 5,821,047; 5,571,698;
5,427,908;
5,516,637; 5,780,225; 5,658,727; 5,733,743 and 5,969,108; each of which is
incorporated
herein by reference in its entirety.
As described in the above references, after phage selection, the antibody
coding
regions from the phage can be isolated and used to generate whole antibodies,
including
human antibodies, or any other desired antigen binding fragment, and expressed
in any
desired host, including mammalian cells, insect cells, plant cells, yeast, and
bacteria, e.g., as
described in detail below. For example, techniques to recombinantly produce
Fab, Fab' and
F(ab')2 fragments can also be employed using methods known in the art such as
those
disclosed in PCT publication WO 92/22324; Mullinax et al., BioTechniques
12(6):864-869
(1992); and Sawai et al., AJRI 34:26-34 (1995); and Better et al., Science
240:1041-1043
(1988) (said references incorporated by reference in their entireties).
Examples of techniques which can be used to produce single-chain Fvs and
antibodies
include those described in U.S. Patents 4,946,778 and 5,258,498; Huston et
al., Methods in
Enzymology 203:46-88 (1991); Shu et al., PNAS 90:7995-7999 (1993); and Skerra
et al.,
Science 240:1038-1040 (1988). For some uses, including in vivo use of
antibodies in
humans and in vitro detection assays, it may be preferable to use chimeric,
humanized, or
human antibodies. A chimeric antibody is a molecule in which different
portions of the
antibody are derived from different animal species, such as antibodies having
a variable
region derived from a murine monoclonal antibody and a human immunoglobulin
constant
region. Methods for producing chimeric antibodies are known in the art. See
e.g., Morrison,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
64
Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Gillies et
al., (1989) J.
Immunol. Methods 125:191-202; U.S. Patent Nos. 5,807,715; 4,816,567; and
4,816397,
which are incorporated herein by reference in their entirety. Humanized,
antibodies are
antibody molecules from non-human species antibody that binds the desired
antigen having
one or more complementarity determining regions (CDRs) from the non-human
species and
a framework regions from a human immunoglobulin molecule. Often, framework
residues in
the human framework regions will be substituted with the corresponding residue
from the
CDR donor antibody to alter, preferably improve, antigen binding. These
framework
substitutions are identified by methods well known in the art, e.g., by
modeling of the
interactions of the CDR and framework residues to identify framework residues
important
for antigen binding and sequence comparison to identify unusual framework
residues at
particular positions. (See, e.g., Queen et al., U.S. Patent No. 5,585,089;
Riechmann et al.,
Nature 332:323 (1988), which are incorporated herein by reference in their
entireties.)
Antibodies can be humanized using a variety of techniques known in the art
including, for
example, CDR-grafting (EP 239,400; PCT publication WO 91/09967; U.S. Patent
Nos.
5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP
519,596;
Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka et al., Protein
Engineering 7(6):805-814 (1994); Roguska. et al., PNAS 91:969-973 (1994)), and
chain
shuffling (U.S. Patent No. 5,565,332).
Completely human antibodies are particularly desirable for therapeutic
treatment of
human patients. Human antibodies can be made by a variety of methods known in
the art
including phage display methods described above using antibody libraries
derived from
human i~nmunoglobulin sequences. See also, U.S. Patent Nos. 4,444,887 and
4,716,111; and
PCT publications WO 98/46645, WO 98/50433, WO 98/24893, WO 98/16654, WO
96/34096, WO 96/33735, and WO 91/10741; each of which is incorporated herein
by
reference in its entirety.
Human antibodies can also be produced using transgenic mice which are
incapable of
expressing functional endogenous immunoglobulins, but which can express human
immunoglobulin genes. For example, the human heavy and light chain
immunoglobulin gene
complexes may be introduced randomly or by homologous recombination into mouse
embryonic stem cells. Alternatively, the human variable region, constant
region, and
diversity region may be introduced into mouse embryonic stem cells in addition
to the human


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
heavy and light chain genes. The mouse heavy and light chain immunoglobulin
genes may
be rendered non-functional separately or simultaneously with the introduction
of human
immunoglobulin loci by homologous recombination. In particular, homozygous
deletion of
the JH region prevents endogenous antibody production. The modified embryonic
stem cells
5 are expanded and microinjected into blastocysts to produce chimeric mice.
The chimeric
mice are then bred to produce homozygous offspring which express human
antibodies. The
transgenic mice are immunized in the normal fashion with a selected antigen,
e.g., all or a
portion of a polypeptide of the invention. Monoclonal antibodies directed
against the
antigen can be obtained from the immunized, transgenic mice using conventional
hybridoma
10 technology. The human immunoglobulin transgenes harbored by the transgenic
mice
rearrange during B cell differentiation, and subsequently undergo class
switching and
somatic mutation. Thus, using such a technique, it is possible to produce
therapeutically
useful IgG, IgA, IgM and IgE antibodies. For an overview of this technology
for producing
human antibodies, see Lonberg and Huszar, Int. Rev. Immunol. 13:65-93 (1995).
For a
15 detailed discussion of this technology for producing human antibodies and
human
monoclonal antibodies and protocols for producing such antibodies, see, e.g.,
PCT
publications WO 98/24893; WO 92/01047; WO 96/34096; WO 96/33735; European
Patent
No. 0 598 877; U.S. Patent Nos. 5,413,923; 5,625,126; 5,633,425; 5,569,825;
5,661,016;
5,545,806; 5,814,318; 5,885,793; 5,916,771; and 5,939,598, which are
incorporated by
20 reference herein in their entirety. In addition, companies such as Abgenix,
Inc. (Freemont,
CA) and Genpharm (San Jose, CA) can be engaged to provide human antibodies
directed
against a selected antigen using technology similar to that described above.
Completely human antibodies which recognize a selected epitope can be
generated
using a technique referred to as "guided selection." In this approach a
selected non-human
25 monoclonal antibody, e.g., a mouse antibody, is used to guide the selection
of a completely
human antibody recognizing the same epitope. (Jespers et al., Biotechnology
12:899-903
(1988)).
Further, antibodies to the polypeptides of the invention can, in turn, be
utilized to
generate anti-idiotype antibodies that "mimic" polypeptides of the invention
using techniques
30 well known to those skilled in the art. (See, e.g., Greenspan & Bona, FASEB
J. 7(5):437-444;
(1989) and Nissinoff, J. Immunol. 147(8):2429-2438 (1991)). For example,
antibodies
which bind to and competitively inhibit polypeptide multimerization and/or
binding of a


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
66
polypeptide of the invention to a ligand can be used to generate anti-
idiotypes that "mimic"
the polypeptide multimerization and/or binding domain and, as a consequence,
bind to and
neutralize polypeptide and/or its ligand. Such neutralizing anti-idiotypes or
Fab fragments of
such anti-idiotypes can be used in therapeutic regimens to neutralize
polypeptide ligand. For
example, such anti-idiotypic antibodies can be used to bind a polypeptide of
the invention
and/or to bind its ligands/receptors, and thereby block its biological
activity.
Polyriucleotides Encoding Antibodies
The invention further provides polynucleotides comprising a nucleotide
sequence
encoding an antibody of the invention and fragments thereof. The invention
also
encompasses polynucleotides that hybridize under stringent or alternatively,
under lower
stringency hybridization conditions, e.g., as defined supra, to
polynucleotides that encode an
antibody, preferably, that specifically binds to a polypeptide of the
invention, preferably, an
antibody that binds to a polypeptide having the amino acid sequence of SEQ >D
NO:Y.
The polynucleotides may be obtained, and the nucleotide sequence of the
polynucleotides determined, by any method known in the art. For example, if
the nucleotide
sequence of the antibody is known, a polynucleotide encoding the antibody may
be
assembled from chemically synthesized oligonucleotides (e.g., as described in
Kutmeier et
al., BioTechniques 17:242 (1994)), which, briefly, involves the synthesis of
overlapping
oligonucleotides containing portions of the sequence encoding the antibody,
annealing and
ligating of those oligonucleotides, and then amplification of the ligated
oligonucleotides by
PCR.
Alternatively, a polynucleotide encoding an antibody may be generated from
nucleic
acid from a suitable source. If a clone containing a nucleic acid encoding a
particular
antibody is not available, but the sequence of the antibody molecule is known,
a nucleic acid
encoding the immunoglobulin may be chemically synthesized or obtained from a
suitable
source (e.g., an antibody cDNA library, or a cDNA library generated from, or
nucleic acid,
preferably poly A+ RNA, isolated from, any tissue or cells expressing the
antibody, such as
hybridoma cells selected to express an antibody of the invention) by PCR
amplification
using synthetic primers hybridizable to the 3' and 5' ends of the sequence or
by cloning using
an oligonucleotide probe specific for the particular gene sequence to
identify, e.g., a cDNA
clone from a cDNA library that encodes the antibody. Amplified nucleic acids
generated by


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
67
PCR may then be cloned into replicable cloning vectors using any method well
known in the
art.
Once the nucleotide sequence and corresponding amino acid sequence of the
antibody
is determined, the nucleotide sequence of the antibody may be manipulated
using methods
well known in the art for the manipulation of nucleotide sequences, e.g.,
recombinant DNA
techniques, site directed mutagenesis, PCR, etc. (see, for example, the
techniques described
in Sambrook et al., 1990, Molecular Cloning, A Laboratory Manual, 2d Ed., Cold
Spring
Harbor Laboratory, Cold Spring Harbor, NY and Ausubel et al., eds., 1998,
Current Protocols
in Molecular Biology, John Wiley & Sons, NY, which are both incorporated by
reference
herein in their entireties ), to generate antibodies having a different amino
acid sequence, for
example to create amino acid substitutions, deletions, and/or insertions.
In a specific embodiment, the amino acid sequence of the heavy and/or light
chain
variable domains may be inspected to identify the sequences of the
complementarity
determining regions (CDRs) by methods that are well know in the art, e.g., by
comparison to
known amino acid sequences of other heavy and light chain variable regions to
determine the
regions of sequence hypervariability. Using routine recombinant DNA
techniques, one or
more of the CDRs may be inserted within framework regions, e.g., into human
framework
regions to humanize a non-human antibody, as described supra. The framework
regions may
be naturally occurring or consensus framework regions, and preferably human
framework
regions (see, e.g., Chothia et al., J. Mol. Biol. 278: 457-479 (1998) for a
listing of human
framework regions). Preferably, the polynucleotide generated by the
combination of the
framework regions and CDRs encodes an antibody that specifically binds a
polypeptide of
the invention. Preferably, as discussed supra, one or more amino acid
substitutions may be
made within the framework regions, and, preferably, the amino acid
substitutions improve
binding of the antibody to its antigen. Additionally, such methods may be used
to make
amino acid substitutions or deletions of one or more variable region cysteine
residues
participating in an intrachain disulfide bond to generate antibody molecules
lacking one or
more intrachain disulfide bonds. Other alterations to the polynucleotide are
encompassed by
the present invention and within the skill of the art.
In addition, techniques developed for the production of "chimeric antibodies"
(Morrison et al., Proc. Natl. Acad. Sci. 81:851-855 (1984); Neuberger et al.,
Nature
312:604-608 (1984); Takeda et al., Nature 314:452-454 (1985)) by splicing
genes from a


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
68
mouse antibody molecule of appropriate antigen specificity together with genes
from a
human antibody molecule of appropriate biological activity can be used. As
described supra,
a chimeric antibody is a molecule in which different portions are derived from
different
animal species, such as those having a variable region derived from a murine
mAb and a
human immunoglobulin constant region, e.g., humanized antibodies.
Alternatively, techniques described for the production of single chain
antibodies (U.S.
Patent No. 4,946,778; Bird, Science 242:423- 42 (1988); Huston et al., Proc.
Natl. Acad. Sci.
USA 85:5879-5883 (1988); and Ward et al., Nature 334:544-54 (1989)) can be
adapted to
produce single chain antibodies. Single chain antibodies are formed by linking
the heavy
and light chain fragments of the Fv region via an amino acid bridge, resulting
in a single
chain polypeptide. Techniques for the assembly of functional Fv fragments in
E. coli may
also be used (Skerra et al., Science 242:1038- 1041 (1988)).
Methods of Producing Antibodies
The antibodies of the invention can be produced by any method known in the art
for
the synthesis of antibodies, in particular, by chemical synthesis or
preferably, by recombinant
expression techniques.
Recombinant expression of an antibody of the invention, or fragment,
derivative or
analog thereof, (e.g., a heavy or light chain of an antibody of the invention
or a single chain
antibody of the invention), requires construction of an expression vector
containing a
polynucleotide that encodes the antibody. Once a polynucleotide encoding an
antibody
molecule or a heavy or light chain of an antibody, or portion thereof
(preferably containing
the heavy or light chain variable domain), of the invention has been obtained,
the vector for
the production of the antibody molecule may be produced by recombinant DNA
technology
using techniques well known in the art. Thus, methods for preparing a protein
by expressing
a polynucleotide containing an antibody encoding nucleotide sequence are
described herein.
Methods which are well known to those skilled in the art can be used to
construct expression
vectors containing antibody coding sequences and appropriate transcriptional
and
translational control signals. These methods include, for example, in vitro
recombinant DNA
techniques, synthetic techniques, and in vivo genetic recombination. The
invention, thus,
provides replicable vectors comprising a nucleotide sequence encoding an
antibody molecule
of the invention, or a heavy or light chain thereof, or a heavy or light chain
variable domain,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
69
operably linked to a promoter. Such vectors may include the nucleotide
sequence encoding
the constant region of the antibody molecule (see, e.g., PCT Publication WO
86/05807; PCT
Publication WO 89/01036; and U.S. Patent No. 5,122,464) and the variable
domain of the
antibody may be cloned into such a vector for expression of the entire heavy
or light chain.
The expression vector is transferred to a host cell by conventional techniques
and the
transfected cells are then cultured by conventional techniques to produce an
antibody of the
invention. Thus, the invention includes host cells containing a polynucleotide
encoding an
antibody of the invention, or a heavy or light chain thereof, or a single
chain antibody of the
invention, operably linked to a heterologous promoter. In preferred
embodiments for the
expression of double-chained antibodies, vectors encoding both the heavy and
light chains
may be co-expressed in the host cell for expression of the entire
immunoglobulin molecule,
as detailed below.
A variety of host-expression vector systems may be utilized to express the
antibody
molecules of the invention. Such host-expression systems represent vehicles by
which the
coding sequences of interest may be produced and subsequently purified, but
also represent
cells which may, when transformed or transfected with the appropriate
nucleotide coding
sequences, express an antibody molecule of the invention in situ. These
include but are not
limited to microorganisms such as bacteria (e.g., E. coli, B. subtilis)
transformed with
recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors
containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia)
transformed with
recombinant yeast expression vectors containing antibody coding sequences;
insect cell
systems infected with recombinant virus expression vectors (e.g., baculovirus)
containing
antibody coding sequences; plant cell systems infected with recombinant virus
expression
vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or
transformed
with recombinant plasmid expression vectors (e.g., Ti plasmid) containing
antibody coding
sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells)
harboring
recombinant expression constructs containing promoters derived from the genome
of
mammalian cells (e.g., metallothionein promoter) or from mammalian viruses
(e.g., the
adenovirus late promoter; the vaccinia virus 7.5K promoter). Preferably,
bacterial cells such
as Escherichia coli, and more preferably, eukaryotic cells, especially for the
expression of
whole recombinant antibody molecule, are used for the expression of a
recombinant antibody
molecule. For example, mammalian cells such as Chinese hamster ovary cells
(CHO), in


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
conjunction with a vector such as the major intermediate early gene promoter
element from
human cytomegalovirus is an effective expression system for antibodies
(Foecking et al.,
Gene 45:101 (1986); Cockett et al., Bio/Technology 8:2 (1990)).
In bacterial systems, a number of expression vectors may be advantageously
selected
5 depending upon the use intended for the antibody molecule being expressed.
For example,
when a large quantity of such a protein is to be produced, for the generation
of
pharmaceutical compositions of an antibody molecule, vectors which direct the
expression of
high levels of fusion protein products that are readily purified may be
desirable. Such vectors
include, but are not limited, to the E. coli expression vector pUR278 (Ruther
et al., EMBO J.
10 2:1791 (1983)), in which the antibody coding sequence may be ligated
individually into the
vector in frame with the lac Z coding region so that a fusion protein is
produced; pIN vectors
(Inouye & Inouye, Nucleic Acids Res. 13:3101-3109 (1985); Van Heeke &
Schuster, J. Biol.
Chem. 24:5503-5509 (1989)); and the like. pGEX vectors may also be used to
express
foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
In general,
15 such fusion proteins are soluble and can easily be purified from lysed
cells by adsorption and
binding to matrix glutathione-agarose beads followed by elution in the
presence of free
glutathione. The pGEX vectors are designed to include thrombin or factor Xa
protease
cleavage sites so that the cloned target gene product can be released from the
GST moiety.
In an insect system, Autographa californica nuclear polyhedrosis virus (AcNPV)
is
20 used as a vector to express foreign genes. The virus grows in Spodoptera
frugiperda cells.
The antibody coding sequence may be cloned individually into non-essential
regions (for
example the polyhedrin gene) of the virus and placed under control of an AcNPV
promoter
(for example the polyhedrin promoter).
In mammalian host cells, a number of viral-based expression systems may be
utilized.
25 In cases where an adenovirus is used as an expression vector, the antibody
coding sequence
of interest may be ligated to an adenovirus transcription/translation control
complex, e.g., the
late promoter and tripartite leader sequence. This chimeric gene may then be
inserted in the
adenovirus genome by in vitro or in vivo recombination. Insertion in a non-
essential region
of the viral genome (e.g., region E1 or E3) will result in a recombinant virus
that is viable and
30 capable of expressing the antibody molecule in infected hosts. (e.g.,'see
Logan & Shenk,
Proc. Natl. Acad. Sci. USA 81:355-359 (1984)). Specific initiation signals may
also be
required for efficient translation of inserted antibody coding sequences.
These signals


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
71
include the ATG initiation codon and adjacent sequences. Furthermore, the
initiation codon
must be in phase with the reading frame of the desired coding sequence to
ensure translation
of the entire insert. These exogenous translational control signals and
initiation codons can
be of a variety of origins, both natural and synthetic. The efficiency of
expression may be
enhanced by the inclusion of appropriate transcription enhancer elements,
transcription
terminators, etc. (see Bittner et al., Methods in Enzyrnol. 153:51-544
(1987)).
In addition, a host cell strain may be chosen which modulates the expression
of the
inserted sequences, or modifies and processes the gene product in the specific
fashion
desired. Such modifications (e.g., glycosylation) and processing (e.g.,
cleavage) of protein
products may be important for the function of the. protein. Different host
cells have
characteristic and specific mechanisms for the post-translational processing
and modification
of proteins and gene products. Appropriate cell lines or host systems can be
chosen to
ensure the correct modification and processing of the foreign protein
expressed. To this end,
eukaryotic host cells which possess the cellular machinery for proper
processing of the
primary transcript, glycosylation, and phosphorylation of the gene product may
be used.
Such mammalian host cells include but are not limited to CHO, VERY, BHK, Hela,
COS,
MDCK, 293, 3T3, WI38, and in particular, breast cancer cell lines such as, for
example,
BT483, Hs578T, HTB2, BT20 and T47D, and normal mammary gland cell line such
as, for
example, CRL7030 and Hs578Bst.
For long-term, high-yield production of recombinant proteins, stable
expression is
preferred. For example, cell lines which stably express the antibody molecule
may be
engineered. Rather than using expression vectors which contain viral origins
of replication,
host cells can be transformed with DNA controlled by appropriate expression
control
elements (e.g., promoter, enhancer, sequences, transcription terminators,
polyadenylation
sites, etc.), and a selectable marker. Following the introduction of the
foreign DNA,
engineered cells may be allowed to grow for 1-2 days in an enriched media, and
then are
switched to a selective media. The selectable marker in the recombinant
plasmid confers
resistance to the selection and allows cells to stably integrate the plasmid
into their
chromosomes and grow to form foci which in turn can be cloned and expanded
into cell lines.
This method may advantageously be used to engineer cell lines which express
the antibody
molecule. Such engineered cell lines may be particularly useful in screening
and evaluation
of compounds that interact directly or indirectly with the antibody molecule.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
72
A number of selection systems may be used, including but not limited to the
herpes
simplex virus thymidine kinase (Wigler et al., Cell 11:223 (1977)),
hypoxanthine-guanine
phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA
48:202
(1992)), and adenine phosphoribosyltransferase (Lowy et al., Cell 22:817
(1980)) genes can
be employed in tk-, hgprt- or aprt- cells, respectively. Also, antimetabolite
resistance can be
used as the basis of selection for the following genes: dhfr, which confers
resistance to
methotrexate (Wigler et al., Natl. Acad. Sci. USA 77:357 (1980); O'Hare et
al., Proc. Natl.
Acad. Sci. USA 78:1527 (1981)); gpt, which confers resistance to mycophenolic
acid
(Mulligan & Berg, Proc. Natl. Acad. Sci. USA 78:2072 (1981)); neo, which
confers
resistance to the aminoglycoside G-418 Clinical Pharmacy 12:488-505; Wu and
Wu,
Biotherapy 3:87-95 (1991); Tolstoshev, Ann. Rev. Pharmacol. Toxicol. 32:573-
596 (1993);
Mulligan, Science 260:926-932 (1993); and Morgan and Anderson, Ann. Rev.
Biochem.
62:191-217 (1993); May, 1993, TIB TECH 11(5):155-215); and hygro, which
confers
resistance to hygromycin (Santerre et al., Gene 30:147 (1984)). Methods
commonly known
in the art of recombinant DNA technology may be routinely applied to select
the desired
recombinant clone, and such methods are described, for example, in Ausubel et
al. (eds.),
Current Protocols in Molecular Biology, John Wiley & Sons, NY (1993);
Kriegler, Gene
Transfer and Expression, A Laboratory Manual, Stockton Press, NY (1990); and
in Chapters
12 and 13, Dracopoli et al. (eds), Current Protocols in Human Genetics, John
Wiley & Sons,
NY (1994); Colberre-Garapin et al., J. Mol. Biol. 150:1 (1981), which are
incorporated by
reference herein in their entireties.
The expression levels of an antibody molecule can be increased by vector
amplification (for a review, see Bebbington and Hentschel, The use of vectors
based on gene
amplification for the expression of cloned genes in mammalian cells in DNA
cloning, Vol.3.
(Academic Press, New York, 1987)). When a marker in the vector system
expressing
antibody is amplifiable, increase in the level of inhibitor present in culture
of host cell will
increase the number of copies of the marker gene. Since the amplified region
is associated
with the antibody gene, production of the antibody will also increase (Grouse
et al., Mol.
Cell. Biol. 3:257 (1983)).
The host cell may be co-transfected with two expression vectors of the
invention, the
first vector encoding a heavy chain derived polypeptide and the second vector
encoding a
light chain derived polypeptide. The two vectors may contain identical
selectable markers


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
73
which enable equal expression of heavy and light chain polypeptides.
Alternatively, a single
vector may be used which encodes, and is capable of expressing, both heavy and
light chain
polypeptides. In such situations, the light chain should be placed before the
heavy chain to
avoid an excess of toxic free heavy chain (Proudfoot, Nature 322:52 (1986);
Kohler, Proc.
Natl. Acad. Sci. USA 77:2197 (1980)). The coding sequences for the heavy and
light chains
may comprise cDNA or genomic DNA.
Once an antibody molecule of the invention has been produced by an animal,
chemically synthesized, or recombinantly expressed, it may be purified by any
method
known in the art for purification of an immunoglobulin molecule, for example,
by
chromatography (e.g., ion exchange, affinity, particularly by affinity for the
specific antigen
after Protein A, and sizing column chromatography), centrifugation,
differential solubility, or
by any other standard technique for the purification of proteins. In addition,
the antibodies of
the present invention or fragments thereof can be fused to heterologous
polypeptide
sequences described herein or otherwise known in the art, to facilitate
purification.
The present invention encompasses antibodies recombinantly fused or chemically
conjugated (including both covalently and non-covalently conjugations) to a
polypeptide (or
portion thereof, preferably at least 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100
amino acids of the
polypeptide) of the present invention to generate fusion proteins. The fusion
does not
necessarily need to be direct, but may occur through linker sequences. The
antibodies may
be specific for antigens other than polypeptides (or portion thereof,
preferably at least 10, 20,
30, 40, S0, 60, 70, 80, 90 or 100 amino acids of the polypeptide) of the
present invention. For
example, antibodies may be used to target the polypeptides of the present
invention to
particular cell types, either in vitro or in vivo, by fusing or conjugating
the polypeptides of
the present invention to antibodies specific for particular cell surface
receptors. Antibodies
fused or conjugated to the polypeptides of the present invention may also be
used in in vitro
immunoassays and purification methods using methods known in the art. See
e.g., Harbor et
al., supra, and PCT publication WO 93/21232; EP 439,095; Naramura et al.,
Immunol. Lett.
39:91-99 (1994); U.S. Patent 5,474,981; Gillies et al., PNAS 89:1428-1432
(1992); Fell et
al., J. Immunol. 146:2446-2452(1991), which are incorporated by reference in
their entireties.
The present invention further includes compositions comprising the
polypeptides of
the present invention fused or conjugated to antibody domains other than the
variable regions.
For example, the polypeptides of the present invention may be fused or
conjugated to an


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
74
antibody Fc region, or portion thereof. The antibody portion fused to a
polypeptide of the
present invention may comprise the constant region, hinge region, CH1 domain,
CH2
domain, and CH3 domain or any combination of whole domains or portions
thereof. The
polypeptides may also be fused or conjugated to the above antibody portions to
form
multimers. For example, Fc portions fused to the polypeptides of the present
invention can
form dimers through disulfide bonding between the Fc portions. Higher
multimeric forms
can be made by fusing the polypeptides to portions of IgA and IgM. Methods for
fusing or
conjugating the polypeptides of the present invention to antibody portions are
known in the
art. See, e.g., U.S. Patent Nos. 5,336,603; 5,622,929; 5,359,046; 5,349,053;
5,447,851;
5,112,946; EP 307,434; EP 367,166; PCT publications WO 96/04388; WO 91/06570;
Ashkenazi et al., Proc. Natl. Acad. Sci. USA 88:10535-10539 (1991); Zheng et
al., J.
Immunol. 154:5590-5600 (1995); and Vil et al., Proc. Natl. Acad. Sci. USA
89:11337-
11341 ( 1992) (said references incorporated by reference in their entireties).
As discussed, supra, the polypeptides corresponding to a polypeptide,
polypeptide
fragment, or a variant of SEQ ID NO:Y may be fused or conjugated to the above
antibody
portions to increase the in vivo half life of the polypeptides or for use in
immunoassays using
methods known in the art. Further, the polypeptides corresponding to SEQ ID
NO:Y may be
fused or conjugated to the above antibody portions to facilitate purification.
One reported
example describes 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. (EP 394,827; Traunecker et al., Nature 331:84-86
(1988).
The polypeptides of the present invention fused or conjugated to an antibody
having
disulfide- linked dimeric structures (due to the IgG) may also be more
efficient in binding
and neutralizing other molecules, than the monomeric secreted protein or
protein fragment
alone. (Fountoulakis et al., J. Biochem. 270:3958-3964 (1995)). In many cases,
the Fc part
in a fusion protein is beneficial in therapy and diagnosis, and thus can
result in, for example,
improved pharmacokinetic properties. (EP A 232,262). Alternatively, deleting
the Fc part
after the fusion protein has been expressed, detected, and purified, would be
desired. For
example, the Fc portion may hinder therapy and diagnosis if the fusion protein
is used as an
antigen for immunizations. In drug discovery, for example, human proteins,
such as hIL-5,
have been fused with Fc portions for the purpose of high-throughput screening
assays to


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
identify antagonists of hIL-5. (See, Bennett et al., J. Molecular Recognition
8:52-58 (1995);
Johanson et al., J. Biol. Chem. 270:9459-9471 (1995).
Moreover, the antibodies or fragments thereof of the present invention can be
fused to
marker sequences, such as a peptide to facilitate purification. In preferred
embodiments, the
5 marker amino acid sequence is a hexa-histidine peptide, such as the tag
provided in a pQE
vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others,
many of
which are commercially available. As described in Gentz et al., Proc. Natl.
Acad. Sci. USA
86:821-824 (1989), for instance, hexa-histidine provides for convenient
purification of the
fusion protein. Other peptide tags useful for purification include, but are
not limited to, the
10 "HA" tag, which corresponds to an epitope derived from the influenza
hemagglutinin protein
(Wilson et al., Cell 37:767 (1984)) and the "flag" tag.
The present invention further encompasses antibodies or fragments thereof
conjugated
to a diagnostic or therapeutic agent. The antibodies can be used
diagnostically to, for
example, monitor the development or progression of a tumor as part of a
clinical testing
15 procedure to, e.g., determine the efficacy of a given treatment regimen.
Detection can be
facilitated by coupling the antibody to a detectable substance. Examples of
detectable
substances include various enzymes, prosthetic groups, fluorescent materials,
luminescent
materials, bioluminescent materials, radioactive materials, positron emitting
metals using
various positron emission tomographies, and nonradioactive paramagnetic metal
ions. The
20 detectable substance may be coupled or conjugated either directly to the
antibody (or
fragment thereof) or indirectly, through an intermediate (such as, for
example, a linker known
in the art) using techniques known in the art. See, for example, U.S. Patent
No. 4,741,900 for
metal ions which can be conjugated to antibodies for use as diagnostics
according to the
present invention. Examples of suitable enzymes include horseradish
peroxidase, alkaline
25 phosphatase, beta-galactosidase, or acetylcholinesterase; examples of
suitable prosthetic
group complexes include streptavidin/biotin and avidin/biotin; examples of
suitable
fluorescent materials include umbelliferone, fluorescein, fluorescein
isothiocyanate,
rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or
phycoerythrin; an example
of a luminescent material includes luminol; examples of bioluminescent
materials include
30 luciferase, luciferin, and aequorin; and examples of suitable radioactive
material include
125I, 131I, 11 lIn or 99Tc.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
76
Further, an antibody or fragment thereof may be conjugated to a therapeutic
moiety
such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic
agent or a radioactive
metal ion, e.g., alpha-emitters such as, for example, 213Bi. A cytotoxin or
cytotoxic agent
includes any agent that is detrimental to cells. Examples include paclitaxol,
cytochalasin B,
gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide,
vincristine,
vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione,
mitoxantrone,
mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine,
tetracaine,
lidocaine, propranolol, and puromycin and analogs or homologs thereof.
Therapeutic agents
include, but are not limited to, antimetabolites (e.g., methotrexate, 6-
mercaptopurine, 6-
thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g.,
mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and
lomustine
(CCNL~, cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin
C, and
cis- dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g.,
daunorubicin
(formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin
(formerly
actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic
agents
(e.g., vincristine and vinblastine).
The conjugates of the invention can be used for modifying a given biological
response, the therapeutic agent or drug moiety is not to be construed as
limited to classical
chemical therapeutic agents. For example, the drug moiety may be a protein or
polypeptide
possessing a desired biological activity. Such proteins may include, for
example, a toxin
such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein
such as tumor
necrosis factor, a-interferon, l3-interferon, nerve growth factor, platelet
derived growth factor,
tissue plasminogen activator, an apoptotic agent, e.g., TNF-alpha, TNF-beta,
AIM I (See,
International Publication No. WO 97/33899), AIM II (See, International
Publication No. WO
97/34911), Fas Ligand (Takahashi et al., Int. Immunol., 6:1567-1574 (1994)),
VEGI (See,
International Publication No. WO 99/23105), a thrombotic agent or an anti-
angiogenic agent,
e.g., angiostatin or endostatin; or, biological response modifiers such as,
for example,
lymphokines, interleukin-1 ("IL-1 "), interleukin-2 ("IL-2"), interleukin-6
("IL-6"),
granulocyte macrophage colony stimulating factor ("GM-CSF"), granulocyte
colony
stimulating factor ("G-CSF"), or other growth factors.
Antibodies may also be attached to solid supports, which are particularly
useful for
immunoassays or purification of the target antigen. Such solid supports
include, but are not


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
77
limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl
chloride or
polypropylene.
Techniques for conjugating such therapeutic moiety to antibodies are well
known,
see, e.g., Arnon et al., "Monoclonal Antibodies For Immunotargeting Of Drugs
In Cancer
Therapy", in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.),
pp. 243-56
(Alan R. Liss, Inc. 1985); Hellstrom et al., "Antibodies For Drug Delivery",
in Controlled
Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker,
Inc. 1987);
Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review",
in
Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et
al. (eds.), pp.
475-506 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic
Use Of
Radiolabeled Antibody In Cancer Therapy", in Monoclonal Antibodies For Cancer
Detection
And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and
Thorpe et al.,
"The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates",
Immunol. Rev.
62:119-58 (1982).
Alternatively, an antibody can be conjugated to a second antibody to form an
antibody
heteroconjugate as described by Segal in U.S. Patent No. 4,676,980, which is
incorporated
herein by reference in its entirety.
An antibody, with or without a therapeutic moiety conjugated to it,
administered alone
or in combination with cytotoxic factors) and/or cytokine(s) can be used as a
therapeutic.
Immunophenotyping
The antibodies of the invention may be utilized for immunophenotyping of cell
lines
and biological samples. The translation product of the gene of the present
invention may be
useful as a cell specific marker, or more specifically as a cellular marker
that is differentially
expressed at various stages of differentiation and/or maturation of particular
cell types.
Monoclonal antibodies directed against a specific epitope, or combination of
epitopes, will
allow for the screening of cellular populations expressing the marker. Various
techniques can
be utilized using monoclonal antibodies to screen for cellular populations
expressing the
marker(s), and include magnetic separation using antibody-coated magnetic
beads, "panning"
with antibody attached to a solid matrix (i.e., plate), and flow cytometry
(See, e.g., U.S.
Patent 5,985,660; and Morrison et al., Cell, 96:737-49 (1999)).


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
78
These techniques allow for the screening of particular populations of cells,
such as
might be found with hematological malignancies (i.e. minimal residual disease
(MRD) in
acute leukemic patients) and "non-self' cells in transplantations to prevent
Graft-versus-Host
Disease (GVHD). Alternatively, these techniques allow for the screening of
hematopoietic
stem and progenitor cells capable of undergoing proliferation and/or
differentiation, as might
be found in human umbilical cord blood.
Assays For Antibody Binding
The antibodies of the invention may be assayed for immunospecific binding by
any
method known in the art. The immunoassays which can be used include but are
not limited
to competitive and non-competitive assay systems using techniques such as
western blots,
radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich"
immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion
precipitin
reactions, immunodiffusion assays, agglutination assays, complement-fixation
assays,
immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, to
name
but a few. Such assays are routine and well known in the art (see, e.g.,
Ausubel et al, eds,
1994, Current Protocols in Molecular Biology, Vol. l, John Wiley & Sons, Inc.,
New York,
which is incorporated by reference herein in its entirety). Exemplary
immunoassays are
described briefly below (but are not intended by way of limitation).
Immunoprecipitation protocols generally comprise lysing a population of cells
in a
lysis buffer such as RIPA buffer (1 % NP-40 or Triton X- 100, 1 % sodium
deoxycholate,
0.1 % SDS, 0.15 M NaCI, 0.01 M sodium phosphate at pH 7.2, 1 % Trasylol)
supplemented
with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF,
aprotinin, sodium
vanadate), adding the antibody of interest to the cell lysate, incubating for
a period of time
(e.g., 1-4 hours) at 4° C, adding protein A and/or protein G sepharose
beads to the cell lysate,
incubating for about an hour or more at 4° C, washing the beads in
lysis buffer and
resuspending the beads in SDS/sample buffer. The ability of the antibody of
interest to
immunoprecipitate a particular antigen can be assessed by, e.g., western blot
analysis. One
of skill in the art would be knowledgeable as to the parameters that can be
modified to
increase the binding of the antibody to an antigen and decrease the background
(e.g., pre-
clearing the cell lysate with sepharose beads). For further discussion
regarding


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
79
immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current
Protocols in
Molecular Biology, Vol. l, John Wiley & Sons, Inc., New York at 10.16.1.
Western blot analysis generally comprises preparing protein samples,
electrophoresis
of the protein samples in a polyacrylamide gel (e.g., 8%- 20% SDS-PAGE
depending on the
molecular weight of the antigen), transferring the protein sample from the
polyacrylamide gel
to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in
blocking
solution (e.g., PBS with 3% BSA or non-fat milk), washing the membrane in
washing buffer
(e.g., PBS-Tween 20), blocking the membrane with primary antibody (the
antibody of
interest) diluted in blocking buffer, washing the membrane in washing buffer,
blocking the
membrane with a secondary antibody (which recognizes the primary antibody,
e.g., an anti-
human antibody) conjugated to an enzymatic substrate (e.g., horseradish
peroxidase or
alkaline phosphatase) or radioactive molecule (e.g., 32P or 125I) diluted in
blocking buffer,
washing the membrane in wash buffer, and detecting the presence of the
antigen. One of skill
in the art would be knowledgeable as to the parameters that can be modified to
increase the
signal detected and to reduce the background noise. For further discussion
regarding western
blot protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in
Molecular Biology,
Vol. 1, John Wiley & Sons, Inc., New York at 10.8.1.
ELISAs comprise preparing antigen, coating the well of a 96 well microtiter
plate
with the antigen, adding the antibody of interest conjugated to a detectable
compound such
as an enzymatic substrate (e.g., horseradish peroxidase or alkaline
phosphatase) to the well
and incubating for a period of time, and detecting the presence of the
antigen. In ELISAs the
antibody of interest does not have to be conjugated to a detectable compound;
instead, a
second antibody (which recognizes the antibody of interest) conjugated to a
detectable
compound may be added to the well. Further, instead of coating the well with
the antigen,
the antibody may be coated to the well. In this case, a second antibody
conjugated to a
detectable compound may be added following the addition of the antigen of
interest to the
coated well. One of skill in the art would be knowledgeable as to the
parameters that can be
modified to increase the signal detected as well as other variations of ELISAs
known in the
art. For further discussion regarding ELISAs see, e.g., Ausubel et al, eds,
1994, Current
Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at
11.2.1.
The binding affinity of an antibody to an antigen and the off rate of an
antibody-
antigen interaction can be determined by competitive binding assays. One
example of a


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
competitive binding assay is a radioimmunoassay comprising the incubation of
labeled
antigen (e.g., 3H or 125I) with the antibody of interest in the presence of
increasing amounts
of unlabeled antigen, and the detection of the antibody bound to the labeled
antigen. The
affinity of the antibody of interest for a particular antigen and the binding
off rates can be
5 determined from the data by scatchard plot analysis. Competition with a
second antibody
can also be determined using radioimmunoassays. In this case, the antigen is
incubated with
antibody of interest conjugated to a labeled compound (e.g., 3H or 125I) in
the presence of
increasing amounts of an unlabeled second antibody.
10 Therapeutic Uses
The present invention is further directed to antibody-based therapies which
involve
administering antibodies of the invention to an animal, preferably a mammal,
and most
preferably a human, patient for treating one or more of the disclosed
diseases, disorders, or
conditions. Therapeutic compounds of the invention include, but are not
limited to,
15 antibodies of the invention (including fragments, analogs and derivatives
thereof as described
herein) and nucleic acids encoding antibodies of the invention (including
fragments, analogs
and derivatives thereof and anti-idiotypic antibodies as described herein).
The antibodies of
the invention can be used to treat, inhibit or prevent diseases, disorders or
conditions
associated with aberrant expression and/or activity of a polypeptide of the
invention,
20 including, but not limited to, any one or more of the diseases, disorders,
or conditions
described herein. The treatment and/or prevention of diseases, disorders, or
conditions
associated with aberrant expression and/or activity of a polypeptide of the
invention
includes, but is not limited to, alleviating symptoms associated with those
diseases, disorders
or conditions. Antibodies of the invention may be provided in pharmaceutically
acceptable
25 compositions as known in the art or as described herein.
A summary of the ways in which the antibodies of the present invention may be
used
therapeutically includes binding polynucleotides or polypeptides of the
present invention
locally or systemically in the body or by direct cytotoxicity of the antibody,
e.g. as mediated
by complement (CDC) or by effector cells (ADCC). Some of these approaches are
described
30 in more detail below. Armed with the teachings provided herein, one of
ordinary skill in the
art will know how to use the antibodies of the present invention for
diagnostic, monitoring or
therapeutic purposes without undue experimentation.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
81
The antibodies of this invention may be advantageously utilized in combination
with
other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic
growth
factors (such as, e.g., IL-2, IL-3 and IL-7), for example, which serve to
increase the number
or activity of effector cells which interact with the antibodies.
S The antibodies of the invention may be administered alone or in combination
with
other types of treatments (e.g., radiation therapy, chemotherapy, hormonal
therapy,
immunotherapy and anti-tumor agents). Generally, administration of products of
a species
origin or species reactivity (in the case of antibodies) that is the same
species as that of the
patient is preferred. Thus, in a preferred embodiment, human antibodies,
fragments
derivatives, analogs, or nucleic acids, are administered to a human patient
for therapy or
prophylaxis.
It is preferred to use high affinity and/or potent in vivo inhibiting and/or
neutralizing
antibodies against polypeptides or polynucleotides of the present invention,
fragments or
regions thereof, for both immunoassays directed to and therapy of disorders
related to
polynucleotides or polypeptides, including fragments thereof, of the present
invention. Such
antibodies, fragments, or regions, will preferably have an affinity for
polynucleotides or
polypeptides of the invention, including fragments thereof. Preferred binding
affinities
include those with a dissociation constant or Kd less than 5 X 10-Z M, 10-2 M,
5 X 10-3 M,
10-3 M, S X 10-4 M, 10-4 M, 5 X 10-5 M, 10-5 M, 5 X 10-6 M, 10-6 M, 5 X 10-7
M, 10-~ M, 5 X
10~g M, 10-$ M, S X 10-9 M, 109 M, 5 X 10-' ° M, 10-' ° M, 5 X
10-'' M, 10-" M, 5 X 10-' 2 M,
10-' Z M, 5 X 10-' 3 M, 10-' 3 M, 5 X 10-' 4 M, 10-' 4 M, 5 X 10-' S M, and 10-
' S M.
Gene Therapy
In a specific embodiment, nucleic acids comprising sequences encoding
antibodies or
functional derivatives thereof, are administered to treat, inhibit or prevent
a disease or
disorder associated with aberrant expression and/or activity of a polypeptide
of the invention,
by way of gene therapy. Gene therapy refers to therapy performed by the
administration to a
subject of an expressed or expressible nucleic acid. In this embodiment of the
invention, the
nucleic acids produce their encoded protein that mediates a therapeutic
effect.
Any of the methods for gene therapy available in the art can be used according
to the
present invention. Exemplary methods are described below.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
82
For general reviews of the methods of gene therapy, see Goldspiel et al.,
Clinical
Pharmacy 12:488-505 (1993); Wu and Wu, Biotherapy 3:87-95 (1991); Tolstoshev,
Ann.
Rev. Pharmacol. Toxicol. 32:573-596 (1993); Mulligan, Science 260:926-932
(1993); and
Morgan and Anderson, Ann. Rev. Biochem. 62:191-217 (1993); May, TIBTECH
11(5):155-
21 S (1993). Methods commonly known in the art of recombinant DNA technology
which can
be used are described in Ausubel et al. (eds.), Current Protocols in Molecular
Biology, John
Wiley & Sons, NY (1993); and Kriegler, Gene Transfer and Expression, A
Laboratory
Manual, Stockton Press, NY ( 1990).
In a preferred aspect, the compound comprises nucleic acid sequences encoding
an
antibody, said nucleic acid sequences being part of expression vectors that
express the
antibody or fragments or chimeric proteins or heavy or light chains thereof in
a suitable host.
In particular, such nucleic acid sequences have promoters operably linked to
the antibody
coding region, said promoter being inducible or constitutive, and, optionally,
tissue-specific.
In another particular embodiment, nucleic acid molecules are used in which the
antibody
coding sequences and any other desired sequences are flanked by regions that
promote
homologous recombination at a desired site in the genome, thus providing for
intrachromosomal expression of the antibody encoding nucleic acids (Koller and
Smithies,
Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); Zijlstra et al., Nature
342:435-438 (1989).
In specific embodiments, the expressed antibody molecule is a single chain
antibody;
alternatively, the nucleic acid sequences include sequences encoding both the
heavy and
light chains, or fragments thereof, of the antibody.
Delivery of the nucleic acids into a patient may be either direct, in which
case the
patient is directly exposed to the nucleic acid or nucleic acid- carrying
vectors, or indirect, in
which case, cells are first transformed with the nucleic acids in vitro, then
transplanted into
the patient. These two approaches are known, respectively, as in vivo or ex
vivo gene
therapy.
In a specific embodiment, the nucleic acid sequences are directly administered
in
vivo, where it is expressed to produce the encoded product. This can be
accomplished by
any of numerous methods known in the art, e.g., by constructing them as part
of an
appropriate nucleic acid expression vector and administering it so that they
become
intracellular, e.g., by infection using defective or attenuated retrovirals or
other viral vectors
(see U.S. Patent No. 4,980,286), or by direct injection of naked DNA, or by
use of


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
83
microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating
with lipids or
cell-surface receptors or transfecting agents, encapsulation in liposomes,
microparticles, or
microcapsules, or by administering them in linkage to a peptide which is known
to enter the
nucleus, by administering it in linkage to a ligand subject to receptor-
mediated endocytosis
(see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)) (which can be used
to target
cell types specifically expressing the receptors), etc. In another embodiment,
nucleic acid-
ligand complexes can be formed in which the ligand comprises a fusogenic viral
peptide to
disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
In yet another
embodiment, the nucleic acid can be targeted in vivo for cell specific uptake
and expression,
by targeting a specific receptor (see, e.g., PCT Publications WO 92/06180; WO
92/22635;
W092/20316; W093/14188, WO 93/20221). Alternatively, the nucleic acid can be
introduced intracellularly and incorporated within host cell DNA for
expression, by
homologous recombination (Koller and Smithies, Proc. Natl. Acad. Sci. USA
86:8932-8935
(1989); Zijlstra et al., Nature 342:435-438 (1989)).
In a specific embodiment, viral vectors that contains nucleic acid sequences
encoding
an antibody of the invention are used. For example, a retroviral vector can be
used (see
Miller et al., Meth. Enzymol. 217:581-599 (1993)). These retroviral vectors
contain the
components necessary for the correct packaging of the viral genome and
integration into the
host cell DNA. The nucleic acid sequences encoding the antibody to be used in
gene therapy
are cloned into one or more vectors, which facilitates delivery of the gene
into a patient.
More detail about retroviral vectors can be found in Boesen et al., Biotherapy
6:291-302
(1994), which describes the use of a retroviral vector to deliver the mdrl
gene to
hematopoietic stem cells in order to make the stem cells more resistant to
chemotherapy.
Other references illustrating the use of retroviral vectors in gene therapy
are: Clowes et al., J.
Clin. Invest. 93:644-651 (1994); Kiem et al., Blood 83:1467-1473 (1994);
Salmons and
Gunzberg, Human Gene Therapy 4:129-141 (1993); and Grossman and Wilson, Curr.
Opin.
in Genetics and Devel. 3:110-114 (1993).
Adenoviruses are other viral vectors that can be used in gene therapy.
Adenoviruses
are especially attractive vehicles for delivering genes to respiratory
epithelia. Adenoviruses
naturally infect respiratory epithelia where they cause a mild disease. Other
targets for
adenovirus-based delivery systems are liver, the central nervous system,
endothelial cells,
and muscle. Adenoviruses have the advantage of being capable of infecting non-
dividing


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
84
cells. Kozarsky and Wilson, Current Opinion in Genetics and Development 3:499-
503
(1993) present a review of adenovirus-based gene therapy. Bout et al., Human
Gene
Therapy 5:3-10 (1994) demonstrated the use of adenovirus vectors to transfer
genes to the
respiratory epithelia of rhesus monkeys. Other instances of the use of
adenoviruses in gene
S therapy can be found in Rosenfeld et al., Science 252:431-434 (1991);
Rosenfeld et al., Cell
68:143- 155 (1992); Mastrangeli et al., J. Clin. Invest. 91:225-234 (1993);
PCT Publication
W094/12649; and Wang, et al., Gene Therapy 2:775-783 (1995). In a preferred
embodiment, adenovirus vectors are used.
Adeno-associated virus (AAV) has also been proposed for use in gene therapy
(Walsh
et al., Proc. Soc. Exp. Biol. Med. 204:289-300 (1993); U.S. Patent No.
5,436,146).
Another approach to gene therapy involves transferring a gene to cells in
tissue
culture by such methods as electroporation, lipofection, calcium phosphate
mediated
transfection, or viral infection. Usually, the method of transfer includes the
transfer of a
selectable marker to the cells. The cells are then placed under selection to
isolate those cells
1 S that have taken up and are expressing the transferred gene. Those cells
are then delivered to a
patient.
In this embodiment, the nucleic acid is introduced into a cell prior to
administration in
vivo of the resulting recombinant cell. Such introduction can be carned out by
any method
known in the art, including but not limited to transfection, electroporation,
microinjection,
infection with a viral or bacteriophage vector containing the nucleic acid
sequences, cell
fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer,
spheroplast
fusion, etc. Numerous techniques are known in the art for the introduction of
foreign genes
into cells (see, e.g., Loeffler and Behr, Meth. Enzymol. 217:599-618 (1993);
Cohen et al.,
Meth. Enzymol. 217:618-644 (1993); Cline, Pharmac. Ther. 29:69-92m (1985) and
may be
used in accordance with the present invention, provided that the necessary
developmental
and physiological functions of the recipient cells are not disrupted. The
technique should
provide for the stable transfer of the nucleic acid to the cell, so that the
nucleic acid is
expressible by the cell and preferably heritable and expressible by its cell
progeny.
The resulting recombinant cells can be delivered to a patient by various
methods
known in the art. Recombinant blood cells (e.g., hematopoietic stem or
progenitor cells) are
preferably administered intravenously. The amount of cells envisioned for use
depends on
the desired effect, patient state, etc., and can be determined by one skilled
in the art.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
Cells into which a nucleic acid can be introduced for purposes of gene therapy
encompass any desired, available cell type, and include but are not limited to
epithelial cells,
endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes;
blood cells such as T
lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils,
5 megakaryocytes, granulocytes; various stem or progenitor cells, in
particular hematopoietic
stem or progenitor cells, e.g., as obtained from bone marrow, umbilical cord
blood,
peripheral blood, fetal liver, etc.
In a preferred embodiment, the cell used for gene therapy is autologous to the
patient.
In an embodiment in which recombinant cells are used in gene therapy, nucleic
acid
10 sequences encoding an antibody are introduced into the cells such that they
are expressible
by the cells or their progeny, and the recombinant cells are then administered
in vivo for
therapeutic effect. In a specific embodiment, stem or progenitor cells are
used. Any stem
and/or progenitor cells which can be isolated and maintained in vitro can
potentially be used
in accordance with this embodiment of the present invention (see e.g. PCT
Publication WO
15 94/08598; Stemple and Anderson, Cell 71:973-985 (1992); Rheinwald, Meth.
Cell Bio.
21 A:229 ( 1980); and Pittelkow and Scott, Mayo Clinic Proc. 61:771 ( 1986)).
In a specific embodiment, the nucleic acid to be introduced for purposes of
gene
therapy comprises an inducible promoter operably linked to the coding region,
such that
expression of the nucleic acid is controllable by controlling the presence or
absence of the
20 appropriate inducer of transcription. Demonstration of Therapeutic or
Prophylactic Activity
The compounds or pharmaceutical compositions of the invention are preferably
tested
in vitro, and then in vivo for the desired therapeutic or prophylactic
activity, prior to use in
humans. For example, in vitro assays to demonstrate the therapeutic or
prophylactic utility of
a compound or pharmaceutical composition include, the effect of a compound on
a cell line
25 or a patient tissue sample. The effect of the compound or composition on
the cell line and/or
tissue sample can be determined utilizing techniques known to those of skill
in the art
including, but not limited to, rosette formation assays and cell lysis assays.
In accordance
with the invention, in vitro assays which can be used to determine whether
administration of
a specific compound is indicated, include in vitro cell culture assays in
which a patient tissue
30 sample is grown in culture, and exposed to or otherwise administered a
compound, and the
effect of such compound upon the tissue sample is observed.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
86
TherapeuticlProphylactic Administration and Composition
The invention provides methods of treatment, inhibition and prophylaxis by
administration to a subject of an effective amount of a compound or
pharmaceutical
composition of the invention, preferably a polypeptide or antibody of the
invention. In a
S preferred aspect, the compound is substantially purified (e.g.,
substantially free from
substances that limit its effect or produce undesired side-effects). The
subject is preferably
an animal, including but not limited to animals such as cows, pigs, horses,
chickens, cats,
dogs, etc., and is preferably a mammal, and most preferably human.
Formulations and methods of administration that can be employed when the
compound comprises a nucleic acid or an immunoglobulin are described above;
additional
appropriate formulations and routes of administration can be selected from
among those
described herein below.
Various delivery systems are known and can be used to administer a compound of
the
invention, e.g., encapsulation in liposomes, microparticles, microcapsules,
recombinant cells
capable of expressing the compound, receptor-mediated endocytosis (see, e.g.,
Wu and Wu,
J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part
of a retroviral or
other vector, etc. Methods of introduction include but are not limited to
intradermal,
intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal,
epidural, and oral
routes. The compounds or compositions may be administered by any convenient
route, for
example by infusion or bolus injection, by absorption through epithelial or
mucocutaneous
linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be
administered
together with other biologically active agents. Administration can be systemic
or local. In
addition, it may be desirable to introduce the pharmaceutical compounds or
compositions of
the invention into the central nervous system by any suitable route, including
intraventricular
and intrathecal injection; intraventricular injection may be facilitated by an
intraventricular
catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
Pulmonary
administration can also be employed, e.g., by use of an inhaler or nebulizer,
and formulation
with an aerosolizing agent.
In a specific embodiment, it may be desirable to administer the pharmaceutical
compounds or compositions of the invention locally to the area in need of
treatment; this may
be achieved by, for example, and not by way of limitation, local infusion
during surgery,
topical application, e.g., in conjunction with a wound dressing after surgery,
by injection, by


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
87
means of a catheter, by means of a suppository, or by means of an implant,
said implant being
of a porous, non-porous, or gelatinous material, including membranes, such as
sialastic
membranes, or fibers. Preferably, when administering a protein, including an
antibody, of
the invention, care must be taken to use materials to which the protein does
not absorb.
In another embodiment, the compound or composition can be delivered in a
vesicle,
in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et
al., in
Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and
Fidler
(eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 317-
327; see
generally ibid.)
In yet another embodiment, the compound or composition can be delivered in a
controlled release system. In one embodiment, a pump may be used (see Langer,
supra;
Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery
88:507 (1980);
Saudek et al., N. Engl. J. Med. 321:574 (1989)). In another embodiment,
polymeric
materials can be used (see Medical Applications of Controlled Release, Langer
and Wise
(eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug
Bioavailability, Drug
Product Design and Performance, Smolen and Ball (eds.), Wiley, New York
(1984); Ranger
and Peppas, J., Macromol. Sci. Rev. Macromol. Chem. 23:61 (1983); see also
Levy et al.,
Science 228:190 (1985); During et al., Ann. Neurol. 25:351 (1989); Howard et
al.,
J.Neurosurg. 71:105 (1989)). In yet another embodiment, a controlled release
system can be
placed in proximity of the therapeutic target, i.e., the brain, thus requiring
only a fraction of
the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled
Release, supra,
vol. 2, pp. 115-138 (1984)).
Other controlled release systems are discussed in the review by Langer
(Science
249:1527-1533 (1990)).
In a specific embodiment where the compound of the invention is a nucleic acid
encoding a protein, the nucleic acid can be administered in vivo to promote
expression of its
encoded protein, by constructing it as part of an appropriate nucleic acid
expression vector
and administering it so that it becomes intracellular, e.g., by use of a
retroviral vector (see
U.S. Patent No. 4,980,286), or by direct injection, or by use of microparticle
bombardment
(e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface
receptors or
transfecting agents, or by administering it in linkage to a homeobox- like
peptide which is
known to enter the nucleus (see e.g., Joliot et al., Proc. Natl. Acad. Sci.
USA 88:1864-1868


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
88
(1991)), etc. Alternatively, a nucleic acid can be introduced intracellularly
and incorporated
within host cell DNA for expression, by homologous recombination.
The present invention also provides pharmaceutical compositions. Such
compositions
comprise a therapeutically effective amount of a compound, and a
pharmaceutically
acceptable carrier. In a specific embodiment, the term "pharmaceutically
acceptable" means
approved by a regulatory agency of the Federal or a state government or listed
in the U.S.
Pharmacopeia or other generally recognized pharmacopeia for use in animals,
and more
particularly in humans. The term "carrier" refers to a diluent, adjuvant,
excipient, or vehicle
with which the therapeutic is administered. Such pharmaceutical carriers can
be sterile
liquids, such as water and oils, including those of petroleum, animal,
vegetable or synthetic
origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
Water is a
preferred carrier when the pharmaceutical composition is administered
intravenously. Saline
solutions and aqueous dextrose and glycerol solutions can also be employed as
liquid carriers,
particularly for injectable solutions. Suitable pharmaceutical excipients
include starch,
glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel,
sodium stearate, glycerol
monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene,
glycol, water,
ethanol and the like. The composition, if desired, can also contain minor
amounts of wetting
or emulsifying agents, or pH buffering agents. These compositions can take the
form of
solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-
release
formulations and the like. The composition can be formulated as a suppository,
with
traditional binders and carriers such as triglycerides. Oral formulation can
include standard
carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium
stearate,
sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable
pharmaceutical Garners are described in "Remington's Pharmaceutical Sciences"
by E.W.
Martin. Such compositions will contain a therapeutically effective amount of
the compound,
preferably in purified form, together with a suitable amount of carrier so as
to provide the
form for proper administration to the patient. The formulation should suit the
mode of
administration.
In a preferred embodiment, the composition is formulated in accordance with
routine
procedures as a pharmaceutical composition adapted for intravenous
administration to
human beings. Typically, compositions for intravenous administration are
solutions in sterile
isotonic aqueous buffer. Where necessary, the composition may also include a
solubilizing


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
89
agent and a local anesthetic such as lignocaine to ease pain at the site of
the injection.
Generally, the ingredients are supplied either separately or mixed together in
unit dosage
form, for example, as a dry lyophilized powder or water free concentrate in a
hermetically
sealed container such as an ampoule or sachette indicating the quantity of
active agent.
Where the composition is to be administered by infusion, it can be dispensed
with an
infusion bottle containing sterile pharmaceutical grade water or saline. Where
the
composition is administered by injection, an ampoule of sterile water for
injection or saline
can be provided so that the ingredients may be mixed prior to administration.
The compounds of the invention can be formulated as neutral or salt forms.
Pharmaceutically acceptable salts include those formed with anions such as
those derived
from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those
formed with
canons such as those derived from sodium, potassium, ammonium, calcium, fernc
hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine,
procaine, etc.
The amount of the compound of the invention which will be effective in the
treatment, inhibition and prevention of a disease or disorder associated with
aberrant
expression and/or activity of a polypeptide of the invention can be determined
by standard
clinical techniques. In addition, in vitro assays may optionally be employed
to help identify
optimal dosage ranges. The precise dose to be employed in the formulation will
also depend
on the route of administration, and the seriousness of the disease or
disorder, and should be
decided according to the judgment of the practitioner and each patient's
circumstances.
Effective doses may be extrapolated from dose-response curves derived from in
vitro or
animal model test systems.
For antibodies, the dosage administered to a patient is typically 0.1 mg/kg to
100
mg/kg of the patient's body weight. Preferably, the dosage administered to a
patient is
between 0.1 mg/kg and 20 mg/kg of the patient's body weight, more preferably 1
mg/kg to 10
mg/kg of the patient's body weight. Generally, human antibodies have a longer
half life
within the human body than antibodies from other species due to the immune
response to the
foreign polypeptides. Thus, lower dosages of human antibodies and less
frequent
administration is often possible. Further, the dosage and frequency of
administration of
antibodies of the invention may be reduced by enhancing uptake and tissue
penetration (e.g.,
into the brain) of the antibodies by modifications such as, for example,
lipidation.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
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. Optionally associated with such containers) can be a notice in
the form
prescribed by a governmental agency regulating the manufacture, use or sale of
S pharmaceuticals or biological products, which notice reflects approval by
the agency of
manufacture, use or sale for human administration.
Diagnosis and Imaging
Labeled antibodies, and derivatives and analogs thereof, which specifically
bind to a
10 polypeptide of interest can be used for diagnostic purposes to detect,
diagnose, or monitor
diseases, disorders, and/or conditions associated with the aberrant expression
and/or activity
of a polypeptide of the invention. The invention provides for the detection of
aberrant
expression of a polypeptide of interest, comprising (a) assaying the
expression of the
polypeptide of interest in cells or body fluid of an individual using one or
more antibodies
15 specific to the polypeptide interest and (b) comparing the level of gene
expression with a
standard gene expression level, whereby an increase or decrease in the assayed
polypeptide
gene expression level compared to the standard expression level is indicative
of aberrant
expression.
The invention provides a diagnostic assay for diagnosing a disorder,
comprising (a)
20 assaying the expression of the polypeptide of interest in cells or body
fluid of an individual
using one or more antibodies specific to the polypeptide interest and (b)
comparing the level
of gene expression with a standard gene expression level, whereby an increase
or decrease in
the assayed polypeptide gene expression level compared to the standard
expression level is
indicative of a particular disorder. With respect to cancer, the presence of a
relatively high
25 amount of transcript in biopsied tissue from an individual may indicate a
predisposition for
the development of the disease, or may provide a means for detecting the
disease prior to the
appearance of actual clinical symptoms. A more definitive diagnosis of this
type may allow
health professionals to employ preventative measures or aggressive treatment
earlier thereby
preventing the development or further progression of the cancer.
30 Antibodies of the invention can be used to assay protein levels in a
biological sample
using classical immunohistological methods known to those of skill in the art
(e.g., see
Jalkanen, et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, et al., J. Cell
. Biol. 105:3087-


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
91
3096 ( 1987)). Other antibody-based methods useful for detecting protein gene
expression
include immunoassays, such as the enzyme linked immunosorbent assay (ELISA)
and the
radioimmunoassay (RIA). Suitable antibody assay labels are known in the art
and include
enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine (125I,
121I), carbon
(14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99Tc);
luminescent labels,
such as luminol; and fluorescent labels, such as fluorescein and rhodamine,
and biotin.
One aspect of the invention is the detection and diagnosis of a disease or
disorder
associated with aberrant expression of a polypeptide of interest in an animal,
preferably a
mammal and most preferably a human. In one embodiment, diagnosis comprises: a)
administering (for example, parenterally, subcutaneously, or
intraperitoneally) to a subject an
effective amount of a labeled molecule which specifically binds to the
polypeptide of
interest; b) waiting for a time interval following the administering for
permitting the labeled
molecule to preferentially concentrate at sites in the subject where the
polypeptide is
expressed (and for unbound labeled molecule to be cleared to background
level); c)
determining background level; and d) detecting the labeled molecule in the
subject, such that
detection of labeled molecule above the background level indicates that the
subject has a
particular disease or disorder associated with aberrant expression of the
polypeptide of
interest. Background level can be determined by various methods including,
comparing the
amount of labeled molecule detected to a standard value previously determined
for a
particular system.
It will be understood in the art that the size of the subject and the imaging
system used
will determine the quantity of imaging moiety needed to produce diagnostic
images. In the
case of a radioisotope moiety, for a human subject, the quantity of
radioactivity injected will
normally range from about 5 to 20 millicuries of 99mTc. The labeled antibody
or antibody
fragment will then preferentially accumulate at the location of cells which
contain the
specific protein. In vivo tumor imaging is described in S.W. Burchiel et al.,
"Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments."
(Chapter 13
in Tumor Imaging: The Radiochemical Detection of Cancer, S.W. Burchiel and B.
A.
Rhodes, eds., Masson Publishing Inc. (1982).
Depending on several variables, including the type of label used and the mode
of
administration, the time interval following the administration for permitting
the labeled
molecule to preferentially concentrate at sites in the subject and for unbound
labeled


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
92
molecule to be cleared to background level is 6 to 48 hours or 6 to 24 hours
or 6 to 12 hours.
In another embodiment the time interval following administration is 5 to 20
days or 5 to 10
days.
In an embodiment, monitoring of the disease or disorder is carned out by
repeating
S the method for diagnosing the disease or disease, for example, one month
after initial
diagnosis, six months after initial diagnosis, one year after initial
diagnosis, etc.
Presence of the labeled molecule can be detected in the patient using methods
known
in the art for in vivo scanning. These methods depend upon the type of label
used. Skilled
artisans will be able to determine the appropriate method for detecting a
particular label.
Methods and devices that may be used in the diagnostic methods of the
invention include, but
are not limited to, computed tomography (CT), whole body scan such as position
emission
tomography (PET), magnetic resonance imaging (MRI), and sonography.
In a specific embodiment, the molecule is labeled with a radioisotope and is
detected
in the patient using a radiation responsive surgical instrument (Thurston et
al., U.S. Patent
No. 5,441,050). In another embodiment, the molecule is labeled with a
fluorescent
compound and is detected in the patient using a fluorescence responsive
scanning instrument.
In another embodiment, the molecule is labeled with a positron emitting metal
and is detected
in the patent using positron emission-tomography. In yet another embodiment,
the molecule
is labeled with a paramagnetic label and is detected in a patient using
magnetic resonance
imaging (MRI).
Kits
The present invention provides kits that can be used in the above methods. In
one
embodiment, a kit comprises an antibody of the invention, preferably a
purified antibody, in
one or more containers. In a specific embodiment, the kits of the present
invention contain a
substantially isolated polypeptide comprising an epitope which is specifically
immunoreactive with an antibody included in the kit. Preferably, the kits of
the present
invention further comprise a control antibody which does not react with the
polypeptide of
interest. In another specific embodiment, the kits of the present invention
contain a means
for detecting the binding of an antibody to a polypeptide of interest (e.g.,
the antibody may be
conjugated to a detectable substrate such as a fluorescent compound, an
enzymatic substrate,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
93
a radioactive compound or a luminescent compound, or a second antibody which
recognizes
the first antibody may be conjugated to a detectable substrate).
In another specific embodiment of the present invention, the kit is a
diagnostic kit for
use in screening serum containing antibodies specific against proliferative
and/or cancerous
polynucleotides and polypeptides. Such a kit may include a control antibody
that does not
react with the polypeptide of interest. Such a kit may include a substantially
isolated
polypeptide antigen comprising an epitope which is specifically immunoreactive
with at least
one anti-polypeptide antigen antibody. Further, such a kit includes means for
detecting the
binding of said antibody to the antigen (e.g., the antibody may be conjugated
to a fluorescent
compound such as fluorescein or rhodamine which can be detected by flow
cytometry). In
specific embodiments, the kit may include a recombinantly produced or
chemically
synthesized polypeptide antigen. The polypeptide antigen of the kit may also
be attached to a
solid support.
In a more specific embodiment the detecting means of the above-described kit
includes a solid support to which said polypeptide antigen is attached. Such a
kit may also
include a non-attached reporter-labeled anti-human antibody. In this
embodiment, binding of
the antibody to the polypeptide antigen can be detected by binding of the said
reporter
labeled antibody.
In an additional embodiment, the invention includes a diagnostic kit for use
in
screening serum containing antigens of the polypeptide of the invention. The
diagnostic kit
includes a substantially isolated antibody specifically immunoreactive with
polypeptide or
polynucleotide antigens, and means for detecting the binding of the
polynucleotide or
polypeptide antigen to the antibody. In one embodiment, the antibody is
attached to a solid
support. In a specific embodiment, the antibody may be a monoclonal antibody.
The
detecting means of the kit may include a second, labeled monoclonal antibody.
Alternatively, or in addition, the detecting means may include a labeled,
competing antigen.
In one diagnostic configuration, test serum is reacted with a solid phase
reagent
having a surface-bound antigen obtained by the methods of the present
invention. After
binding with specific antigen antibody to the reagent and removing unbound
serum
components by washing, the reagent is reacted with reporter-labeled anti-human
antibody to
bind reporter to the reagent in proportion to the amount of bound anti-antigen
antibody on the
solid support. The reagent is again washed to remove unbound labeled antibody,
and the


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
94
amount of reporter associated with the reagent is determined. Typically, the
reporter is an
enzyme which is detected by incubating the solid phase in the presence of a
suitable
fluorometric, luminescent or colorimetric substrate (Sigma, St. Louis, MO).
The solid surface reagent in the above assay is prepared by known techniques
for
attaching protein material to solid support material, such as polymeric beads,
dip sticks, 96-
well plate or filter material. These attachment methods generally include non-
specific
adsorption of the protein to the support or covalent attachment of the
protein, typically
through a free amine group, to a chemically reactive group on the solid
support, such as an
activated carboxyl, hydroxyl, or aldehyde group. Alternatively, streptavidin
coated plates can
be used in conjunction with biotinylated antigen(s).
Thus, the invention provides an assay system or kit for carrying out this
diagnostic
method. The kit generally includes a support with surface- bound recombinant
antigens, and a
reporter-labeled anti-human antibody for detecting surface-bound anti-antigen
antibody.
Uses of the Polynucleotides
Each of the polynucleotides identified herein can be used in numerous ways as
reagents. The following description should be considered exemplary and
utilizes known
techniques.
The polynucleotides of the present invention are useful for chromosome
identification. There exists an ongoing need to identify new chromosome
markers, since few
chromosome marking reagents, based on actual sequence data (repeat
polymorphisms), are
presently available. Each sequence is specifically targeted to and can
hybridize with a
particular location on an individual human chromosome, thus each
polynucleotide of the
present invention can routinely be used as a chromosome marker using
techniques known in
the art.
Briefly, sequences can be mapped to chromosomes by preparing PCR primers
(preferably at least 15 by (e.g., 15-25 bp) from the sequences shown in SEQ ID
NO:X.
Primers can optionally be selected using computer analysis so that primers do
not span more
than one predicted exon in the genomic DNA. These primers are then used for
PCR
screening of somatic cell hybrids containing individual human chromosomes.
Only those


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
hybrids containing the human gene corresponding to SEQ ID NO:X will yield an
amplified
fragment.
Similarly, somatic hybrids provide a rapid method of PCR mapping the
polynucleotides to particular chromosomes. Three or more clones can be
assigned per day
5 using a single thermal cycler. Moreover, sublocalization of the
polynucleotides can be
achieved with panels of specific chromosome fragments. Other gene mapping
strategies that
can be used include in situ hybridization, prescreening with labeled flow-
sorted
chromosomes, preselection by hybridization to construct chromosome specific-
cDNA
libraries, and computer mapping techniques (See, e.g., Shuler, Trends
Biotechnol 16:456-459
10 (1998) which is hereby incorporated by reference in its entirety).
Precise chromosomal location of the polynucleotides can also be achieved using
fluorescence in situ hybridization (FISH) of a metaphase chromosomal spread.
This
technique uses polynucleotides as short as 500 or 600 bases; however,
polynucleotides 2,000
4,000 by are preferred. For a review of this technique, see Verma et al.,
"Human
15 Chromosomes: a Manual of Basic Techniques," Pergamon Press, New York
(1988).
For chromosome mapping, the polynucleotides can be used individually (to mark
a
single chromosome or a single site on that chromosome) or in panels (for
marking multiple
sites and/or multiple chromosomes).
Thus, the present invention also provides a method for chromosomal
localization
20 which involves (a) preparing PCR primers from the polynucleotide sequences
in Table 1 and
SEQ ID NO:X and (b) screening somatic cell hybrids containing individual
chromosomes.
The polynucleotides of the present invention would likewise be useful for
radiation
hybrid mapping, HAPPY mapping, and long range restriction mapping. For a
review of these
techniques and others known in the art, see, e.g. Dear, "Genome Mapping: A
Practical
25 Approach," IRL Press at Oxford University Press, London (1997); Aydin, J.
Mol. Med.
77:691-694 (1999); Hacia et al., Mol. Psychiatry 3:483-492 (1998); Hernck et
al.,
Chromosome Res. 7:409-423 (1999); Hamilton et al., Methods Cell Biol. 62:265-
280 (2000);
and/or Ott, J. Hered. 90:68-70 (1999) each of which is hereby incorporated by
reference in its
entirety.
30 Once a polynucleotide has been mapped to a precise chromosomal location,
the
physical position of the polynucleotide can be used in linkage analysis.
Linkage analysis
establishes coinheritance between a chromosomal location and presentation of a
particular


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
96
disease. (Disease mapping data are found, for example, in V. McKusick,
Mendelian
Inheritance in Man (available on line through Johns Hopkins University Welch
Medical
Library).) Assuming 1 megabase mapping resolution and one gene per 20 kb, a
cDNA
precisely localized to a chromosomal region associated with the disease could
be one of 50
500 potential causative genes.
Thus, once coinheritance is established, differences in a polynucleotide of
the
invention and the corresponding gene between affected and unaffected
individuals can be
examined. First, visible structural alterations in the chromosomes, such as
deletions or
translocations, are examined in chromosome spreads or by PCR. If no structural
alterations
exist, the presence of point mutations are ascertained. Mutations observed in
some or all
affected individuals, but not in normal individuals, indicates that the
mutation may cause the
disease. However, complete sequencing of the polypeptide and the corresponding
gene from
several normal individuals is required to distinguish the mutation from a
polymorphism. If a
new polymorphism is identified, this polymorphic polypeptide can be used for
further linkage
analysis.
Furthermore, increased or decreased expression of the gene in affected
individuals as
compared to unaffected individuals can be assessed using the polynucleotides
of the
invention. Any of these alterations (altered expression, chromosomal
rearrangement, or
mutation) can be used as a diagnostic or prognostic marker.
Thus, the invention also provides a diagnostic method useful during diagnosis
of a
disorder, involving measuring the expression level of polynucleotides of the
present invention
in cells or body fluid from an individual and comparing the measured gene
expression level
with a standard level of polynucleotide expression level, whereby an increase
or decrease in
the gene expression level compared to the standard is indicative of a
disorder.
In still another embodiment, the invention includes a kit for analyzing
samples for the
presence of proliferative and/or cancerous polynucleotides derived from a test
subject. In a
general embodiment, the kit includes at least one polynucleotide probe
containing a
nucleotide sequence that will specifically hybridize with a polynucleotide of
the invention
and a suitable container. In a specific embodiment, the kit includes two
polynucleotide probes
defining an internal region of the polynucleotide of the invention, where each
probe has one
strand containing a 31'mer-end internal to the region. In a further
embodiment, the probes
may be useful as primers for polymerase chain reaction amplification.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
97
Where a diagnosis of a related disorder, including, for example, diagnosis of
a tumor,
has already been made according to conventional methods, the present invention
is useful as
a prognostic indicator, whereby patients exhibiting enhanced or depressed
polynucleotide of
the invention expression will experience a worse clinical outcome relative to
patients
expressing the gene at a level nearer the standard level.
By ~"measuring the expression level of polynucleotides of the invention" is
intended
qualitatively or quantitatively measuring or estimating the level of the
polypeptide of the
invention or the level of the mRNA encoding the polypeptide of the invention
in a first
biological sample either directly (e.g., by determining or estimating absolute
protein level or
mRNA level) or relatively (e.g., by comparing to the polypeptide level or mRNA
level in a
second biological sample). Preferably, the polypeptide level or mRNA level in
the first
biological sample is measured or estimated and compared to a standard
polypeptide level or
mRNA level, the standard being taken from a second biological sample obtained
from an
individual not having the related disorder or being determined by averaging
levels from a
1 S population of individuals not having a related disorder. As will be
appreciated in the art,
once a standard polypeptide level or mRNA level is known, it can be used
repeatedly as a
standard for comparison.
By "biological sample" is intended any biological sample obtained from an
individual,
body fluid, cell line, tissue culture, or other source which contains
polypeptide of the present
invention or the corresponding mRNA. As indicated, biological samples include
body fluids
(such as semen, lymph, sera, plasma, urine, synovial fluid and spinal fluid)
which contain the
polypeptide of the present invention, and tissue sources found to express the
polypeptide of
the present invention. Methods for obtaining tissue biopsies and body fluids
from mammals
are well known in the art. Where the biological sample is to include mRNA, a
tissue biopsy
is the preferred source. '
The methods) provided above may preferrably be applied in a diagnostic method
and/or kits in which polynucleotides and/or polypeptides of the invention are
attached to a
solid support. In one exemplary method, the support may be a "gene chip" or a
"biological
chip" as described in US Patents 5,837,832, 5,874,219, and 5,856,174. Further,
such a gene
chip with polynucleotides of the invention attached may be used to identify
polymorphisms
between the isolated polynucleotide sequences of the invention, with
polynucleotides isolated
from a test subject. The knowledge of such polymorphisms (i.e. their location,
as well as,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
98
their existence) would be beneficial in identifying disease loci for many
disorders, such as for
example, in neural disorders, immune system disorders, muscular disorders,
reproductive
disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular
disorders, renal
disorders, proliferative disorders, and/or cancerous diseases and conditions.
Such a method is
described in US Patents 5,858,659 and 5,856,104. The US Patents referenced
supra are
hereby incorporated by reference in their entirety herein.
The present invention encompasses polynucleotides of the present invention
that are
chemically synthesized, or reproduced as peptide nucleic acids (PNA); or
according to other
methods known in the art. The use of PNAs would serve as the preferred form if
the
polynucleotides of the invention are incorporated onto a solid support, or
gene chip. For the
purposes of the present invention, a peptide nucleic acid (PNA) is a polyamide
type of DNA
analog and the monomeric units for adenine, guanine, thymine and cytosine are
available
commercially (Perceptive Biosystems). Certain components of DNA, such as
phosphorus,
phosphorus oxides, or deoxyribose derivatives, are not present in PNAs. As
disclosed by P.
E. Nielsen, M. Egholm, R. H. Berg and O. Buchardt, Science 254, 1497 (1991);
and M.
Egholin, O. Buchardt, L.Christensen, C. Behrens, S. M. Freier, D. A. Driver,
R. H. Berg, S.
K. Kim, B. Norden, and P. E. Nielsen, Nature 365, 666 (1993), PNAs bind
specifically and
tightly to complementary DNA strands and are not degraded by nucleases. In
fact, PNA binds
more strongly to DNA than DNA itself does. This is probably because there is
no
electrostatic repulsion between the two strands, and also the polyamide
backbone is more
flexible. Because of this, PNA/DNA duplexes bind under a wider range of
stringency
conditions than DNA/DNA duplexes, making it easier to perform multiplex
hybridization.
Smaller probes can be used than with DNA due to the strong binding. In
addition, it is more
likely that single base mismatches can be determined with PNA/DNA
hybridization because
a single mismatch in a PNA/DNA 15-mer lowers the melting point (Tm) by
8°-20° C,
vs. 4°-16° C for the DNA/DNA 15-mer duplex. Also, the absence of
charge groups in PNA
means that hybridization can be done at low ionic strengths and reduce
possible interference
by salt during the analysis.
The present invention have uses which include, but are not limited to,
detecting
cancer in mammals. In particular the invention is useful during diagnosis of
pathological cell
proliferative neoplasias which include, but are not limited to: acute
myelogenous leukemias
including acute monocytic leukemia, acute myeloblastic leukemia, acute
promyelocytic


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
99
leukemia, acute myelomonocytic leukemia, acute erythroleukemia, acute
megakaryocytic
leukemia, and acute undifferentiated leukemia, etc.; and chronic myelogenous
leukemias
including chronic myelomonocytic leukemia, chronic granulocytic leukemia, etc.
Preferred
mammals include monkeys, apes, cats, dogs, cows, pigs, horses, rabbits and
humans.
Particularly preferred are humans.
Pathological cell proliferative disorders are often associated with
inappropriate
activation of proto-oncogenes. (Gelmann, E. P. et al., "The Etiology of Acute
Leukemia:
Molecular Genetics and Viral Oncology," in Neoplastic Diseases of the Blood,
Vol 1.,
Wiernik, P. H. et al. eds., 161-182 (1985)). Neoplasias are now believed to
result from the
qualitative alteration of a normal cellular gene product, or from the
quantitative modification
of gene expression by insertion into the chromosome of a viral sequence, by
chromosomal
translocation of a gene to a more actively transcribed region, or by some
other mechanism.
(Gelmann et al., supra) It is likely that mutated or altered expression of
specific genes is
involved in the pathogenesis of some leukemias, among other tissues and cell
types.
(Gelmann et al., supra) Indeed, the human counterparts of the oncogenes
involved in some
animal neoplasias have been amplified or translocated in some cases of human
leukemia and
carcinoma. (Gelmann et al., supra)
For example, c-myc expression is highly amplified in the non-lymphocytic
leukemia
cell line HL-60. When HL-60 cells are chemically induced to stop
proliferation, the level of
c-myc is found to be downregulated. (International Publication Number WO
91/15580).
However, it has been shown that exposure of HL-60 cells to a DNA construct
that is
complementary to the 5' end of c-myc or c-myb blocks translation of the
corresponding
mRNAs which downregulates expression of the c-myc or c-myb proteins and causes
arrest of
cell proliferation and differentiation of the treated cells. (International
Publication Number
WO 91/15580; Wickstrom et al., Proc. Natl. Acad. Sci. 85:1028 (1988); Anfossi
et al., Proc.
Natl. Acad. Sci. 86:3379 (1989)). However, the skilled artisan would
appreciate the present
invention's usefulness is not be limited to treatment of proliferative
disorders of
hematopoietic cells and tissues, in light of the numerous cells and cell types
of varying
origins which are known to exhibit proliferative phenotypes.
In addition to the foregoing, a polynucleotide of the present invention can be
used to
control gene expression through triple helix formation or through antisense
DNA or RNA.
Antisense techniques are discussed, for example, in Okano, J. Neurochem. 56:
560 (1991);


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
100
"Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press,
Boca Raton,
FL (1988). Triple helix formation is discussed in, for instance Lee et al.,
Nucleic Acids
Research 6: 3073 (1979); Cooney et al., Science 241: 456 (1988); and Dervan et
al., Science
251: 1360 (1991). Both methods rely on binding of the polynucleotide to a
complementary
DNA or RNA. For these techniques, preferred polynucleotides are usually
oligonucleotides
20 to 40 bases in length and complementary to either the region of the gene
involved in
transcription (triple helix - see Lee et al., Nucl. Acids Res. 6:3073 (1979);
Cooney et al.,
Science 241:456 (1988); and Dervan et al., Science 251:1360 (1991) ) or to the
mRNA itself
(antisense - Okano, J. Neurochem. 56:560 (1991); Oligodeoxy-nucleotides as
Antisense
Inhibitors of Gene Expression, CRC Press, Boca Raton, FL (1988).) Triple helix
formation
optimally results in a shut-off of RNA transcription from DNA, while antisense
RNA
hybridization blocks translation of an mRNA molecule into polypeptide. The
oligonucleotide
described above can also be delivered to cells such that the antisense RNA or
DNA may be
expressed in vivo to inhibit production of polypeptide of the present
invention antigens. Both
1 S techniques are effective in model systems, and the information disclosed
herein can be used
to design antisense or triple helix polynucleotides in an effort to treat
disease, and in
particular, for the treatment of proliferative diseases and/or conditions.
Polynucleotides of the present invention are also useful in gene therapy. One
goal of
gene therapy is to insert a normal gene into an organism having a defective
gene, in an effort
to correct the genetic defect. The polynucleotides disclosed in the present
invention offer a
means of targeting such genetic defects in a highly accurate manner. Another
goal is to insert
a new gene that was not present in the host genome, thereby producing a new
trait in the host
cell.
The polynucleotides are also useful for identifying individuals from minute
biological
samples. The United States military, for example, is considering the use of
restriction
fragment length polymorphism (RFLP) for identification of its personnel. In
this technique,
an individual's genomic DNA is digested with one or more restriction enzymes,
and probed
on a Southern blot to yield unique bands for identifying personnel. This
method does not
suffer from the current limitations of "Dog Tags" which can be lost, switched,
or stolen,
making positive identification difficult. The polynucleotides of the present
invention can be
used as additional DNA markers for RFLP.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
101
The polynucleotides of the present invention can also be used as an
alternative to
RFLP, by determining the actual base-by-base DNA sequence of selected portions
of an
individual's genome. These sequences can be used to prepare PCR primers for
amplifying
and isolating such selected DNA, which can then be sequenced. Using this
technique,
individuals can be identified because each individual will have a unique set
of DNA
sequences. Once an unique ID database is established for an individual,
positive
identification of that individual, living or dead, can be made from extremely
small tissue
samples.
Forensic biology also benefits from using DNA-based identification techniques
as
disclosed herein. DNA sequences taken from very small biological samples such
as tissues,
e.g., hair or skin, or body fluids, e.g., blood, saliva, semen, synovial
fluid, amniotic fluid,
breast milk, lymph, pulmonary sputum or surfactant, urine, fecal matter, etc.,
can be
amplified using PCR. In one prior art technique, gene sequences amplified from
polymorphic loci, such as DQa class II HLA gene, are used in forensic biology
to identify
individuals. (Erlich, H., PCR Technology, Freeman and Co. (1992).) Once these
specific
polymorphic loci are amplified, they are digested with one or more restriction
enzymes,
yielding an identifying set of bands on a Southern blot probed with DNA
corresponding to
the DQa class II HLA gene. Similarly, polynucleotides of the present invention
can be used
as polymorphic markers for forensic purposes.
There is also a need for reagents capable of identifying the source of a
particular
tissue. Such need arises, for example, in forensics when presented with tissue
of unknown
origin. Appropriate reagents can comprise, for example, DNA probes or primers
prepared
from the sequences of the present invention. Panels of such reagents can
identify tissue by
species and/or by organ type. In a similar fashion, these reagents can be used
to screen tissue
cultures for contamination.
The polynucleotides of the present invention are also useful as hybridization
probes
for differential identification of the tissues) or cell types) present in a
biological sample.
Similarly, polypeptides and antibodies directed to polypeptides of the present
invention are
useful to provide immunological probes for differential identification of the
tissues) (e.g.,
immunohistochemistry assays) or cell types) (e.g., immunocytochemistry
assays). In
addition, for a number of disorders of the above tissues or cells,
significantly higher or lower
levels of gene expression of the polynucleotides/polypeptides of the present
invention may be


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
102
detected in certain tissues (e.g., tissues expressing polypeptides and/or
polynucleotides of the
present invention and/or cancerous and/or wounded tissues) or bodily fluids
(e.g., serum,
plasma, urine, synovial fluid or spinal fluid) taken from an individual having
such a disorder,
relative to a "standard" gene expression level, i.e., the expression level in
healthy tissue from
an individual not having the disorder.
Thus, the invention provides a diagnostic method of a disorder, which
involves: (a)
assaying gene expression level in cells or body fluid of an individual; (b)
comparing the gene
expression level with a standard gene expression level, whereby an increase or
decrease in
the assayed gene expression level compared to the standard expression level is
indicative of a
disorder.
In the very least, the polynucleotides of the present invention can be used as
molecular weight markers on Southern gels, as diagnostic probes for the
presence of a
specific mRNA in a particular cell type, as a probe to "subtract-out" known
sequences in the
process of discovering novel polynucleotides, for selecting and making
oligomers for
attachment to a "gene chip" or other support, to raise anti-DNA antibodies
using DNA
immunization techniques, and as an antigen to elicit an immune response.
Uses of the Polypeptides
Each of the polypeptides identified herein can be used in numerous ways. The
following description should be considered exemplary and utilizes known
techniques.
Polypeptides and antibodies directed to polypeptides of the present invention
are
useful to provide immunological probes for differential identification of the
tissues) (e.g.,
immunohistochemistry assays such as, for example, ABC immunoperoxidase (Hsu et
al., J.
Histochem. Cytochem. 29:577-580 (1981)) or cell types) (e.g.,
immunocytochemistry
assays).
Antibodies can be used to assay levels of polypeptides encoded by
polynucleotides of
the invention in a biological sample using classical immunohistological
methods known to
those of skill in the art (e.g., see Jalkanen, et al., J. Cell. Biol. 101:976-
985 (1985); Jalkanen,
et al., J. Cell. Biol. 105:3087-3096 (1987)). Other antibody-based methods
useful for
detecting protein gene expression include immunoassays, such as the enzyme
linked
immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable antibody
assay
labels are known in the art and include enzyme labels, such as, glucose
oxidase;


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
103
radioisotopes, such as iodine (13'h l2sf 123f 121I)~ carbon ('4C), sulfur
(3sS), tritium (3H),
indium (llsmln, 113",In, 1'zIn, Illln), and technetium (9~Tc, 99mTc), thallium
(z°1T1), gallium
(6gGa, 67Ga), palladium ('°3Pd), molybdenum (99Mo), xenon ('33Xe),
fluorine (18F), Is3Sm,
177Lu' 159Gd' 149Pm' 140La' 175' 1 GGH~' 90Y' 47SC' 1 s6Re~ 1 saRe~ 142Pr' l
Os~' 97Ru;
luminescent labels, such as luminol; and fluorescent labels, such as
fluorescein and
rhodamine, and biotin.
In addition to assaying levels of polypeptide of the present invention in a
biological
sample, proteins can also be detected in vivo by imaging. Antibody labels or
markers for in
vivo imaging of protein include those detectable by X-radiography, NMR or ESR.
For X-
radiography, suitable labels include radioisotopes such as barium or cesium,
which emit
detectable radiation but are not overtly harmful to the subject. Suitable
markers for NMR and
ESR include those with a detectable characteristic spin, such as deuterium,
which may be
incorporated into the antibody by labeling of nutrients for the relevant
hybridoma.
A protein-specific antibody or antibody fragment which has been labeled with
an
appropriate detectable imaging moiety, such as a radioisotope (for
example,'3'I,'lz~~ 99mTc,
(131f l2sl' 123I' 121I)~ carbon (14C), Sulfur (3sS), tritium (3H), indium
(llsm~' 113m~' 112' 1110'
and technetium (~~Tc, 99mTc), thallium (z°1Ti), gallium (6gGa, 67Ga),
palladium (1°3Pd),
molybdenum (99Mo), xenon (133Xe), fluorine (18F, ls3Sm, 177Lu, ls9Gd, 149Pm,
l4oLa~ 17s~~
166Ho~ 901, 47Sc~ IB~Re, 188Re, l4zPr, losRh, 97Ru), a radio-opaque substance,
or a material
detectable by nuclear magnetic resonance, is introduced (for example,
parenterally,
subcutaneously or intraperitoneally) into the mammal to be examined for immune
system
disorder. It will be understood in the art that the size of the subject and
the imaging system
used will determine the quantity of imaging moiety needed to produce
diagnostic images. In
the case of a radioisotope moiety, for a human subject, the quantity of
radioactivity injected
will normally range from about 5 to 20 millicuries of ~~mTc. The labeled
antibody or
antibody fragment will then preferentially accumulate at the location of cells
which express
the polypeptide encoded by a polynucleotide of the invention. In vivo tumor
imaging is
described in S.W. Burchiel et al., "Immunopharmacokinetics of Radiolabeled
Antibodies and
Their Fragments" (Chapter 13 in Tumor Imaging. The Radiochemical Detection of
Cancer,
S.W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982)).
In one embodiment, the invention provides a method for the specific delivery
of
compositions of the invention to cells by administering polypeptides of the
invention (e.g.,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
104
polypeptides encoded by polynucleotides of the invention and/or antibodies)
that are
associated with heterologous polypeptides or nucleic acids. In one example,
the invention
provides a method for delivering a therapeutic protein into the targeted cell.
In another
example, the invention provides a method for delivering a single stranded
nucleic acid (e.g.,
antisense or ribozymes) or double stranded nucleic acid (e.g., DNA that can
integrate into the
cell's genome or replicate episomally and that can be transcribed) into the
targeted cell.
In another embodiment, the invention provides a method for the specific
destruction
of cells (e.g., the destruction of tumor cells) by administering polypeptides
of the invention in
association with toxins or cytotoxic prodrugs.
By "toxin" is meant one or more compounds that bind and activate endogenous
cytotoxic effector systems, radioisotopes, holotoxins, modified toxins,
catalytic subunits of
toxins, or any molecules or enzymes not normally present in or on the surface
of a cell that
under defined conditions cause the cell's death. Toxins that may be used
according to the
methods of the invention include, but are not limited to, radioisotopes known
in the art,
compounds such as, for example, antibodies (or complement fixing containing
portions
thereof) that bind an inherent or induced endogenous cytotoxic effector
system, thymidine
kinase, endonuclease, RNAse, alpha toxin, ricin, abrin, Pseudomonas exotoxin
A, diphtheria
toxin, saporin, momordin, gelonin, pokeweed antiviral protein, alpha-sarcin
and cholera
toxin. "Toxin" also includes a cytostatic or cytocidal agent, a therapeutic
agent or a
radioactive metal ion, e.g., alpha-emitters such as, for example, 213$i, or
other radioisotopes
such as, for example, 103Pd, 133xe, 1311, 68Ge, 57CO, 65Zn, 855r, 32P, 35s,
90Y, 1535m, 153Gd,
169, SiCr, s4Mn, 7sSe, 1135n, 9°Yttrium, 117Tin, lg6Rhenium,
166Holmium, and 188Rhenium;
luminescent labels, such as luminol; and fluorescent labels, such as
fluorescein and
rhodamine, and biotin.
Techniques known in the art may be applied to label polypeptides of the
invention
(including antibodies). Such techniques include, but are not limited to, the
use of
bifunctional conjugating agents (see e.g., U.S. Patent Nos. 5,756,065;
5,714,631; 5,696,239;
5,652,361; 5,505,931; 5,489,425; 5,435,990; 5,428,139; 5,342,604; 5,274,119;
4,994,560;
and 5,808,003; the contents of each of which are hereby incorporated by
reference in its
entirety).
Thus, the invention provides a diagnostic method of a disorder, which involves
(a)
assaying the expression level of a polypeptide of the present invention in
cells or body fluid


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
105
of an individual; and (b) comparing the assayed polypeptide expression level
with a standard
polypeptide expression level, whereby an increase or decrease in the assayed
polypeptide
expression level compared to the standard expression level is indicative of a
disorder. With
respect to cancer, the presence of a relatively high amount of transcript in
biopsied tissue
from an individual may indicate a predisposition for the development of the
disease, or may
provide a means for detecting the disease prior to the appearance of actual
clinical symptoms.
A more definitive diagnosis of this type may allow health professionals to
employ
preventative measures or aggressive treatment earlier thereby preventing the
development or
further progression of the cancer.
Moreover, polypeptides of the present invention can be used to treat or
prevent
diseases or conditions such as, for example, neural disorders, immune system
disorders,
muscular disorders, reproductive disorders, gastrointestinal disorders,
pulmonary disorders,
cardiovascular disorders, renal disorders, proliferative disorders, and/or
cancerous diseases
and conditions. For example, patients can be administered a polypeptide of the
present
invention in an effort to replace absent or decreased levels of the
polypeptide (e.g., insulin),
to supplement absent or decreased levels of a different polypeptide (e.g.,
hemoglobin S for
hemoglobin B, SOD, catalase, DNA repair proteins), to inhibit the activity of
a polypeptide
(e.g., an oncogene or tumor supressor), to activate the activity of a
polypeptide (e.g., by
binding to a receptor), to reduce the activity of a membrane bound receptor by
competing
with it for free ligand (e.g., soluble TNF receptors used in reducing
inflammation), or to
bring about a desired response (e.g., blood vessel growth inhibition,
enhancement of the
immune response to proliferative cells or tissues).
Similarly, antibodies directed to a polypeptide of the present invention can
also be
used to treat disease (as described supra, and elsewhere herein). For example,
administration
of an antibody directed to a polypeptide of the present invention can bind,
and/or neutralize
the polypeptide, and/or reduce overproduction of the polypeptide. Similarly,
administration
of an antibody can activate the polypeptide, such as by binding to a
polypeptide bound to a
membrane (receptor).
At the very least, the polypeptides of the present invention can be used as
molecular
weight markers on SDS-PAGE gels or on molecular sieve gel filtration columns
using
methods well known to those of skill in the art. Polypeptides can also be used
to raise
antibodies, which in turn are used to measure protein expression from a
recombinant cell, as a


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
106
way of assessing transformation of the host cell. Moreover, the polypeptides
of the present
invention can be used to test the following biological activities.
Gene Therapy Methods
Another aspect of the present invention is to gene therapy methods for
treating or
preventing disorders, diseases and conditions. The gene therapy methods relate
to the
introduction of nucleic acid (DNA, RNA and antisense DNA or RNA) sequences
into an
animal to achieve expression of the polypeptide of the present invention. This
method
requires a polynucleotide which codes for a polypeptide of the present
invention operatively
linked to a promoter and any other genetic elements necessary for the
expression of the
polypeptide by the target tissue. Such gene therapy and delivery techniques
are known in the
art, see, for example, W090/11092, which is herein incorporated by reference.
Thus, for example, cells from a patient may be engineered with a
polynucleotide
(DNA or RNA) comprising a promoter operably linked to a polynucleotide of the
present
invention ex vivo, with the engineered cells then being provided to a patient
to be treated
with the polypeptide of the present invention. Such methods are well-known in
the art. For
example, see Belldegrun, A., et al., J. Natl. Cancer Inst. 85: 207-216 (1993);
Ferrantini, M. et
al., Cancer Research 53: 1107-1112 (1993); Ferrantini, M. et al., J.
Immunology 153: 4604-
4615 (1994); Kaido, T., et al., Int. J. Cancer 60: 221-229 (1995); Ogura, H.,
et al., Cancer
Research 50: 5102-5106 (1990); Santodonato, L., et al., Human Gene Therapy 7:1-
10 (1996);
Santodonato, L., et al., Gene Therapy 4:1246-1255 (1997); and Zhang, J.-F. et
al., Cancer
Gene Therapy 3: 31-38 (1996)), which are herein incorporated by reference. In
one
embodiment, the cells which are engineered are arterial cells. The arterial
cells may be
reintroduced into the patient through direct injection to the artery, the
tissues surrounding the
artery, or through catheter injection.
As discussed in more detail below, the polynucleotide constructs can be
delivered by
any method that delivers injectable materials to the cells of an animal, such
as, injection into
the interstitial space of tissues (heart, muscle, skin, lung, liver, and the
like). The
polynucleotide constructs may be delivered in a pharmaceutically acceptable
liquid or
aqueous carrier.
In one embodiment, the polynucleotide of the present invention is delivered as
a
naked polynucleotide. The term "naked" polynucleotide, DNA or RNA refers to
sequences


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
107
that are free from any delivery vehicle that acts to assist, promote or
facilitate entry into the
cell, including viral sequences, viral particles, liposome formulations,
lipofectin or
precipitating agents and the like. However, the polynucleotide of the present
invention can
also be delivered in liposome formulations and lipofectin formulations and the
like can be
S prepared by methods well known to those skilled in the art. Such methods are
described, for
example, in U.S. Patent Nos. 5,593,972, 5,589,466, and 5,580,859, which are
herein
incorporated by reference.
The polynucleotide vector constructs used in the gene therapy method are
preferably
constructs that will not integrate into the host genome nor will they contain
sequences that
allow for replication. Appropriate vectors include pWLNEO, pSV2CAT, pOG44,
pXTl and
pSG available from Stratagene; pSVK3, pBPV, pMSG and pSVL available from
Pharmacia;
and pEFI/V5, pcDNA3.1, and pRc/CMV2 available from Invitrogen. Other suitable
vectors
will be readily apparent to the skilled artisan.
Any strong promoter known to those skilled in the art can be used for driving
the
1 S expression of the polynucleotide sequence. Suitable promoters include
adenoviral promoters,
such as the adenoviral major late promoter; or heterologous promoters, such as
the
cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV)
promoter; inducible
promoters, such as the MMT promoter, the metallothionein promoter; heat shock
promoters;
the albumin promoter; the ApoAI promoter; human globin promoters; viral
thymidine kinase
promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral
LTRs; the b-
acon promoter; and human growth hormone promoters. The promoter also may be
the native
promoter for the polynucleotide of the present invention.
Unlike other gene therapy techniques, one major advantage of introducing naked
nucleic acid sequences into target cells is the transitory nature of the
polynucleotide synthesis
in the cells. Studies have shown that non-replicating DNA sequences can be
introduced into
cells to provide production of the desired polypeptide for periods of up to
six months.
The polynucleotide construct can be delivered to the interstitial space of
tissues within
the an animal, including of muscle, skin, brain, lung, liver, spleen, bone
marrow, thymus,
heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach,
intestine, testis,
ovary, uterus, rectum, nervous system, eye, gland, and connective tissue.
Interstitial space of
the tissues comprises the intercellular, fluid, mucopolysaccharide matrix
among the reticular
fibers of organ tissues, elastic fibers in the walls of vessels or chambers,
collagen fibers of


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
108
fibrous tissues, or that same matrix within connective tissue ensheathing
muscle cells or in the
lacunae of bone. It is similarly the space occupied by the plasma of the
circulation and the
lymph fluid of the lymphatic channels. Delivery to the interstitial space of
muscle tissue is
preferred for the reasons discussed below. They may be conveniently delivered
by injection
into the tissues comprising these cells. They are preferably delivered to and
expressed in
persistent, non-dividing cells which are differentiated, although delivery and
expression may
be achieved in non-differentiated or less completely differentiated cells,
such as, for example,
stem cells of blood or skin fibroblasts. In vivo muscle cells are particularly
competent in their
ability to take up and express polynucleotides.
For the naked nucleic acid sequence injection, an effective dosage amount of
DNA or
RNA will be in the range of from about 0.05 mg/kg body weight to about 50
mg/kg body
weight. Preferably the dosage will be from about 0.005 mg/kg to about 20 mg/kg
and more
preferably from about 0.05 mg/kg to about 5 mg/kg. Of course, as the artisan
of ordinary skill
will appreciate, this dosage will vary according to the tissue site of
injection. The appropriate
and effective dosage of nucleic acid sequence can readily be determined by
those of ordinary
skill in the art and may depend on the condition being treated and the route
of administration.
The preferred route of administration is by the parenteral route of injection
into the
interstitial space of tissues. However, other parenteral routes may also be
used, such as,
inhalation of an aerosol formulation particularly for delivery to lungs or
bronchial tissues,
throat or mucous membranes of the nose. In addition, naked DNA constructs can
be
delivered to arteries during angioplasty by the catheter used in the
procedure.
The naked polynucleotides are delivered by any method known in the art,
including,
but not limited to, direct needle injection at the delivery site, intravenous
injection, topical
administration, catheter infusion, and so-called "gene guns". These delivery
methods are
known in the art.
The constructs may also be delivered with delivery vehicles such as viral
sequences,
viral particles, liposome formulations, lipofectin, precipitating agents, etc.
Such methods of
delivery are known in the art.
In certain embodiments, the polynucleotide constructs are complexed in a
liposome
preparation. Liposomal preparations for use in the instant invention include
cationic
(positively charged), anionic (negatively charged) and neutral preparations.
However,
cationic liposomes are particularly preferred because a tight charge complex
can be formed


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
109
between the cationic liposome and the polyanionic nucleic acid. Cationic
liposomes have
been shown to mediate intracellular delivery of plasmid DNA (Felgner et al.,
Proc. Natl.
Acad. Sci. USA (1987) 84:7413-7416, which is herein incorporated by
reference); mRNA
(Malone et al., Proc. Natl. Acad. Sci. USA (1989) 86:6077-6081, which is
herein
incorporated by reference); and purified transcription factors (Debs et al.,
J. Biol. Chem.
(1990) 265:10189-10192, which is herein incorporated by reference), in
functional form.
Cationic liposomes are readily available. For example,
N[1-2,3-dioleyloxy)propyl]-N,N,N-triethylammonium (DOTMA) liposomes are
particularly
useful and are available under the trademark Lipofectin, from GIBCO BRL, Grand
Island,
N.Y. (See, also, Felgner et al., Proc. Natl Acad. Sci. USA (1987) 84:7413-
7416, which is
herein incorporated by reference). Other commercially available liposomes
include
transfectace (DDAB/DOPE) and DOTAP/DOPE (Boehringer).
Other cationic liposomes can be prepared from readily available materials
using
techniques well known in the art. See, e.g. PCT Publication No. WO 90/11092
(which is
herein incorporated by reference) for a description of the synthesis of DOTAP
(1,2
bis(oleoyloxy)-3-(trimethylammonio)propane) liposomes. Preparation of DOTMA
liposomes
is explained in the literature, see, e.g., P. Felgner et al., Proc. Natl.
Acad. Sci. USA
84:7413-7417, which is herein incorporated by reference. Similar methods can
be used to
prepare liposomes from other cationic lipid materials.
Similarly, anionic and neutral liposomes are readily available, such as from
Avanti
Polar Lipids (Birmingham, Ala.), or can be easily prepared using readily
available materials.
Such materials include phosphatidyl, choline, cholesterol, phosphatidyl
ethanolamine,
dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG),
dioleoylphoshatidyl ethanolamine (DOPE), among others. These materials can
also be mixed
with the DOTMA and DOTAP starting materials in appropriate ratios. Methods for
making
liposomes using these materials are well known in the art.
For example, commercially dioleoylphosphatidyl choline (DOPC),
dioleoylphosphatidyl glycerol (DOPG), and dioleoylphosphatidyl ethanolamine
(DOPE) can
be used in various combinations to make conventional liposomes, with or
without the
addition of cholesterol. Thus, for example, DOPG/DOPC vesicles can be prepared
by drying
50 mg each of DOPG and DOPC under a stream of nitrogen gas into a sonication
vial. The
sample is placed under a vacuum pump overnight and is hydrated the following
day with


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
110
deionized water. The sample is then sonicated for 2 hours in a capped vial,
using a Heat
Systems model 350 sonicator equipped with an inverted cup (bath type) probe at
the
maximum setting while the bath is circulated at 15EC. Alternatively,
negatively charged
vesicles can be prepared without sonication to produce multilamellar vesicles
or by extrusion
through nucleopore membranes to produce unilamellar vesicles of discrete size.
Other
methods are known and available to those of skill in the art.
The liposomes can comprise multilamellar vesicles (MLVs), small unilamellar
vesicles (SUVs), or large unilamellar vesicles (LUVs), with SUVs being
preferred. The
various liposome-nucleic acid complexes are prepared using methods well known
in the art.
See, e.g., Straubinger et al., Methods of Immunology (1983), 101:512-527,
which is herein
incorporated by reference. For example, MLVs containing nucleic acid can be
prepared by
depositing a thin film of phospholipid on the walls of a glass tube and
subsequently hydrating
with a solution of the material to be encapsulated. SUVs are prepared by
extended sonication
of MLVs to produce a homogeneous population of unilamellar liposomes. The
material to be
entrapped is added to a suspension of preformed MLVs and then sonicated. When
using
liposomes containing cationic lipids, the dried lipid film is resuspended in
an appropriate
solution such as sterile water or an isotonic buffer solution such as 10 mM
Tris/NaCI,
sonicated, and then the preformed liposomes are mixed directly with the DNA.
The liposome
and DNA form a very stable complex due to binding of the positively charged
liposomes to
the cationic DNA. SUVs find use with small nucleic acid fragments. LUVs are
prepared by a
number of methods, well known in the art. Commonly used methods include Ca2+-
EDTA
chelation (Papahadjopoulos et al., Biochim. Biophys. Acta (1975) 394:483;
Wilson et al.,
Cell (1979) 17:77); ether injection (Deamer, D. and Bangham, A., Biochim.
Biophys. Acta
(1976) 443:629; Ostro et al., Biochem. Biophys. Res. Commun. (1977) 76:836;
Fraley et al.,
Proc. Natl. Acad. Sci. USA (1979) 76:3348); detergent dialysis (Enoch, H. and
Strittmatter,
P., Proc. Natl. Acad. Sci. USA (1979) 76:145); and reverse-phase evaporation
(REV) (Fraley
et al., J. Biol. Chem. (1980) 255:10431; Szoka, F. and Papahadjopoulos, D.,
Proc. Natl. Acad.
Sci. USA (1978) 75:145; Schaefer-Ridder et al., Science (1982) 215:166), which
are herein
incorporated by reference.
Generally, the ratio of DNA to liposomes will be from about 10:1 to about
1:10.
Preferably, the ration will be from about 5:1 to about 1:5. More preferably,
the ration will be
about 3:1 to about 1:3. Still more preferably, the ratio will be about 1:1.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
111
U.S. Patent No. 5,676,954 (which is herein incorporated by reference) reports
on the
injection of genetic material, complexed with cationic liposomes carriers,
into mice. U.S.
Patent Nos. 4,897,355, 4,946,787, 5,049,386, 5,459,127, 5,589,466, 5,693,622,
5,580,859,
5,703,055, and international publication no. WO 94/9469 (which are herein
incorporated by
reference) provide cationic lipids for use in transfecting DNA into cells and
mammals. U.S.
Patent Nos. 5,589,466, 5,693,622, 5,580,859, 5,703,055, and international
publication no.
WO 94/9469 (which are herein incorporated by reference) provide methods for
delivering
DNA-cationic lipid complexes to mammals.
In certain embodiments, cells are engineered, ex vivo or in vivo, using a
retroviral
particle containing RNA which comprises a sequence encoding a polypeptide of
the present
invention. Retroviruses from which the retroviral plasmid vectors may be
derived include,
but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus,
Rous sarcoma
Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus,
human
immunodeficiency virus, Myeloproliferative Sarcoma Virus, and mammary tumor
virus.
The retroviral plasmid vector is employed to transduce packaging cell lines to
form
producer cell lines. Examples of packaging cells which may be transfected
include, but are
not limited to, the PESO1, PA317, R-2, R-AM, PA12, T19-14X, VT-19-17-H2, RCRE,
RCRIP, GP+E-86, GP+envAml2, and DAN cell lines as described in Miller, Human
Gene
Therapy 1:5-14 (1990), which is incorporated herein by reference in its
entirety. The vector
may transduce the packaging cells through any means known in the art. Such
means include,
but are not limited to, electroporation, the use of liposomes, and CaP04
precipitation. In one
alternative, the retroviral plasmid vector may be encapsulated into a
liposome, or coupled to a
lipid, and then administered to a host.
The producer cell line generates infectious retroviral vector particles which
include
polynucleotide encoding a polypeptide of the present invention. Such
retroviral vector
particles then may be employed, to transduce eukaryotic cells, either in vitro
or in vivo. The
transduced eukaryotic cells will express a polypeptide of the present
invention.
In certain other embodiments, cells are engineered, ex vivo or in vivo, with
polynucleotide contained in an adenovirus vector. Adenovirus can be
manipulated such that
it encodes and expresses a polypeptide of the present invention, and at the
same time is
inactivated in terms of its ability to replicate in a normal lytic viral life
cycle. Adenovirus
expression is achieved without integration of the viral DNA into the host cell
chromosome,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
112
thereby alleviating concerns about insertional mutagenesis. Furthermore,
adenoviruses have
been used as live enteric vaccines for many years with an excellent safety
profile (Schwartz,
A. R. et al. (1974) Am. Rev. Respir. Dis.109:233-238). Finally, adenovirus
mediated gene
transfer has been demonstrated in a number of instances including transfer of
alpha-1-antitrypsin and CFTR to the lungs of cotton rats (Rosenfeld, M. A. et
al. (1991)
Science 252:431-434; Rosenfeld et al., (1992) Cell 68:143-155). Furthermore,
extensive
studies to attempt to establish adenovirus as a causative agent in human
cancer were
uniformly negative (Green, M. et al. (1979) Proc. Natl. Acad. Sci. USA
76:6606).
Suitable adenoviral vectors useful in the present invention are described, for
example,
in Kozarsky and Wilson, Curr. Opin. Genet. bevel. 3:499-503 (1993); Rosenfeld
et al., Cell
68:143-155 (1992); Engelhardt et al., Human Genet. Ther. 4:759-769 (1993);
Yang et al.,
Nature Genet. 7:362-369 (1994); Wilson et al., Nature 365:691-692 (1993); and
U.S. Patent
No. 5,652,224, which are herein incorporated by reference. For example, the
adenovirus
vector Ad2 is useful and can be grown in human 293 cells. These cells contain
the E1 region
of adenovirus and constitutively express Ela and Elb, which complement the
defective
adenoviruses by providing the products of the genes deleted from the vector.
In addition to
Ad2, other varieties of adenovirus (e.g., Ad3, AdS, and Ad7) are also useful
in the present
invention.
Preferably, the adenoviruses used in the present invention are replication
deficient.
Replication deficient adenoviruses require the aid of a helper virus and/or
packaging cell line
to form infectious particles. The resulting virus is capable of infecting
cells and can express a
polynucleotide of interest which is operably linked to a promoter, but cannot
replicate in most
cells. Replication deficient adenoviruses may be deleted in one or more of all
or a portion of
the following genes: Ela, Elb, E3, E4, E2a, or Ll through L5.
In certain other embodiments, the cells are engineered, ex vivo or in vivo,
using an
adeno-associated virus (AAV). AAVs are naturally occurnng defective viruses
that require
helper viruses to produce infectious particles (Muzyczka, N., Curr. Topics in
Microbiol.
Immunol. 158:97 (1992)). It is also one of the few viruses that may integrate
its DNA into
non-dividing cells. Vectors containing as little as 300 base pairs of AAV can
be packaged and
can integrate, but space for exogenous DNA is limited to about 4.5 kb. Methods
for
producing and using such AAVs are known in the art. See, for example, U.S.
Patent Nos.
5,139,941, 5,173,414, 5,354,678, 5,436,146, 5,474,935, 5,478,745, and
5,589,377.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
113
For example, an appropriate AAV vector for use in the present invention will
include
all the sequences necessary for DNA replication, encapsidation, and host-cell
integration.
The polynucleotide construct is inserted into the AAV vector using standard
cloning
methods, such as those found in Sambrook et al., Molecular Cloning: A
Laboratory Manual,
S Cold Spring Harbor Press (1989). The recombinant AAV vector is then
transfected into
packaging cells which are infected with a helper virus, using any standard
technique,
including lipofection, electroporation, calcium phosphate precipitation, etc.
Appropriate
helper viruses include adenoviruses, cytomegaloviruses, vaccinia viruses, or
herpes viruses.
Once the packaging cells are transfected and infected, they will produce
infectious AAV viral
particles which contain the polynucleotide construct. These viral particles
are then used to
transduce eukaryotic cells, either ex vivo or in vivo. The transduced cells
will contain the
polynucleotide construct integrated into its genome, and will express a
polypeptide of the
invention.
Another method of gene therapy involves operably associating heterologous
control
regions and endogenous polynucleotide sequences (e.g. encoding a polypeptide
of the present
invention) via homologous recombination (see, e.g., U.S. Patent No. 5,641,670,
issued June
24, 1997; International Publication No. WO 96/29411, published September 26,
1996;
International Publication No. WO 94/12650, published August 4, 1994; Koller et
al., Proc.
Natl. Acad. Sci. USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-
438 (1989).
This method involves the activation of a gene which is present in the target
cells, but which is
not normally expressed in the cells, or is expressed at a lower level than
desired.
Polynucleotide constructs are made, using standard techniques known in the
art,
which contain the promoter with targeting sequences flanking the promoter.
Suitable
promoters are described herein. The targeting sequence is sufficiently
complementary to an
endogenous sequence to permit homologous recombination of the promoter-
targeting
sequence with the endogenous sequence. The targeting sequence will be
sufficiently near the
5' end of the desired endogenous polynucleotide sequence so the promoter will
be operably
linked to the endogenous sequence upon homologous recombination.
The promoter and the targeting sequences can be amplified using PCR.
Preferably,
the amplified promoter contains distinct restriction enzyme sites on the 5'
and 3' ends.
Preferably, the 3' end of the first targeting sequence contains the same
restriction enzyme site
as the 5' end of the amplified promoter and the S' end of the second targeting
sequence


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
114
contains the same restriction site as the 3' end of the amplified promoter.
The amplified
promoter and targeting sequences are digested and ligated together.
The promoter-targeting sequence construct is delivered to the cells, either as
naked
polynucleotide, or in conjunction with transfection-facilitating agents, such
as liposomes,
S viral sequences, viral particles, whole viruses, lipofection, precipitating
agents, etc., described
in more detail above. The P promoter-targeting sequence can be delivered by
any method,
included direct needle injection, intravenous injection, topical
administration, catheter
infusion, particle accelerators, etc. The methods are described in more detail
below.
The promoter-targeting sequence construct is taken up by cells. Homologous
recombination between the construct and the endogenous sequence takes place,
such that an
endogenous sequence is placed under the control of the promoter. The promoter
then drives
the expression of the endogenous sequence.
Preferably, the polynucleotide encoding a polypeptide of the present invention
contains a secretory signal sequence that facilitates secretion of the
protein. Typically, the
signal sequence is positioned in the coding region of the polynucleotide to be
expressed
towards or at the 5' end of the coding region. The signal sequence may be
homologous or
heterologous to the polynucleotide of interest and may be homologous or
heterologous to the
cells to be transfected. Additionally, the signal sequence may be chemically
synthesized
using methods known in the art.
Any mode of administration of any of the above-described polynucleotides
constructs
can be used so long as the mode results in the expression of one or more
molecules in an
amount sufficient to provide a therapeutic effect. This includes direct needle
injection,
systemic injection, catheter infusion, biolistic injectors, particle
accelerators (i.e., "gene
guns"), gelfoam sponge depots, other commercially available depot materials,
osmotic pumps
(e.g., Alza minipumps), oral or suppositorial solid (tablet or pill)
pharmaceutical
formulations, and decanting or topical applications during surgery. For
example, direct
injection of naked calcium phosphate-precipitated plasmid into rat liver and
rat spleen or a
protein-coated plasmid into the portal vein has resulted in gene expression of
the foreign gene
in the rat livers (Kaneda et al., Science 243:375 (1989)).
A preferred method of local administration is by direct injection. Preferably,
a
recombinant molecule of the present invention complexed with a delivery
vehicle is
administered by direct injection into or locally within the area of arteries.
Administration of a


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
115
composition locally within the area of arteries refers to injecting the
composition centimeters
and preferably, millimeters within arteries.
Another method of local administration is to contact a polynucleotide
construct of the
present invention in or around a surgical wound. For example, a patient can
undergo surgery
and the polynucleotide construct can be coated on the surface of tissue inside
the wound or
the construct can be injected into areas of tissue inside the wound.
Therapeutic compositions useful in systemic administration, include
recombinant
molecules of the present invention complexed to a targeted delivery vehicle of
the present
invention. Suitable delivery vehicles for use with systemic administration
comprise
liposomes comprising ligands for targeting the vehicle to a particular site.
Preferred methods of systemic administration, include intravenous injection,
aerosol,
oral and percutaneous (topical) delivery. Intravenous injections can be
performed using
methods standard in the art. Aerosol delivery can also be performed using
methods standard
in the art (see, for example, Stribling et al., Proc. Natl. Acad. Sci. USA
189:11277-11281,
1992, which is incorporated herein by reference). Oral delivery can be
performed by
complexing a polynucleotide construct of the present invention to a Garner
capable of
withstanding degradation by digestive enzymes in the gut of an animal.
Examples of such
Garners, include plastic capsules or tablets, such as those known in the art.
Topical delivery
can be performed by mixing a polynucleotide construct of the present invention
with a
lipophilic reagent (e.g., DMSO) that is capable of passing into the skin.
Determining an effective amount of substance to be delivered can depend upon a
number of factors including, for example, the chemical structure and
biological activity of the
substance, the age and weight of the animal, the precise condition requiring
treatment and its
severity, and the route of administration. The frequency of treatments depends
upon a
number of factors, such as the amount of polynucleotide constructs
administered per dose, as
well as the health and history of the subject. The precise amount, number of
doses, and
timing of doses will be determined by the attending physician or veterinarian.
Therapeutic compositions of the present invention can be administered to any
animal,
preferably to mammals and birds. Preferred mammals include humans, dogs, cats,
mice, rats,
rabbits sheep, cattle, horses and pigs, with humans being particularly
preferred.
Biological Activities


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
116
Polynucleotides or polypeptides, or agonists or antagonists of the present
invention,
can be used in assays to test for one or more biological activities. If these
polynucleotides or
polypeptides, or agonists or antagonists of the present invention, do exhibit
activity in a
particular assay, it is likely that these molecules may be involved in the
diseases associated
with the biological activity. Thus, the polynucleotides and polypeptides, and
agonists or
antagonists could be used to treat the associated disease.
Members of the steroid hormone receptor family of proteins are believed to be
involved in biological activities associated with the transduction of hormone
signals across
cellular membranes, with the result that these receptors bind with high
affinity to specific
response elements in the promoter regions of target genes, known as hormone
responsive
elements (HREs), inducing transcription of certain genes. Accordingly,
compositions of the
invention (including polynucleotides, polypeptides and antibodies of the
invention, and
fragments and variants thereof) may be used in the diagnosis, detection and/or
treatment of
diseases and/or disorders associated with aberrant signal transduction
activity. In preferred
1 S embodiments, compositions of the invention (including polynucleotides,
polypeptides and
antibodies of the invention, and fragments and variants thereof) may be used
in the diagnosis,
detection and/or treatment of diseases and/or disorders relating to cancers
(e.g., breast
cancers, and/or as described under "Immune Activity," "Anti-angiogenesis
Activity" and
"Diseases at the Cellular Level" below). Thus, polynucleotides, translation
products and
antibodies of the invention are useful in the diagnosis, detection and/or
treatment of diseases
and/or disorders associated with activities that include, but are not limited
to, diseases and/or
disorders involving signal transduction and/or cancers.
More generally, polynucleotides, translation products and antibodies
corresponding to
this gene may be useful for the diagnosis, detection and/or treatment of
diseases and/or
disorders associated with the following systems.
Immune Activity
Polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of
the
present invention may be useful in treating, preventing, and/or diagnosing
diseases, disorders,
and/or conditions of the immune system, by, for example, activating or
inhibiting the
proliferation, differentiation, or mobilization (chemotaxis) of immune cells.
Immune cells


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
117
develop through a process called hematopoiesis, producing myeloid (platelets,
red blood
cells, neutrophils, and macrophages) and lymphoid (B and T lymphocytes) cells
from
pluripotent stem cells. The etiology of these immune diseases, disorders,
and/or conditions
may be genetic, somatic, such as cancer and some autoimmune diseases, acquired
(e.g., by
chemotherapy or toxins), or infectious. Moreover, polynucleotides,
polypeptides, antibodies,
and/or agonists or antagonists of the present invention can be used as a
marker or detector of
a particular immune system disease or disorder.
Polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of
the
present invention may be useful in treating, preventing, and/or diagnosing
diseases, disorders,
and/or conditions of hematopoietic cells. Polynucleotides, polypeptides,
antibodies, and/or
agonists or antagonists of the present invention could be used to increase
differentiation and
proliferation of hematopoietic cells, including the pluripotent stem cells, in
an effort to treat
or prevent those diseases, disorders, and/or conditions associated with a
decrease in certain
(or many) types hematopoietic cells. Examples of immunologic deficiency
syndromes
include, but are not limited to: blood protein diseases, disorders, and/or
conditions (e.g.,
agammaglobulinemia, dysgammaglobulinemia), ataxia telangiectasia, common
variable
immunodeficiency, Digeorge Syndrome, HIV infection, HTLV-BLV infection,
leukocyte
adhesion deficiency syndrome, lymphopenia, phagocyte bactericidal dysfunction,
severe
combined immunodeficiency (SCIDs), Wiskott-Aldrich Disorder, anemia,
thrombocytopenia, or hemoglobinuria.
Moreover, polynucleotides, polypeptides, antibodies, and/or agonists or
antagonists of
the present invention could also be used to modulate hemostatic (the stopping
of bleeding) or
thrombolytic activity (clot formation). For example, by increasing hemostatic
or
thrombolytic activity, polynucleotides or polypeptides, and/or agonists or
antagonists of the
present invention could be used to treat or prevent blood coagulation
diseases, disorders,
and/or conditions (e.g., afibrinogenemia, factor deficiencies), blood platelet
diseases,
disorders, and/or conditions (e.g., thrombocytopenia), or wounds resulting
from trauma,
surgery, or other causes. Alternatively, polynucleotides, polypeptides,
antibodies, and/or
agonists or antagonists of the present invention that can decrease hemostatic
or thrombolytic
activity could be used to inhibit or dissolve clotting. These molecules could
be important in
the treatment or prevention of heart attacks (infarction), strokes, or
scarnng.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
118
The polynucleotides, polypeptides, antibodies, and/or agonists or antagonists
of the
present invention may be useful in treating, preventing, and/or diagnosing
autoimmune
disorders. Many autoimmune disorders result from inappropriate recognition of
self as
foreign material by immune cells. This inappropriate recognition results in an
immune
response leading to the destruction of the host tissue. Therefore, the
administration of
polynucleotides and polypeptides of the invention that can inhibit an immune
response,
particularly the proliferation, differentiation, or chemotaxis of T-cells, may
be an effective
therapy in preventing autoimmune disorders.
Autoimmune diseases or disorders that may be treated, prevented, and/or
diagnosed
by polynucleotides, polypeptides, antibodies, and/or agonists or antagonists
of the present
invention include, but are not limited to, one or more of the following:
autoimmune
hemolytic anemia, autoimmune neonatal thrombocytopenia, idiopathic
thrombocytopenia
purpura, autoimmunocytopenia, hemolytic anemia, antiphospholipid syndrome,
dermatitis,
allergic encephalomyelitis, myocarditis, relapsing polychondritis, rheumatic
heart disease,
glomerulonephritis (e.g, IgA nephropathy), Multiple Sclerosis, Neuritis,
Uveitis Ophthalmia,
Polyendocrinopathies, Purpura (e.g., Henloch-Scoenlein purpura), Reiter's
Disease, Stiff
Man Syndrome, Autoimmune Pulmonary Inflammation, Autism, Guillain-Barre
Syndrome,
insulin dependent diabetes mellitis, and autoimmune inflammatory eye,
autoimmune
thyroiditis, hypothyroidism (i.e., Hashimoto's thyroiditis, systemic lupus
erhythematosus,
Goodpasture's syndrome, Pemphigus, Receptor autoimmunities such as, for
example, (a)
Graves' Disease, (b) Myasthenia Gravis, and (c) insulin resistance, autoimmune
hemolytic
anemia, autoimmune thrombocytopenic purpura, rheumatoid arthritis,
schleroderma with
anti-collagen antibodies, mixed connective tissue disease,
polymyositis/dermatomyositis,
pernicious anemia, idiopathic Addison's disease, infertility,
glomerulonephritis such as
primary glomerulonephritis and IgA nephropathy, bullous pemphigoid, Sjogren's
syndrome,
diabetes millitus, and adrenergic drug resistance (including adrenergic drug
resistance with
asthma or cystic fibrosis), chronic active hepatitis, primary biliary
cirrhosis, other endocrine
gland failure, vitiligo, vasculitis, post-MI, cardiotomy syndrome, urticaria,
atopic dermatitis,
asthma, inflammatory myopathies, and other inflammatory, granulamatous,
degenerative, and
atrophic disorders.
Additional autoimmune disorders (that are probable) that may be treated,
prevented,
and/or diagnosed with the compositions of the invention include, but are not
limited to,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
119
rheumatoid arthritis (often characterized, e.g., by immune complexes in
joints), scleroderma
with anti-collagen antibodies (often characterized, e.g., by nucleolar and
other nuclear
antibodies), mixed connective tissue disease (often characterized, e.g., by
antibodies to
extractable nuclear antigens (e.g., ribonucleoprotein)), polymyositis (often
characterized, e.g.,
S by nonhistone ANA), pernicious anemia (often characterized, e.g., by
antiparietal cell,
microsomes, and intrinsic factor antibodies), idiopathic Addison's disease
(often
characterized, e.g., by humoral and cell-mediated adrenal cytotoxicity,
infertility (often
characterized, e.g., by antispermatozoal antibodies), glomerulonephritis
(often characterized,
e.g., by glomerular basement membrane antibodies or immune complexes), bullous
pemphigoid (often characterized, e.g., by IgG and complement in basement
membrane),
Sjogren's syndrome (often characterized, e.g., by multiple tissue antibodies,
and/or a specific
nonhistone ANA (SS-B)), diabetes millitus (often characterized, e.g., by cell-
mediated and
humoral islet cell antibodies), and adrenergic drug resistance (including
adrenergic drug
resistance with asthma or cystic fibrosis) (often characterized, e.g., by beta-
adrenergic
receptor antibodies).
Additional autoimmune disorders (that are possible) that may be treated,
prevented,
and/or diagnosed with the compositions of the invention include, but are not
limited to,
chronic active hepatitis (often characterized, e.g., by smooth muscle
antibodies), primary
biliary cirrhosis (often characterized, e.g., by mitchondrial antibodies),
other endocrine gland
failure (often characterized, e.g., by specific tissue antibodies in some
cases), vitiligo (often
characterized, e.g., by melanocyte antibodies), vasculitis (often
characterized, e.g., by Ig and
complement in vessel walls and/or low serum complement), post-MI (often
characterized,
e.g., by myocardial antibodies), cardiotomy syndrome (often characterized,
e.g., by
myocardial antibodies), urticaria (often characterized, e.g., by IgG and IgM
antibodies to
IgE), atopic dermatitis (often characterized, e.g., by IgG and IgM antibodies
to IgE), asthma
(often characterized, e.g., by IgG and IgM antibodies to IgE), and many other
inflammatory,
granulamatous, degenerative, and atrophic disorders.
In a preferred embodiment, the autoimmune diseases and disorders and/or
conditions
associated with the diseases and disorders recited above are treated,
prevented, and/or
diagnosed using for example, antagonists or agonists, polypeptides or
polynucleotides, or
antibodies of the present invention.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
120
In a preferred embodiment polynucleotides, polypeptides, antibodies, and/or
agonists
or antagonists of the present invention could be used as an agent to boost
immunoresponsiveness among B cell and/or T cell immunodeficient individuals.
B cell immunodeficiencies that may be ameliorated or treated by administering
the
polypeptides or polynucleotides of the invention, and/or agonists thereof,
include, but are not
limited to, severe combined immunodeficiency (SCID)-X linked, SCID-autosomal,
adenosine
deaminase deficiency (ADA deficiency), X-linked agammaglobulinemia (XLA),
Bruton's
disease, congenital agammaglobulinemia, X-linked infantile agammaglobulinemia,
acquired
agammaglobulinemia, adult onset agammaglobulinemia, late-onset
agammaglobulinemia,
dysgammaglobulinemia, hypogammaglobulinemia, transient hypogammaglobulinemia
of
infancy, unspecified hypogammaglobulinemia, agammaglobulinemia, common
variable
immunodeficiency (CVI) (acquired), Wiskott-Aldrich Syndrome (WAS), X-linked
immunodeficiency with hyper IgM, non X-linked immunodeficiency with hyper IgM,
selective IgA deficiency, IgG subclass deficiency (with or without IgA
deficiency), antibody
deficiency with normal or elevated Igs, immunodeficiency with thymoma, Ig
heavy chain
deletions, kappa chain deficiency, B cell lymphoproliferative disorder (BLPD),
selective
IgM immunodeficiency, recessive agammaglobulinemia (Swiss type), reticular
dysgenesis,
neonatal neutropenia, severe congenital leukopenia, thymic alymophoplasia-
aplasia or
dysplasia with immunodeficiency, ataxia-telangiectasia, short limbed dwarfism,
X-linked
lymphoproliferative syndrome (XLP), Nezelof syndrome-combined immunodeficiency
with
Igs, purine nucleoside phosphorylase deficiency (PNP), MHC Class II deficiency
(Bare
Lymphocyte Syndrome) and severe combined immunodeficiency.
T cell deficiencies that may be ameliorated or treated by administering the
polypeptides or polynucleotides of the invention, and/or agonists thereof
include, but are not
limited to, for example, DiGeorge anomaly, thymic hypoplasia, third and fourth
pharyngeal
pouch syndrome, 22q11.2 deletion, chronic mucocutaneous candidiasis, natural
killer cell
deficiency (NK), idiopathic CD4+ T-lyrnphocytopenia, immunodeficiency with
predominant
T cell defect (unspecified), and unspecified immunodeficiency of cell mediated
immunity. In
specific embodiments, DiGeorge anomaly or conditions associated with DiGeorge
anomaly
are ameliorated or treated by, for example, administering the polypeptides or
polynucleotides
of the invention, or antagonists or agonists thereof.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
121
Other immunodeficiencies that may be ameliorated or treated by administering
polypeptides or polynucleotides of the invention, and/or agonists thereof,
include, but are not
limited to, severe 'combined immunodeficiency (SCID; e.g., X-linked SCID,
autosomal
SLID, and adenosine deaminase deficiency), ataxia-telangiectasia, Wiskott-
Aldrich
syndrome, short-limber dwarfism, X-linked lymphoproliferative syndrome (XLP),
Nezelof
syndrome (e.g., purine nucleoside phosphorylase deficiency), MHC Class II
deficiency. In
specific embodiments, ataxia-telangiectasia or conditions associated with
ataxia-
telangiectasia are ameliorated or treated by administering the polypeptides or
polynucleotides
of the invention, and/or agonists thereof.
In a specific preferred embodiment, rheumatoid arthritis is treated,
prevented, and/or
diagnosed using polynucleotides, polypeptides, antibodies, and/or agonists or
antagonists of
the present invention. In another specific preferred embodiment, systemic
lupus erythemosus
is treated, prevented, and/or diagnosed using polynucleotides, polypeptides,
antibodies,
and/or agonists or antagonists of the present invention. In another specific
preferred
embodiment, idiopathic thrombocytopenia purpura is treated, prevented, and/or
diagnosed
using polynucleotides, polypeptides, antibodies, and/or agonists or
antagonists of the present
invention. In another specific preferred embodiment IgA nephropathy is
treated, prevented,
and/or diagnosed using polynucleotides, polypeptides, antibodies, and/or
agonists or
antagonists of the present invention. In a preferred embodiment, the
autoimmune diseases and
disorders and/or conditions associated with the diseases and disorders recited
above are
treated, prevented, and/or diagnosed using antibodies against the protein of
the invention.
Similarly, allergic reactions and conditions, such as asthma (particularly
allergic
asthma) or other respiratory problems, may also be treated, prevented, and/or
diagnosed using
polypeptides, antibodies, or polynucleotides of the invention, and/or agonists
or antagonists
thereof. Moreover, these molecules can be used to treat, prevent, and/or
diagnose
anaphylaxis, hypersensitivity to an antigenic molecule, or blood group
incompatibility.
Moreover, inflammatory conditions may also be treated, diagnosed, and/or
prevented
with polynucleotides, polypeptides, antibodies, and/or agonists or antagonists
of the present
invention. Such inflammatory conditions include, but are not limited to, for
example,
respiratory disorders (such as, e.g., asthma and allergy); gastrointestinal
disorders (such as,
e.g., inflammatory bowel disease); cancers (such as, e.g., gastric, ovarian,
lung, bladder, liver,
and breast); CNS disorders (such as, e.g., multiple sclerosis, blood-brain
barrier permeability,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
122
ischemic brain injury andlor stroke, traumatic brain injury, neurodegenerative
disorders (such
as, e.g., Parkinson's disease and Alzheimer's disease), AIDS-related dementia,
and prion
disease); cardiovascular disorders (such as, e.g., atherosclerosis,
myocarditis, cardiovascular
disease, and cardiopulmonary bypass complications); as well as many additional
diseases,
S conditions, and disorders that are characterized by inflammation (such as,
e.g., chronic
hepatitis (B and C), rheumatoid arthritis, gout, trauma, septic shock,
pancreatitis, sarcoidosis,
dermatitis, renal ischemia-reperfusion injury, Grave's disease, systemic lupus
erythematosis,
diabetes mellitus (i.e., type 1 diabetes), and allogenic transplant
rejection).
In specific embodiments, polypeptides, antibodies, or polynucleotides of the
invention, and/or agonists or antagonists thereof, are useful to treat,
diagnose, and/or prevent
transplantation rejections, graft-versus-host disease, autoimmune and
inflammatory diseases
(e.g., immune complex-induced vasculitis, glomerulonephritis, hemolytic
anemia, myasthenia
gravis, type II collagen-induced arthritis, experimental allergic and
hyperacute xenograft
rejection, rheumatoid arthritis, and systemic lupus erythematosus (SLE). Organ
rejection
occurs by host immune cell destruction of the transplanted tissue through an
immune
response. Similarly, an immune response is also involved in GVHD, but, in this
case, the
foreign transplanted immune cells destroy the host tissues. Polypeptides,
antibodies, or
polynucleotides of the invention, and/or agonists or antagonists thereof, that
inhibit an
immune response, particularly the activation, proliferation, differentiation,
or chemotaxis of
T-cells, may be an effective therapy in preventing organ rejection or GVHD.
Similarly, polynucleotides, polypeptides, antibodies, and/or agonists or
antagonists of
the present invention may also be used to modulate and/or diagnose
inflammation. For
example, since polypeptides, antibodies, or polynucleotides of the invention,
and/or agonists
or antagonists of the invention may inhibit the activation, proliferation
and/or differentiation
of cells involved in an inflammatory response, these molecules can be used to
treat, diagnose,
or prognose, inflammatory conditions, both chronic and acute conditions,
including, but not
limited to, inflammation associated with infection (e.g., septic shock,
sepsis, or systemic
inflammatory response syndrome (SIRS)), ischemia-reperfusion injury, endotoxin
lethality,
arthritis, complement-mediated hyperacute rejection, nephritis, cytokine or
chemokine
induced lung injury, inflammatory bowel disease, Crohn's disease, and
resulting from over
production of cytokines (e.g., TNF or IL-1.).


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
123
Polypeptides, antibodies, polynucleotides and/or agonists or antagonists of
the
invention can be used to treat, detect, and/or prevent infectious agents. For
example, by
increasing the immune response, particularly increasing the proliferation
activation and/or
differentiation of B and/or T cells, infectious diseases may be treated,
detected, and/or
prevented. The immune response may be increased by either enhancing an
existing immune
response, or by initiating a new immune response. Alternatively,
polynucleotides,
polypeptides, antibodies, and/or agonists or antagonists of the present
invention may also
directly inhibit the infectious agent (refer to section of application listing
infectious agents,
etc), without necessarily eliciting an immune response.
Additional preferred embodiments of the invention include, but are not limited
to, the
use of polypeptides, antibodies, polynucleotides and/or agonists or
antagonists in the
following applications:
Administration to an animal (e.g., mouse, rat, rabbit, hamster, guinea pig,
pigs, micro-
pig, chicken, camel, goat, horse, cow, sheep, dog, cat, non-human primate, and
human, most
preferably human) to boost the immune system to produce increased quantities
of one or
more antibodies (e.g., IgG, IgA, IgM, and IgE), to induce higher affinity
antibody production
(e.g., IgG, IgA, IgM, and IgE), and/or to increase an immune response.
Administration to an animal (including, but not limited to, those listed
above, and also
including transgenic animals) incapable of producing functional endogenous
antibody
molecules or having an otherwise compromised endogenous immune system, but
which is
capable of producing human immunoglobulin molecules by means of a
reconstituted or
partially reconstituted immune system from another animal (see, e.g.,
published PCT
Application Nos. W098/24893, WO/9634096, WO/9633735, and WO/9110741.
A vaccine adjuvant that enhances immune responsiveness to specific antigen.
An adjuvant to enhance tumor-specific immune responses.
An adj uvant to enhance anti-viral immune responses. Anti-viral immune
responses
that may be enhanced using the compositions of the invention as an adjuvant,
include virus
and virus associated diseases or symptoms described herein or otherwise known
in the art. In
specific embodiments, the compositions of the invention are used as an
adjuvant to enhance
an immune response to a virus, disease, or symptom selected from the group
consisting of
AIDS, meningitis, Dengue, EBV, and hepatitis (e.g., hepatitis B). In another
specific
embodiment, the compositions of the invention are used as an adjuvant to
enhance an


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
124
immune response to a virus, disease, or symptom selected from the group
consisting of:
HIV/AIDS, Respiratory syncytial virus, Dengue, Rotavirus, Japanese B
encephalitis,
Influenza A and B, Parainfluenza, Measles, Cytomegalovirus, Rabies, Junin,
Chikungunya,
Rift Valley fever, Herpes simplex, and yellow fever.
S An adjuvant to enhance anti-bacterial or anti-fungal immune responses. Anti-
bacterial
or anti-fungal immune responses that may be enhanced using the compositions of
the
invention as an adjuvant, include bacteria or fungus and bacteria or fungus
associated
diseases or symptoms described herein or otherwise known in the art. In
specific
embodiments, the compositions of the invention are used as an adjuvant to
enhance an
immune response to a bacteria or fungus, disease, or symptom selected from the
group
consisting of: tetanus, Diphtheria, botulism, and meningitis type B. In
another specific
embodiment, the compositions of the invention are used as an adjuvant to
enhance an
immune response to a bacteria or fungus, disease, or symptom selected from the
group
consisting of Vibrio cholerae, Mycobacterium leprae, Salmonella typhi,
Salmonella
1 S paratyphi, Meisseria meningitidis, Streptococcus pneumoniae, Group B
streptococcus,
Shigella spp., Enterotoxigenic Escherichia coli, Enterohemorrhagic E. coli,
Borrelia
burgdorferi, and Plasmodium (malaria).
An adjuvant to enhance anti-parasitic immune responses. Anti-parasitic immune
responses that may be enhanced using the compositions of the invention as an
adjuvant,
include parasite and parasite associated diseases or symptoms described herein
or otherwise
known in the art. In specific embodiments, the compositions of the invention
are used as an
adjuvant to enhance an immune response to a parasite. In another specific
embodiment, the
compositions of the invention are used as an adjuvant to enhance an immune
response to
Plasmodium (malaria).
As a stimulator of B cell responsiveness to pathogens.
As an activator of T cells.
As an agent that elevates the immune status of an individual prior to their
receipt of
immunosuppressive therapies.
As an agent to induce higher affinity antibodies.
As an agent to increase serum immunoglobulin concentrations.
As an agent to accelerate recovery of immunocompromised individuals.
As an agent to boost immunoresponsiveness among aged populations.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
125
As an immune system enhancer prior to, during, or after bone marrow transplant
and/or other transplants (e.g., allogeneic or xenogeneic organ
transplantation). With respect
to transplantation, compositions of the invention may be administered prior
to, concomitant
with, and/or after transplantation. In a specific embodiment, compositions of
the invention
are administered after transplantation, prior to the beginning of recovery of
T-cell
populations. In another specific embodiment, compositions of the invention are
first
administered after transplantation after the beginning of recovery of T cell
populations, but
prior to full recovery of B cell populations.
As an agent to boost immunoresponsiveness among individuals having an acquired
loss of B cell function. Conditions resulting in an acquired loss of B cell
function that may
be ameliorated or treated by administering the polypeptides, antibodies,
polynucleotides
and/or agonists or antagonists thereof, include, but are not limited to, HIV
Infection, AIDS,
bone marrow transplant, and B cell chronic lymphocytic leukemia (CLL).
As an agent to boost immunoresponsiveness among individuals having a temporary
immune deficiency. Conditions resulting in a temporary immune deficiency that
may be
ameliorated or treated by administering the polypeptides, antibodies,
polynucleotides and/or
agonists or antagonists thereof, include, but are not limited to, recovery
from viral infections
(e.g., influenza), conditions associated with malnutrition, recovery from
infectious
mononucleosis, or conditions associated with stress, recovery from measles,
recovery from
blood transfusion, recovery from surgery.
As a regulator of antigen presentation by monocytes, dendritic cells, and/or B-
cells.
In one embodiment, polynucleotides, polypeptides, antibodies, and/or agonists
or antagonists
of the present invention enhance antigen presentation or antagonizes antigen
presentation in
vitro or in vivo. Moreover, in related embodiments, said enhancement or
antagonization of
antigen presentation may be useful as an anti-tumor treatment or to modulate
the immune
system.
As an agent to direct an individuals immune system towards development of a
humoral response (i.e. TH2) as opposed to a TH1 cellular response.
As a means to induce tumor proliferation and thus make it more susceptible to
anti-
neoplastic agents. For example, multiple myeloma is a slowly dividing disease
and is thus
refractory to virtually all anti-neoplastic regimens. If these cells were
forced to proliferate
more rapidly their susceptibility profile would likely change.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
126
As a stimulator of B cell production in pathologies such as AIDS, chronic
lymphocyte
disorder and/or Common Variable Immunodificiency.
As a therapy for generation and/or regeneration of lymphoid tissues following
surgery, trauma or genetic defect.
As a gene-based therapy for genetically inherited disorders resulting in
immuno-
incompetence such as observed among SCID patients.
As an antigen for the generation of antibodies to inhibit or enhance immune
mediated
responses against polypeptides of the invention.
As a means of activating T cells.
As a means of activating monocytes/macrophages to defend against parasitic
diseases
that effect monocytes such as Leshmania.
As pretreatment of bone marrow samples prior to transplant. Such treatment
would
increase B cell representation and thus accelerate recover.
As a means of regulating secreted cytokines that are elicited by polypeptides
of the
invention.
Additionally, polypeptides or polynucleotides of the invention, and/or
agonists
thereof, may be used to treat or prevent IgE-mediated allergic reactions. Such
allergic
reactions include, but are not limited to, asthma, rhinitis, and eczema.
All of the above described applications as they may apply to veterinary
medicine.
Antagonists of the invention include, for example, binding and/or inhibitory
antibodies, antisense nucleic acids, ribozymes or soluble forms of the steroid
hormone
receptor (e.g., a steroid hormone receptor-Fc fusion protein) (see e.g.,
Example 9). These
would be expected to reverse many of the activities of the ligand described
above as well as
find clinical or practical application as:
A means of blocking various aspects of immune responses to foreign agents or
self.
Examples include autoimmune disorders such as lupus, and arthritis, as well as
immunoresponsiveness to skin allergies, inflammation, bowel disease, injury
and pathogens.
A therapy for preventing the B cell proliferation and Ig secretion associated
with
autoimmune diseases such as idiopathic thrombocytopenic purpura, systemic
lupus
erythramatosus and MS.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
127
An inhibitor of B and/or T cell migration in endothelial cells. This activity
disrupts
tissue architecture or cognate responses and is useful, for example in
disrupting immune
responses, and blocking sepsis.
An inhibitor of graft versus host disease or transplant rejection.
A therapy for B cell and/or T cell malignancies such as ALL, Hodgkins disease,
non-
Hodgkins lymphoma, Chronic lymphocyte leukemia, plasmacytomas, multiple
myeloma,
Burkitt's lymphoma, and EBV-transformed diseases.
A therapy for chronic hypergammaglobulinemeia evident in such diseases as
monoclonalgammopathy of undetermined significance (MGUS), Waldenstrom's
disease,
related idiopathic monoclonalgammopathies, and plasmacytomas.
A therapy for decreasing cellular proliferation of Large B-cell Lymphomas.
A means of decreasing the involvement of B cells and Ig associated with
Chronic
Myelogenous Leukemia.
An immunosuppressive agent(s).
Polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of
the
present invention may be used to modulate IgE concentrations in vitro or in
vivo.
In another embodiment, administration of polypeptides, antibodies,
polynucleotides
and/or agonists or antagonists of the invention, may be used to treat or
prevent IgE-mediated
allergic reactions including, but not limited to, asthma, rhinitis, and
eczema.
The agonists and antagonists may be employed in a composition with a
pharmaceutically acceptable carrier, e.g., as described herein.
The agonists or antagonists may be employed for instance to inhibit
polypeptide
chemotaxis and activation of macrophages and their precursors, and of
neutrophils, basophils,
B lymphocytes and some T-cell subsets, e.g., activated and CD8 cytotoxic T
cells and natural
killer cells, in certain auto-immune and chronic inflammatory and infective
diseases.
Examples of autoimmune diseases are described herein and include multiple
sclerosis, and
insulin-dependent diabetes. The antagonists or agonists may also be employed
to treat
infectious diseases including silicosis, sarcoidosis, idiopathic pulmonary
fibrosis by, for
example, preventing the recruitment and activation of mononuclear phagocytes.
They may
also be employed to treat idiopathic hyper-eosinophilic syndrome by, for
example, preventing
eosinophil production and migration. The antagonists or agonists or may also
be employed


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
128
for treating atherosclerosis, for example, by preventing monocyte infiltration
in the artery
wall.
Antibodies against polypeptides of the invention may be employed to treat
ARDS.
Agonists and/or antagonists of the invention also have uses in stimulating
wound and
tissue repair, stimulating angiogenesis, stimulating the repair of vascular or
lymphatic
diseases or disorders. Additionally, agonists and antagonists of the invention
may be used to
stimulate the regeneration of mucosal surfaces.
In a specific embodiment, polynucleotides or polypeptides, and/or agonists
thereof
are used to treat or prevent a disorder characterized by primary or acquired
immunodeficiency, deficient serum immunoglobulin production, recurrent
infections, and/or
immune system dysfunction. Moreover, polynucleotides or polypeptides, and/or
agonists
thereof may be used to treat or prevent infections of the joints, bones, skin,
and/or parotid
glands, blood-borne infections (e.g., sepsis, meningitis, septic arthritis,
and/or osteomyelitis),
autoimmune diseases (e.g., those disclosed herein), inflammatory disorders,
and
malignancies, and/or any disease or disorder or condition associated with
these infections,
diseases, disorders and/or malignancies) including, but not limited to, CV>D,
other primary
immune deficiencies, HIV disease, CLL, recurrent bronchitis, sinusitis, otitis
media,
conjunctivitis, pneumonia, hepatitis, meningitis, herpes zoster (e.g., severe
herpes zoster),
and/or pneumocystis carnii.
In another embodiment, polynucleotides, polypeptides, antibodies, and/or
agonists or
antagonists of the present invention are used to treat, and/or diagnose an
individual having
common variable immunodeficiency disease ("CVID"; also known as "acquired
agammaglobulinemia" and "acquired hypogammaglobulinemia") or a subset of this
disease.
In a specific embodiment, polynucleotides, polypeptides, antibodies, and/or
agonists
or antagonists of the present invention may be used to treat, diagnose, and/or
prevent (1)
cancers or neoplasms and (2) autoimmune cell or tissue-related cancers or
neoplasms. In a
preferred embodiment, polynucleotides, polypeptides, antibodies, and/or
agonists or
antagonists of the present invention conjugated to a toxin or a radioactive
isotope, as
described herein, may be used to treat, diagnose, and/or prevent acute
myelogeneous
leukemia. In a further preferred embodiment, polynucleotides, polypeptides,
antibodies,
and/or agonists or antagonists of the present invention conjugated to a toxin
or a radioactive
isotope, as described herein, may be used to treat, diagnose, and/or prevent,
chronic


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
129
myelogeneous leukemia, multiple myeloma, non-Hodgkins lymphoma, and/or
Hodgkins
disease.
In another specific embodiment, polynucleotides or polypeptides, and/or
agonists or
antagonists of the invention may be used to treat, diagnose, prognose, and/or
prevent
selective IgA deficiency, myeloperoxidase deficiency, C2 deficiency, ataxia-
telangiectasia,
DiGeorge anomaly, common variable immunodeficiency (CVI), X-linked
agammaglobulinemia, severe combined immunodeficiency (SCID), chronic
granulomatous
disease (CGD), and Wiskott-Aldrich syndrome.
Examples of autoimmune disorders that can be treated or detected are described
above
and also include, but are not limited to: Addison's Disease, hemolytic anemia,
antiphospholipid syndrome, rheumatoid arthritis, dermatitis, allergic
encephalomyelitis,
glomerulonephritis, Goodpasture's Syndrome, Graves' Disease, Multiple
Sclerosis,
Myasthenia Gravis, Neuritis, Ophthalmia, Bullous Pemphigoid, Pemphigus,
Polyendocrinopathies, Purpura, Reiter's Disease, Stiff Man Syndrome,
Autoimmune
Thyroiditis, Systemic Lupus Erythematosus, Autoimmune Pulmonary Inflammation,
Guillain-Barre Syndrome, insulin dependent diabetes mellitis, and autoimmune
inflammatory
eye disease.
In a preferred embodiment, the autoimmune diseases and disorders andlor
conditions
associated with the diseases and disorders recited above are treated,
prevented, and/or
diagnosed using steroid hormone receptor antibodies and/or anti-steroid
hormone receptor
antibodies and/or a soluble steroid hormone receptor polypeptide of the
invention.
In specific embodiments, the compositions of the invention are used as an
agent to
boost immunoresponsiveness among B cell immunodeficient individuals, such as,
for
example, an individual who has undergone a partial or complete splenectomy.
Additionally, polynucleotides, polypeptides, and/or antagonists of the
invention may
affect apoptosis, and therefore, would be useful in treating a number of
diseases associated
with increased cell survival or the inhibition of apoptosis. For example,
diseases associated
with increased cell survival or the inhibition of apoptosis that could be
treated or detected by
polynucleotides, polypeptides, andlor antagonists of the invention, include
cancers (such as
follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent
tumors,
including, but not limited to colon cancer, cardiac tumors, pancreatic cancer,
melanoma,
retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular
cancer, stomach


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
130
cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma,
osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate
cancer,
Kaposi's sarcoma and ovarian cancer); autoimmune disorders (such as, multiple
sclerosis,
Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's
disease, Crohn's
disease, polymyositis, systemic lupus erythematosus and immune-related
glomerulonephritis
and rheumatoid arthritis) and viral infections (such as herpes viruses, pox
viruses and
adenoviruses), inflammation, graft v. host disease, acute graft rejection, and
chronic graft
rejection. In preferred embodiments, polynucleotides, polypeptides, and/or
antagonists of the
invention are used to inhibit growth, progression, and/or metastisis of
cancers, in particular
those listed above.
Additional diseases or conditions associated with increased cell survival that
could be
treated or detected by polynucleotides, polypeptides, and/or antagonists of
the invention,
include, but are not limited to, progression, and/or metastases of
malignancies and related
disorders such as leukemia (including acute leukemias (e.g., acute lymphocytic
leukemia,
acute myelocytic leukemia (including myeloblastic, promyelocytic,
myelomonocytic,
monocytic, and erythroleukemia)) and chronic leukemias (e.g., chronic
myelocytic
(granulocytic) leukemia and chronic lymphocytic leukemia)), polycythemia vera,
lymphomas
(e.g., Hodgkin's disease and non-Hodgkin's disease), multiple myeloma,
Waldenstrom's
macroglobulinemia, heavy chain disease, and solid tumors including, but not
limited to,
sarcomas and carcinomas such as fibrosarcoma, myxosarcoma, liposarcoma,
chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma,
lymphangiosarcoma, lyrnphangioendotheliosarcoma, synovioma, mesothelioma,
Ewing's
tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer,
breast
cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell
carcinoma,
adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary
carcinoma,
papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma,
bronchogenic
carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma,
choriocarcinoma,
seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular
tumor, lung
carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma,
glioma,
astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma,
hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma,
neuroblastoma, and retinoblastoma.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
131
Diseases associated with increased apoptosis that could be treated or detected
by
polynucleotides, polypeptides, and/or antagonists of the invention, include
AIDS;
neurodegenerative disorders (such as Alzheimer's disease, Parkinson's disease,
Amyotrophic
lateral sclerosis, Retinitis pigmentosa, Cerebellar degeneration and brain
tumor or prior
associated disease); autoimmune disorders (such as, multiple sclerosis,
Sjogren's syndrome,
Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease,
polymyositis,
systemic lupus erythematosus and immune-related glomerulonephritis and
rheumatoid
arthritis) myelodysplastic syndromes (such as aplastic anemia), graft v. host
disease, ischemic
injury (such as that caused by myocardial infarction, stroke and reperfusion
injury), liver
injury (e.g., hepatitis related liver injury, ischemia/reperfusion injury,
cholestosis (bile duct
injury) and liver cancer); toxin-induced liver disease (such as that caused by
alcohol), septic
shock, cachexia and anorexia.
Hyperproliferative diseases and/or disorders that could be detected and/or
treated by
polynucleotides, polypeptides, and/or antagonists of the invention, include,
but are not
limited to neoplasms located in the: liver, abdomen, bone, breast, digestive
system, pancreas,
peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles,
ovary, thymus,
thyroid), eye, head and neck, nervous (central and peripheral), lymphatic
system, pelvic, skin,
soft tissue, spleen, thoracic, and urogenital.
Similarly, other hyperproliferative disorders can also be treated or detected
by
polynucleotides, polypeptides, and/or antagonists of the invention. Examples
of such
hyperproliferative disorders include, but are not limited to:
hypergammaglobulinemia,
lymphoproliferative disorders, paraproteinemias, purpura, sarcoidosis, Sezary
Syndrome,
Waldenstron's Macroglobulinemia, Gaucher's Disease, histiocytosis, and any
other
hyperproliferative disease, besides neoplasia, located in an organ system
listed above.
Hyperproliferative Disorders
Polynucleotides or polypeptides, or agonists or antagonists of the present
invention
can be used to treat or detect hyperproliferative disorders, including
neoplasms.
Polynucleotides or polypeptides, or agonists or antagonists of the present
invention may
inhibit the proliferation of the disorder through direct or indirect
interactions. Alternatively,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
132
Polynucleotides or polypeptides, or agonists or antagonists of the present
invention may
proliferate other cells which can inhibit the hyperproliferative disorder.
For example, by increasing an immune response, particularly increasing
antigenic
qualities of the hyperproliferative disorder or by proliferating,
differentiating, or mobilizing
T-cells, hyperproliferative disorders can be treated. This immune response may
be increased
by either enhancing an existing immune response, or by initiating a new immune
response.
Alternatively, decreasing an immune response may also be a method of treating
hyperproliferative disorders, such as a chemotherapeutic agent.
Examples of hyperproliferative disorders that can be treated or detected by
Polynucleotides or polypeptides, or agonists or antagonists of the present
invention include,
but are not limited to neoplasms located in the: colon, abdomen, bone, breast,
digestive
system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid,
pituitary,
testicles, ovary, thymus, thyroid), eye, head and neck, nervous (central and
peripheral),
lymphatic system, pelvic, skin, soft tissue, spleen, thoracic, and urogenital.
Similarly, other hyperproliferative disorders can also be treated or detected
by
polynucleotides or polypeptides, or agonists or antagonists of the present
invention.
Examples of such hyperproliferative disorders include, but are not limited to:
hypergammaglobulinemia, lymphoproliferative disorders, paraproteinemias,
purpura,
sarcoidosis, Sezary Syndrome, Waldenstron's Macroglobulinemia, Gaucher's
Disease,
histiocytosis, and any other hyperproliferative disease, besides neoplasia,
located in an organ
system listed above.
One preferred embodiment utilizes polynucleotides of the present invention to
inhibit
aberrant cellular division, by gene therapy using the present invention,
and/or protein fusions
or fragments thereof.
Thus, the present invention provides a method for treating cell proliferative
disorders
by inserting into an abnormally proliferating cell a polynucleotide of the
present invention,
wherein said polynucleotide represses said expression.
Another embodiment of the present invention provides a method of treating cell-

proliferative disorders in individuals comprising administration of one or
more active gene
copies of the present invention to an abnormally proliferating cell or cells.
In a preferred
embodiment, polynucleotides of the present invention is a DNA construct
comprising a
recombinant expression vector effective in expressing a DNA sequence encoding
said


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
133
polynucleotides. In another preferred embodiment of the present invention, the
DNA
construct encoding the poynucleotides of the present invention is inserted
into cells to be
treated utilizing a retrovirus, or more preferrably an adenoviral vector (See
G J. Nabel, et. al.,
PNAS 1999 96: 324-326, which is hereby incorporated by reference). In a most
preferred
embodiment, the viral vector is defective and will not transform non-
proliferating cells, only
proliferating cells. Moreover, in a preferred embodiment, the polynucleotides
of the present
invention inserted into proliferating cells either alone, or in combination
with or fused to
other polynucleotides, can then be modulated via an external stimulus (i.e.
magnetic, specific
small molecule, chemical, or drug administration, etc.), which acts upon the
promoter
upstream of said polynucleotides to induce expression of the encoded protein
product. As
such the beneficial therapeutic affect of the present invention may be
expressly modulated
(i.e. to increase, decrease, or inhibit expression of the present invention)
based upon said
external stimulus.
The polynucleotides encoding a polypeptide of the present invention may be
administered along with other polynucleotides encoding an angiogenic protein.
Examples of
angiogenic proteins include, but are not limited to, acidic and basic
fibroblast growth factors,
VEGF-1, VEGF-2, VEGF-3, epidermal growth factor alpha and beta, platelet-
derived
endothelial cell growth factor, platelet-derived growth factor, tumor necrosis
factor alpha,
hepatocyte growth factor, insulin like growth factor, colony stimulating
factor, macrophage
colony stimulating factor, granulocyte/macrophage colony stimulating factor,
and nitric oxide
synthase.
Polynucleotides of the present invention may be useful in repressing
expression of
oncogenic genes or antigens. By "repressing expression of the oncogenic genes
" is intended
the suppression of the transcription of the gene, the degradation of the gene
transcript (pre-
message RNA), the inhibition of splicing, the destruction of the messenger
RNA, the
prevention of the post-translational modifications of the protein, the
destruction of the
protein, or the inhibition of the normal function of the protein.
For local administration to abnormally proliferating cells, polynucleotides of
the
present invention may be administered by any method known to those of skill in
the art
including, but not limited to transfection, electroporation, microinjection of
cells, or in
vehicles such as liposomes, lipofectin, or as naked polynucleotides, or any
other method
described throughout the specification. The polynucleotide of the present
invention may be


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
134
delivered by known gene delivery systems such as, but not limited to,
retroviral vectors
(Gilboa, J. Virology 44:845 (1982); Hocke, Nature 320:275 (1986); Wilson, et
al., Proc. Natl.
Acad. Sci. U.S.A. 85:3014), vaccinia virus system (Chakrabarty et al., Mol.
Cell Biol. 5:3403
(1985) or other efficient DNA delivery systems (Yates et al., Nature 313:812
(1985)) known
S to those skilled in the art. These references are exemplary only and are
hereby incorporated
by reference. In order to specifically deliver or transfect cells which are
abnormally
proliferating and spare non-dividing cells, it is preferable to utilize a
retrovirus, or adenoviral
(as described in the art and elsewhere herein) delivery system known to those
of skill in the
art. Since host DNA replication is required for retroviral DNA to integrate
and the retrovirus
will be unable to self replicate due to the lack of the retrovirus genes
needed for its life cycle.
Utilizing such a retroviral delivery system for polynucleotides of the present
invention will
target said gene and constructs to abnormally proliferating cells and will
spare the non-
dividing normal cells.
The polynucleotides of the present invention may be delivered directly to cell
proliferative disorder/disease sites in internal organs, body cavities and the
like by use of
imaging devices used to guide an injecting needle directly to the disease
site. The
polynucleotides of the present invention may also be administered to disease
sites at the time
of surgical intervention.
By "cell proliferative disease" is meant any human or animal disease or
disorder,
affecting any one or any combination of organs, cavities, or body parts, which
is
characterized by single or multiple local abnormal proliferations of cells,
groups of cells, or
tissues, whether benign or malignant.
Any amount of the polynucleotides of the present invention may be administered
as
long as it has a biologically inhibiting effect on the proliferation of the
treated cells.
Moreover, it is possible to administer more than one of the polynucleotide of
the present
invention simultaneously to the same site. By "biologically inhibiting" is
meant partial or
total growth inhibition as well as decreases in the rate of proliferation or
growth of the cells.
The biologically inhibitory dose may be determined by assessing the effects of
the
polynucleotides of the present invention on target malignant or abnormally
proliferating cell
growth in tissue culture, tumor growth in animals and cell cultures, or any
other method
known to one of ordinary skill in the art.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
135
The present invention is further directed to antibody-based therapies which
involve
administering of anti-polypeptides and anti-polynucleotide antibodies to a
mammalian,
preferably human, patient for treating one or more of the described disorders.
Methods for
producing anti-polypeptides and anti-polynucleotide antibodies polyclonal and
monoclonal
antibodies are described in detail elsewhere herein. Such antibodies may be
provided in
pharmaceutically acceptable compositions as known in the art or as described
herein.
A summary of the ways in which the antibodies of the present invention may be
used
therapeutically includes binding polynucleotides or polypeptides of the
present invention
locally or systemically in the body or by direct cytotoxicity of the antibody,
e.g. as mediated
by complement (CDC) or by effector cells (ADCC). Some of these approaches are
described
in more detail below. Armed with the teachings provided herein, one of
ordinary skill in the
art will know how to use the antibodies of the present invention for
diagnostic, monitoring or
therapeutic purposes without undue experimentation.
In particular, the antibodies, fragments and derivatives of the present
invention are
useful for treating a subject having or developing cell proliferative and/or
differentiation
disorders as described herein. Such treatment comprises administering a single
or multiple
doses of the antibody, or a fragment, derivative, or a conjugate thereof.
The antibodies of this invention may be advantageously utilized in combination
with
other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic
growth
factors, for example., which serve to increase the number or activity of
effector cells which
interact with the antibodies.
It is preferred to use high affinity and/or potent in vivo inhibiting and/or
neutralizing
antibodies against polypeptides or polynucleotides of the present invention,
fragments or
regions thereof, for both immunoassays directed to and therapy of disorders
related to
polynucleotides or polypeptides, including fragements thereof, of the present
invention. Such
antibodies, fragments, or regions, will preferably have an affinity for
polynucleotides or
polypeptides, including fragements thereof. Preferred binding affinities
include those with a
dissociation constant or Kd less than SX10-6M, 10-6M, SX10-~M, 10-7M, SXlO~gM,
10-8M,
SX10-~M, 10-9M, 5X10-'°M, 10~'°M, SX10-"M, 10-''M, SX10-'2M, 10-
'2M, SX10-'3M, 10-
'3M, SX10-'4M, 10-'4M, SX10-'SM, and 10-'SM.
Moreover, polypeptides of the present invention are useful in inhibiting the
angiogenesis of proliferative cells or tissues, either alone, as a protein
fusion, or in


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
136
combination with other polypeptides directly or indirectly, as described
elsewhere herein. In a
most preferred embodiment, said anti-angiogenesis effect may be achieved
indirectly, for
example, through the inhibition of hematopoietic, tumor-specific cells, such
as tumor-
associated macrophages (See Joseph IB, et al. J Natl Cancer Inst, 90(21):1648-
53 (1998),
which is hereby incorporated by reference). Antibodies directed to
polypeptides or
polynucleotides of the present invention may also result in inhibition of
angiogenesis directly,
or indirectly (See Witte L, et al., Cancer Metastasis Rev. 17(2):155-61
(1998), which is
hereby incorporated by reference)).
Polypeptides, including protein fusions, of the present invention, or
fragments thereof
may be useful in inhibiting proliferative cells or tissues through the
induction of apoptosis.
Said polypeptides may act either directly, or indirectly to induce apoptosis
of proliferative
cells and tissues, for example in the activation of a death-domain receptor,
such as tumor
necrosis factor (TNF) receptor-1, CD95 (Fas/APO-1), TNF-receptor-related
apoptosis-
mediated protein (TRAMP) and TNF-related apoptosis-inducing ligand (TRAIL)
receptor-1
and -2 (See Schulze-Osthoff K, et.al., Eur J Biochem 254(3):439-59 (1998),
which is hereby
incorporated by reference). Moreover, in another preferred embodiment of the
present
invention, said polypeptides may induce apoptosis through other mechanisms,
such as in the
activation of other proteins which will activate apoptosis, or through
stimulating the
expression of said proteins, either alone or in combination with small
molecule drugs or
adjuviants, such as apoptonin, galectins, thioredoxins, antiinflammatory
proteins (See for
example, Mutat Res 400(1-2):447-55 (1998), Med Hypotheses.50(5):423-33 (1998),
Chem
Biol Interact. Apr 24;111-112:23-34 (1998), J Mol Med.76(6):402-12 (1998), Int
J Tissue
React;20(1):3-15 (1998), which are all hereby incorporated by reference).
Polypeptides, including protein fusions to, or fragments thereof, of the
present
invention are useful in inhibiting the metastasis of proliferative cells or
tissues. Inhibition
may occur as a direct result of administering polypeptides, or antibodies
directed to said
polypeptides as described elsewere herein, or indirectly, such as activating
the expression of
proteins known to inhibit metastasis, for example alpha 4 integrins, (See,
e.g., Curr Top
Microbiol Immunol 1998;231:125-41, which is hereby incorporated by reference).
Such
thereapeutic affects of the present invention may be achieved either alone, or
in combination
with small molecule drugs or adjuvants.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
137
In another embodiment, the invention provides a method of delivering
compositions
containing the polypeptides of the invention (e.g., compositions containing
polypeptides or
polypeptide antibodes associated with heterologous polypeptides, heterologous
nucleic acids,
toxins, or prodrugs) to targeted cells expressing the polypeptide of the
present invention.
Polypeptides or polypeptide antibodes of the invention may be associated with
with
heterologous polypeptides, heterologous nucleic acids, toxins, or prodrugs via
hydrophobic,
hydrophilic, ionic and/or covalent interactions.
Polypeptides, protein fusions to, or fragments thereof, of the present
invention are
useful in enhancing the immunogenicity and/or antigenicity of proliferating
cells or tissues,
either directly, such as would occur if the polypeptides of the present
invention 'vaccinated'
the immune response to respond to proliferative antigens and immunogens, or
indirectly, such
as in activating the expression of proteins known to enhance the immune
response (e.g.
chemokines), to said antigens and immunogens.
Cardiovascular Disorders
Polynucleotides or polypeptides, or agonists or antagonists of the present
invention,
may be used to treat cardiovascular disorders, including peripheral artery
disease, such as
limb ischemia.
Cardiovascular disorders include cardiovascular abnormalities, such as arterio-
arterial
fistula, arteriovenous fistula, cerebral arteriovenous malformations,
congenital heart defects,
pulmonary atresia, and Scimitar Syndrome. Congenital heart defects include
aortic
coarctation, cor triatriatum, coronary vessel anomalies, crisscross heart,
dextrocardia, patent
ductus arteriosus, Ebstein's anomaly, Eisenmenger complex, hypoplastic left
heart syndrome,
levocardia, tetralogy of fallot, transposition of great vessels, double outlet
right ventricle,
tricuspid atresia, persistent truncus arteriosus, and heart septal defects,
such as
aortopulmonary septal defect, endocardial cushion defects, Lutembacher's
Syndrome, trilogy
of Fallot, ventricular heart septal defects.
Cardiovascular disorders also include heart disease, such as arrhythmias,
carcinoid
heart disease, high cardiac output, low cardiac output, cardiac tamponade,
endocarditis
(including bacterial), heart aneurysm, cardiac arrest, congestive heart
failure, congestive
cardiomyopathy, paroxysmal dyspnea, cardiac edema, heart hypertrophy,
congestive
cardiomyopathy, left ventricular hypertrophy, right ventricular hypertrophy,
post-infarction


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
138
heart rupture, ventricular septal rupture, heart valve diseases, myocardial
diseases,
myocardial ischemia, pericardial effusion, pericarditis (including
constrictive and
tuberculous), pneumopericardium, postpericardiotomy syndrome, pulmonary heart
disease,
rheumatic heart disease, ventricular dysfunction, hyperemia, cardiovascular
pregnancy
complications, Scimitar Syndrome, cardiovascular syphilis, and cardiovascular
tuberculosis.
Arrhythmias include sinus arrhythmia, atrial fibrillation, atrial flutter,
bradycardia,
extrasystole, Adams-Stokes Syndrome, bundle-branch block, sinoatrial block,
long QT
syndrome, parasystole, Lown-Ganong-Levine Syndrome, Mahaim-type pre-excitation
syndrome, Wolff Parkinson-White syndrome, sick sinus syndrome, tachycardias,
and
ventricular fibrillation. Tachycardias include paroxysmal tachycardia,
supraventricular
tachycardia, accelerated idioventricular rhythm, atrioventricular nodal
reentry tachycardia,
ectopic atrial tachycardia, ectopic functional tachycardia, sinoatrial nodal
reentry tachycardia,
sinus tachycardia, Torsades de Pointes, and ventricular tachycardia.
Heart valve disease include aortic valve insufficiency, aortic valve stenosis,
hear
murmurs, aortic valve prolapse, mitral valve prolapse, tricuspid valve
prolapse, mitral valve
insufficiency, mitral valve stenosis, pulmonary atresia, pulmonary valve
insufficiency,
pulmonary valve stenosis, tricuspid atresia, tricuspid valve insufficiency,
and tricuspid valve
stenosis.
Myocardial diseases include alcoholic cardiomyopathy, congestive
cardiomyopathy,
hypertrophic cardiomyopathy, aortic subvalvular stenosis, pulmonary
subvalvular stenosis,
restrictive cardiomyopathy, Chagas cardiomyopathy, endocardial fibroelastosis,
endomyocardial fibrosis, Kearns Syndrome, myocardial reperfusion injury, and
myocarditis.
Myocardial ischemias include coronary disease, such as angina pectoris,
coronary
aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasospasm,
myocardial
infarction and myocardial stunning.
Cardiovascular diseases also include vascular diseases such as aneurysms,
angiodysplasia, angiomatosis, bacillary angiomatosis, Hippel-Lindau Disease,
Klippel-
Trenaunay-Weber Syndrome, Sturge-Weber Syndrome, angioneurotic edema, aortic
diseases,
Takayasu's Arteritis, aortitis, Leriche's Syndrome, arterial occlusive
diseases, arteritis,
enarteritis, polyarteritis nodosa, cerebrovascular disorders, diabetic
angiopathies, diabetic
retinopathy, embolisms, thrombosis, erythromelalgia, hemorrhoids, hepatic veno-
occlusive
disease, hypertension, hypotension, ischemia, peripheral vascular diseases,
phlebitis,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
139
pulmonary veno-occlusive disease, Raynaud's disease, CREST syndrome, retinal
vein
occlusion, Scimitar syndrome, superior vena cava syndrome, telangiectasia,
atacia
telangiectasia, hereditary hemorrhagic telangiectasia, varicocele, varicose
veins, varicose
ulcer, vasculitis, and venous insufficiency.
Aneurysms include dissecting aneurysms, false aneurysms, infected aneurysms,
ruptured aneurysms, aortic aneurysms, cerebral aneurysms, coronary aneurysms,
heart
aneurysms, and iliac aneurysms.
Arterial occlusive diseases include arteriosclerosis, intermittent
claudication, carotid
stenosis, fibromuscular dysplasias, mesenteric vascular occlusion, Moyamoya
disease, renal
artery obstruction, retinal artery occlusion, and thromboangiitis obliterans.
Cerebrovascular disorders include carotid artery diseases, cerebral amyloid
angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis,
cerebral
arteriovenous malformation, cerebral artery diseases, cerebral embolism and
thrombosis,
carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, cerebral
hemorrhage,
epidural hematoma, subdural hematoma, subaraxhnoid hemorrhage, cerebral
infarction,
cerebral ischemia (including transient), subclavian steal syndrome,
periventricular
leukomalacia, vascular headache, cluster headache, migraine, and
vertebrobasilar
insufficiency.
Embolisms include air embolisms, amniotic fluid embolisms, cholesterol
embolisms,
blue toe syndrome, fat embolisms, pulmonary embolisms, and thromoboembolisms.
Thrombosis include coronary thrombosis, hepatic vein thrombosis, retinal vein
occlusion,
carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, and
thrombophlebitis.
Ischemia includes cerebral ischemia, ischemic colitis, compartment syndromes,
anterior compartment syndrome, myocardial ischemia, reperfusion injuries, and
peripheral
limb ischemia. Vasculitis includes aortitis, arteritis, Behcet's Syndrome,
Churg-Strauss
Syndrome, mucocutaneous lymph node syndrome, thromboangiitis obliterans,
hypersensitivity vasculitis, Schoenlein-Henoch purpura, allergic cutaneous
vasculitis, and
Wegener's granulomatosis.
Polynucleotides or polypeptides, or agonists or antagonists of the present
invention,
are especially effective for the treatment of critical limb ischemia and
coronary disease.
Polypeptides may be administered using any method known in the art, including,
but
not limited to, direct needle injection at the delivery site, intravenous
injection, topical


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
140
administration, catheter infusion, biolistic injectors, particle accelerators,
gelfoam sponge
depots, other commercially available depot materials, osmotic pumps, oral or
suppositorial
solid pharmaceutical formulations, decanting or topical applications during
surgery, aerosol
delivery. Such methods are known in the art. Polypeptides may be administered
as part of a
Therapeutic, described in more detail below. Methods of delivering
polynucleotides are
described in more detail herein.
Anti-Angiogenesis Activity
The naturally occurnng balance between endogenous stimulators and inhibitors
of
angiogenesis is one in which inhibitory influences predominate. Rastinejad et
al., Cell
56:345-355 (1989). In those rare instances in which neovascularization occurs
under normal
physiological conditions, such as wound healing, organ regeneration, embryonic
development, and female reproductive processes, angiogenesis is stringently
regulated and
spatially and temporally delimited. Under conditions of pathological
angiogenesis such as
that characterizing solid tumor growth, these regulatory controls fail.
Unregulated
angiogenesis becomes pathologic and sustains progression of many neoplastic
and non-
neoplastic diseases. A number of serious diseases are dominated by abnormal
neovascularization including solid tumor growth and metastases, arthritis,
some types of eye
disorders, and psoriasis. See, e.g., reviews by Moses et al., Biotech. 9:630-
634 (1991);
Folkman et al., N. Engl. J. Med., 333:1757-1763 (1995); Auerbach et al., J.
Microvasc. Res.
29:401-411 (1985); Folkman, Advances in Cancer Research, eds. Klein and
Weinhouse,
Academic Press, New York, pp. 175-203 (1985); Patz, Am. J. Opthalmol. 94:715-
743
(1982); and Folkman et al., Science 221:719-725 (1983). In a number of
pathological
conditions, the process of angiogenesis contributes to the disease state. For
example,
significant data have accumulated which suggest that the growth of solid
tumors is dependent
on angiogenesis. Folkman and Klagsbrun, Science 235:442-447 (1987).
The present invention provides for treatment of diseases or disorders
associated with
neovascularization by administration of the polynucleotides and/or
polypeptides of the
invention, as well as agonists or antagonists of the present invention.
Malignant and
metastatic conditions which can be treated with the polynucleotides and
polypeptides, or
agonists or antagonists of the invention include, but are not limited to,
malignancies, solid
tumors, and cancers described herein and otherwise known in the art (for a
review of such


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
141
disorders, see Fishman et al., Medicine, 2d Ed., J. B. Lippincott Co.,
Philadelphia
(1985)).Thus, the present invention provides a method of treating an
angiogenesis-related
disease and/or disorder, comprising administering to an individual in need
thereof a
therapeutically effective amount of a polynucleotide, polypeptide, antagonist
and/or agonist
of the invention. For example, polynucleotides, polypeptides, antagonists
and/or agonists
may be utilized in a variety of additional methods in order to therapeutically
treat a cancer or
tumor. Cancers which may be treated with polynucleotides, polypeptides,
antagonists and/or
agonists include, but are not limited to solid tumors, including prostate,
lung, breast, ovarian,
stomach, pancreas, larynx, esophagus, testes, liver, parotid, biliary tract,
colon, rectum,
cervix, uterus, endometrium, kidney, bladder, thyroid cancer; primary tumors
and metastases;
melanomas; glioblastoma; Kaposi's sarcoma; leiomyosarcoma; non- small cell
lung cancer;
colorectal cancer; advanced malignancies; and blood born tumors such as
leukemias. For
example, polynucleotides, polypeptides, antagonists and/or agonists may be
delivered
topically, in order to treat cancers such as skin cancer, head and neck
tumors, breast tumors,
1 S and Kaposi's sarcoma.
Within yet other aspects, polynucleotides, polypeptides, antagonists and/or
agonists
may be utilized to treat superficial forms of bladder cancer by, for example,
intravesical
administration. Polynucleotides, polypeptides, antagonists and/or agonists may
be delivered
directly into the tumor, or near the tumor site, via injection or a catheter.
Of course, as the
artisan of ordinary skill will appreciate, the appropriate mode of
administration will vary
according to the cancer to be treated. Other modes of delivery are discussed
herein.
Polynucleotides, polypeptides, antagonists and/or agonists may be useful in
treating
other disorders, besides cancers, which involve angiogenesis. These disorders
include, but
are not limited to: benign tumors, for example hemangiomas, acoustic neuromas,
neurofibromas, trachomas, and pyogenic granulomas; artheroscleric plaques;
ocular
- angiogenic diseases, for example, diabetic retinopathy, retinopathy of
prematurity, macular
degeneration, corneal graft rejection, neovascular glaucoma, retrolental
fibroplasia, rubeosis,
retinoblastoma, uvietis and Pterygia (abnormal blood vessel growth) of the
eye; rheumatoid
arthritis; psoriasis; delayed wound healing; endometriosis; vasculogenesis;
granulations;
hypertrophic scars (keloids); nonunion fractures; scleroderma; trachoma;
vascular adhesions;
myocardial angiogenesis; coronary collaterals; cerebral collaterals;
arteriovenous
malformations; ischemic limb angiogenesis; Osler-Webber Syndrome; plaque


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
142
neovascularization; telangiectasia; hemophiliac joints; angiofibroma;
fibromuscular
dysplasia; wound granulation; Crohn's disease; and atherosclerosis.
For example, within one aspect of the present invention methods are provided
for
treating hypertrophic scars and keloids, comprising the step of administering
a
polynucleotide, polypeptide, antagonist and/or agonist of the invention to a
hypertrophic scar
or keloid.
Within one embodiment of the present invention polynucleotides, polypeptides,
antagonists and/or agonists are directly injected into a hypertrophic scar or
keloid, in order to
prevent the progression of these lesions. This therapy is of particular value
in the
prophylactic treatment of conditions which are known to result in the
development of
hypertrophic scars and keloids (e.g., burns), and is preferably initiated
after the proliferative
phase has had time to progress (approximately 14 days after the initial
injury), but before
hypertrophic scar or keloid development. As noted above, the present invention
also
provides methods for treating neovascular diseases of the eye, including for
example, corneal
neovascularization, neovascular glaucoma, proliferative diabetic retinopathy,
retrolental
fibroplasia and macular degeneration.
Moreover, Ocular disorders associated with neovascularization which can be
treated
with the polynucleotides and polypeptides of the present invention (including
agonists and/or
antagonists) include, but are not limited to: neovascular glaucoma, diabetic
retinopathy,
retinoblastoma, retrolental fibroplasia, uveitis, retinopathy of prematurity
macular
degeneration, corneal graft neovascularization, as well as other eye
inflammatory diseases,
ocular tumors and diseases associated with choroidal or iris
neovascularization. See, e.g.,
reviews by Waltman et al., Am. J. Ophthal. 85:704-710 (1978) and Gartner et
al., Surv.
Ophthal. 22:291-312 (1978).
Thus, within one aspect of the present invention methods are provided for
treating
neovascular diseases of the eye such as corneal neovascularization (including
corneal graft
neovascularization), comprising the step of administering to a patient a
therapeutically
effective amount of a compound (as described above) to the cornea, such that
the formation
of blood vessels is inhibited. Briefly, the cornea is a tissue which normally
lacks blood
vessels. In certain pathological conditions however, capillaries may extend
into the cornea
from the pericorneal vascular plexus of the limbus. When the cornea becomes
vascularized,
it also becomes clouded, resulting in a decline in the patient's visual
acuity. Visual loss may


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
143
become complete if the cornea completely opacitates. A wide variety of
disorders can result
in corneal neovascularization, including for example, corneal infections
(e.g., trachoma,
herpes simplex keratitis, leishmaniasis and onchocerciasis), immunological
processes (e.g.,
graft rejection and Stevens-Johnson's syndrome), alkali burns, trauma,
inflammation (of any
cause), toxic and nutritional deficiency states, and as a complication of
wearing contact
lenses.
Within particularly preferred embodiments of the invention, may be prepared
for
topical administration in saline (combined with any of the preservatives and
antimicrobial
agents commonly used in ocular preparations), and administered in eyedrop
form. The
solution or suspension may be prepared in its pure form and administered
several times daily.
Alternatively, anti-angiogenic compositions, prepared as described above, may
also be
administered directly to the cornea. Within preferred embodiments, the anti-
angiogenic
composition is prepared with a muco-adhesive polymer which binds to cornea.
Within
further embodiments, the anti-angiogenic factors or anti-angiogenic
compositions may be
utilized as an adjunct to conventional steroid therapy. Topical therapy may
also be useful
prophylactically in corneal lesions which are known to have a high probability
of inducing an
angiogenic response (such as chemical burns). In these instances the
treatment, likely in
combination with steroids, may be instituted immediately to help prevent
subsequent
complications.
Within other embodiments, the compounds described above may be injected
directly
into the corneal stroma by an ophthalmologist under microscopic guidance. The
preferred
site of injection may vary with the morphology of the individual lesion, but
the goal of the
administration would be to place the composition at the advancing front of the
vasculature
(i.e., interspersed between the blood vessels and the normal cornea). In most
cases this
would involve perilimbic corneal injection to "protect" the cornea from the
advancing blood
vessels. This method may also be utilized shortly after a corneal insult in
order to
prophylactically prevent corneal neovascularization. In this situation the
material could be
injected in the perilimbic cornea interspersed between the corneal lesion and
its undesired
potential limbic blood supply. Such methods may also be utilized in a similar
fashion to
prevent capillary invasion of transplanted corneas. In a sustained-release
form injections
might only be required 2-3 times per year. A steroid could also be added to
the injection
solution to reduce inflammation resulting from the injection itself.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
144
Within another aspect of the present invention, methods are provided for
treating
neovascular glaucoma, comprising the step of administering to a patient a
therapeutically
effective amount of a polynucleotide, polypeptide, antagonist and/or agonist
to the eye, such
that the formation of blood vessels is inhibited. In one embodiment, the
compound may be
administered topically to the eye in order to treat early forms of neovascular
glaucoma.
Within other embodiments, the compound may be implanted by injection into the
region of
the anterior chamber angle. Within other embodiments, the compound may also be
placed in
any location such that the compound is continuously released into the aqueous
humor.
Within another aspect of the present invention, methods are provided for
treating
proliferative diabetic retinopathy, comprising the step of administering to a
patient a
therapeutically effective amount of a polynucleotide, polypeptide, antagonist
and/or agonist
to the eyes, such that the formation of blood vessels is inhibited.
Within particularly preferred embodiments of the invention, proliferative
diabetic
retinopathy may be treated by injection into the aqueous humor or the
vitreous, in order to
1 S increase the local concentration of the polynucleotide, polypeptide,
antagonist and/or agonist
in the retina. Preferably, this treatment should be initiated prior to the
acquisition of severe
disease requiring photocoagulation.
Within another aspect of the present invention, methods are provided for
treating
retrolental fibroplasia, comprising the step of administering to a patient a
therapeutically
effective amount of a polynucleotide, polypeptide, antagonist and/or agonist
to the eye, such
that the formation of blood vessels is inhibited. The compound may be
administered
topically, via intravitreous injection andlor via intraocular implants.
Additionally, disorders which can be treated with the polynucleotides,
polypeptides,
agonists and/or agonists include, but are not limited to, hemangioma,
arthritis, psoriasis,
angiofibroma, atherosclerotic plaques, delayed wound healing, granulations,
hemophilic
joints, hypertrophic scars, nonunion fractures, Osler-Weber syndrome, pyogenic
granuloma,
scleroderma, trachoma, and vascular adhesions.
Moreover, disorders and/or states, which can be treated with be treated with
the the
polynucleotides, polypeptides, agonists and/or agonists include, but are not
limited to, solid
tumors, blood born tumors such as leukemias, tumor metastasis, Kaposi's
sarcoma, benign
tumors, for example hemangiomas, acoustic neuromas, neurofibromas, trachomas,
and
pyogenic granulomas, rheumatoid arthritis, psoriasis, ocular angiogenic
diseases, for


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
145
example, diabetic retinopathy, retinopathy of prematurity, macular
degeneration, corneal
graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis,
retinoblastoma, and
uvietis, delayed wound healing, endometriosis, vascluogenesis, granulations,
hypertrophic
scars (keloids), nonunion fractures, scleroderma, trachoma, vascular
adhesions, myocardial
angiogenesis, coronary collaterals, cerebral collaterals, arteriovenous
malformations,
ischemic limb angiogenesis, Osler-Webber Syndrome, plaque neovascularization,
telangiectasia, hemophiliac joints, angiofibroma fibromuscular dysplasia,
wound granulation,
Crohn's disease, atherosclerosis, birth control agent by preventing
vascularization required for
embryo implantation controlling menstruation, diseases that have angiogenesis
as a
pathologic consequence such as cat scratch disease (Rochele minalia quintosa),
ulcers
(Helicobacter pylori), Bartonellosis and bacillary angiomatosis.
In one aspect of the birth control method, an amount of the compound
sufficient to
block embryo implantation is administered before or after intercourse and
fertilization have
occurred, thus providing an effective method of birth control, possibly a
"morning a$er"
method. Polynucleotides, polypeptides, agonists and/or agonists may also be
used in
controlling menstruation or administered as either a peritoneal lavage fluid
or for peritoneal
implantation in the treatment of endometriosis.
Polynucleotides, polypeptides, agonists and/or agonists of the present
invention may
be incorporated into surgical sutures in order to prevent stitch granulomas.
Polynucleotides, polypeptides, agonists and/or agonists may be utilized in a
wide
variety of surgical procedures. For example, within one aspect of the present
invention a
compositions (in the form of, for example, a spray or film) may be utilized to
coat or spray an
area prior to removal of a tumor, in order to isolate normal surrounding
tissues from
malignant tissue, and/or to prevent the spread of disease to surrounding
tissues. Within other
aspects of the present invention, compositions (e.g., in the form of a spray)
may be delivered
via endoscopic procedures in order to coat tumors, or inhibit angiogenesis in
a desired locale.
Within yet other aspects of the present invention, surgical meshes which have
been coated
with anti- angiogenic compositions of the present invention may be utilized in
any procedure
wherein a surgical mesh might be utilized. For example, within one embodiment
of the
invention a surgical mesh laden with an anti-angiogenic composition may be
utilized during
abdominal cancer resection surgery (e.g., subsequent to colon resection) in
order to provide
support to the structure, and to release an amount of the anti-angiogenic
factor.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
146
Within further aspects of the present invention, methods are provided for
treating
tumor excision sites, comprising administering a polynucleotide, polypeptide,
agonist and/or
agonist to the resection margins of a tumor subsequent to excision, such that
the local
recurrence of cancer and the formation of new blood vessels at the site is
inhibited. Within
one embodiment of the invention, the anti-angiogenic compound is administered
directly to
the tumor excision site (e.g., applied by swabbing, brushing or otherwise
coating the
resection margins of the tumor with the anti-angiogenic compound).
Alternatively, the anti-
angiogenic compounds may be incorporated into known surgical pastes prior to
administration. Within particularly preferred embodiments of the invention,
the anti-
angiogenic compounds are applied after hepatic resections for malignancy, and
after
neurosurgical operations.
Within one aspect of the present invention, polynucleotides, polypeptides,
agonists
and/or agonists may be administered to the resection margin of a wide variety
of tumors,
including for example, breast, colon, brain and hepatic tumors. For example,
within one
embodiment of the invention, anti-angiogenic compounds may be administered to
the site of
a neurological tumor subsequent to excision, such that the formation of new
blood vessels at
the site are inhibited.
The polynucleotides, polypeptides, agonists and/or agonists of the present
invention
may also be administered along with other anti-angiogenic factors.
Representative examples
of other anti-angiogenic factors include: Anti-Invasive Factor, retinoic acid
and derivatives
thereof, paclitaxel, Suramin, Tissue Inhibitor of Metalloproteinase-1, Tissue
Inhibitor of
Metalloproteinase-2, Plasminogen Activator Inhibitor-1, Plasminogen Activator
Inhibitor-2,
and various forms of the lighter "d group" transition metals.
Lighter "d group" transition metals include, for example, vanadium,
molybdenum,
tungsten, titanium, niobium, and tantalum species. Such transition metal
species may form
transition metal complexes. Suitable complexes of the above-mentioned
transition metal
species include oxo transition metal complexes.
Representative examples of vanadium complexes include oxo vanadium complexes
such as vanadate and vanadyl complexes. Suitable vanadate complexes include
metavanadate and orthovanadate complexes such as, for example, ammonium
metavanadate,
sodium metavanadate, and sodium orthovanadate. Suitable vanadyl complexes
include, for


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
147
example, vanadyl acetylacetonate and vanadyl sulfate including vanadyl sulfate
hydrates such
as vanadyl sulfate mono- and trihydrates.
Representative examples of tungsten and molybdenum complexes also include oxo
complexes. Suitable oxo tungsten complexes include tungstate and tungsten
oxide
complexes. Suitable tungstate complexes include ammonium tungstate, calcium
tungstate,
sodium tungstate dihydrate, and tungstic acid. Suitable tungsten oxides
include tungsten (IV)
oxide and tungsten (VI) oxide. Suitable oxo molybdenum complexes include
molybdate,
molybdenum oxide, and molybdenyl complexes. Suitable molybdate complexes
include
ammonium molybdate and its hydrates, sodium molybdate and its hydrates, and
potassium
molybdate and its hydrates. Suitable molybdenum oxides include molybdenum (VI)
oxide,
molybdenum (VI) oxide, and molybdic acid. Suitable molybdenyl complexes
include, for
example, molybdenyl acetylacetonate. Other suitable tungsten and molybdenum
complexes
include hydroxo derivatives derived from, for example, glycerol, tartaric
acid, and sugars.
A wide variety of other anti-angiogenic factors may also be utilized within
the context
of the present invention. Representative examples include platelet factor 4;
protamine
sulphate; sulphated chitin derivatives (prepared from queen crab shells),
(Murata et al.,
Cancer Res. 51:22-26, 1991); Sulphated Polysaccharide Peptidoglycan Complex
(SP- PG)
(the function of this compound may be enhanced by the presence of steroids
such as estrogen,
and tamoxifen citrate); Staurosporine; modulators of matrix metabolism,
including for
example, proline analogs, cishydroxyproline, d,L-3,4-dehydroproline,
Thiaproline,
alpha,alpha-dipyridyl, aminopropionitrile fumarate; 4-propyl-5-(4-pyridinyl)-
2(3H)-
oxazolone; Methotrexate; Mitoxantrone; Heparin; Interferons; 2 Macroglobulin-
serum;
ChIMP-3 (Pavloff et al., J. Bio. Chem. 267:17321-17326, 1992); Chymostatin
(Tomkinson et
al., Biochem J. 286:475-480, 1992); Cyclodextrin Tetradecasulfate; Eponemycin;
Camptothecin; Fumagillin (Ingber et al., Nature 348:555-557, 1990); Gold
Sodium
Thiomalate ("GST"; Matsubara and Ziff, J. Clin. Invest. 79:1440-1446, 1987);
anticollagenase-serum; alpha2-antiplasmin (Holmes et al., J. Biol. Chem.
262(4):1659-1664,
1987); Bisantrene (National Cancer Institute); Lobenzarit disodium (N-(2)-
carboxyphenyl-4-
chloroanthronilic acid disodium or "CCA"; Takeuchi et al., Agents Actions
36:312-316,
1992); Thalidomide; Angostatic steroid; AGM-1470; carboxynaminolmidazole; and
metalloproteinase inhibitors such as BB94.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
148
Diseases at the Cellular Level
Diseases associated with increased cell survival or the inhibition of
apoptosis that
could be treated or detected by polynucleotides or polypeptides, as well as
antagonists or
agonists of the present invention, include cancers (such as follicular
lymphomas, carcinomas
with p53 mutations, and hormone-dependent tumors, including, but not limited
to colon
cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma,
glioblastoma, lung
cancer, intestinal cancer, testicular cancer, stomach cancer, neuroblastoma,
myxoma, myoma,
lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma,
chondrosarcoma,
adenoma, breast cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer);
autoimmune
disorders (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's
thyroiditis, biliary
cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus
erythematosus and
immune-related glomerulonephritis and rheumatoid arthritis) and viral
infections (such as
herpes viruses, pox viruses and adenoviruses), inflammation, graft v. host
disease, acute graft
rejection, and chronic graft rejection. In preferred embodiments,
polynucleotides,
polypeptides, and/or antagonists of the invention are used to inhibit growth,
progression,
and/or metasis of cancers, in particular those listed above.
Additional diseases or conditions associated with increased cell survival that
could be
treated or detected by polynucleotides or polypeptides, or agonists or
antagonists of the
present invention include, but are not limited to, progression, and/or
metastases of
malignancies and related disorders such as leukemia (including acute leukemias
(e.g., acute
lymphocytic leukemia, acute myelocytic leukemia (including myeloblastic,
promyelocytic,
myelomonocytic, monocytic, and erythroleukemia)) and chronic leukemias (e.g.,
chronic
myelocytic (granulocytic) leukemia and chronic lyrnphocytic leukemia)),
polycythemia vera,
lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple
myeloma,
Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors
including, but not
limited to, sarcomas and carcinomas such as fibrosarcoma, myxosarcoma,
liposarcoma,
chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma,
lymphangiosarcoma, lyrnphangioendotheliosarcoma, synovioma, mesothelioma,
Ewing's
tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer,
breast
cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell
carcinoma,
adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary
carcinoma,
papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma,
bronchogenic


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
149
carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma,
choriocarcinoma,
seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular
tumor, lung
carcinoma, small cell lung carcinoma, , bladder carcinoma, epithelial
carcinoma, glioma,
astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma,
hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma,
neuroblastoma, and retinoblastoma.
Diseases associated with increased apoptosis that could be treated or detected
by
polynucleotides or polypeptides, as well as agonists or antagonists of the
present invention,
include AIDS; neurodegenerative disorders (such as Alzheimer's disease,
Parkinson's disease,
Amyotrophic lateral sclerosis, Retinitis pigmentosa, Cerebellar degeneration
and brain tumor
or prior associated disease); autoimmune disorders (such as, multiple
sclerosis, Sjogren's
syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease,
Crohn's disease,
polymyositis, systemic lupus erythematosus and immune-related
glomerulonephritis and
rheumatoid arthritis) myelodysplastic syndromes (such as aplastic anemia),
graft v. host
disease, ischemic injury (such as that caused by myocardial infarction, stroke
and reperfusion
injury), liver injury (e.g., hepatitis related liver injury,
ischemia/reperfusion injury,
cholestosis (bile duct injury) and liver cancer); toxin-induced liver disease
(such as that
caused by alcohol), septic shock, cachexia and anorexia.
Wound Healing and Epithelial Cell Proliferation
. In accordance with yet a further aspect of the present invention, there is
provided a
process for utilizing polynucleotides or polypeptides, as well as agonists or
antagonists of the
present invention, for therapeutic purposes, for example, to stimulate
epithelial cell
proliferation and basal keratinocytes for the purpose of wound healing, and to
stimulate hair
follicle production and healing of dermal wounds. Polynucleotides or
polypeptides, as well as
agonists or antagonists of the present invention, may be clinically useful in
stimulating
wound healing including surgical wounds, excisional wounds, deep wounds
involving
damage of the dermis and epidermis, eye tissue wounds, dental tissue wounds,
oral cavity
wounds, diabetic ulcers, dermal ulcers, cubitus ulcers, arterial ulcers,
venous stasis ulcers,
burns resulting from heat exposure or chemicals, and other abnormal wound
healing
conditions such as uremia, malnutrition, vitamin deficiencies and
complications associted
with systemic treatment with steroids, radiation therapy and antineoplastic
drugs and


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
150
antimetabolites. Polynucleotides or polypeptides, as well as agonists or
antagonists of the
present invention, could be used to promote dermal reestablishment subsequent
to dermal
loss
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention, could be used to increase the adherence of skin grafts to a wound
bed and to
stimulate re-epithelialization from the wound bed. The following are types of
grafts that
polynucleotides or polypeptides, agonists or antagonists of the present
invention, could be
used to increase adherence to a wound bed: autografts, artificial skin,
allografts, autodermic
graft, autoepdermic grafts, avacular grafts, Blair-Brown grafts, bone graft,
brephoplastic
grafts, cutis graft, delayed graft, dermic graft, epidermic graft, fascia
graft, full thickness
graft, heterologous graft, xenograft, homologous graft, hyperplastic graft,
lamellar graft,
mesh graft, mucosal graft, Ollier-Thiersch graft, omenpal graft, patch graft,
pedicle graft,
penetrating graft, split skin graft, thick split graft. Polynucleotides or
polypeptides, as well as
agonists or antagonists of the present invention, can be used to promote skin
strength and to
improve the appearance of aged skin.
It is believed that polynucleotides or polypeptides, as well as agonists or
antagonists of
the present invention, will also produce changes in hepatocyte proliferation,
and epithelial
cell proliferation in the lung, breast, pancreas, stomach, small intesting,
and large intestine.
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present invention,
could promote proliferation of epithelial cells such as sebocytes, hair
follicles, hepatocytes,
type II pneumocytes, mucin-producing goblet cells, and other epithelial cells
and their
progenitors contained within the skin, lung, liver, and gastrointestinal
tract. Polynucleotides
or polypeptides, agonists or antagonists of the present invention, may promote
proliferation
of endothelial cells, keratinocytes, and basal keratinocytes.
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention, could also be used to reduce the side effects of gut toxicity that
result from
radiation, chemotherapy treatments or viral infections. Polynucleotides or
polypeptides, as
well as agonists or antagonists of the present invention, may have a
cytoprotective effect on
the small intestine mucosa. Polynucleotides or polypeptides, as well as
agonists or
antagonists of the present invention, may also stimulate healing of mucositis
(mouth ulcers)
that result from chemotherapy and viral infections.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
151
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention, could further be used in full regeneration of skin in full and
partial thickness skin
defects, including burns, (i.e., repopulation of hair follicles, sweat glands,
and sebaceous
glands), treatment of other skin defects such as psoriasis. Polynucleotides or
polypeptides, as
S well as agonists or antagonists of the present invention, could be used to
treat epidermolysis
bullosa, a defect in adherence of the epidermis to the underlying dermis which
results in
frequent, open and painful blisters by accelerating reepithelialization of
these lesions.
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present invention,
could also be used to treat gastric and doudenal ulcers and help heal by scar
formation of the
mucosal lining and regeneration of glandular mucosa and duodenal mucosal
lining more
rapidly. Inflamamatory bowel diseases, such as Crohn's disease and ulcerative
colitis, are
diseases which result in destruction of the mucosal surface of the small or
large intestine,
respectively. Thus, polynucleotides or polypeptides, as well as agonists or
antagonists of the
present invention, could be used to promote the resurfacing of the mucosal
surface to aid
more rapid healing and to prevent progression of inflammatory bowel disease.
Treatment
with polynucleotides or polypeptides, agonists or antagonists of the present
invention, is
expected to have a significant effect on the production of mucus throughout
the
gastrointestinal tract and could be used to protect the intestinal mucosa from
injurious
substances that are ingested or following surgery. Polynucleotides or
polypeptides, as well as
agonists or antagonists of the present invention, could be used to treat
diseases associate with
the under expression.
Moreover, polynucleotides or polypeptides, as well as agonists or antagonists
of the
present invention, could be used to prevent and heal damage to the lungs due
to various
pathological states. Polynucleotides or polypeptides, as well as agonists or
antagonists of the
present invention, which could stimulate proliferation and differentiation and
promote the
repair of alveoli and brochiolar epithelium to prevent or treat acute or
chronic lung damage.
For example, emphysema, which results in the progressive loss of aveoli, and
inhalation
injuries, i.e., resulting from smoke inhalation and burns, that cause necrosis
of the bronchiolar
' epithelium and alveoli could be effectively treated using polynucleotides or
polypeptides,
agonists or antagonists of the present invention. Also, polynucleotides or
polypeptides, as
well as agonists or antagonists of the present invention, could be used to
stimulate the
proliferation of and differentiation of type II pneumocytes, which may help
treat or prevent


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
152
disease such as hyaline membrane diseases, such as infant respiratory distress
syndrome and
bronchopulmonary displasia, in premature infants.
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention, could stimulate the proliferation and differentiation of
hepatocytes and, thus, could
be used to alleviate or treat liver diseases and pathologies such as fulminant
liver failure
caused by cirrhosis, liver damage caused by viral hepatitis and toxic
substances (i.e.,
acetaminophen, carbon tetraholoride and other hepatotoxins known in the art).
In addition, polynucleotides or polypeptides,.as well as agonists or
antagonists of the
present invention, could be used treat or prevent the onset of diabetes
mellitus. In patients
with newly diagnosed Types I and II diabetes, where some islet cell function
remains,
polynucleotides or polypeptides, as well as agonists or antagonists of the
present invention,
could be used to maintain the islet function so as to alleviate, delay or
prevent permanent
manifestation of the disease. Also, polynucleotides or polypeptides, as well
as agonists or
antagonists of the present invention, could be used as an auxiliary in islet
cell transplantation
to improve ~or promote islet cell function.
Neurological Diseases
In accordance with yet a further aspect of the present invention, there is
provided a
process for utilizing polynucleotides or polypeptides, as well as agonists or
antagonists of the
present invention, for therapeutic purposes, for example, to stimulate
neurological cell
proliferation and/or differentiation. Therefore, polynucleotides,
polypeptides, agonists and/or
antagonists of the invention may be used to treat and/or detect neurologic
diseases.
Moreover, polynucleotides or polypeptides, or agonists or antagonists of the
invention, can be
used as a marker or detector of a particular nervous system disease or
disorder.
Examples of neurologic diseases which can be treated or detected with
polynucleotides, polypeptides, agonists, and/or antagonists of the present
invention include
brain diseases, such as metabolic brain diseases which includes
phenylketonuria such as
maternal phenylketonuria, pyruvate carboxylase deficiency, pyruvate
dehydrogenase
complex deficiency, Wernicke's Encephalopathy, brain edema, brain neoplasms
such as
cerebellar neoplasms which include infratentorial neoplasms, cerebral
ventricle neoplasms
such as choroid plexus neoplasms, hypothalamic neoplasms, supratentorial
neoplasms,
canavan disease, cerebellar diseases such as cerebellar ataxia which include
spinocerebellar


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
153
degeneration such as ataxia telangiectasia, cerebellar dyssynergia,
Friederich's Ataxia,
Machado-Joseph Disease, olivopontocerebellar atrophy, cerebellar neoplasms
such as
infratentorial neoplasms, diffuse cerebral sclerosis such as encephalitis
periaxialis, globoid
cell leukodystrophy, metachromatic leukodystrophy and subacute sclerosing
panencephalitis,
cerebrovascular disorders (such as carotid artery diseases which include
carotid artery
thrombosis, carotid stenosis and Moyamoya Disease, cerebral amyloid
angiopathy, cerebral
aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arteriovenous
malformations,
cerebral artery diseases, cerebral embolism and thrombosis such as carotid
artery thrombosis,
sinus thrombosis and Wallenberg's Syndrome, cerebral hemorrhage such as
epidural
hematoma, subdural hematoma and subarachnoid hemorrhage, cerebral infarction,
cerebral
ischemia such as transient cerebral ischemia, Subclavian Steal Syndrome and
vertebrobasilar
insufficiency, vascular dementia such as mufti-infarct dementia,
periventricular leukomalacia,
vascular headache such as cluster headache, migraine, dementia such as AIDS
Dementia
Complex, presenile dementia such as Alzheimer's Disease and Creutzfeldt-Jakob
Syndrome,
senile dementia such as Alzheimer's Disease and progressive supranuclear
palsy, vascular
dementia such as mufti-infarct dementia, encephalitis which include
encephalitis periaxialis,
viral encephalitis such as epidemic encephalitis, Japanese Encephalitis, St.
Louis
Encephalitis, tick-borne encephalitis and West Nile Fever, acute disseminated
encephalomyelitis, meningoencephalitis such as uveomeningoencephalitic
syndrome,
Postencephalitic Parkinson Disease and subacute sclerosing panencephalitis,
encephalomalacia such as periventricular leukomalacia, epilepsy such as
generalized epilepsy
which includes infantile spasms, absence epilepsy, myoclonic epilepsy which
includes
MERRF Syndrome, tonic-clonic epilepsy, partial epilepsy such as complex
partial epilepsy,
frontal lobe epilepsy and temporal lobe epilepsy, post-traumatic epilepsy,
status epilepticus
such as Epilepsia Partialis Continua, Hallervorden-Spatz Syndrome,
hydrocephalus such as
Dandy-Walker Syndrome and normal pressure hydrocephalus, hypothalamic diseases
such as
hypothalamic neoplasms, cerebral malaria, narcolepsy which includes cataplexy,
bulbar
poliomyelitis, cerebri pseudotumor, Rett Syndrome, Reye's Syndrome, thalamic
diseases,
cerebral toxoplasmosis, intracranial tuberculoma and Zellweger Syndrome,
central nervous
system infections such as AIDS Dementia Complex, Brain Abscess, subdural
empyema,
encephalomyelitis such as Equine Encephalomyelitis, Venezuelan Equine
Encephalomyelitis,
Necrotizing Hemorrhagic Encephalomyelitis, Visna, cerebral malaria, meningitis
such as


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
154
arachnoiditis, aseptic meningtitis such as viral meningtitis which includes
lymphocytic
choriomeningitis. Bacterial meningtitis which includes Haemophilus
Meningtitis, Listeria
Meningtitis, Meningococcal Meningtitis such as Waterhouse-Friderichsen
Syndrome,
Pneumococcal Meningtitis and meningeal tuberculosis, fungal meningitis such as
Cryptococcal Meningtitis, subdural effusion, meningoencephalitis such as
uvemeningoencephalitic syndrome, myelitis such as transverse myelitis,
neurosyphilis such
as tabes dorsalis, poliomyelitis which includes bulbar poliomyelitis and
postpoliomyelitis
syndrome, prion diseases (such as Creutzfeldt-Jakob Syndrome, Bovine
Spongiform
Encephalopathy, Gerstmann-Straussler Syndrome, Kuru, Scrapie) cerebral
toxoplasmosis,
central nervous system neoplasms such as brain neoplasms that include
cerebellear
neoplasms such as infratentorial neoplasms, cerebral ventricle neoplasms such
as choroid
plexus neoplasms, hypothalamic neoplasms and supratentorial neoplasms,
meningeal
neoplasms, spinal cord neoplasms which include epidural neoplasms,
demyelinating diseases
such as Canavan Diseases, diffuse cerebral sceloris which includes
adrenoleukodystrophy,
encephalitis periaxialis, globoid cell leukodystrophy, diffuse cerebral
sclerosis such as
metachromatic leukodystrophy, allergic encephalomyelitis, necrotizing
hemorrhagic
encephalomyelitis, progressive multifocal leukoencephalopathy, multiple
sclerosis, central
pontine myelinolysis, transverse myelitis, neuromyelitis optics, Scrapie,
Swayback, Chronic
Fatigue Syndrome, Visna, High Pressure Nervous Syndrome, Meningism, spinal
cord
diseases such as amyotonia congenita, amyotrophic lateral sclerosis, spinal
muscular atrophy
such as Werdnig-Hoffinann Disease, spinal cord compression, spinal cord
neoplasms such as
epidural neoplasms, syringomyelia, Tabes Dorsalis, Stiff Man Syndrome, mental
retardation
such as Angelman Syndrome, Cri-du-Chat Syndrome, De Lange's Syndrome, Down
Syndrome, Gangliosidoses such as gangliosidoses G(M1), Sandhoff Disease, Tay-
Sachs
Disease, Hartnup Disease, homocystinuria, Laurence-Moon- Biedl Syndrome, Lesch-
Nyhan
Syndrome, Maple Syrup Urine Disease, mucolipidosis such as fucosidosis,
neuronal ceroid-
lipofuscinosis, oculocerebrorenal syndrome, phenylketonuria such as maternal
phenylketonuria, Prader-Willi Syndrome, Rett Syndrome, Rubinstein-Taybi
Syndrome,
Tuberous Sclerosis, WAGR Syndrome, nervous system abnormalities such as
holoprosencephaly, neural tube defects such as anencephaly which includes
hydrangencephaly, Arnold-Chairi Deformity, encephalocele, meningocele,
meningomyelocele, spinal dysraphism such as spins bifida cystica and spins
bifida occults,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
155
hereditary motor and sensory neuropathies which include Charcot-Marie Disease,
Hereditary
optic atrophy, Refsum's Disease, hereditary spastic paraplegia, Werdnig-
Hoffmann Disease,
Hereditary Sensory and Autonomic Neuropathies such as Congenital Analgesia and
Familial
Dysautonomia, Neurologic manifestations (such as agnosia that include
Gerstmann's
Syndrome, Amnesia such as retrograde amnesia, apraxia, neurogenic bladder,
cataplexy,
communicative disorders such as hearing disorders that includes deafness,
partial hearing
loss, loudness recruitment and tinnitus, language disorders such as aphasia
which include
agraphia, anomia, broca aphasia, and Wernicke Aphasia, Dyslexia such as
Acquired
Dyslexia, language development disorders, speech disorders such as aphasia
which includes
anomia, broca aphasia and Wernicke Aphasia, articulation disorders,
communicative
disorders such as speech disorders which include dysarthria, echolalia, mutism
and stuttering,
voice disorders such as aphonia and hoarseness, decerebrate state, delirium,
fasciculation,
hallucinations, meningism, movement disorders such as angelman syndrome,
ataxia,
athetosis, chorea, dystonia, hypokinesia, muscle hypotonia, myoclonus, tic,
torticollis and
1 S tremor, muscle hypertonia such as muscle rigidity such as stiff man
syndrome, muscle
spasticity, paralysis such as facial paralysis which includes Herpes Zoster
Oticus,
Gastroparesis, Hemiplegia, ophthalmoplegia such as diplopia, Duane's Syndrome,
Homer's
Syndrome, Chronic progressive external ophthalmoplegia such as Kearns
Syndrome, Bulbar
Paralysis, Tropical Spastic Paraparesis, Paraplegia such as Brown-Sequard
Syndrome,
quadriplegia, respiratory paralysis and vocal cord paralysis, paresis, phantom
limb, taste
disorders such as ageusia and dysgeusia, vision disorders such as amblyopia,
blindness, color
vision defects, diplopia, hemianopsia, scotoma and subnormal vision, sleep
disorders such as
hypersomnia which includes Kleine-Levin Syndrome, insomnia, and somnambulism,
spasm
such as trismus, unconsciousness such as coma, persistent vegetative state and
syncope and
vertigo, neuromuscular diseases such as amyotonia congenita, amyotrophic
lateral sclerosis,
Lambert-Eaton Myasthenic Syndrome, motor neuron disease, muscular atrophy such
as
spinal muscular atrophy, Charcot-Marie Disease and Werdnig-Hoffmann Disease,
Postpoliomyelitis Syndrome, Muscular Dystrophy, Myasthenia Gravis, Myotonia
Atrophica,
Myotonia Confenita, Nemaline Myopathy, Familial Periodic Paralysis, Multiplex
Paramyloclonus, Tropical Spastic Paraparesis and Stiff Man Syndrome,
peripheral nervous
system diseases such as acrodynia, amyloid neuropathies, autonomic nervous
system diseases
such as Adie's Syndrome, Barre-Lieou Syndrome, Familial Dysautonomia, Homer's


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
156
Syndrome, Reflex Sympathetic Dystrophy and Shy-Drager Syndrome, Cranial Nerve
Diseases such as Acoustic Nerve Diseases such as Acoustic Neuroma which
includes
Neurofibromatosis 2, Facial Nerve Diseases such as Facial Neuralgia,Melkersson-
Rosenthal
Syndrome, ocular motility disorders which includes amblyopia, nystagmus,
oculomotor nerve
paralysis, ophthalmoplegia such as Duane's Syndrome, Homer's Syndrome, Chronic
Progressive External Ophthalmoplegia which includes Kearns Syndrome,
Strabismus such as
Esotropia and Exotropia, Oculomotor Nerve Paralysis, Optic Nerve Diseases such
as Optic
Atrophy which includes Hereditary Optic Atrophy, Optic Disk Drusen, Optic
Neuritis such as
Neuromyelitis Optica, Papilledema, Trigeminal Neuralgia, Vocal Cord Paralysis,
Demyelinating Diseases such as Neuromyelitis Optica and Swayback, Diabetic
neuropathies
such as diabetic foot, nerve compression syndromes such as carpal tunnel
syndrome, tarsal
tunnel syndrome, thoracic outlet syndrome such as cervical rib syndrome, ~
ulnar nerve
compression syndrome, neuralgia such as causalgia, cervico-brachial neuralgia,
facial
neuralgia and trigeminal neuralgia, neuritis such as experimental allergic
neuritis, optic
neuritis, polyneuritis, polyradiculoneuritis and radiculities such as
polyradiculitis, hereditary
motor and sensory neuropathies such as Charcot-Marie Disease, Hereditary Optic
Atrophy,
Refsum's Disease, Hereditary Spastic Paraplegia and Werdnig-Hoffmann Disease,
Hereditary
Sensory and Autonomic Neuropathies which include Congenital Analgesia and
Familial
Dysautonomia, POEMS Syndrome, Sciatica, Gustatory Sweating and Tetany).
Infectious Disease
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention can be used to treat or detect infectious agents. For example, by
increasing the
immune response, particularly increasing the proliferation and differentiation
of B and/or T
cells, infectious diseases may be treated. The immune response may be
increased by either
enhancing an existing immune response, or by initiating a new immune response.
Alternatively, polynucleotides or polypeptides, as well as agonists or
antagonists of the
present invention may also directly inhibit the infectious agent, without
necessarily eliciting
an immune response.
Viruses are one example of an infectious agent that can cause disease or
symptoms
that can be treated or detected by a polynucleotide or polypeptide and/or
agonist or antagonist
of the present invention. Examples of viruses, include, but are not limited to
Examples of


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
157
viruses, include, but are not limited to the following DNA and RNA viruses and
viral
families: Arbovirus, Adenoviridae, Arenaviridae, Arterivirus, Birnaviridae,
Bunyaviridae,
Caliciviridae, Circoviridae, Coronaviridae, Dengue, EBV, HIV, Flaviviridae,
Hepadnaviridae
(Hepatitis), Herpesviridae (such as, Cytomegalovirus, Herpes Simplex, Herpes
Zoster),
Mononegavirus (e.g., Paramyxoviridae, Morbillivirus, Rhabdoviridae),
Orthomyxoviridae
(e.g., Influenza A, Influenza B, and parainfluenza), Papiloma virus,
Papovaviridae,
Parvoviridae, Picornaviridae, Poxviridae (such as Smallpox or Vaccinia),
Reoviridae (e.g.,
Rotavirus), Retroviridae (HTLV-I, HTLV-II, Lentivirus), and Togaviridae (e.g.,
Rubivirus).
Viruses falling within these families can cause a variety of diseases or
symptoms, including,
but not limited to: arthritis, bronchiollitis, respiratory syncytial virus,
encephalitis, eye
infections (e.g., conjunctivitis, keratitis), chronic fatigue syndrome,
hepatitis (A, B, C, E,
Chronic Active, Delta), Japanese B encephalitis, Junin, Chikungunya, Rift
Valley fever,
yellow fever, meningitis, opportunistic infections (e.g., AIDS), pneumonia,
Burkitt's
Lymphoma, chickenpox, hemorrhagic fever, Measles, Mumps, Parainfluenza,
Rabies, the
~ common cold, Polio, leukemia, Rubella, sexually transmitted diseases, skin
diseases (e.g.,
Kaposi's, warts), and viremia. polynucleotides or polypeptides, or agonists or
antagonists of
the invention, can be used to treat or detect any of these symptoms or
diseases. In specific
embodiments, polynucleotides, polypeptides, or agonists or antagonists of the
invention are
used to treat: meningitis, Dengue, EBV, and/or hepatitis (e.g., hepatitis B).
In an additional
specific embodiment polynucleotides, polypeptides, or agonists or antagonists
of the
invention are used to treat patients nonresponsive to one or more other
commercially
available hepatitis vaccines. In a further specific embodiment
polynucleotides, polypeptides,
or agonists or antagonists of the invention are used to treat AIDS.
Similarly, bacterial or fungal agents that can cause disease or symptoms and
that can
be treated or detected by a polynucleotide or polypeptide and/or agonist or
antagonist of the
present invention include, but not limited to, include, but not limited to,
the following Gram-
Negative and Gram-positive bacteria and bacterial families and fungi:
Actinomycetales (e.g.,
Corynebacterium, Mycobacterium, Norcardia), Cryptococcus neoformans,
Aspergillosis,
Bacillaceae (e.g., Anthrax, Clostridium), Bacteroidaceae, Blastomycosis,
Bordetella, Borrelia
(e.g., Borrelia burgdorferi, Brucellosis, Candidiasis, Campylobacter,
Coccidioidomycosis,
Cryptococcosis, Dermatocycoses, E. coli (e.g., Enterotoxigenic E. coli and
Enterohemorrhagic E. coli), Enterobacteriaceae (Klebsiella, Salmonella (e.g.,
Salmonella


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
158
typhi, and Salmonella paratyphi), Serratia, Yersinia), Erysipelothrix,
Helicobacter,
Legionellosis, Leptospirosis, Listeria, Mycoplasmatales, Mycobacterium leprae,
Vibrio
cholerae, Neisseriaceae (e.g., Acinetobacter, Gonorrhea, Menigococcal),
Meisseria
meningitidis, Pasteurellacea Infections (e.g., Actinobacillus, Heamophilus
(e.g., Heamophilus
influenza type B), Pasteurella), Pseudomonas, Rickettsiaceae, Chlamydiaceae,
Syphilis,
Shigella spp., Staphylococcal, Meningiococcal, Pneumococcal and Streptococcal
(e.g.,
Streptococcus pneumoniae and Group B Streptococcus). These bacterial or fungal
families
can cause the following diseases or symptoms, including, but not limited to:
bacteremia,
endocarditis, eye infections (conjunctivitis, tuberculosis, uveitis),
gingivitis, opportunistic
infections (e.g., A117S related infections), paronychia, prosthesis-related
infections, Reiter's
Disease, respiratory tract infections, such as Whooping Cough or Empyema,
sepsis, Lyrne
Disease, Cat-Scratch Disease, Dysentery, Paratyphoid Fever, food poisoning,
Typhoid,
pneumonia, Gonorrhea, meningitis (e.g., mengitis types A and B), Chlamydia,
Syphilis,
Diphtheria, Leprosy, Paratuberculosis, Tuberculosis, Lupus, Botulism,
gangrene, tetanus,
1 S impetigo, Rheumatic Fever, Scarlet Fever, sexually transmitted diseases,
skin diseases (e.g.,
cellulitis, dermatocycoses), toxemia, urinary tract infections, wound
infections.
Polynucleotides or polypeptides, agonists or antagonists of the invention, can
be used to treat
or detect any of these symptoms or diseases. In specific embodiments,
Ppolynucleotides,
polypeptides, agonists or antagonists of the invention are used to treat:
tetanus, Diptheria,
botulism, and/or meningitis type B.
Moreover, parasitic agents causing disease or symptoms that can be treated or
detected by a polynucleotide or polypeptide and/or agonist or antagonist of
the present
invention include, but not limited to, the following families or class:
Amebiasis, Babesiosis,
Coccidiosis, Cryptosporidiosis, Dientamoebiasis, Dourine, Ectoparasitic,
Giardiasis,
Helminthiasis, Leishmaniasis, Theileriasis, Toxoplasmosis, Trypanosomiasis,
and
Trichomonas and Sporozoans (e.g., Plasmodium virax, Plasmodium falciparium,
Plasmodium
malariae and Plasmodium ovate). These parasites can cause a variety of
diseases or
symptoms, including, but not limited to: Scabies, Trombiculiasis, eye
infections, intestinal
disease (e.g., dysentery, giardiasis), liver disease, lung disease,
opportunistic infections (e.g.,
AIDS related), malaria, pregnancy complications, and toxoplasmosis.
polynucleotides or
polypeptides, or agonists or antagonists of the invention, can be used to
treat or detect any of
these symptoms or diseases.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
159
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention of the present invention could either be by administering an
effective amount of a
polypeptide to the patient, or by removing cells from the patient, supplying
the cells with a
polynucleotide of the present invention, and returning the engineered cells to
the patient (ex
vivo therapy). Moreover, the polypeptide or polynucleotide of the present
invention can be
used as an antigen in a vaccine to raise an immune response against infectious
disease.
l~eneration
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention can be used to differentiate, proliferate, and attract cells,
leading to the regeneration
of tissues. (See, Science 276:59-87 (1997).) The regeneration of tissues could
be used to
repair, replace, or protect tissue damaged by congenital defects, trauma
(wounds, burns,
incisions, or ulcers), age, disease (e.g. osteoporosis, osteocarthritis,
periodontal disease, liver
failure), surgery, including cosmetic plastic surgery, fibrosis, reperfusion
injury, or systemic
cytokine damage.
Tissues that could be regenerated using the present invention include organs
(e.g.,
pancreas, liver, intestine, kidney, skin, endothelium), muscle (smooth,
skeletal or cardiac),
vasculature (including vascular and lymphatics), nervous, hematopoietic, and
skeletal (bone,
cartilage, tendon, and ligament) tissue. Preferably, regeneration occurs
without or decreased
scarring. Regeneration also may include angiogenesis.
Moreover, polynucleotides or polypeptides, as well as agonists or antagonists
of the
present invention, may increase regeneration of tissues difficult to heal. For
example,
increased tendon/ligament regeneration would quicken recovery time after
damage.
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present invention
could also be used prophylactically in an effort to avoid damage. Specific
diseases that could
be treated include of tendinitis, carpal tunnel syndrome, and other tendon or
ligament defects.
A further example of tissue regeneration of non-healing wounds includes
pressure ulcers,
ulcers associatedwith vascular insufficiency, surgical, and traumatic wounds.
Similarly, nerve and brain tissue could also be regenerated by using
polynucleotides
or polypeptides, as well as agonists or antagonists of the present invention,
to proliferate and
differentiate nerve cells. Diseases that could be treated using this method
include central and
peripheral nervous system diseases, neuropathies, or mechanical and traumatic
disorders


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
160
(e.g., spinal cord disorders, head trauma, cerebrovascular disease, and
stoke). Specifically,
diseases associated with peripheral nerve injuries, peripheral neuropathy
(e.g., resulting from
chemotherapy or other medical therapies), localized neuropathies, and central
nervous system
diseases (e.g., Alzheimer's disease, Parkinson's disease, Huntington's
disease, amyotrophic
S lateral sclerosis, and Shy-Drager syndrome), could all be treated using the
polynucleotides or
polypeptides, as well as agonists or antagonists of the present invention.
Chemotaxis
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention may have chemotaxis activity. A chemotaxic molecule attracts or
mobilizes cells
(e.g., monocytes, fibroblasts, neutrophils, T-cells, mast cells, eosinophils,
epithelial and/or
endothelial cells) to a particular site in the body, such as inflammation,
infection, or site of
hyperproliferation. The mobilized cells can then fight off and/or heal the
particular trauma or
abnormality.
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention may increase chemotaxic activity of particular cells. These
chemotactic molecules
can then be used to treat inflammation, infection, hyperproliferative
disorders, or any immune
system disorder by increasing the number of cells targeted to a particular
location in the body.
For example, chemotaxic molecules can be used to treat wounds and other trauma
to tissues
by attracting immune cells to the injured location. Chemotactic molecules of
the present
invention can also attract fibroblasts, which can be used to treat wounds.
It is also contemplated that polynucleotides or polypeptides, as well as
agonists or
antagonists of the present invention may inhibit chemotactic activity. These
molecules could
also be used to treat disorders. Thus, polynucleotides or polypeptides, as
well as agonists or
antagonists of the present invention could be used as an inhibitor of
chemotaxis.
Binding Activity
A polypeptide of the present invention may be used to screen for molecules
that bind
to the polypeptide or for molecules to which the polypeptide binds. The
binding of the
polypeptide and the molecule may activate (agonist), increase, inhibit
(antagonist), or
decrease activity of the polypeptide or the molecule bound. Examples of such
molecules
include antibodies, oligonucleotides, proteins (e.g., receptors),or small
molecules.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
161
Preferably, the molecule is closely related to the natural ligand of the
polypeptide,
e.g., a fragment of the ligand, or a natural substrate, a ligand, a structural
or functional
mimetic. (See, Coligan et al., Current Protocols in Immunology 1(2):Chapter 5
(1991).)
Similarly, the molecule can be closely related to the natural receptor to
which the polypeptide
binds, or at least, a fragment of the receptor capable of being bound by the
polypeptide (e.g.,
active site). In either case, the molecule can be rationally designed using
known techniques.
Preferably, the screening for these molecules involves producing appropriate
cells
which express the polypeptide. Preferred cells include cells from mammals,
yeast,
Drosophila, or E. coli. Cells expressing the polypeptide (or cell membrane
containing the
expressed polypeptide) are then preferably contacted with a test compound
potentially
containing the molecule to observe binding, stimulation, or inhibition of
activity of either the
polypeptide or the molecule.
The assay may simply test binding of a candidate compound to the polypeptide,
wherein binding is detected by a label, or in an assay involving competition
with a labeled
competitor. Further, the assay may test whether the candidate compound results
in a signal
generated by binding to the polypeptide.
Alternatively, the assay can be carned out using cell-free preparations,
polypeptide/molecule affixed to a solid support, chemical libraries, or
natural product
mixtures. The assay may also simply comprise the steps of mixing a candidate
compound
with a solution containing a polypeptide, measuring polypeptide/molecule
activity or
binding, and comparing the polypeptide/molecule activity or binding to a
standard.
Preferably, an ELISA assay can measure polypeptide level or activity in a
sample
(e.g., biological sample) using a monoclonal or polyclonal antibody. The
antibody can
measure polypeptide level or activity by either binding, directly or
indirectly, to the
polypeptide or by competing with the polypeptide for a substrate.
Additionally, the receptor to which the polypeptide of the present invention
binds can
be identified by numerous methods known to those of skill in the art, for
example, ligand
panning and FACS sorting (Coligan, et al., Current Protocols in Immun., 1 (2),
Chapter 5,
(1991)). For example, expression cloning is employed wherein polyadenylated
RNA is
prepared from a cell responsive to the polypeptides, for example, NIH3T3 cells
which are
known to contain multiple receptors for the FGF family proteins, and SC-3
cells, and a cDNA
library created from this RNA is divided into pools and used to transfect COS
cells or other


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
162
cells that are not responsive to the polypeptides. Transfected cells which are
grown on glass
slides are exposed to the polypeptide of the present invention, after they
have been labelled.
The polypeptides can be labeled by a variety of means including iodination or
inclusion of a
recognition site for a site-specific protein kinase.
Following fixation and incubation, the slides are subjected to auto-
radiographic
analysis. Positive pools are identified and sub-pools are prepared and re-
transfected using an
iterative sub-pooling and re-screening process, eventually yielding a single
clones that
encodes the putative receptor.
As an alternative approach for receptor identification, the labeled
polypeptides can be
photoaffinity linked with cell membrane or extract preparations that express
the receptor
molecule. Cross-linked material is resolved by PAGE analysis and exposed to X-
ray film.
The labeled complex containing the receptors of the polypeptides can be
excised, resolved
into peptide fragments, and subjected to protein microsequencing. The amino
acid sequence
obtained from microsequencing would be used to design a set of degenerate
oligonucleotide
probes to screen a cDNA library to identify the genes encoding the putative
receptors.
Moreover, the techniques of gene-shuffling, motif shuffling, exon-shuffling,
and/or
codon-shuffling (collectively referred to as "DNA shuffling") may be employed
to modulate
the activities of the polypeptide of the present invention thereby effectively
generating
agonists and antagonists of the polypeptide of the present invention. See
generally, U.S.
Patent Nos. 5,605,793, 5,811,238, 5,830,721, 5,834,252, and 5,837,458, and
Patten, P. A., et
al., Curr. Opinion Biotechnol. 8:724-33 (1997); Harayama, S. Trends
Biotechnol. 16(2):76-
82 (1998); Hansson, L. O., et al., J. Mol. Biol. 287:265-76 (1999); and
Lorenzo, M. M. and
Blasco, R. Biotechniques 24(2):308-13 (1998) (each of these patents and
publications are
hereby incorporated by reference). In one embodiment, alteration of
polynucleotides and
corresponding polypeptides may be achieved by DNA shuffling. DNA shuffling
involves the
assembly of two or more DNA segments into a desired molecule by homologous, or
site-
specific, recombination. In another embodiment, polynucleotides and
corresponding
polypeptides may be alterred by being subjected to random mutagenesis by error-
prone PCR,
random nucleotide insertion or other methods prior to recombination. In
another
embodiment, one or more components, motifs, sections, parts, domains,
fragments, etc., of
the polypeptide of the present invention may be recombined with one or more
components,
motifs, sections, parts, domains, fragments, etc. of one or more heterologous
molecules. In


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
163
preferred embodiments, the heterologous molecules are family members. In
further preferred
embodiments, the heterologous molecule is a growth factor such as, for
example,
platelet-derived growth factor (PDGF), insulin-like growth factor (IGF-I),
transforming
growth factor (TGF)-alpha, epidermal growth factor (EGF), fibroblast growth
factor (FGF),
TGF-beta, bone morphogenetic protein (BMP)-2, BMP-4, BMP-5, BMP-6, BMP-7,
activins
A and B, decapentaplegic(dpp), 60A, OP-2, dorsalin, growth differentiation
factors (GDFs),
nodal, MIS, inhibin-alpha, TGF-betal, TGF-beta2, TGF-beta3, TGF-betas, and
glial-derived
neurotrophic factor (GDNF).
Other preferred fragments are biologically active fragments of the polypeptide
of the
present invention. Biologically active fragments are those exhibiting activity
similar, but not
necessarily identical, to an activity of the polypeptide of the present
invention. The
biological activity of the fragments may include an improved desired activity,
or a decreased
undesirable activity.
Additionally, this invention provides a method of screening compounds to
identify
those which modulate the action of the polypeptide of the present invention.
An example of
such an assay comprises combining a mammalian fibroblast cell, a the
polypeptide of the
present invention, the compound to be screened and 3 [H] thymidine under cell
culture
conditions where the fibroblast cell would normally proliferate. A control
assay may be
performed in the absence of the compound to be screened and compared to the
amount of
fibroblast proliferation in the presence of the compound to determine if the
compound
stimulates proliferation by determining the uptake of 3[H] thymidine in each
case. The
amount of fibroblast cell proliferation is measured by liquid scintillation
chromatography
which measures the incorporation of 3[H] thymidine. Both agonist and
antagonist
compounds may be identified by this procedure.
In another method, a mammalian cell or membrane preparation expressing a
receptor
for a polypeptide of the present invention is incubated with a labeled
polypeptide of the
present invention in the presence of the compound. The ability of the compound
to enhance
or block this interaction could then be measured. Alternatively, the response
of a known
second messenger system following interaction of a compound to be screened and
the
receptor is measured and the ability of the compound to bind to the receptor
and elicit a
second messenger response is measured to determine if the compound is a
potential agonist


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
164
or antagonist. Such second messenger systems include but are not limited to,
cAMP
guanylate cyclase, ion channels or phosphoinositide hydrolysis.
All of these above assays can be used as diagnostic or prognostic markers. The
molecules discovered using these assays can be used to treat disease or to
bring about a
particular result in a patient (e.g., blood vessel growth) by activating or
inhibiting the
polypeptide/molecule. Moreover, the assays can discover agents which may
inhibit or
enhance the production of the polypeptides of the invention from suitably
manipulated cells
or tissues.
Therefore, the invention includes a method of identifying compounds which bind
to a
polypeptide of the invention comprising the steps o~ (a) incubating a
candidate binding
compound with a polypeptide of the present invention; and (b) determining if
binding has
occurred. Moreover, the invention includes a method of identifying
agonists/antagonists
comprising the steps o~ (a) incubating a candidate compound with a polypeptide
of the
present invention, (b) assaying a biological activity, and (b) determining if
a biological
activity of the polypeptide has been altered.
Targeted Delivery
In another embodiment, the invention provides a method of delivering
compositions
to targeted cells expressing a receptor for a polypeptide of the invention, or
cells expressing
a cell bound form of a polypeptide of the invention.
As discussed herein, polypeptides or antibodies of the invention may be
associated
with heterologous polypeptides, heterologous nucleic acids, toxins, or
prodrugs via
hydrophobic, hydrophilic, ionic and/or covalent interactions. In one
embodiment, the
invention provides a method for the specific delivery of compositions of the
invention to cells
by administering polypeptides of the invention (including antibodies) that are
associated with
heterologous polypeptides or nucleic acids. In one example, the invention
provides a method
for delivering a therapeutic protein into the targeted cell. In another
example, the invention
provides a method for delivering a single stranded nucleic acid (e.g.,
antisense or ribozymes)
or double stranded nucleic acid (e.g., DNA that can integrate into the cell's
genome or
replicate episomally and that can be transcribed) into the targeted cell.
In another embodiment, the invention provides a method for the specific
destruction
of cells (e.g., the destruction of tumor cells) by administering polypeptides
of the invention


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
165
(e.g., polypeptides of the invention or antibodies of the invention) in
association with toxins
or cytotoxic prodrugs.
By "toxin" is meant compounds that bind and activate endogenous cytotoxic
effector
systems, radioisotopes, holotoxins, modified toxins, catalytic subunits of
toxins, or any
molecules or enzymes not normally present in or on the surface of a cell that
under defined
conditions cause the cell's death. Toxins that may be used according to the
methods of the
invention include, but are not limited to, radioisotopes known in the art,
compounds such as,
for example, antibodies (or complement fixing containing portions thereof)
that bind an
inherent or induced endogenous cytotoxic effector system, thymidine kinase,
endonuclease,
RNAse, alpha toxin, ricin, abrin, Pseudomonas exotoxin A, diphtheria toxin,
saporin,
momordin, gelonin, pokeweed antiviral protein, alpha-sarcin and cholera toxin.
By
"cytotoxic prodrug" is meant a non-toxic compound that is converted by an
enzyme,
normally present in the cell, into a cytotoxic compound. Cytotoxic prodrugs
that may be
used according to the methods of the invention include, but are not limited
to, glutamyl
1 S derivatives of benzoic acid mustard alkylating agent, phosphate
derivatives of etoposide or
mitomycin C, cytosine arabinoside, daunorubisin, and phenoxyacetamide
derivatives of
doxorubicin.
Drug Screening
Further contemplated is the use of the polypeptides of the present invention,
or the
polynucleotides encoding these polypeptides, to screen for molecules which
modify the
activities of the polypeptides of the present invention. Such a method would
include
contacting the polypeptide of the present invention with a selected compounds)
suspected of
having antagonist or agonist activity, and assaying the activity of these
polypeptides
following binding.
This invention is particularly useful for screening therapeutic compounds by
using the
polypeptides of the present invention, or binding fragments thereof, in any of
a variety of
drug screening techniques. The polypeptide or fragment employed in such a test
may be
affixed to a solid support, expressed on a cell surface, free in solution, or
located
intracellularly. One method of drug screening utilizes eukaryotic or
prokaryotic host cells
which are stably transformed with recombinant nucleic acids expressing the
polypeptide or
fragment. Drugs are screened against such transformed cells in competitive
binding assays.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
166
One may measure, for example, the formulation of complexes between the agent
being tested
and a polypeptide of the present invention.
Thus, the present invention provides methods of screening for drugs or any
other
agents which affect activities mediated by the polypeptides of the present
invention. These
S methods comprise contacting such an agent with a polypeptide of the present
invention or a
fragment thereof and assaying for the presence of a complex between the agent
and the
polypeptide or a fragment thereof, by methods well known in the art. In such a
competitive
binding assay, the agents to screen are typically labeled. Following
incubation, free agent is
separated from that present in bound form, and the amount of free or
uncomplexed label is a
measure of the ability of a particular agent to bind to the polypeptides of
the present
invention.
Another technique for drug screening provides high throughput screening for
compounds having suitable binding affinity to the polypeptides of the present
invention, and
is described in great detail in European Patent Application 84/03564,
published on September
13, 1984, which is incorporated herein by reference herein. Briefly stated,
large numbers of
different small peptide test compounds are synthesized on a solid substrate,
such as plastic
pins or some other surface. The peptide test compounds are reacted with
polypeptides of the
present invention and washed. Bound polypeptides are then detected by methods
well known
in the art. Purified polypeptides are coated directly onto plates for use in
the aforementioned
drug screening techniques. In addition, non-neutralizing antibodies may be
used to capture
the peptide and immobilize it on the solid support.
This invention also contemplates the use of competitive drug screening assays
in
which neutralizing antibodies capable of binding polypeptides of the present
invention
specifically compete with a test compound for binding to the polypeptides or
fragments
thereof. In this manner, the antibodies are used to detect the presence of any
peptide which
shares one or more antigenic epitopes with a polypeptide of the invention.
Antisense And Ribozyme (Anta~onists~
In specific embodiments, antagonists according to the present invention are
nucleic
acids corresponding to the sequences contained in SEQ ID NO:X, or the
complementary
strand thereof, and/or to nucleotide sequences contained in the cDNA plasmid:Z
identified in
Table 1. In one embodiment, antisense sequence is generated internally, by the
organism, in


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
167
another embodiment, the antisense sequence is separately administered (see,
for example,
O'Connor, J., Neurochem. 56:560 (1991). Oligodeoxynucleotides as Antisense
Inhibitors of
Gene Expression, CRC Press, Boca Raton, FL (1988). Antisense technology can be
used to
control gene expression through antisense DNA or RNA, or through triple-helix
formation.
Antisense techniques are discussed for example, in Okano, J., Neurochem.
56:560 (1991);
Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press,
Boca Raton,
FL (1988). Triple helix formation is discussed in, for instance, Lee et al.,
Nucleic Acids
Research 6:3073 (1979); Cooney et al., Science 241:456 (1988); and Dervan et
al., Science
251:1300 (1991). The methods are based on binding of a polynucleotide to a
complementary
DNA or RNA.
For example, the use of c-myc and c-myb antisense RNA constructs to inhibit
the
growth of the non-lymphocytic leukemia cell line HL-60 and other cell lines
was previously
described. (Wickstrom et al. (1988); Anfossi et al. (1989)). These experiments
were
performed in vitro by incubating cells with the oligoribonucleotide. A similar
procedure for
in vivo use is described in WO 91/15580. Briefly, a pair of oligonucleotides
for a given
antisense RNA is produced as follows: A sequence complimentary to the first 15
bases of the
open reading frame is flanked by an EcoRl site on the 5 end and a HindIII site
on the 3 end.
Next, the pair of oligonucleotides is heated at 90°C for one minute and
then annealed in 2X
ligation buffer (20mM TRIS HCl pH 7.5, IOmM MgCl2, IOMM dithiothreitol (DTT)
and 0.2
mM ATP) and then ligated to the EcoRl/Hind III site of the retroviral vector
PMV7 (WO
91/15580).
For example, the 5' coding portion of a polynucleotide that encodes the
polypeptide of
the present invention may be used to design an antisense RNA oligonucleotide
of from about
10 to 40 base pairs in length. A DNA oligonucleotide is designed to be
complementary to a
region of the gene involved in transcription thereby preventing transcription
and the
production of the receptor. The antisense RNA oligonucleotide hybridizes to
the mRNA in
vivo and blocks translation of the mRNA molecule into receptor polypeptide.
In one embodiment, the antisense nucleic acid of the invention is produced
intracellularly by transcription from an exogenous sequence. For example, a
vector or a
portion thereof, is transcribed, producing an antisense nucleic acid (RNA) of
the invention.
Such a vector would contain a sequence encoding the antisense nucleic acid.
Such a vector
can remain episomal or become chromosomally integrated, as long as it can be
transcribed to


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
168
produce the desired antisense RNA. Such vectors can be constructed by
recombinant DNA
technology methods standard in the art. Vectors can be plasmid, viral, or
others known in the
art, used for replication. and expression in vertebrate cells. Expression of
the sequence
encoding the polypeptide of the present invnetion or fragments thereof, can be
by any
promoter known in the art to act in vertebrate, preferably human cells. Such
promoters can
be inducible or constitutive. Such promoters include, but are not limited to,
the SV40 early
promoter region (Bernoist and Chambon, Nature 29:304-310 (1981), the promoter
contained
in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto et al., Cell
22:787-797
(1980), the herpes thymidine promoter (Wagner et al., Proc. Natl. Acad. Sci.
U.S.A. 78:1441
1445 ( 1981 ), the regulatory sequences of the metallothionein gene (Brinster,
et al., Nature
296:39-42 (1982)), etc.
The antisense nucleic acids of the invention comprise a sequence complementary
to at
least a portion of an RNA transcript of a gene of the present invention.
However, absolute
complementarity, although preferred, is not required. A sequence
"complementary to at least
a portion of an RNA," referred to herein, means a sequence having sufficient
complementarity to be able to hybridize with the RNA, forming a stable duplex;
in the case
of double stranded antisense nucleic acids, a single strand of the duplex DNA
may thus be
tested, or triplex formation may be assayed. The ability to hybridize will
depend on both the
degree of complementarity and the length of the antisense nucleic acid.
Generally, the larger
the hybridizing nucleic acid, the more base mismatches with a RNA it may
contain and still
form a stable duplex (or triplex as the case may be). One skilled in the art
can ascertain a
tolerable degree of mismatch by use of standard procedures to determine the
melting point of
the hybridized complex.
Oligonucleotides that are complementary to the 5' end of the message, e.g.,
the 5'
untranslated sequence up to and including the AUG initiation codon, should
work most
efficiently at inhibiting translation. However, sequences complementary to the
3'
untranslated sequences of mRNAs have been shown to be effective at inhibiting
translation of
mRNAs as well. See generally, Wagner, R., 1994, Nature 372:333-335. Thus,
oligonucleotides complementary to either the 5'- or 3'- non- translated, non-
coding regions of
polynucleotide sequences described herein could be used in an antisense
approach to inhibit
translation of endogenous mRNA. Oligonucleotides complementary to the 5'
untranslated
region of the mRNA should include the complement of the AUG start codon.
Antisense


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
169
oligonucleotides complementary to mRNA coding regions are less efficient
inhibitors of
translation but could be used in accordance with the invention. Whether
designed to
hybridize to the 5'-, 3'- or coding region of mRNA of the present invention,
antisense nucleic
acids should be at least six nucleotides in length, and are preferably
oligonucleotides ranging
from 6 to about 50 nucleotides in length. In specific aspects the
oligonucleotide is at least 10
nucleotides, at least 17 nucleotides, at least 25 nucleotides or at least 50
nucleotides.
The polynucleotides of the invention can be DNA or RNA or chimeric mixtures or
derivatives or modified versions thereof, single-stranded or double-stranded.
The
oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate
backbone, for
example, to improve stability of the molecule, hybridization, etc. The
oligonucleotide may
include other appended groups such as peptides (e.g., for targeting host cell
receptors in
vivo), or agents facilitating transport across the cell membrane (see, e.g.,
Letsinger et al.,
1989, Proc. Natl. Acad. Sci. U.S.A. 86:6553-6556; Lemaitre et al., 1987, Proc.
Natl. Acad.
Sci. 84:648-652; PCT Publication No. W088/09810, published December 15, 1988)
or the
blood-brain barrier (see, e.g., PCT Publication No. W089/10134, published
April 25, 1988),
hybridization-triggered cleavage agents. (See, e.g., Krol et al., 1988,
BioTechniques 6:958-
976) or intercalating agents. (See, e.g., Zon, 1988, Pharm. Res. 5:539-549).
To this end, the
oligonucleotide may be conjugated to another molecule, e.g., a peptide,
hybridization
triggered cross-linking agent, transport agent, hybridization-triggered
cleavage agent, etc.
The antisense oligonucleotide may comprise at least one modified base moiety
which
is selected from the group including, but not limited to, 5-fluorouracil, 5-
bromouracil,
5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine,
5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine,
5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine,
inosine,
N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine,
2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-
adenine,
7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil,
beta-
D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-
methylthio-N6-
isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil,
queosine,
2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-
methyluracil, uracil-
5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-
thiouracil, 3-(3-amino-
3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
170
The antisense oligonucleotide may also comprise at least one modified sugar
moiety
selected from the group including, but not limited to, arabinose, 2-
fluoroarabinose, xylulose,
and hexose.
In yet another embodiment, the antisense oligonucleotide comprises at least
one
modified phosphate backbone selected from the group including, but not limited
to, a
phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a
phosphoramidate, a
phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, and a
formacetal or
analog thereof.
In yet another embodiment, the antisense oligonucleotide is an a-anomeric
oligonucleotide. An a-anomeric oligonucleotide forms specific double-stranded
hybrids with
complementary RNA in which, contrary to the usual b-units, the strands run
parallel to each
other (Gautier et al., 1987, Nucl. Acids Res. 15:6625-6641). The
oligonucleotide is a 2'-0-
methylribonucleotide (moue et al., 1987, Nucl. Acids Res. 15:6131-6148), or a
chimeric
RNA-DNA analogue (moue et al., 1987, FEBS Lett. 215:327-330).
Polynucleotides of the invention may be synthesized by standard methods known
in
the art, e.g. by use of an automated DNA synthesizer (such as are commercially
available
from Biosearch, Applied Biosystems, etc.)., As examples, phosphorothioate
oligonucleotides
may be synthesized by the method of Stein et al. (1988, Nucl. Acids Res.
16:3209),
methylphosphonate oligonucleotides can be prepared by use of controlled pore
glass polymer
supports (Sarin et al., 1988, Proc. Natl. Acad. Sci. U.S.A. 85:7448-7451),
etc.
While antisense nucleotides complementary to the coding region sequence could
be
used, those complementary to the transcribed untranslated region are most
preferred.
Potential antagonists according to the invention also include catalytic RNA,
or a
ribozyme (See, e.g., PCT International Publication WO 90/11364, published
October 4, 1990;
Sarver et al, Science 247:1222-1225 (1990). While ribozymes that cleave mRNA
at site
specific recognition sequences can be used to destroy mRNAs, the use of
hammerhead
ribozymes is preferred. Hammerhead ribozymes cleave mRNAs at locations
dictated by
flanking regions that form complementary base pairs with the target mRNA. The
sole
requirement is that the target mRNA have the following sequence of two bases:
5'-UG-3'.
The construction and production of hammerhead ribozymes is well known in the
art and is
described more fully in Haseloff and Gerlach, Nature 334:585-591 (1988). There
are
numerous potential hammerhead ribozyme cleavage sites within the nucleotide
sequence of


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
171
SEQ >D NO:X. Preferably, the ribozyme is engineered so that the cleavage
recognition site is
located near the 5' end of the mRNA; i.e., to increase efficiency and minimize
the
intracellular accumulation of non-functional mRNA transcripts.
As in the antisense approach, the ribozymes of the invention can be composed
of
modified oligonucleotides (e.~. for improved stability, targeting, etc.) and
should be delivered
to cells which express polypeptides of the present invention in vivo. DNA
constructs
encoding the ribozyme may be introduced into the cell in the same manner as
described
above for the introduction of antisense encoding DNA. A preferred method of
delivery
involves using a DNA construct "encoding" the ribozyme under the control of a
strong
constitutive promoter, such as, for example, pol III or pol II promoter, so
that transfected
cells will produce sufficient quantities of the ribozyme to destroy endogenous
messages and
inhibit translation. Since ribozymes unlike antisense molecules, are
catalytic, a lower
intracellular concentration is required for efficiency.
Antagonist/agonist compounds may be employed to inhibit the cell growth and
proliferation effects of the polypeptides of the present invention on
neoplastic cells and
tissues, i.e. stimulation of angiogenesis of tumors, and, therefore, retard or
prevent abnormal
cellular growth and proliferation, for example, in tumor formation or growth.
The antagonist/agonist may also be employed to prevent hyper-vascular
diseases, and
prevent the proliferation of epithelial lens cells after extracapsular
cataract surgery.
Prevention of the mitogenic activity of the polypeptides of the present
invention may also be
desirous in cases such as restenosis after balloon angioplasty.
The antagonist/agonist may also be employed to prevent the growth of scar
tissue
during wound healing.
The antagonist/agonist may also be employed to treat the diseases described
herein.
Thus, the invention provides a method of treating disorders or diseases,
including but
not limited to the disorders or diseases listed throughout this application,
associated with
overexpression of a polynucleotide of the present invention by administering
to a patient (a)
an antisense molecule directed to the polynucleotide of the present invention,
and/or (b) a
ribozyme directed to the polynucleotide of the present invention.
Binding Peptides and Other Molecules
The invention also encompasses screening methods for identifying polypeptides
and


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
172
nonpolypeptides that bind steroid hormone receptor polypeptides, and the
steroid hormone
receptor binding molecules identified thereby. These binding molecules are
useful, for
example, as agonists and antagonists of the steroid hormone receptor
polypeptides. Such
agonists and antagonists can be used, in accordance with the invention, in the
therapeutic
embodiments described in detail, below.
This method comprises the steps o~
a. contacting steroid hormone receptor polypeptides or steroid hormone
receptor-like
polypeptides with a plurality of molecules; and
b. identifying a molecule that binds the steroid hormone receptor polypeptides
or
steroid hormone receptor-like polypeptides.
The step of contacting the steroid hormone receptor polypeptides or steroid
hormone
receptor-like polypeptides with the plurality of molecules may be effected in
a number of
ways. For example, one may contemplate immobilizing the steroid hormone
receptor
polypeptides or steroid hormone receptor-like polypeptides on a solid support
and bringing a
1 S solution of the plurality of molecules in contact with the immobilized
steroid hormone
receptor polypeptides or steroid hormone receptor-like polypeptides. Such a
procedure would
be akin to an affinity chromatographic process, with the affinity matrix being
comprised of
the immobilized steroid hormone receptor polypeptides or steroid hormone
receptor-like
polypeptides. The molecules having a selective affinity for the steroid
hormone receptor
polypeptides or steroid hormone receptor-like polypeptides can then be
purified by affinity
selection. The nature of the solid support, process for attachment of the
steroid hormone
receptor polypeptides or steroid hormone receptor-like polypeptides to the
solid support,
solvent, and conditions of the affinity isolation or selection are largely
conventional and well
known to those of ordinary skill in the art.
Alternatively, one may also separate a plurality of polypeptides into
substantially
separate fractions comprising a subset of or individual polypeptides. For
instance, one can
separate the plurality of polypeptides by gel electrophoresis, column
chromatography, or like
method known to those of ordinary skill for the separation of polypeptides.
The individual
polypeptides can also be produced by a transformed host cell in such a way as
to be
expressed on or about its outer surface (e.g., a recombinant phage).
Individual isolates can
then be "probed" by the steroid hormone receptor polypeptides or steroid
hormone receptor-
like polypeptides, optionally in the presence of an inducer should one be
required for


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
173
expression, to determine if any selective affinity interaction takes place
between the steroid
hormone receptor polypeptides or steroid hormone receptor-like polypeptides
and the
individual clone. Prior to contacting the steroid hormone receptor
polypeptides or steroid
hormone receptor-like polypeptides with each fraction comprising individual
polypeptides,
the polypeptides could first be transferred to a solid support for additional
convenience. Such
a solid support may simply be a piece of filter membrane, such as one made of
nitrocellulose
or nylon. In this manner, positive clones could be identified from a
collection of transformed
host cells of an expression library, which harbor a DNA construct encoding a
polypeptide
having a selective affinity for steroid hormone receptor polypeptides or
steroid hormone
receptor-like polypeptides. Furthermore, the amino acid sequence of the
polypeptide having a
selective affinity for the steroid hormone receptor polypeptides or steroid
hormone receptor-
like polypeptides can be determined directly by conventional means or the
coding sequence
of the DNA encoding the polypeptide can frequently be determined more
conveniently. The
primary sequence can then be deduced from the corresponding DNA sequence. If
the amino
acid sequence is to be determined from the polypeptide itself, one may use
microsequencing
techniques. The sequencing technique may include mass spectroscopy.
In certain situations, it may be desirable to wash away any unbound steroid
hormone
receptor polypeptides or steroid hormone receptor-like polypeptides, or
alterntatively,
unbound polypeptides, from a mixture of the steroid hormone receptor
polypeptides or
steroid hormone receptor-like polypeptides and the plurality of polypeptides
prior to
attempting to determine or to detect the presence of a selective affinity
interaction. Such a
wash step may be particularly desirable when the steroid hormone receptor
polypeptides or
steroid hormone receptor-like polypeptides or the plurality of polypeptides is
bound to a solid
support.
The plurality of molecules provided according to this method may be provided
by
way of diversity libraries, such as random or combinatorial peptide or
nonpeptide libraries
which can be screened for molecules that specifically bind steroid hormone
receptor
polypeptides. Many libraries are known in the art that can be used, e.g.,
chemically
synthesized libraries, recombinant (e.g., phage display libraries), and in
vitro translation-
based libraries. Examples of chemically synthesized libraries are described in
Fodor et al.,
1991, Science 251:767-773; Houghten et al., 1991, Nature 354:84-86; Lam et
al., 1991,
Nature 354:82-84; Medynski, 1994, Bio/Technology 12:709-710;Gallop et al.,
1994, J.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
174
Medicinal Chemistry 37(9):1233-1251; Ohlmeyer et al., 1993, Proc. Natl. Acad.
Sci. USA
90:10922-10926; Erb et al., 1994, Proc. Natl. Acad. Sci. USA 91:11422-11426;
Houghten et
al., 1992, Biotechniques 13:412; Jayawickreme et al., 1994, Proc. Natl. Acad.
Sci. USA
91:1614-1618; Salmon et al., 1993, Proc. Natl. Acad. Sci. USA 90:11708-11712;
PCT
Publication No. WO 93/20242; and Brenner and Lerner, 1992, Proc. Natl. Acad.
Sci. USA
89:5381-5383.
Examples of phage display libraries are described in Scott and Smith, 1990,
Science
249:386-390; Devlin et al., 1990, Science, 249:404-406; Christian, R. B., et
al., 1992, J. Mol.
Biol. 227:711-718); Lenstra, 1992, J. Immunol. Meth. 152:149-157; Kay et al.,
1993, Gene
128:59-65; and PCT Publication No. WO 94/18318 dated Aug. 18, 1994.
In vitro translation-based libraries include but are not limited to those
described in
PCT Publication No. WO 91/05058 dated Apr. 18, 1991; and Mattheakis et al.,
1994, Proc.
Natl. Acad. Sci. USA 91:9022-9026.
By way of examples of nonpeptide libraries, a benzodiazepine library (see
e.g., Bunin
et al., 1994, Proc. Natl. Acad. Sci. USA 91:4708-4712) can be adapted for use.
Peptoid
libraries (Simon et al., 1992, Proc. Natl. Acad. Sci. USA 89:9367-9371) can
also be used.
Another example of a library that can be used, in which the amide
functionalities in peptides
have been permethylated to generate a chemically transformed combinatorial
library, is
described by Ostresh et al. (1994, Proc. Natl. Acad. Sci. USA 91:11138-11142).
The variety of non-peptide libraries that are useful in the present invention
is great.
For example, Ecker and Crooke, 1995, Bio/Technology 13:351-360 list
benzodiazepines,
hydantoins, piperazinediones, biphenyls, sugar analogs, beta-mercaptoketones,
arylacetic
acids, acylpiperidines, benzopyrans, cubanes, xanthines, aminimides, and
oxazolones as
among the chemical species that form the basis of various libraries.
Non-peptide libraries can be classified broadly into two types: decorated
monomers
and oligomers. Decorated monomer libraries employ a relatively simple scaffold
structure
upon which a variety functional groups is added. Often the scaffold will be a
molecule with a
known useful pharmacological activity. For example, the scaffold might be the
benzodiazepine structure.
Non-peptide oligomer libraries utilize a large number of monomers that are
assembled
together in ways that create new shapes that depend on the order of the
monomers. Among
the monomer units that have been used are carbamates, pyrrolinones, and
morpholinos.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
175
Peptoids, peptide-like oligomers in which the side chain is attached to the
alpha amino group
rather than the alpha carbon, form the basis of another version of non-peptide
oligomer
libraries. The first non-peptide oligomer libraries utilized a single type of
monomer and thus
contained a repeating backbone. Recent libraries have utilized more than one
monomer,
S giving the libraries added flexibility.
Screening the libraries can be accomplished by any of a variety of commonly
known
methods. See, e.g., the following references, which disclose screening of
peptide libraries:
Parmley and Smith, 1989, Adv. Exp. Med. Biol. 251:215-218; Scott and Smith,
1990,
Science 249:386-390; Fowlkes et al., 1992; BioTechniques 13:422-427; Oldenburg
et al.,
1992, Proc. Natl. Acad. Sci. USA 89:5393-5397; Yu et al., 1994, Cell 76:933-
945; Staudt et
al., 1988, Science 241:577-580; Bock et al., 1992, Nature 355:564-566; Tuerk
et al., 1992,
Proc. Natl. Acad. Sci. USA 89:6988-6992; Ellington et al., 1992, Nature
355:850-852; U.S.
Pat. No. 5,096,815, U.S. Pat. No. 5,223,409, and U.S. Pat. No. 5,198,346, all
to Ladner et al.;
Rebar and Pabo, 1993, Science 263:671-673; and CT Publication No. WO 94/18318.
In a specific embodiment, screening to identify a molecule that binds steroid
hormone
receptor polypeptides can be carried out by contacting the library members
with a steroid
hormone receptor polypeptides or steroid hormone receptor-like polypeptides
immobilized on
a solid phase and harvesting those library members that bind to the steroid
hormone receptor
polypeptides or steroid hormone receptor-like polypeptides. Examples of such
screening
methods, termed "panning" techniques are described by way of example in
Parmley and
Smith, 1988, Gene 73:305-318; Fowlkes et al., 1992, BioTechniques 13:422-427;
PCT
Publication No. WO 94/18318; and in references cited herein.
In another embodiment, the two-hybrid system for selecting interacting
proteins in
yeast (Fields and Song, 1989, Nature 340:245-246; Chien et al., 1991, Proc.
Natl. Acad. Sci.
USA 88:9578-9582) can be used to identify molecules that specifically bind to
steroid
hormone receptor polypeptides or steroid hormone receptor-like polypeptides.
Where the steroid hormone receptor binding molecule is a polypeptide, the
polypeptide can be conveniently selected from any peptide library, including
random peptide
libraries, combinatorial peptide libraries, or biased peptide libraries. The
term "biased" is
used herein to mean that the method of generating the library is manipulated
so as to restrict
one or more parameters that govern the diversity of the resulting collection
of molecules, in
this case peptides.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
176
Thus, a truly random peptide library would generate a collection of peptides
in which
the probability of finding a particular amino acid at a given position of the
peptide is the same
for all 20 amino acids. A bias can be introduced into the library, however, by
specifying, for
example, that a lysine occur every fifth amino acid or that positions 4, 8,
and 9 of a
decapeptide library be fixed to include only arginine. Clearly, many types of
biases can be
contemplated, and the present invention is not restricted to any particular
bias. Furthermore,
the present invention contemplates specific types of peptide libraries, such
as phage displayed
peptide libraries and those that utilize a DNA construct comprising a lambda
phage vector
with a DNA insert.
As mentioned above, in the case of a steroid hormone receptor binding molecule
that
is a polypeptide, the polypeptide may have about 6 to less than about 60 amino
acid residues,
preferably about 6 to about 10 amino acid residues, and most preferably, about
6 to about 22
amino acids. In another embodiment, a steroid hormone receptor binding
polypeptide has in
the range of 15-100 amino acids, or 20-50 amino acids.
The selected steroid hormone receptor binding polypeptide can be obtained by
chemical synthesis or recombinant expression.
Other Activities
A polypeptide, polynucleotide, agonist, or antagonist of the present
invention, as a
result of the ability to stimulate vascular endothelial cell growth, may be
employed in
treatment for stimulating re-vascularization of ischemic tissues due to
various disease
conditions such as thrombosis, arteriosclerosis, and other cardiovascular
conditions. The
polypeptide, polynucleotide, agonist, or antagonist of the present invention
may also be
employed to stimulate angiogenesis and limb regeneration, as discussed above.
A polypeptide, polynucleotide, agonist, or antagonist of the present invention
may
also be employed for treating wounds due to injuries, burns, post-operative
tissue repair, and
ulcers since they are mitogenic to various cells of different origins, such as
fibroblast cells
and skeletal muscle cells, and therefore, facilitate the repair or replacement
of damaged or
diseased tissue.
A polypeptide, polynucleotide, agonist, or antagonist of the present invention
may
also be employed stimulate neuronal growth and to treat and prevent neuronal
damage which
occurs in certain neuronal disorders or neuro-degenerative conditions such as
Alzheimer's


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
177
disease, Parkinson's disease, and AIDS-related complex. A polypeptide,
polynucleotide,
agonist, or antagonist of the present invention may have the ability to
stimulate chondrocyte
growth, therefore, they may be employed to enhance bone and periodontal
regeneration and
aid in tissue transplants or bone grafts.
A polypeptide, polynucleotide, agonist, or antagonist of the present invention
may be
also be employed to prevent skin aging due to sunburn by stimulating
keratinocyte growth.
A polypeptide, polynucleotide, agonist, or antagonist of the present invention
may
also be employed for preventing hair loss, since FGF family members activate
hair-forming
cells and promotes melanocyte growth. Along the same lines, a polypeptide,
polynucleotide,
agonist, or antagonist of the present invention may be employed to stimulate
growth and
differentiation of hematopoietic cells and bone marrow cells when used in
combination with
other cytokines.
A polypeptide, polynucleotide, agonist, or antagonist of the present invention
may
also be employed to maintain organs before transplantation or for supporting
cell culture of
primary tissues. A polypeptide, polynucleotide, agonist, or antagonist of the
present
invention may also be employed for inducing tissue of mesodermal origin to
differentiate in
early embryos.
A polypeptide, polynucleotide, agonist, or antagonist of the present invention
may
also increase or decrease the differentiation or proliferation of embryonic
stem cells, besides,
as discussed above, hematopoietic lineage.
A polypeptide, polynucleotide, agonist, or antagonist of the present invention
may
also be used to modulate mammalian characteristics, such as body height,
weight, hair color,
eye color, skin, percentage of adipose tissue, pigmentation, size, and shape
(e.g., cosmetic
surgery). Similarly, a polypeptide, polynucleotide, agonist, or antagonist of
the present
invention may be used to modulate mammalian metabolism affecting catabolism,
anabolism,
processing, utilization, and storage of energy.
A polypeptide, polynucleotide, agonist, or antagonist of the present invention
may be
used to change a mammal's mental state or physical state by influencing
biorhythms,
caricadic rhythms, depression (including depressive disorders), tendency for
violence,
tolerance for pain, reproductive capabilities (preferably by Activin or
Inhibin-like activity),
hormonal or endocrine levels, appetite, libido, memory, stress, or other
cognitive qualities.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
178
A polypeptide, polynucleotide, agonist, or antagonist of the present invention
may
also be used as a food additive or preservative, such as to increase or
decrease storage
capabilities, fat content, lipid, protein, carbohydrate, vitamins, minerals,
cofactors or other
nutritional components.
The above-recited applications have uses in a wide variety of hosts. Such
hosts
include, but are not limited to, human, murine, rabbit, goat, guinea pig,
camel, horse, mouse,
rat, hamster, pig, micro-pig, chicken, goat, cow, sheep, dog, cat, non-human
primate, and
human. In specific embodiments, the host is a mouse, rabbit, goat, guinea pig,
chicken, rat,
hamster, pig, sheep, dog or cat. In preferred embodiments, the host is a
mammal. In most
preferred embodiments, the host is a human.
Other Preferred Embodiments
Other preferred embodiments of the claimed invention include an isolated
nucleic
acid molecule comprising a nucleotide sequence which is at least 95% identical
to a sequence
of at least about 50 contiguous nucleotides in the nucleotide sequence of SEQ
117 NO:X or
the complementary strand thereto, and/or cDNA plasmid:Z.
Also preferred is a nucleic acid molecule wherein said sequence of contiguous
nucleotides is included in the nucleotide sequence of SEQ ID NO:X in the range
of positions
identified for SEQ ID NO:X in Table 1.
Also preferred is an isolated nucleic acid molecule comprising a nucleotide
sequence
which is at least 95% identical to a sequence of at least about 150 contiguous
nucleotides in
the nucleotide sequence of SEQ ID NO:X or the complementary strand thereto,
and/or cDNA
plasmid:Z.
Further preferred is an isolated nucleic acid molecule comprising a nucleotide
sequence which is at least 95% identical to a sequence of at least about 500
contiguous
nucleotides in the nucleotide sequence of SEQ ID NO:X or the complementary
strand thereto,
and/or cDNA plasmid:Z.
A further preferred embodiment is a nucleic acid molecule comprising a
nucleotide
sequence which is at least 95% identical to the nucleotide sequence of SEQ ID
NO:X in the
range of positions identified for SEQ ID NO:X in Table 1.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
179
A further preferred embodiment is an isolated nucleic acid molecule comprising
a
nucleotide sequence which is at least 95% identical to the complete nucleotide
sequence of
SEQ ID NO:X or the complementary strand thereto, and/or cDNA plasmid:Z.
Also preferred is an isolated nucleic acid molecule which hybridizes under
stringent
hybridization conditions to a nucleic acid molecule comprising a nucleotide
sequence of SEQ
ID NO:X or the complementary strand thereto and/or cDNA plasmid:Z, wherein
said nucleic
acid molecule which hybridizes does not hybridize under stringent
hybridization conditions
to a nucleic acid molecule having a nucleotide sequence consisting of only A
residues or of
only T residues.
Also preferred is a composition of matter comprising a DNA molecule which
comprises cDNA plasmid:Z.
Also preferred is an isolated nucleic acid molecule comprising a nucleotide
sequence
which is at least 95% identical to a sequence of at least 50 contiguous
nucleotides in the
nucleotide sequence of cDNA plasmid:Z.
Also preferred is an isolated nucleic acid molecule, wherein said sequence of
at least
50 contiguous nucleotides is included in the nucleotide sequence of an open
reading frame
sequence encoded by cDNA plasmid:Z.
Also preferred is an isolated nucleic acid molecule comprising a nucleotide
sequence
which is at least 95% identical to sequence of at least 150 contiguous
nucleotides in the
nucleotide sequence encoded by cDNA plasmid:Z.
A further preferred embodiment is an isolated nucleic acid molecule comprising
a
nucleotide sequence which is at least 95% identical to sequence of at least
S00 contiguous
nucleotides in the nucleotide sequence encoded by cDNA plasmid:Z.
A further preferred embodiment is an isolated nucleic acid molecule comprising
a
nucleotide sequence which is at least 95% identical to the complete nucleotide
sequence
encoded by cDNA plasmid:Z.
A further preferred embodiment is a method for detecting in a biological
sample a
nucleic acid molecule comprising a nucleotide sequence which is at least 95%
identical to a
sequence of at least 50 contiguous nucleotides in a sequence selected from the
group
consisting o~ a nucleotide sequence of SEQ ID NO:X or the complementary strand
thereto
and a nucleotide sequence encoded by cDNA plasmid:Z; which method comprises a
step of
comparing a nucleotide sequence of at least one nucleic acid molecule in said
sample with a


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
180
sequence selected from said group and determining whether the sequence of said
nucleic acid
molecule in said sample is at least 95% identical to said selected sequence.
Also preferred is the above method wherein said step of comparing sequences
comprises determining the extent of nucleic acid hybridization between nucleic
acid
molecules in said sample and a nucleic acid molecule comprising said sequence
selected from
said group. Similarly, also preferred is the above method wherein said step of
comparing
sequences is performed by comparing the nucleotide sequence determined from a
nucleic
acid molecule in said sample with said sequence selected from said group. The
nucleic acid
molecules can comprise DNA molecules or RNA molecules.
A further preferred embodiment is a method for identifying the species, tissue
or cell
type of a biological sample which method comprises a step of detecting nucleic
acid
molecules in said sample, if any, comprising a nucleotide sequence that is at
least 95%
identical to a sequence of at least 50 contiguous nucleotides in a sequence
selected from the
group consisting of a nucleotide sequence of SEQ m NO:X or the complementary
strand
thereto and a nucleotide sequence encoded by cDNA plasmid:Z.
The method for identifying the species, tissue or cell type of a biological
sample can
comprise a step of detecting nucleic acid molecules comprising a nucleotide
sequence in a
panel of at least two nucleotide sequences, wherein at least one sequence in
said panel is at
least 95% identical to a sequence of at least 50 contiguous nucleotides in a
sequence selected
from said group.
Also preferred is a method for diagnosing in a subject a pathological
condition
associated with abnormal structure or expression of a nucleotide sequence of
SEQ ID NO:X
or the complementary strand thereto or cDNA plasmid:Z which encodes a protein,
wherein
the method comprises a step of detecting in a biological sample obtained from
said subject
nucleic acid molecules, if any, comprising a nucleotide sequence that is at
least 95% identical
to a sequence of at least 50 contiguous nucleotides in a sequence selected
from the group
consisting o~ a nucleotide sequence of SEQ m NO:X or the complementary strand
thereto
and a nucleotide sequence of cDNA plasmid:Z.
The method for diagnosing a pathological condition can comprise a step of
detecting
nucleic acid molecules comprising a nucleotide sequence in a panel of at least
two nucleotide
sequences, wherein at least one sequence in said panel is at least 95%
identical to a sequence
of at least 50 contiguous nucleotides in a sequence selected from said group.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
181
Also preferred is a composition of matter comprising isolated nucleic acid
molecules
wherein the nucleotide sequences of said nucleic acid molecules comprise a
panel of at least
two nucleotide sequences, wherein at least one sequence in said panel is at
least 95%
identical to a sequence of at least 50 contiguous nucleotides in a sequence
selected from the
group consisting o~ a nucleotide sequence of SEQ ID NO:X or the complementary
strand
thereto and a nucleotide sequence encoded by cDNA plasmid:Z. The nucleic acid
molecules
can comprise DNA molecules or RNA molecules.
Also preferred is an isolated polypeptide comprising an amino acid sequence at
least
90% identical to a sequence of at least about 10 contiguous amino acids in the
polypeptide
sequence of SEQ )D NO:Y; a polypeptide encoded by SEQ 117 NO:X or the
complementary
strand thereto and/or a polypeptide encoded by cDNA plasmid:Z.
Also preferred is an isolated polypeptide comprising an amino acid sequence at
least
95% identical to a sequence of at least about 30 contiguous amino acids in the
amino acid
sequence of SEQ ID NO:Y; a polypeptide encoded by SEQ ID NO:X or the
complementary
strand thereto and/or a polypeptide encoded by cDNA plasmid:Z.
Further preferred is an isolated polypeptide comprising an amino acid sequence
at
least 95% identical to a sequence of at least about 100 contiguous amino acids
in the amino
acid sequence of SEQ ID NO:Y; a polypeptide encoded by SEQ ID NO:X or the
complementary strand thereto and/or a polypeptide encoded by cDNA plasmid:Z.
Further preferred is an isolated polypeptide comprising an amino acid sequence
at
least 95% identical to the complete amino acid sequence of SEQ ID NO:Y; a
polypeptide
encoded by SEQ ID NO:X or the complementary strand thereto and/or a
polypeptide encoded
by cDNA plasmid:Z.
Further preferred is an isolated polypeptide comprising an amino acid sequence
at
least 90% identical to a sequence of at least about 10 contiguous amino acids
in the complete
amino acid sequence of a polypeptide encoded by cDNA plasmid:Z
Also preferred is a polypeptide wherein said sequence of contiguous amino
acids is
included in the amino acid sequence of a portion of said polypeptide encoded
by cDNA
plasmid:Z; a polypeptide encoded by SEQ ID NO:X or the complementary strand
thereto
and/or the polypeptide sequence of SEQ ID NO:Y.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
182
Also preferred is an isolated polypeptide comprising an amino acid sequence at
least
95% identical to a sequence of at least about 30 contiguous amino acids in the
amino acid
sequence of a polypeptide encoded by cDNA plasmid:Z.
Also preferred is an isolated polypeptide comprising an amino acid sequence at
least
95% identical to a sequence of at least about 100 contiguous amino acids in
the amino acid
sequence of a polypeptide encoded by cDNA plasmid:Z.
Also preferred is an isolated polypeptide comprising an amino acid sequence at
least
95% identical to the amino acid sequence of a polypeptide encoded by cDNA
plasmid:Z.
Further preferred is an isolated antibody which binds specifically to a
polypeptide
comprising an amino acid sequence that is at least 90% identical to a sequence
of at least 10
contiguous amino acids in a sequence selected from the group consisting of a
polypeptide
sequence of SEQ >D NO:Y; a polypeptide encoded by SEQ ID NO:X or the
complementary
strand thereto and a polypeptide encoded by cDNA plasmid:Z.
Further preferred is a method for detecting in a biological sample a
polypeptide
comprising an amino acid sequence which is at least 90% identical to a
sequence of at least
10 contiguous amino acids in a sequence selected from the group consisting of:
a polypeptide
sequence of SEQ ID NO:Y; a polypeptide encoded by SEQ ID NO:X or the
complementary
strand thereto and a polypeptide encoded by cDNA plasmid:Z; which method
comprises a
step of comparing an amino acid sequence of at least one polypeptide molecule
in said
sample with a sequence selected from said group and determining whether the
sequence of
said polypeptide molecule in said sample is at least 90% identical to said
sequence of at least
10 contiguous amino acids.
Also preferred is the above method wherein said step of comparing an amino
acid
sequence of at least one polypeptide molecule in said sample with a sequence
selected from
said group comprises determining the extent of specific binding of
polypeptides in said
sample to an antibody which binds specifically to a polypeptide comprising an
amino acid
sequence that is at least 90% identical to a sequence of at least 10
contiguous amino acids in a
sequence selected from the group consisting of a polypeptide sequence of SEQ m
NO:Y; a
polypeptide encoded by SEQ ID NO:X or the complementary strand thereto and a
polypeptide encoded by cDNA plasmid:Z.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
183
Also preferred is the above method wherein said step of comparing sequences is
performed by comparing the amino acid sequence determined from a polypeptide
molecule in
said sample with said sequence selected from said group.
Also preferred is a method for identifying the species, tissue or cell type of
a
biological sample which method comprises a step of detecting polypeptide
molecules in said
sample, if any, comprising an amino acid sequence that is at least 90%
identical to a sequence
of at least 10 contiguous amino acids in a sequence selected from the group
consisting of
polypeptide sequence of SEQ ID NO:Y; a polypeptide encoded by SEQ ID NO:X or
the
complementary strand thereto and a polypeptide encoded by cDNA plasmid:Z.
Also preferred is the above method for identifying the species, tissue or cell
type of a
biological sample, which method comprises a step of detecting polypeptide
molecules
comprising an amino acid sequence in a panel of at least two amino acid
sequences, wherein
at least one sequence in said panel is at least 90% identical to a sequence of
at least 10
contiguous amino acids in a sequence selected from the above group.
Also preferred is a method for diagnosing in a subject a pathological
condition
associated with abnormal structure or expression of a nucleic acid sequence
identified in
Table 1 encoding a polypeptide, which method comprises a step of detecting in
a biological
sample obtained from said subject polypeptide molecules comprising an amino
acid sequence
in a panel of at least two amino acid sequences, wherein at least one sequence
in said panel is
at least 90% identical to a sequence of at least 10 contiguous amino acids in
a sequence
selected from the group consisting o~ polypeptide sequence of SEQ 117 NO:Y; a
polypeptide
encoded by SEQ ID NO:X or the complementary strand thereto and a polypeptide
encoded
by cDNA plasmid:Z.
In any of these methods, the step of detecting said polypeptide molecules
includes
using an antibody.
Also preferred is an isolated nucleic acid molecule comprising a nucleotide
sequence
which is at least 95% identical to a nucleotide sequence encoding a
polypeptide wherein said
polypeptide comprises an amino acid sequence that is at least 90% identical to
a sequence of
at least 10 contiguous amino acids in a sequence selected from the group
consisting of:
polypeptide sequence of SEQ ID NO:Y; a polypeptide encoded by SEQ ID NO:X or
the
complementary strand thereto and a polypeptide encoded by cDNA plasmid:Z.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
184
Also preferred is an isolated nucleic acid molecule, wherein said nucleotide
sequence
encoding a polypeptide has been optimized for expression of said polypeptide
in a
prokaryotic host.
Also preferred is an isolated nucleic acid molecule, wherein said polypeptide
comprises an amino acid sequence selected from the group consisting o~
polypeptide
sequence of SEQ ID NO:Y; a polypeptide encoded by SEQ ID NO:X or the
complementary
strand thereto and a polypeptide encoded by cDNA plasmid:Z.
Further preferred is a method of making a recombinant vector comprising
inserting
any of the above isolated nucleic acid molecule into a vector. Also preferred
is the
recombinant vector produced by this method. Also preferred is a method of
making a
recombinant host cell comprising introducing the vector into a host cell, as
well as the
recombinant host cell produced by this method.
Also preferred is a method of making an isolated polypeptide comprising
culturing
this recombinant host cell under conditions such that said polypeptide is
expressed and
recovering said polypeptide. Also preferred is this method of making an
isolated
polypeptide, wherein said recombinant host cell is a eukaryotic cell and said
polypeptide is a
human protein comprising an amino acid sequence selected from the group
consisting of
polypeptide sequence of SEQ ID NO:Y; a polypeptide encoded by SEQ ID NO:X or
the
complementary strand thereto and a polypeptide encoded by cDNA plasmid:Z. The
isolated
polypeptide produced by this method is also preferred.
Also preferred is a method of treatment of an individual in need of an
increased level
of a protein activity, which method comprises administering to such an
individual a
Therapeutic comprising an amount of an isolated polypeptide, polynucleotide,
immunogenic
fragment or analogue thereof, binding agent, antibody, or antigen binding
fragment of the
claimed invention effective to increase the level of said protein activity in
said individual.
Also preferred is a method of treatment of an individual in need of a
decreased level
of a protein activity, which method comprised administering to such an
individual a
Therapeutic comprising an amount of an isolated polypeptide, polynucleotide,
immunogenic
fragment or analogue thereof, binding agent, antibody, or antigen binding
fragment of the
claimed invention effective to decrease the level of said protein activity in
said individual.
In specific embodiments of the invention, for each "Contig 1D" listed in the
fourth
column of Table 2, preferably excluded are one or more polynucleotides
comprising, or


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
185
alternatively consisting of, a nucleotide sequence referenced in the fifth
column of Table 2
and described by the general formula of a-b, whereas a and b are uniquely
determined for the
corresponding SEQ ID NO:X referred to in column 3 of Table 2. Further specific
embodiments are directed to polynucleotide sequences excluding one, two,
three, four, or
more of the specific polynucleotide sequences referred to in the fifth column
of Table 2. In no
way is this listing meant to encompass all of the sequences which may be
excluded by the
general formula, it is just a representative example. All references available
through these
accessions are hereby incorporated by reference in their entirety.
TABLE 2
Gene cDNA NT Contig Public Accession Numbers


No. Clone ID SEQ ID
ID


NO:
X


1 HWHQR64 2 790187 AI744466, AW009708, AI858982,


AA527985, AI150051, AI366789,


AI097089, AI192808, AI288593,
AI265968,


AI369388, AI494313, AW070581,


AW050705, AI346159, AA404504,


AW005035, AA973927, AI016283,


AA809633, 847884, AI269185, AI269077,


AA639397, AI680157, AA928427,


AI339180, AI028315, AA909756,


AW382959, AA730141, AI573114,
848001,


AW439806, AA594507, AI520841,


AI092728, AI318289, AW026051,


AI890896, AI202178, N78584, AI168713,


AA627624, AI371395, 802105, AA026211,


AA380416, 868665, AA552066, AI379769,


AI217243, AI933984, AJ239444,
AI654784,


AA308302, 868718, AW364625, and


N72772.


2 HHPFB87 3 1027768 AW241826, AI613111, AA903235,
and


AA970041.


3 HOUFT74 4 790183 AI478927, AW071645, AI566751,


AI637656, AA628416, AI655849,


AW136578, AI342003, AW136580,


AW136585, N94614, AA454157, AI138477,


AI376851, AA846238, AI292352,


AA047446, AI767841, W45047, AI004357,


SUBSTITUTE SHEET (RULE 26)


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
186
H71118, H42483, N92184, AA934748,


AI652351, AA373943, AW104994,


AA160181, AA046636, AA946892,


AI970182, AA833797, 862724, AW339069,


AW090555, AI933958, 863679, AI373725,


AI952944, AI811605, AA741383,
N64387,


AA370734, AI635804, and AL049996.


4 HPWSB15 5 838458 AA906222, AA862519, AW383156,


AI140607, AA723622, AA758692,


AW372067, AW451516, AI569507,


AI937131, AA416872, AI198079,


AI912434, AW 161202, AI796568,


AI581139, AI689557, and AI277260.


TABLE 3
Clone ID Library Code


NO: Z


HWHQR64 H0052 H0136H0171 H0264H0265 H0318 H0424 H0457 H0488


H0518


H0556 H0559H0587 H0597H0635 H0677 H0682 L0513 L0640


L0659


L0663 L0664L0666 L0743L0747 L0748 L0750 L0754 L0755
L0756


L0758 L0759L0764 L0766L0768 L0769 L0771 L0772 L0773
L0774


L0775 L0776L0777 L0794L0809 50053 50132 50358 S0360
S0418


T0041 T0109TO110


HHPFB87 H0040 H0051


HOUFT74 H0013 H0032H0042 H0090H0144 H0346 H0375 H0393 H0402


H0423


H0441 H0520H0521 H0529H0551 H0560 H0575 H0586 H0587


H0589


H0591 H0600H0624 H0635H0638 H0641 H0657 H0667 H0682


H0689


L0362 L0471L0638 L0648L0659 L0663 L0664 L0666 L0731
L0744


L0748 L0752L0754 L0755L0756 L0757 L0758 L0759 L0766
L0767


L0768 L0775L0789 L0791L0794 L0803 L0804 L0806 50026
S0027


50028 5004650146 50192
S0214
S0344
S0358
50412
50420
T0039



HPWSB15 HO144 H0519H0553 HO570L0774 L0779 50174


~HNGAY56 S0052


TABLE 4
Library Library Description


Code


H0013 Human 8 Week Whole Embryo


SUBSTITUTE SHEET (RULE 26)


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
187
H0032 Human Prostate


H0040 Human Testes Tumor


H0042 Human Adult Pulmonary


H0051 Human Hi ocam us


H0052 Human Cerebellum


H0090 Human T-Cell Lym homa


H0136 Supt Cells, cyclohexamide treated


H0144 Nine Week Old Early Stage Human


H0171 12 Week Old Earl Stage Human, II


H0264 human tonsils


H0265 Activated T-Cell (l2hs)/Thiouridine labelledEco


H0318 HUMAN B CELL LYMPHOMA


H0346 Brain-medulloblastoma


H0375 Human Lung


H0393 Fetal Liver, subtraction II


H0402 CD34 depleted Buffy Coat (Cord Blood), re-excision


H0423 T-Cell PHA 24 hrs


H0424 Human Pituitary, subt IX


H0441 H. Kidney Cortex, subtracted'


H0457 Human Eosino hils


H0488 Human Tonsils, Lib 2


H0518 BMC stimulated w/ poly I/C


H0519 NTERA2, control


H0520 NTERA2 + retinoic acid, 14 days


H0521 Primary Dendritic Cells, lib 1


H0529 Myoloid Progenitor Cell Line


HO551 Human Thymus Stromal Cells


H0553 Human Placenta


H0556 Activated T-cell(12h)/Thiouridine-re-excision


H0559 HL-60, PMA 4H, re-excision


H0560 KMH2


H0570 Human Fetal Brain, normalized CSOOH


H0575 Human Adult Pulmonary,re-excision


H0586 Healing groin wound, 6.5 hours post incision


H0587 Healing groin wound, 7.5 hours post incision


H0589 CD34 positive cells (cord blood),re-ex


H0591 Human T-cell lymphoma,re-excision


H0597 Human Colon, re-excision


H0600 Healing Abdomen wound,70&90 min post incision


H0624 12 Week Early Stage Human II, Reexcision


H0635 Human Activated T-Cells, re-excision


H0638 CD40 activated monocyte dendridic cells


H0641 LPS activated derived dendritic cells


H0657 B-cells (stimulated)


H0667 Stromal cells(HBM3.18)


SUBSTITUTE SHEET (RULE 26)


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
188
H0677 TNFR degenerate oligo


H0682 Ovarian cancer, Serous Pa illary Adenocarcinoma


H0689 Ovarian Cancer


L0362 Stratagene ovarian cancer (#937219)


L0471 Human fetal heart, Lambda ZAP Ex ress


L0513 NCI CGAP Ov37


L0638 NCI CGAP_Brn35


L0640 NCI CLAP Brl8


L0648 NCI CGAP Eso2


L0659 NCI CGAP Panl


L0663 NCI_CGAP_Ut2


L0664 NCI CGAP Ut3


L0666 NCI_CGAP Utl


L0731 Soares regnant_uterus NbHPU


L0743 Soares breast 2NbHBst


L0744 Soares breast 3NbHBst


L0747 Soares fetal_heart NbHHI9W


L0748 Soares fetal liver s teen 1NFLS


L0750 Soares_fetal_lung NbHLI9W


L0752 Soares_parathyroid_tumor NbHPA


L0754 Soares placenta Nb2HP


L0755 Soares lacenta 8to9weeks 2NbHP8to9W


L0756 Soares_multiple sclerosis_2NbHMSP


L0757 Soares_senescent fibroblasts NbHSF


L0758 Soares_testis_NHT


L0759 Soares total fetus_Nb2HF8 9w


L0764 NCI_CGAP Co3


L0766 NCI_CGAP_GCB 1


L0767 NCI CGAP GC3


L0768 NCI_CGAP GC4


L0769 NCI CGAP Brn25


L0771 NCI_CGAP_Co8


L0772 NCI CGAP_CoIO


L0773 NCI CLAP Co9


L0774 NCI_CGAP_Kid3


L0775 NCI_CGAP_KidS


L0776 NCI_CGAP Lu5


L0777 Soares NhHMPu_Sl


L0779 Soares NFL_T_GBC S 1


L0789 NCI_CGAP_Sub3


L0791 NCI_CGAP Subs


L0794 NCI_CGAP GC6


L0803 NCI_CGAP Kidll


L0804 NCI_CGAP_Kidl2


L0806 NCI CGAP Lu 19


SUBSTITUTE SHEET (RULE 26)


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
189
L0809 NCI CGAP Pr28


S0026 Stromal cell TF274


50027 Smooth muscle, serum treated


50028 Smooth muscle,control


50046 Endothelial-induced


50052 neutro hils control


50053 Neutrophils IL-1 and LPS induced


S0132 E ithelial-TNFa and INF induced


50146 rostate-edited


50174 Prostate-BPH subtracted II


50192 Synovial Fibroblasts (control)


50214 Human Osteoclastoma, re-excision


50344 Macro hage-oxLDL, re-excision


50358 Colon Normal III


50360 Colon Tumor II


S0412 Temporal cortex-Alzheizmer, subtracted


50418 CHME Cell Line,treated 5 hrs


S0420 CHME Cell Line,untreated


T0039 HSA 172 Cells


T0041 Jurkat T-cell G1 phase


T0109 Human (HCC) cell line liver (mouse) metastasis, remake


TO110 Human colon carcinoma (HCC) cell line, remake


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.
15
SUBSTITUTE SHEET (RULE 26)


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
190
l0 Examples
Example 1: Isolation of a Selected cDNA Clone From the Deposited Sample
Each cDNA clone in a cited ATCC deposit is contained in a plasmid vector.
Table 1
identifies the vectors used to construct the cDNA library from which each
clone was isolated.
In many cases, the vector used to construct the library is a phage vector from
which a plasmid
has been excised. The table immediately below correlates the related plasmid
for each phage
vector used in constructing the cDNA library. For example, where a particular
clone is
identified in Table 1 as being isolated in the vector "Lambda Zap," the
corresponding
deposited clone is in "pBluescript."
Vector Used to Construct Library Correspondin. Deposited Plasmid
Lambda Zap pBluescript (pBS)
Uni-Zap XR pBluescript (pBS)
Zap Express pBK
lafinid BA plafinid BA
pSport 1 pSport 1
pCMVSport 2.0 pCMVSport 2.0
pCMVSport 3.0 pCMVSport 3.0
pCR~2.1 pCR~2.1
Vectors Lambda Zap (U.S. Patent Nos. 5,128,256 and 5,286,636), Uni-Zap XR
(U.S.
Patent Nos. 5,128, 256 and 5,286,636), Zap Express (U.S. Patent Nos. 5,128,256
and
5,286,636), pBluescript (pBS) (Short et al., Nucleic Acids Res., 16:7583-7600
(1988); Alting-


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
191
Mees et al., Nucleic Acids Res., 17:9494 (1989)) and pBK (Alting-Mees et al.,
Strategies,
5:58-61 (1992)) are commercially available from Stratagene Cloning Systems,
Inc., 11011 N.
Torrey Pines Road, La Jolla, CA, 92037. pBS contains an ampicillin resistance
gene and
pBK contains a neomycin resistance gene. Both can be transformed into E. coli
strain XL-1
Blue, also available from Stratagene. pBS comes in 4 forms SK+, SK-, KS+ and
KS. The S
and K refers to the orientation of the polylinker to the T7 and T3 primer
sequences which
flank the polylinker region ("S" is for SacI and "K" is for Kpnl which are the
first sites on
each respective end of the linker). "+" or "-" refer to the orientation of the
fl origin of
replication ("ori"), such that in one orientation, single stranded rescue
initiated from the fl on
generates sense strand DNA and in the other, antisense.
Vectors pSportl, pCMVSport 2.0 and pCMVSport 3.0, were obtained from Life
Technologies, Inc., P. O. Box 6009, Gaithersburg, MD 20897. All Sport vectors
contain an
ampicillin resistance gene and may be transformed into E. coli strain DH10B,
also available
from Life Technologies. (See, for instance, Gruber, C. E., et al., Focus 15:59
(1993).)
Vector lafmid BA (Bento Soares, Columbia University, NY) contains an
ampicillin resistance
gene and can be transformed into E. coli strain XL-1 Blue. Vector pCR~2.1,
which is
available from Invitrogen, 1600 Faraday Avenue, Carlsbad, CA 92008, contains
an
ampicillin resistance gene and may be transformed into E. coli strain DH10B,
available from
Life Technologies. (See, for instance, Clark, Nuc. Acids Res., 16:9677-9686
(1988) and
Mead et al., BiolTechnology, 9 (1991).) Preferably, a polynucleotide of the
present invention
does not comprise the phage vector sequences identified for the particular
clone in Table 1, as
well as the corresponding plasmid vector sequences designated above.
The deposited material in the sample assigned the ATCC Deposit Number cited in
Table 1 for any given cDNA clone also may contain one or more additional
plasmids, each
comprising a cDNA clone different from that given clone. Thus, deposits
sharing the same
ATCC Deposit Number contain at least a plasmid for each cDNA clone identified
in Table 1.
Typically, each ATCC deposit sample cited in Table 1 comprises a mixture of
approximately
equal amounts (by weight) of about 50 plasmid DNAs, each containing a
different cDNA
clone; but such a deposit sample may include plasmids for more or less than 50
cDNA
clones, up to about 500 cDNA clones.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
192
Two approaches can be used to isolate a particular clone from the deposited
sample of
plasmid DNAs cited for that clone in Table 1. First, a plasmid is directly
isolated by
screening the clones using a polynucleotide probe corresponding to SEQ m NO:X.
Particularly, a specific polynucleotide with 30-40 nucleotides is synthesized
using an
Applied Biosystems DNA synthesizer according to the sequence reported. The
oligonucleotide is labeled, for instance, with 32P-y-ATP using T4
polynucleotide kinase and
purified according to routine methods. (E.g., Maniatis et al., Molecular
Cloning. A
Laboratory Manual, Cold Spring Harbor Press, Cold Spring, NY (1982).) The
plasmid
mixture is transformed into a suitable host, as indicated above (such as XL-1
Blue
(Stratagene)) using techniques known to those of skill in the art, such as
those provided by
the vector supplier or in related publications or patents cited above. The
transformants are
plated on 1.5% agar plates (containing the appropriate selection agent, e.g.,
ampicillin) to a
density of about 1 SO transformants (colonies) per plate. These plates are
screened using
Nylon membranes according to routine methods for bacterial colony screening
(e.g.,
Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Edit., (1989),
Cold Spring
Harbor Laboratory Press, pages 1.93 to 1.104), or other techniques known to
those of skill in
the art.
Alternatively, two primers of 17-20 nucleotides derived from both ends of the
SEQ >D
NO:X (i.e., within the region of SEQ >D NO:X bounded by the S' NT and the 3'
NT of the
clone defined in Table 1) are synthesized and used to amplify the desired cDNA
using the
deposited cDNA plasmid as a template. The polymerise chain reaction is carried
out under
routine conditions, for instance, in 25 p1 of reaction mixture with 0.5 ug of
the above cDNA
template. A convenient reaction mixture is 1.5-5 mM MgCl2, 0.01% (w/v)
gelatin, 20 pM
each of dATP, dCTP, dGTP, dTTP, 25 pmol of each primer and 0.25 Unit of Taq
polymerise. Thirty five cycles of PCR (denaturation at 94°C for 1 min;
annealing at 55°C for
1 min; elongation at 72°C for 1 min) are performed with a Perkin-Elmer
Cetus automated
thermal cycler. The amplified product is analyzed by agarose gel
electrophoresis and the
DNA band with expected molecular weight is excised and purified. The PCR
product is
verified to be the selected sequence by subcloning and sequencing the DNA
product.
Several methods are available for the identification of the 5' or 3' non-
coding portions
of a gene which may not be present in the deposited clone. These methods
include but are not
limited to, filter probing, clone enrichment using specific probes, and
protocols similar or


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
193
identical to 5' and 3' "RACE" protocols which are well known in the art. For
instance, a
method similar to 5' RACE is available for generating the missing 5' end of a
desired full-
length transcript. (Fromont-Racine et al., Nucleic Acids Res., 21(7):1683-1684
(1993).)
Briefly, a specific RNA oligonucleotide is ligated to the S' ends of a
population of
RNA presumably containing full-length gene RNA transcripts. A primer set
containing a
primer specific to the ligated RNA oligonucleotide and a primer specific to a
known sequence
of the gene of interest is used to PCR amplify the 5' portion of the desired
full-length gene.
This amplified product may then be sequenced and used to generate the full
length gene.
This above method starts with total RNA isolated from the desired source,
although
poly-A+ RNA can be used. The RNA preparation can then be treated with
phosphatase if
necessary to eliminate S' phosphate groups on degraded or damaged RNA which
may
interfere with the later RNA ligase step. The phosphatase should then be
inactivated and the
RNA treated with tobacco acid pyrophosphatase in order to remove the cap
structure present
at the 5' ends of messenger RNAs. This reaction leaves a 5' phosphate group at
the 5' end of
the cap cleaved RNA which can then be ligated to an RNA oligonucleotide using
T4 RNA
ligase.
This modified RNA preparation is used as a template for first strand cDNA
synthesis
using a gene specific oligonucleotide. The first strand synthesis reaction is
used as a template
for PCR amplification of the desired S' end using a primer specific to the
ligated RNA
oligonucleotide and a primer specific to the known sequence of the gene of
interest. The
resultant product is then sequenced and analyzed to confirm that the 5' end
sequence belongs
to the desired gene.
Example 2: Isolation of Genomic Clones Corresponding to a Polynucleotide
A human genomic P1 library (Genomic Systems, Inc.) is screened by PCR using
primers selected for the cDNA sequence corresponding to SEQ ID NO:X.,
according to the
method described in Example 1. (See also, Sambrook.)
Example 3: Tissue Distribution of Polypeptide
Tissue distribution of mRNA expression of polynucleotides of the present
invention is
determined using protocols for Northern blot analysis, described by, among
others, Sambrook
et al. For example, a cDNA probe produced by the method described in Example 1
is labeled


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
194
with P3z using the rediprimeTM DNA labeling system (Amersham Life Science),
according to
manufacturer's instructions. After labeling, the probe is purified using
CHROMA SPIN
100TM column (Clontech Laboratories, Inc.), according to manufacturer's
protocol number
PT1200-1. The purified labeled probe is then used to examine various human
tissues for
mRNA expression.
Multiple Tissue Northern (MTN) blots containing various human tissues (H) or
human immune system tissues (IM) (Clontech) are examined with the labeled
probe using
ExpressHybTM 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 the films developed according to
standard procedures.
Example 4: Chromosomal Mapping of the Polynucleotides
An oligonucleotide primer set is designed according to the sequence at the S'
end of
SEQ m NO:X. This primer preferably spans about 100 nucleotides. This primer
set is then
used in a polymerase chain reaction under the following set of conditions : 30
seconds, 95°C;
1 minute, 56°C; 1 minute, 70°C. This cycle is repeated 32 times
followed by one S minute
cycle at 70°C. Human, mouse, and hamster DNA is used as template in
addition to a somatic
cell hybrid panel containing individual chromosomes or chromosome fragments
(Bios, Inc).
The reactions is analyzed on either 8% polyacrylamide gels or 3.5 % agarose
gels.
Chromosome mapping is determined by the presence of an approximately 100 by
PCR
fragment in the particular somatic cell hybrid.
Example 5: Bacterial Expression of a Polypeptide
A polynucleotide encoding a polypeptide of the present invention is amplified
using
PCR oligonucleotide primers corresponding to the 5' and 3' ends of the DNA
sequence, as
outlined in Example 1, to synthesize insertion fragments. The primers used to
amplify the
cDNA insert should preferably contain restriction sites, such as BamHI and
XbaI and
initiation/stop codons, if necessary, to clone the amplified product into the
expression vector.
For example, BamHI and XbaI correspond to the restriction enzyme sites on the
bacterial
expression vector pQE-9. (Qiagen, Inc., Chatsworth, CA). This plasmid vector
encodes
antibiotic resistance (Amps, a bacterial origin of replication (ori), an IPTG-
regulatable


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
195
promoter/operator (P/0), a ribosome binding site (RBS), a 6-histidine tag (6-
His), and
restriction enzyme cloning sites.
The pQE-9 vector is digested with BamHI and XbaI and the amplified fragment is
ligated into the pQE-9 vector maintaining the reading frame initiated at the
bacterial RBS.
The ligation mixture is then used to transform the E. coli strain M15/rep4
(Qiagen, Inc.)
which contains multiple copies of the plasmid pREP4, which expresses the lacI
repressor and
also confers kanamycin resistance (Kan~. Transformants are identified by their
ability to
grow on LB plates and ampicillin/kanamycin resistant colonies are selected.
Plasmid DNA is
isolated and confirmed by restriction analysis.
Clones containing the desired constructs are grown overnight (0/N) in liquid
culture
in LB media supplemented with both Amp (100 ug/ml) and Kan (25 ug/ml). The O/N
culture
is used to inoculate a large culture at a ratio of 1:100 to 1:250. The cells
are grown to an
optical density 600 (O.D.boo) of between 0.4 and 0.6. IPTG (Isopropyl-B-D-
thiogalacto
pyranoside) is then added to a final concentration of 1 mM. IPTG induces by
inactivating the
lacI repressor, clearing the P/O leading to increased gene expression.
Cells are grown for an extra 3 to 4 hours. Cells are then harvested by
centrifugation
(20 mins at 6000Xg). The cell pellet is solubilized in the chaotropic agent 6
Molar Guanidine
HCl by stirnng for 3-4 hours at 4°C. The cell debris is removed by
centrifugation, and the
supernatant containing the polypeptide is loaded onto a nickel-nitrilo-tri-
acetic acid ("Ni-
NTA") affinity resin column (available from QIAGEN, Inc., supra). Proteins
with a 6 x His
tag bind to the Ni-NTA resin with high affinity and can be purified in a
simple one-step
procedure (for details see: The QIAexpressionist (1995) QIAGEN, Inc., supra).
Briefly, the supernatant is loaded onto the column in 6 M guanidine-HCI, pH 8,
the
column is first washed with 10 volumes of 6 M guanidine-HCI, pH 8, then washed
with
10 volumes of 6 M guanidine-HCl pH 6, and finally the polypeptide is eluted
with 6 M
guanidine-HCI, pH 5.
The purified protein is then renatured by dialyzing it against phosphate-
buffered
saline (PBS) or 50 mM Na-acetate, pH 6 buffer plus 200 mM NaCI. Alternatively,
the
protein can be successfully refolded while immobilized on the Ni-NTA column.
The
recommended conditions are as follows: renature using a linear 6M-1M urea
gradient in S00
mM NaCI, 20% glycerol, 20 mM Tris/HCl pH 7.4, containing protease inhibitors.
The


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
196
renaturation should be performed over a period of 1.5 hours or more. After
renaturation the
proteins are eluted by the addition of 250 mM immidazole. Immidazole is
removed by a final
dialyzing step against PBS or 50 mM sodium acetate pH 6 buffer plus 200 mM
NaCI. The
purified protein is stored at 4° C or frozen at -80° C.
In addition to the above expression vector, the present invention further
includes an
expression vector comprising phage operator and promoter elements operatively
linked to a
polynucleotide of the present invention, called pHE4a. (ATCC Accession Number
209645,
deposited on February 25, 1998.) This vector contains: 1) a
neomycinphosphotransferase
gene as a selection marker, 2) an E. coli origin of replication, 3) a T5 phage
promoter
sequence, 4) two lac operator sequences, 5) a Shine-Delgarno sequence, and 6)
the lactose
operon repressor gene (lacIq). The origin of replication (oriC) is derived
from pUCl9 (LTI,
Gaithersburg, MD). The promoter sequence and operator sequences are made
synthetically.
DNA can be inserted into the pHEa by restricting the vector with NdeI and
XbaI,
BamHI, XhoI, or Asp718, running the restricted product on a gel, and isolating
the larger
fragment (the stuffer fragment should be about 310 base pairs). The DNA insert
is generated
according to the PCR protocol described in Example 1, using PCR primers having
restriction
sites for NdeI (5' primer) and XbaI, BamHI, XhoI, or Asp718 (3' primer). The
PCR insert is
gel purified and restricted with compatible enzymes. The insert and vector are
ligated
according to standard protocols.
The engineered vector could easily be substituted in the above protocol to
express
protein in a bacterial system.
Example 6: Purification of a Polypeptide from an Inclusion Body
The following alternative method can be used to purify a polypeptide expressed
in E
coli when it is present in the form of inclusion bodies. Unless otherwise
specified, all of the
following steps are conducted at 4-10°C.
Upon completion of the production phase of the E. coli fermentation, the cell
culture
is cooled to 4-10°C and the cells harvested by continuous
centrifugation at 15,000 rpm
(Heraeus Sepatech). On the basis of the expected yield of protein per unit
weight of cell
paste and the amount of purified protein required, an appropriate amount of
cell paste, by
weight, is suspended in a buffer solution containing 100 mM Tris, 50 mM EDTA,
pH 7.4.
The cells are dispersed to a homogeneous suspension using a high shear mixer.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
197
The cells are then lysed by passing the solution through a microfluidizer
(Microfuidics, Corp. or APV Gaulin, Inc.) twice at 4000-6000 psi. The
homogenate is then
mixed with NaCI solution to a final concentration of 0.5 M NaCI, followed by
centrifugation
at 7000 xg for 15 min. The resultant pellet is washed again using O.SM NaCI,
100 mM Tris,
50 mM EDTA, pH 7.4.
The resulting washed inclusion bodies are solubilized with 1.5 M guanidine
hydrochloride (GuHCI) for 2-4 hours. After 7000 xg centrifugation for 15 min.,
the pellet is
discarded and the polypeptide containing supernatant is incubated at
4°C overnight to allow
further GuHCI extraction.
Following high speed centrifugation (30,000 xg) to remove insoluble particles,
the
GuHCI solubilized protein is refolded by quickly mixing the GuHCI extract with
20 volumes
of buffer containing 50 mM sodium, pH 4.5, 150 mM NaCI, 2 mM EDTA by vigorous
stirnng. The refolded diluted protein solution is kept at 4°C without
mixing for 12 hours
prior to further purification steps.
1 S To clarify the refolded polypeptide solution, a previously prepared
tangential filtration
unit equipped with 0.16 pm membrane filter with appropriate surface area
(e.g., Filtron),
equilibrated with 40 mM sodium acetate, pH 6.0 is employed. The filtered
sample is loaded
onto a cation exchange resin (e.g., Poros HS-S0, Perseptive Biosystems). The
column is
washed with 40 mM sodium acetate, pH 6.0 and eluted with 250 mM, S00 mM, 1000
mM,
and 1500 mM NaCI in the same buffer, in a stepwise manner. The absorbance at
280 nm of
the effluent is continuously monitored. Fractions are collected and further
analyzed by SDS-
PAGE.
Fractions containing the polypeptide are then pooled and mixed with 4 volumes
of
water. The diluted sample is then loaded onto a previously prepared set of
tandem columns
of strong anion (Poros HQ-50, Perseptive Biosystems) and weak anion (Poros CM-
20,
Perseptive Biosystems) exchange resins. The columns are equilibrated with 40
mM sodium
acetate, pH 6Ø Both columns are washed with 40 mM sodium acetate, pH 6.0,
200 mM
NaCI. The CM-20 column is then eluted using a 10 column volume linear gradient
ranging
from 0.2 M NaCI, 50 mM sodium acetate, pH 6.0 to 1.0 M NaCI, 50 mM sodium
acetate, pH
6.5. Fractions are collected under constant AZ$o monitoring of the effluent.
Fractions
containing the polypeptide (determined, for instance, by 16% SDS-PAGE) are
then pooled.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
198
The resultant polypeptide should exhibit greater than 95% purity after the
above
refolding and purification steps. No major contaminant bands should be
observed from
Commassie blue stained 16% SDS-PAGE gel when 5 p.g of purified protein is
loaded. The
purified protein can also be tested for endotoxin/LPS contamination, and
typically the LPS
content is less than 0.1 ng/ml according to LAL assays.
Example 7: Cloning and Expression of a Polypeptide in a Baculovirus Expression
System
In this example, the plasmid shuttle vector pA2 is used to insert a
polynucleotide into
a baculovirus to express a polypeptide. This expression vector contains the
strong polyhedrin
promoter of the Autographa californica nuclear polyhedrosis virus (AcMNPV)
followed by
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 a viable virus that express the cloned polynucleotide.
Many other baculovirus vectors can be 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).
Specifically, the cDNA sequence contained in the deposited clone is amplified
using
the PCR protocol described in Example 1 using primers with appropriate
restriction sites and
initiation/stop codons. If the naturally occurring signal sequence is used to
produce the
secreted protein, the pA2 vector does not need a second signal peptide.
Alternatively, the
vector can be modified (pA2 GP) to include a baculovirus leader sequence,
using the
standard methods 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).


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
199
The amplified fragment is isolated from a 1 % agarose gel using a commercially
available kit ("Geneclean," BIO 101 Inc., La Jolla, Ca.). The fragment then is
digested with
appropriate restriction enzymes and again purified on a 1 % agarose gel.
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.).
The fragment and the dephosphorylated plasmid 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 containing the plasmid are identified by digesting DNA from
individual
colonies and analyzing the digestion product by gel electrophoresis. The
sequence of the
cloned fragment is confirmed by DNA sequencing.
Five ~g of a plasmid containing the polynucleotide is co-transfected with 1.0
~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). One pg of BaculoGoldTM virus DNA and
5 pg
of the plasmid are mixed in a sterile well of a microtiter plate containing 50
p1 of serum-free
Grace's medium (Life Technologies Inc., Gaithersburg, MD). Afterwards, 10 p1
Lipofectin
plus 90 p1 Grace's medium are added, mixed and incubated for 15 minutes at
room
temperature. Then the transfection mixture is added drop-wise to Sf~7 insect
cells (ATCC
CRL 1711) seeded in a 35 mm tissue culture plate with 1 ml Grace's medium
without serum.
The plate is then incubated for 5 hours at 27° C. The transfection
solution is then removed
from the plate and 1 ml of Grace's insect medium supplemented with 10% fetal
calf serum is
added. Cultivation is then continued at 27° C for four days.
After four days the supernatant is collected and a plaque assay is performed,
as
described by Summers and Smith, supra. An agarose gel with "Blue Gal" (Life
Technologies
Inc., Gaithersburg) 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., Gaithersburg, page 9-10.) After appropriate
incubation, blue stained
plaques are picked with the tip of a micropipettor (e.g., Eppendorf). The agar
containing the


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
200
recombinant viruses is then resuspended in a microcentrifuge tube containing
200 ~1 of
Grace's medium and the suspension containing the recombinant baculovirus is
used to infect
Sf~ 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.
To verify the expression of the polypeptide, Sf~ cells are grown in Grace's
medium
supplemented with 10% heat-inactivated FBS. The cells are infected with the
recombinant
baculovirus containing the polynucleotide at a multiplicity of infection
("MOI") of about 2.
If radiolabeled proteins are desired, 6 hours later the medium is removed and
is replaced with
SF900 II medium minus methionine and cysteine (available from Life
Technologies Inc.,
Rockville, MD). After 42 hours, 5 ~Ci of 35S-methionine and 5 ~Ci 35S-cysteine
(available
from Amersham) are added. The cells are further incubated for 16 hours and
then are
harvested by centrifugation. The proteins in the supernatant as well as the
intracellular
proteins are analyzed by SDS-PAGE followed by autoradiography (if
radiolabeled).
Microsequencing of the amino acid sequence of the amino terminus of purified
protein may be used to determine the amino terminal sequence of the produced
protein.
Example 8: Expression of a Polyneptide in Mammalian Cells
The polypeptide of the present invention can be expressed in a mammalian cell.
A
typical mammalian expression vector contains a promoter element, which
mediates the
initiation of transcription of mRNA, a protein 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 is 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 pSVL and pMSG (Pharmacia, Uppsala, Sweden),
pRSVcat
(ATCC 37152), pSV2dhfr (ATCC 37146), pBCI2MI (ATCC 67109), pCMVSport 2.0, and
pCMVSport 3Ø 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 CV1, quail QC1-3
cells, mouse
L cells and Chinese hamster ovary (CHO) cells.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
201
Alternatively, the polypeptide can be expressed in stable cell lines
containing the
polynucleotide integrated into a chromosome. The co-transfection with a
selectable marker
such as dhfr, gpt, neomycin, hygromycin allows the identification and
isolation of the
transfected cells.
S The transfected gene can also be amplified to express large amounts of the
encoded
protein. The DHFR (dihydrofolate reductase) marker is useful in developing
cell lines that
carry several hundred or even several thousand copies of the gene of interest.
(See, e.g., Alt et
al., J. Biol. Chem., 253:1357-1370 (1978); Hamlin et al., Biochem. et Biophys.
Acta,
1097:107-143 (1990); Page et al., Biotechnology, 9:64-68 (1991)). Another
useful selection
marker is the enzyme glutamine synthase (GS) (Murphy et al., Biochem J.,
227:277-279
(1991); Bebbington et al., BiolTechnology, 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 genes) integrated into a
chromosome.
Chinese hamster ovary (CHO) and NSO cells are often used for the production of
proteins.
Derivatives of the plasmid pSV2-dhfr (ATCC Accession No.: 37146), the
expression
vectors pC4 (ATCC Accession No.: 209646) and pC6 (ATCC Accession No.:209647)
contain the strong promoter (LTR) of the Rous Sarcoma Virus (Cullen et al.,
Molecular and
Cellular Biology, 438-447 (March, 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 also contain the 3' intron, the polyadenylation and termination signal
of the rat
preproinsulin gene, and the mouse DHFR gene under control of the SV40 early
promoter.
Specifically, the plasmid pC6, for example, is digested with appropriate
restriction
enzymes and then dephosphorylated using calf intestinal phosphates by
procedures known in
the art. The vector is then isolated from a 1% agarose gel.
A polynucleotide of the present invention is amplified according to the
protocol
outlined in Example 1 using primers with appropriate restrictions sites and
initiation/stop
codons, if necessary. The vector can be modified to include a heterologous
signal sequence if
necessary for secretion. (See, e.g., WO 96/34891.)
The amplified fragment is isolated from a 1 % agarose gel using a commercially
available kit ("Geneclean," BIO 101 Inc., La Jolla, Ca.). The fragment then is
digested with
appropriate restriction enzymes and again purified on a 1% agarose gel.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
202
The amplified fragment is then digested with the same restriction enzyme and
purified
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 pC6 using, for
instance, restriction
enzyme analysis.
Chinese hamster ovary cells lacking an active DHFR gene is used for
transfection.
Five pg of the expression plasmid pC6 is cotransfected with 0.5 pg of the
plasmid pSVneo
using lipofectin (Felgner et al., supra). The plasmid pSV2-neo contains a
dominant
selectable marker, the neo gene from Tn5 ~ encoding an enzyme that confers
resistance to a
group of antibiotics including 6418. The cells are seeded in alpha minus MEM
supplemented with 1 mg/ml 6418. 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 metothrexate plus 1 mg/ml 6418. 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 pM, 2 pM, 5 pM, 10
mM, 20 mM).
The same procedure is repeated until clones are obtained which grow at a
concentration of
100 - 200 p.M. Expression of the desired gene product is analyzed, for
instance, by SDS
PAGE and Western blot or by reversed phase HPLC analysis.
Example 9: Protein Fusions
The polypeptides of the present invention are preferably fused to other
proteins.
These fusion proteins can be used for a variety of applications. For example,
fusion of the
present polypeptides to His-tag, HA-tag, protein A, IgG domains, and maltose
binding
protein facilitates purification. (See Example 5; see also EP A 394,827;
Traunecker, et al.,
Nature, 331:84-86 (1988)) The polypeptides can also be fused to heterologous
polypeptide
sequences to facilitate secretion and intracellular trafficking (e.g., KDEL).
Moreover, fusion
to IgG-1, IgG-3, and albumin increases the halflife time in vivo. Nuclear
localization signals
fused to the polypeptides of the present invention can target the protein to a
specific
subcellular localization, while covalent heterodimer or homodimers can
increase or decrease
the activity of a fusion protein. Fusion proteins can also create chimeric
molecules having


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
203
more than one function. Finally, fusion proteins can increase solubility
and/or stability of the
fused protein compared to the non-fused protein. All of the types of fusion
proteins described
above can be made by modifying the following protocol, which outlines the
fusion of a
polypeptide to an IgG molecule, or the protocol described in Example 5.
Briefly, the human Fc portion of the IgG molecule can be PCR amplified, using
primers that span the 5' and 3' ends of the sequence described below. These
primers also
should have convenient restriction enzyme sites that will facilitate cloning
into an expression
vector, preferably a mammalian expression vector, and initiation/stop codons,
if necessary.
For example, if pC4 (Accession No.: 209646) is used, the human Fc portion can
be
ligated into the BamHI cloning site. Note that the 3' BamHI site should be
destroyed. Next,
the vector containing the human Fc portion is re-restricted with BamHI,
linearizing the
vector, and a polynucleotide of the present invention, isolated by the PCR
protocol described
in Example 1, is ligated into this BamHI site. Note that the polynucleotide is
cloned without
a stop codon, otherwise a fusion protein will not be produced.
If the naturally occurring signal sequence is used to produce the secreted
protein, pC4
does not need a second signal peptide. Alternatively, if the naturally
occurring signal
sequence is not used, the vector can be modified to include a heterologous
signal sequence.
(See, e.g., WO 96/34891.)
Human I~G Fc re ig on:
GGGATCCGGAGCCCAAATCTTCTGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAATTCG
AGGGTGCACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACTC
CTGAGGTCACATGCGTGGTGGTGGACGTAAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTAC
GTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGT
ACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGC
AAGGTCTCCAACAAAGCCCTCCCAACCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCC
CCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGATGAGCTGACCAAGAACCAGGTCAGCC
TGACCTGCCTGGTCAAAGGCTTCTATCCAAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAG
CCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCTACAGC
AAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGA
GGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAATGAGTGCGACGGC
CGCGACTCTAGAGGAT (SEQ ID NO:1)
Example 10: Formulating a Polypeptide


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
204
The polypeptide composition will be formulated and dosed in a fashion
consistent
with good medical practice, taking into account the clinical condition of the
individual patient
(especially the side effects of treatment with the secreted polypeptide
alone), the site of
delivery, the method of administration, the scheduling of administration, and
other factors
known to practitioners. The "effective amount" for purposes herein is thus
determined by
such considerations.
As a general proposition, the total pharmaceutically effective amount of
polypeptide
administered parenterally per dose will be in the range of about 1 ~g/kg/day
to 10 mg/kg/day
of patient body weight, although, as noted above, this will be subject to
therapeutic
discretion. More preferably, this dose is at least 0.01 mg/kg/day, and most
preferably for
humans between about 0.01 and 1 mg/kg/day for the hormone. If given
continuously, the
polypeptide is typically administered at a dose rate of about 1 ~g/kg/hour to
about 50
~g/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 the polypeptide of the invention are
administered orally, rectally, parenterally, intracistemally, intravaginally,
intraperitoneally,
topically (as by powders, ointments, gels, drops or transdermal patch),
bucally, or as an oral
or nasal spray. "Pharmaceutically acceptable carrier" refers to a non-toxic
solid, semisolid or
liquid filler, diluent, encapsulating material or formulation auxiliary of any
type. The term
"parenteral" as used herein refers to modes of administration which include
intravenous,
intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular
injection and
infusion.
The polypeptide 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 mirocapsules. 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 et al., Biopolymers, 22:547-556 (1983)), poly
(2-
hydroxyethyl methacrylate) (Langer et al., J. Biomed. Mater. Res. 15:167-277
(1981), and
Langer, Chem. Tech., 12:98-105 (1982)), ethylene vinyl acetate (R. Langer et
al.) or poly-D-
(-)-3-hydroxybutyric acid (EP 133,988). Sustained-release compositions also
include


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
205
liposomally entrapped polypeptides. Liposomes containing the secreted
polypeptide 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; EP 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
secreted polypeptide therapy.
For parenteral administration, in one embodiment, the polypeptide 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
Garner, 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 the polypeptide
uniformly and
intimately with liquid Garners or finely divided solid carriers or both. Then,
if necessary, the
product is shaped into the desired formulation. Preferably the Garner is a
parenteral carrier,
more preferably a solution that is isotonic with the blood of the recipient.
Examples of such
Garner 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.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
206
The polypeptide 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 polypeptide salts.
S Any polypeptide to be used for therapeutic administration can be sterile.
Sterility is
readily accomplished by filtration through sterile filtration membranes (e.g.,
0.2 micron
membranes). Therapeutic polypeptide 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.
Polypeptides 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 S ml
of sterile-filtered 1 % (w/v) aqueous polypeptide solution, 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 containers) 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, the polypeptides of the present invention may be
employed in
conjunction with other therapeutic compounds.
Example 11: Method of Treating Decreased Levels of the Polypeptide
It will be appreciated that conditions caused by a decrease in the standard or
normal
expression level of a polypeptide in an individual can be treated by
administering the
polypeptide of the present invention, preferably in the secreted and/or
soluble form. Thus,
the invention also provides a method of treatment of an individual in need of
an increased
level of the polypeptide comprising administering to such an individual a
pharmaceutical
composition comprising an amount of the polypeptide to increase the activity
level of the
polypeptide in such an individual.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
207
For example, a patient with decreased levels of a polypeptide receives a daily
dose
0.1-100 ug/kg of the polypeptide for six consecutive days. Preferably, the
polypeptide is in
the secreted form. The exact details of the dosing scheme, based on
administration and
formulation, are provided in Example 10.
Example 12: Method of Treating Increased Levels of the Polypeptide
Antisense technology is used to inhibit production of a polypeptide of the
present
invention. This technology is one example of a method of decreasing levels of
a polypeptide,
preferably a secreted form, due to a variety of etiologies, such as cancer.
For example, a patient diagnosed with abnormally increased levels of a
polypeptide is
administered intravenously antisense polynucleotides at 0.5, 1.0, 1.5, 2.0 and
3.0 mg/kg day
for 21 days. This treatment is repeated after a 7-day rest period if the
treatment was well
tolerated. The formulation of the antisense polynucleotide is provided in
Example 10.
Example 13: Method of Treatment Using Gene Therapy - Ex Vivo
One method of gene therapy transplants fibroblasts, which are capable of
expressing a
polypeptide, onto a patient. Generally, fibroblasts are obtained from a
subject by skin biopsy.
The resulting tissue is placed in tissue-culture medium and separated into
small pieces. Small
chunks of the tissue are placed on a wet surface of a tissue culture flask,
approximately ten
pieces are placed in each flask. The flask is turned upside down, closed tight
and left at room
temperature over night. After 24 hours at room temperature, the flask is
inverted and the
chunks of tissue remain fixed to the bottom of the flask and fresh media
(e.g., Ham's F12
media, with 10% FBS, penicillin and streptomycin) is added. The flasks are
then incubated at
37°C for approximately one week.
At this time, fresh media is added and subsequently changed every several
days.
After an additional two weeks in culture, a monolayer of fibroblasts emerge.
The monolayer
is trypsinized and scaled into larger flasks.
pMV-7 (Kirschmeier, P.T. et al., DNA, 7:219-25 (1988)), flanked by the long
terminal repeats of the Moloney murine sarcoma virus, is digested with EcoRI
and HindIII
and subsequently treated with calf intestinal phosphatase. The linear vector
is fractionated on
agarose gel and purified, using glass beads.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
208
The cDNA encoding a polypeptide of the present invention can be amplified
using
PCR primers which correspond to the 5' and 3' end sequences respectively as
set forth in
Example 1 using primers and having appropriate restriction sites and
initiation/stop codons, if
necessary. Preferably, the 5' primer contains an EcoRI site and the 3' primer
includes a
HindIII site. Equal quantities of the Moloney murine sarcoma virus linear
backbone and the
amplified EcoRI and HindIII fragment are added together, in the presence of T4
DNA ligase.
The resulting mixture is maintained under conditions appropriate for ligation
of the two
fragments. The ligation mixture is then used to transform bacteria HB101,
which are then
plated onto agar containing kanamycin for the purpose of confirming that the
vector has the
gene of interest properly inserted.
The amphotropic pA317 or GP+aml2 packaging cells are grown in tissue culture
to
confluent density in Dulbecco's Modified Eagles Medium (DMEM) with 10% calf
serum
(CS), penicillin and streptomycin. The MSV vector containing the gene is then
added to the
media and the packaging cells transduced with the vector. The packaging cells
now produce
infectious viral particles containing the gene (the packaging cells are now
referred to as
producer cells).
Fresh media is added to the transduced producer cells, and subsequently, the
media is
harvested from a 10 cm plate of confluent producer cells. The spent media,
containing the
infectious viral particles, is filtered through a millipore filter to remove
detached producer
cells and this media is then used to infect fibroblast cells. Media is removed
from a sub-
confluent plate of fibroblasts and quickly replaced with the media from the
producer cells.
This media is removed and replaced with fresh media. If the titer of virus is
high, then
virtually all fibroblasts will be infected and no selection is required. If
the titer is very low,
then it is necessary to use a retroviral vector that has a selectable marker,
such as neo or his.
Once the fibroblasts have been efficiently infected, the fibroblasts are
analyzed to determine
whether protein is produced.
The engineered fibroblasts are then transplanted onto the host, either alone
or after
having been grown to confluence on cytodex 3 microcarrier beads.
Example 14: Gene Therapy Using Endogenous steroid hormone receptor Genes
Another method of gene therapy according to the present invention involves
operably
associating the endogenous steroid hormone receptor gene sequence with a
promoter via


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
209
homologous recombination as described, for example, in U.S. Patent NO:
5,641,670, issued
June 24, 1997; International Publication NO: WO 96/2941 l, published September
26, 1996;
International Publication NO: WO 94/12650, published August 4, 1994; Koller et
al., Proc.
Natl. Acad. Sci. USA, 86:8932-8935 (1989); and Zijlstra et al., Nature,
342:435-438 (1989).
This method involves the activation of a gene which is present in the target
cells, but which is
not expressed in the cells, or is expressed at a lower level than desired.
Polynucleotide constructs are made which contain a promoter and targeting
sequences, which are homologous to the 5' non-coding sequence of the
endogenous steroid
hormone receptor gene, flanking the promoter. The targeting sequence will be
sufficiently
near the 5' end of steroid hormone receptor gene so the promoter will be
operably linked to
the endogenous sequence upon homologous recombination. The promoter and the
targeting
sequences can be amplified using PCR. Preferably, the amplified promoter
contains distinct
restriction enzyme sites on the 5' and 3' ends. Preferably, the 3' end of the
first targeting
sequence contains the same restriction enzyme site as the 5' end of the
amplified promoter
and the 5' end of the second targeting sequence contains the same restriction
site as the 3' end
of the amplified promoter.
The amplified promoter and the amplified targeting sequences are digested with
the
appropriate restriction enzymes and subsequently treated with calf intestinal
phosphatase.
The digested promoter and digested targeting sequences are added together in
the presence of
T4 DNA ligase. The resulting mixture is maintained under conditions
appropriate for ligation
of the two fragments. The construct is size fractionated on an agarose gel
then purified by
phenol extraction and ethanol precipitation.
In this Example, the polynucleotide constructs are administered as naked
polynucleotides via electroporation. However, the polynucleotide constructs
may also be
administered with transfection-facilitating agents, such as liposomes, viral
sequences, viral
particles, precipitating agents, etc. Such methods of delivery are known in
the art.
Once the cells are transfected, homologous recombination will take place which
results in the promoter being operably linked to the endogenous steroid
hormone receptor
gene sequence. This results in the expression of steroid hormone receptor in
the cell.
Expression may be detected by immunological staining, or any other method
known in the
art.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
210
Fibroblasts are obtained from a subject by skin biopsy. The resulting tissue
is placed
in DMEM + 10% fetal calf serum. Exponentially growing or early stationary
phase
fibroblasts are trypsinized and rinsed from the plastic surface with nutrient
medium. An
aliquot of the cell suspension is removed for counting, and the remaining
cells are subjected
S to centrifugation. The supernatant is aspirated and the pellet is
resuspended in S ml of
electroporation buffer (20 mM HEPES pH 7.3, 137 mM NaCI, 5 mM KCI, 0.7 mM Na2
HPOa, 6 mM dextrose). The cells are recentrifuged, the supernatant aspirated,
and the cells
resuspended in electroporation buffer containing 1 mg/ml acetylated bovine
serum albumin.
The final cell suspension contains approximately 3X106 cells/ml.
Electroporation should be
performed immediately following resuspension.
Plasmid DNA is prepared according to standard techniques. For example, to
construct a plasmid for targeting to the steroid hormone receptor locus,
plasmid pUC 18 (MBI
Fermentas, Amherst, NY) is digested with HindIII. The CMV promoter is
amplified by PCR
with an XbaI site on the 5' end and a BamHI site on the 3'end. Two steroid
hormone receptor
1 S non-coding gene sequences are amplified via PCR: one steroid hormone
receptor non-coding
sequence (steroid hormone receptor fragment 1 ) is amplified with a HindIII
site at the S' end
and an Xba site at the 3'end; the other steroid hormone receptor non-coding
sequence (steroid
hormone receptor fragment 2) is amplified with a BamHI site at the 5'end and a
HindIII site
at the 3'end. The CMV promoter and steroid hormone receptor fragments are
digested with
the appropriate enzymes (CMV promoter - XbaI and BamHI; steroid hormone
receptor
fragment 1 - XbaI; steroid hormone receptor fragment 2 - BamHI) and ligated
together. The
resulting ligation product is digested with HindIII, and ligated with the
HindIII-digested
pUCl8 plasmid.
Plasmid DNA is added to a sterile cuvette with a 0.4 cm electrode gap (Bio-
Rad). The
final DNA concentration is generally at least 120 pg/ml. 0.5 ml of the cell
suspension
(containing approximately 1.5.X106 cells) is then added to the cuvette, and
the cell
suspension and DNA solutions are gently mixed. Electroporation is performed
with a
Gene-Pulser apparatus (Bio-Rad). Capacitance and voltage are set at 960 pF and
250-300 V,
respectively. As voltage increases, cell survival decreases, but the
percentage of surviving
cells that stably incorporate the introduced DNA into their genome increases
dramatically.
Given these parameters, a pulse time of approximately 14-20 mSec should be
observed.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
211
Electroporated cells are maintained at room temperature for approximately 5
min, and
the contents of the cuvette are then gently removed with a sterile transfer
pipette. The cells
are added directly to 10 ml of prewarmed nutrient media (DMEM with 15% calf
serum) in a
cm dish and incubated at 37 degree C. The following day, the media is
aspirated and
5 replaced with 10 ml of fresh media and incubated for a further 16-24 hours.
The engineered fibroblasts are then injected into the host, either alone or
after having
been grown to confluence on cytodex 3 microcarrier beads. The fibroblasts now
produce the
protein product. The fibroblasts can then be introduced into a patient as
described above.
10 Example 15: Method of Treatment Using Gene Therapy - In Vivo
Another aspect of the present invention is using in vivo gene therapy methods
to treat
disorders, diseases and conditions. The gene therapy method relates to the
introduction of
naked nucleic acid (DNA, RNA, and antisense DNA or RNA) steroid hormone
receptor
sequences into an animal to increase or decrease the expression of the steroid
hormone
receptor polypeptide. The steroid hormone receptor polynucleotide may be
operatively
linked to a promoter or any other genetic elements necessary for the
expression of the steroid
hormone receptor polypeptide by the target tissue. Such gene therapy and
delivery techniques
and methods are known in the art, see, for example, W090/11092, W098/11779;
U.S. Patent
NO: 5693622, 5705151, 5580859; Tabata et al., Cardiovasc. Res. 35(3):470-479
(1997),
Chao J et al., Pharmacol. Res., 35(6):517-522 (1997), Wolff, Neuromuscul.
Disord. 7(5):314-
318 (1997), Schwartz et al., Gene Ther., 3(5):405-411 (1996), Tsurumi Y. et
al., Circulation,
94(12):3281-3290 (1996) (incorporated herein by reference).
The steroid hormone receptor polynucleotide constructs may be delivered by any
method that delivers injectable materials to the cells of an animal, such as,
injection into the
interstitial space of tissues (heart, muscle, skin, lung, liver, intestine and
the like). The steroid
hormone receptor polynucleotide constructs can be delivered in a
pharmaceutically
acceptable liquid or aqueous carrier.
The term "naked" polynucleotide, DNA or RNA, refers to sequences that are free
from any delivery vehicle that acts to assist, promote, or facilitate entry
into the cell,
including viral sequences, viral particles, liposome formulations, lipofectin
or precipitating
agents and the like. However, the steroid hormone receptor polynucleotides may
also be
delivered in liposome formulations (such as those taught in Felgner et al.,
Ann. NYAcad. Sci.,


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
212
772:126-139 (1995) and Abdallah et al., Biol. Cell , 85(1):1-7 (1995)) which
can be prepared
by methods well known to those skilled in the art.
The steroid hormone receptor polynucleotide vector constructs used in the gene
therapy method are preferably constructs that will not integrate into the host
genome nor will
they contain sequences that allow for replication. Any strong promoter known
to those
skilled in the art can be used for driving the expression of DNA. Unlike other
gene therapies
techniques, one major advantage of introducing naked nucleic acid sequences
into target cells
is the transitory nature of the polynucleotide synthesis in the cells. Studies
have shown that
non-replicating DNA sequences can be introduced into cells to provide
production of the
desired polypeptide for periods of up to six months.
The polynucleotide constructs can be delivered to the interstitial space of
tissues
within the an animal, including of muscle, skin, brain, lung, liver, spleen,
bone marrow,
thymus, heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder,
stomach,
intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, and
connective tissue.
Interstitial space of the tissues comprises the intercellular fluid,
mucopolysaccharide matrix
among the reticular fibers of organ tissues, elastic fibers in the walls of
vessels or chambers,
collagen fibers of fibrous tissues, or that same matrix within connective
tissue ensheathing
muscle cells or in the lacunae of bone. It is similarly the space occupied by
the plasma of the
circulation and the lymph fluid of the lymphatic channels. Delivery to the
interstitial space of
muscle tissue is preferred for the reasons discussed below. They may be
conveniently
delivered by injection into the tissues comprising these cells. They are
preferably delivered
to and expressed in persistent, non-dividing cells which are differentiated,
although delivery
and expression may be achieved in non-differentiated or less completely
differentiated cells,
such as, for example, stem cells of blood or skin fibroblasts. In vivo muscle
cells are
particularly competent in their ability to take up and express
polynucleotides.
For the naked steroid hormone receptor polynucleotide injection, an effective
dosage
amount of DNA or RNA will be in the range of from about 0.05 g/kg body weight
to about
50 mg/kg body weight. Preferably the dosage will be from about 0.005 mg/kg to
about 20
mg/kg and more preferably from about 0.05 mg/kg to about 5 mg/kg. Of course,
as the
artisan of ordinary skill will appreciate, this dosage will vary according to
the tissue site of
injection. The appropriate and effective dosage of nucleic acid sequence can
readily be
determined by those of ordinary skill in the art and may depend on the
condition being treated


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
213
and the route of administration. The preferred route of administration is by
the parenteral
route of injection into the interstitial space of tissues. However, other
parenteral routes may
also be used, such as, inhalation of an aerosol formulation particularly for
delivery to lungs or
bronchial tissues, throat or mucous membranes of the nose. In addition, naked
steroid
hormone receptor polynucleotide constructs can be delivered to arteries during
angioplasty by
the catheter used in the procedure.
The dose response effects of injected steroid hormone receptor polynucleotide
in
muscle in vivo is determined as follows. Suitable steroid hormone receptor
template DNA
for production of mRNA coding for steroid hormone receptor polypeptide is
prepared in
accordance with a standard recombinant DNA methodology. The template DNA,
which may
be either circular or linear, is either used as naked DNA or complexed with
liposomes. The
quadriceps muscles of mice are then injected with various amounts of the
template DNA.
Five to six week old female and male Balb/C mice are anesthetized by
intraperitoneal
injection with 0.3 ml of 2.5% Avertin. A 1.5 cm incision is made on the
anterior thigh, and
the quadriceps muscle is directly visualized. The steroid hormone receptor
template DNA is
injected in 0.1 ml of carrier in a 1 cc syringe through a 27 gauge needle over
one minute,
approximately 0.5 cm from the distal insertion site of the muscle into the
knee and about 0.2
cm deep. A suture is placed over the injection site for future localization,
and the skin is
closed with stainless steel clips.
After an appropriate incubation time (e.g., 7 days) muscle extracts are
prepared by
excising the entire quadriceps. Every fifth 15 um cross-section of the
individual quadriceps
muscles is histochemically stained for steroid hormone receptor protein
expression. A time
course for steroid hormone receptor protein expression may be done in a
similar fashion
except that quadriceps from different mice are harvested at different times.
Persistence of
steroid hormone receptor DNA in muscle following injection may be determined
by Southern
blot analysis after preparing total cellular DNA and HIRT supernatants from
injected and
control mice. The results of the above experimentation in mice can be use to
extrapolate
proper dosages and other treatment parameters in humans and other animals
using steroid
hormone receptor naked DNA.
Example 16: Production of an Antibody
a) Hybridoma Technology


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
214
The antibodies of the present invention can be prepared by a variety of
methods.
(See, Current Protocols, Chapter 2.) As one example of such methods, cells
expressing
steroid hormone receptor polypeptide(s) are administered to an animal to
induce the
production of sera containing polyclonal antibodies. In a preferred method, a
preparation of
steroid hormone receptor polypeptide(s) is prepared and purified to render it
substantially free
of natural contaminants. Such a preparation is then introduced into an animal
in order to
produce polyclonal antisera of greater specific activity.
Monoclonal antibodies specific for steroid hormone receptor polypeptide(s) are
prepared using hybridoma technology. (Kohler et al., Nature 256:495 (1975);
Kohler et al.,
Eur. J. Immunol. 6:511 (1976); Kohler et al., Eur. J. Immunol. 6:292 (1976);
Hammerling et
al., in: Monoclonal Antibodies and T-Cell Hybridomas, Elsevier, N.Y., pp. 563-
681 (1981)).
In general, an animal (preferably a mouse) is immunized with steroid hormone
receptor
polypeptide(s) or, more preferably, with a secreted steroid hormone receptor
polypeptide-
expressing cell. Such polypeptide-expressing cells are cultured in any
suitable tissue culture
medium, preferably in Earle's modified Eagle's medium supplemented with 10%
fetal bovine
serum (inactivated at about 56°C), and supplemented with about 10 g/1
of nonessential amino
acids, about 1,000 U/ml of penicillin, and about 100 ~ g/ml of streptomycin.
The splenocytes of such mice are extracted and fused with a suitable myeloma
cell
line. Any suitable myeloma cell line may be employed in accordance with the
present
invention; however, it is preferable to employ the parent myeloma cell line
(SP20), available
from the ATCC. After fusion, the resulting hybridoma cells are selectively
maintained in
HAT medium, and then cloned by limiting dilution as described by Wands et al.
(Gastroenterology 80:225-232 (1981)). The hybridoma cells obtained through
such a
selection are then assayed to identify clones which secrete antibodies capable
of binding the
steroid hormone receptor polypeptide(s).
Alternatively, additional antibodies capable of binding to steroid hormone
receptor
polypeptide(s) can be produced in a two-step procedure using anti-idiotypic
antibodies. Such
a method makes use of the fact that antibodies are themselves antigens, and
therefore, it is
possible to obtain an antibody which binds to a second antibody. In accordance
with this
method, protein specific antibodies are used to immunize an animal, preferably
a mouse. The
splenocytes of such an animal are then used to produce hybridoma cells, and
the hybridoma
cells are screened to identify clones which produce an antibody whose ability
to bind to the


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
215
steroid hormone receptor protein-specific antibody can be blocked by steroid
hormone
receptor polypeptide(s). Such antibodies comprise anti-idiotypic antibodies to
the steroid
hormone receptor protein-specific antibody and are used to immunize an animal
to induce
formation of further steroid hormone receptor protein-specific antibodies.
For in vivo use of antibodies in humans, an antibody is "humanized". Such
antibodies
can be produced using genetic constructs derived from hybridoma cells
producing the
monoclonal antibodies described above. Methods for producing chimeric and
humanized
antibodies are known in the art and are discussed herein. (See, for review,
Morrison, Science
229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Cabilly et al., U.S.
Patent No.
4,816,567; Taniguchi et al., EP 171496; Mornson et al., EP 173494; Neuberger
et al., WO
8601533; Robinson et al., WO 8702671; Boulianne et al., Nature 312:643 (1984);
Neuberger
et al., Nature 314:268 (1985).)
b) Isolation Of Antibody Fragments Directed Against steroid hormone receptor
Polypeptide(s) From A Library Of scFvs
Naturally occurring V-genes isolated from human PBLs are constructed into a
library
of antibody fragments which contain reactivities against steroid hormone
receptor
polypeptide(s) to which the donor may or may not have been exposed (see e.g.,
U.S. Patent
5,885,793 incorporated herein by reference in its entirety).
Rescue of the Library.
A library of scFvs is constructed from the RNA of human PBLs as described in
PCT
publication WO 92/01047. To rescue phage displaying antibody fragments,
approximately
109 E. coli harboring the phagemid are used to inoculate 50 ml of 2xTY
containing 1
glucose and 100 ~g/ml of ampicillin (2xTY-AMP-GLU) and grown to an O.D. of 0.8
with
shaking. Five ml of this culture is used to innoculate 50 ml of 2xTY-AMP-GLU,
2 x 108 TU
of delta gene 3 helper (M13 delta gene III, see PCT publication WO 92/01047)
are added and
the culture incubated at 37°C for 45 minutes without shaking and then
at 37°C for 45
minutes with shaking. The culture is centrifuged at 4000 r.p.m. for 10 min.
and the pellet
resuspended in 2 liters of 2xTY containing 100 pg/ml ampicillin and 50 ug/ml
kanamycin
and grown overnight. Phage are prepared as described in PCT publication WO
92/01047.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
216
M13 delta gene III is prepared as follows: M13 delta gene III helper phage
does not
encode gene III protein, hence the phage(mid) displaying antibody fragments
have a greater
avidity of binding to antigen. Infectious M13 delta gene III particles are
made by growing
the helper phage in cells harboring a pUCl9 derivative supplying the wild type
gene III
protein during phage morphogenesis. The culture is incubated for 1 hour at
37° C without
shaking and then for a further hour at 37°C with shaking. Cells are
spun down (IEC-Centra
8,400 r.p.m. for 10 min), resuspended in 300 ml 2xTY broth containing 100 ~g
ampicillin/ml
and 25 pg kanamycin/ml (2xTY-AMP-KAN) and grown overnight, shaking at
37°C. Phage
particles are purified and concentrated from the culture medium by two PEG-
precipitations
(Sambrook et al., 1990), resuspended in 2 ml PBS and passed through a 0.45 p.m
filter
(Minisart NML; Sartorius) to give a final concentration of approximately 1013
transducing
units/ml (ampicillin-resistant clones).
Panning of the Library.
Immunotubes (Nunc) are coated overnight in PBS with 4 ml of either 100 ~g/ml
or 10
~g/ml of a polypeptide of the present invention. Tubes are blocked with 2%
Marvel-PBS for
2 hours at 37°C and then washed 3 times in PBS. Approximately 1013 TU
of phage is
applied to the tube and incubated for 30 minutes at room temperature tumbling
on an over
and under turntable and then left to stand for another 1.5 hours. Tubes are
washed 10 times
with PBS 0.1% Tween-20 and 10 times with PBS. Phage are eluted by adding 1 ml
of 100
mM triethylamine and rotating 15 minutes on an under and over turntable after
which the
solution is immediately neutralized with 0.5 ml of 1.0M Tris-HCI, pH 7.4.
Phage are then
used to infect 10 ml of mid-log E. coli TG1 by incubating eluted phage with
bacteria for 30
minutes at 37°C. The E. coli are then plated on TYE plates containing
1% glucose and 100
~g/ml ampicillin. The resulting bacterial library is then rescued with delta
gene 3 helper
phage as described above to prepare phage for a subsequent round of selection.
This process
is then repeated for a total of 4 rounds of affinity purification with tube-
washing increased to
20 times with PBS, 0.1% Tween-20 and 20 times with PBS for rounds 3 and 4.
Characterization of Binders.
Eluted phage from the 3rd and 4th rounds of selection are used to infect E.
coli HB
2151 and soluble scFv is produced (Marks, et al., 1991) from single colonies
for assay.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
217
ELISAs are performed with microtitre plates coated with either 10 pg/ml of the
polypeptide
of the present invention in 50 mM bicarbonate pH 9.6. Clones positive in ELISA
are further
characterized by PCR fingerprinting (see, e.g., PCT publication WO 92/01047)
and then by
sequencing. These ELISA positive clones may also be further characterized by
techniques
known in the art, such as, for example, epitope mapping, binding affinity,
receptor signal
transduction, ability to block or competitively inhibit antibody/antigen
binding, and
competitive agonistic or antagonistic activity.
Example 17: Interaction of Steroid Hormone Receptors with other Signaling
Molecules
The purified Steroid Hormone Receptor (SHR) polypeptides of the invention are
research tools useful for the identification, characterization and
purification of proteins that
interact with SHRs, or other proteins in the signal transduction pathway in
which SHRs
function, such as receptor proteins, or other signal transduction pathway
proteins. Briefly,
labeled steroid hormone receptor protein is useful as a reagent for the
purification of
molecules with which it interacts. In one embodiment of affinity purification,
steroid
hormone receptor protein is covalently coupled to a chromatography column.
Cell-free
extract derived from putative target cells, such as liver or kidney cells, is
passed over the
column. Molecules which bind SHRs are retained in the column. The SHR protein-
complex
is recovered from the column, dissociated, and the recovered SHR interacting
protein
subjected to N-terminal protein sequencing. This amino acid sequence is then
used to identify
the captured molecule or to design degenerate oligonucleotide probes for
cloning the relevant
gene from an appropriate cDNA library.
Example 18: f3-Gal Assav of Steroid Hormone Receptor-Interacting Clones
The yeast two hybrid system as described by Seol et al., (Seol. W., et al, Mol
Endocrinology, 9:72-84 (1995)), which is herein incorporated by refernce in
its entirety,
among other assays known in the art, may be employed to assay for the
interaction of Steroid
Hormone Receptors (SHR) of the present invention with other members of the
hormone
receptor superfamily or other signal transduction molecules. Briefly,
expression vectors for
generating two types of fusion proteins are generated: one fusion protein
contains the LexA
DNA binding domain fused to a steroid hormone receptor or signal transduction
molecule of
interest and the other type of fusion protein contains the B42 trancriptional
activation domain


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
218
fused to the SHR of the invention. The EGY48 [MATalpha, leu2, trill ura3 his3
LEU2::pLexop6-LEU2 (DUAS LEU2)] yeast strain (in which the chromosomal LEU2
gene
is under the control of Lex-A operators) is successively transformed with the
8H18-341acZ
reporter plasmid (in which lacZ expression is under the control of Lex-A
operators), a Lex-A-
and a B-42- fusion protein expression vector . The LacZ vector conatins the
URA3 gene; the
Lex-A fusion protein vector contains the HIS3 gene, and the B42 expression
vector contains
the TRP1 gene. At least two separate colonies from plates containing glucose
but lacking
uracil, histidine, and tryptophan are selected randomly for each coexpressing
strain and used
to inoculate liquid media containing galactose to induce expression of the B42
fusion, but not
uracil, histidine, or tryptophan. Cultures are assayed for (3-gal, as (3-gal
expression is an
indicator of interaction between the steroid hormone receptor or signal
transduction molecule
of interest and the SHR of the present invention.
Example 19: Gel Shift Assay
In order to determine if the Steroid Hormone Receptors (SHRs) of the present
invention are involved in binding to DNA response elements, the gel shift
assay described by
Seol et al. is employed (Seol, W., et al, (1995)). Briefly, double-stranded
oligonucleotides
containing a known or putative SHR binding sequence, such as the Ec response
element
(EIRE) from the Drosophila heat shock protein (hsp)27 promoter containing the
conserved
hexameric sequence AGGTCA, are end labeled using [32P]ATP and kinase. In
addition to
naturally occurring response elements, it would be clear to one skilled in the
art to generate
synthetic oligonucleotides containing direct or inverted repeats of the
hexameric sequence
AGGTCA, separated from between 0-5 bp. SHR proteins are preincubated with 20
~1 gel
shift assay buffer (lOmM Tris, pH 8.0, either 40 or 150 mM KCI, 0.05% NP-40,
10%
glycerol, 1mM dithiothreitol, 2.5 mM MgCl2, and 5 ng poly-dl-dC) for 10 min.
on ice. This
mixture is then combined with the indicated labeled probe and incubated for 20
min. at room
temperature. The mixtures are analyzed by 6% nondenaturing polyacrylamide gel
electrophoresis using O.SX Tris-borate buffer at 4°C, followed by
autoradiography. Migration
of the probe incubated with the SHR is compared to migration of the untreated
probe. Slower
migration of the probe incubated with SHR is an indicator that the SHR binds
the DNA
response element.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
219
Example 20: Reporter Gene Assays for SHRs
In order to assay for transcriptional activation of reporter genes by Steroid
Hormone
Receptors (SHRs) of the present invention, the reporter gene assay as
described by Kumar et
al is used (Kumar, M.B., et al., J. Biol Chem., 274:22155-64 (1999), which is
herein
incorporated by reference in its entirety). The reporter constructs are
generated as described
by Kumar et al (Kumar, M.B., et al., (1999)), placing the SHR binding sites,
in addition to
any other regulatory sequences of interest, upstream of the luciferase
reporter gene. Briefly,
cells are cultured in modified Eagle's medium containing 10% fetal bovine
serum (v/v). Cells
are transfected with a total of 750 ng of DNA using LipofectAMINE reagent
(Life
Technologies, Inc.) in 24-well plates for reporter gene assays. The
transfected DNA mixture
include 100 ng each of reporter plasmid and the (3-galactosidase internal
control plasmid-
pCMV/LacZ (Invitrogen, San Diego, CA) along with variable amounts of control
plasmid
and SHR expression plasmid. Cells may then be treated with substances known to
activate
SHR after transfection, for a period of time. Cells are harvested, and
extracts are assayed for
luciferase activity. Luciferase activity is normalized to the observed (3-
galactosidase activity.
All transfections are performed in triplicate.
It would be clear to one skilled in the art that alternative reporter systems
could be
employed to detect reporter gene activation by the SHRs of the present
invention. For
example, one can use GFP or GFP fusion proteins, whose expression is under the
control of
SHR binding sites, as a reporter gene. COS-1 cells seeded on 2mm coverslips on
6-mm
dishes are transfected with a total of 400 ng of GFP or GFP fusion plasmid
containing GFP or
GFP fusion along with the SHR expression plasmid or control plasmid. Cells are
then treated
with substances known or suspected to activate SHR after transfection for a
period of time.
The cells are washed with phosphate-buffered saline and visualized using
fluorescence
microscopy 24 h after transfection.
Certain steroid hormone receptor polynucleotides and polypeptides of the
present
invention, including antibodies, as well as their uses, were disclosed in U.S.
provisional
application serial numbers 60/152,932 and 60/189,032, each of which is herein
incorporated
by reference in its entirety.
It will be clear that the invention may be practiced otherwise than as
particularly
described in the foregoing description and examples. Numerous modifications
and variations


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
220
of the present invention are possible in light of the above teachings and,
therefore, are within
the scope of the appended claims.
The entire disclosure of each document cited (including patents, patent
applications,
journal articles, abstracts, laboratory manuals, books, or other disclosures)
in the Background
of the Invention, Detailed Description, and Examples is hereby incorporated
herein by
reference. Further, the hard copy of the sequence listing submitted herewith
and the
corresponding computer readable form are both incorporated herein by reference
in their
entireties.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
221
Applicant's or agent's file PT016PCT IntemationalapplicationNo (UNASSIGNED
reference number
INDICATIONS RELATING TO A DEPOSTTED MICROORGANISM
(PCT Rule 13bis)
A. The indications made below
relate to the microorganism
referred to in the description


on page 20 , line N/A .


B. IDENTIF'ICATIONOFDEPOSIT Further
deposits are identified on an
additional sheet


Nameofdepositaryinstitution American
Type Culture Collection


Address of depositaty institution
(including postal code and country)


10801 University Boulevard


Manassas, Virginia 20110-2209


United States of America


Date ofdeposit Accession Number


09 September 1999 PTA-629


C. ADDITIONAL INDICATIONS (leave
blank if not applicable) This
information is continued on
an additional sheet



D. DESIGNATED STATES FOR WHICH
INDICATIONS ARE MADE (ifthe
indications are not forall designated
States)


Europe


In respect to those designations
in which a European Patent is
sought a sample of the deposited


microorganism will be made available
until the publication of the
mention of the grant of the
European


patent or until the date on which
application has been refused
or withdrawn or is deemed to
be withdrawn,


only by the issue of such a sample
to an expert nominated by the
person requesting the sample
(Rule 28 (4)


EPC).


E. SEPARATE FURNISHING OFINDICATIONS(leaveblankifnotapplicable)


The indications listed below
will be submitted to the International
Bureau later (spec~thegeneralnatureoftlu'indicationse.g.,
'Accession


Number of Deposit")



For receiving Office use only For International Bureau use only
This sheet was received with the international application ~ This sheet was
received by the International Bureau on:
Authorized officer
Form PCT/RO/134


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
222
ATCC Deposit No. PTA-629
Page 2
CANADA
The applicant requests that, until either a Canadian patent has been issued on
the basis of an
application or the application has been refused, or is abandoned and no longer
subject to
reinstatement, or is withdrawn, the Commissioner of Patents only authorizes
the furnishing of
a sample of the deposited biological material referred to in the application
to an independent
expert nominated by the Commissioner, the applicant must, by a written
statement, inform the
International Bureau accordingly before completion of technical preparations
for publication
of the international application.
NORWAY
The applicant hereby requests that the application has been laid open to
public inspection (by
the Norwegian Patent Office), or has been finally decided upon by the
Norwegian Patent
Office without having been laid open inspection, the furnishing of a sample
shall only be
effected to an expert in the art. The request to this effect shall be filed by
the applicant with
the Norwegian Patent Office not later than at the time when the application is
made available
to the public under Sections 22 and 33(3) of the Norwegian Patents Act. If
such a request has
been filed by the applicant, any request made by a third party for the
furnishing of a sample
shall indicate the expert to be used. That expert may be any person entered on
the list of
recognized experts drawn up by the Norwegian Patent Office or any person
approved by the
applicant in the individual case.
AUSTRALIA
The applicant hereby gives notice that the furnishing of a sample of a
microorganism shall
only be effected prior to the grant of a patent, or prior to the lapsing,
refusal or withdrawal of
the application, to a person who is a skilled addressee without an interest in
the invention
(Regulation 3.25(3) of the Australian Patents Regulations).
FINLAND
The applicant hereby requests that, until the application has been laid open
to public
inspection (by the National Board of Patents and Regulations), or has been
finally decided
upon by the National Board of Patents and Registration without having been
laid open to
public inspection, the furnishing of a sample shall only be effected to an
expert in the art.
UNITED KINGDOM
The applicant hereby requests that the furnishing of a sample of a
microorganism shall only
be made available to an expert. The request to this effect must be filed by
the applicant with
the International Bureau before the completion of the technical preparations
for the
international publication of the application.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
223
ATCC Deposit No. PTA-629
Page 3
DENMARK
The applicant hereby requests that, until the application has been laid open
to public
inspection (by the Danish Patent Office), or has been finally decided upon by
the Danish
Patent office without having been laid open to public inspection, the
furnishing of a sample
shall only be effected to an expert in the art. The request to this effect
shall be filed by the
applicant with the Danish Patent Office not later that at the time when the
application is made
available to the public under Sections 22 and 33(3) of the Danish Patents Act.
If such a
request has been filed by the applicant, any request made by a third party for
the furnishing of
a sample shall indicate the expert to be used. That expert may be any person
entered on a list
of recognized experts drawn up by the Danish Patent Office or any person by
the applicant in
the individual case.
SWEDEN
The applicant hereby requests that, until the application has been laid open
to public
inspection (by the Swedish Patent Office), or has been finally decided upon by
the Swedish
Patent Office without having been laid open to public inspection, the
furnishing of a sample
shall only be effected to an expert in the art. The request to this effect
shall be filed by the
applicant with the International Bureau before the expiration of 16 months
from the priority
date (preferably on the Form PCT/RO/134 reproduced in annex Z of Volume I of
the PCT
Applicant's Guide). If such a request has been filed by the applicant any
request made by a
third party for the furnishing of a sample shall indicate the expert to be
used. That expert may
be any person entered on a list of recognized experts drawn up by the Swedish
Patent Office
or any person approved by a applicant in the individual case.
NETHERLANDS
The applicant hereby requests that until the date of a grant of a Netherlands
patent or until the
date on which the application is refused or withdrawn or lapsed, the
microorganism shall be
made available as provided in the 31 F( 1 ) of the Patent Rules only by the
issue of a sample to
an expert. The request to this effect must be furnished by the applicant with
the Netherlands
Industrial Property Office before the date on which the application is made
available to the
public under Section 22C or Section 25 of the Patents Act of the Kingdom of
the Netherlands,
whichever of the two dates occurs earlier.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
224.
Applicant's or agent's file PT016PCT International applicationNc UNASSIGNED
reference number
INDICATIONS RELATING TO A DEPOSITED MICROORGANLSM
(PCT Rule 13bls)
A. The indications made below
relate to the microorganism
referred to in the description


on page 20 , line N/A .


B. IDENTIFICATIONOFDEPOSIT Further
deposits are identified on an
additional sheet


Nameofdepositaryinstitution American
Type Culture Collection


Address of depositary institution
(including postal code and country)


10801 University Boulevard


Manassas, Virginia 20110-2209


United States of America


Date ofdeposit Accession Number


13 March 2000 PTA-1477


C. ADDITIONAL INDICATIONS (leave
blank if not applicable) This
information is continued on
an additional sheet



D. DESIGNATED STATES FOR WHICH
INDICATIONS ARE MADE (if the
indications are not foralldesignated
States)


Europe


In respect to those designations
in which a European Patent is
sought a sample of the deposited


microorganism will be made available
until the publication of the
mention of the grant of the
European


patent or until the date on which
application has been refused
or withdrawn or is deemed to
be withdrawn,


only by the issue of such a sample
to an expert nominated by the
person requesting the sample
(Rule 28 (4)


EPC).


E. SEPARATE FURNISHING OF INDICATIONS
(leave blank ifnot applicable)


The indications listed below
will be submitted to the International
Bureau later (spec~thegeneralnatureoftheindicationse.g.,
'Accession


Number of Deposit")



For receiving Office use only For International B ureau use only
This sheet was received with the international application ~ This sheet was
received by the International Bureau on:
Authorized officer 1 / Authorizedofficer
Fotm PCT/R0/134 (luly 1992)


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
225
ATCC Deposit No. PTA-1477
Page 2
CANADA
The applicant requests that, until either a Canadian patent has been issued on
the basis of an
application or the application has been refused, or is abandoned and no longer
subject to
reinstatement, or is withdrawn, the Commissioner of Patents only authorizes
the furnishing of
a sample of the deposited biological material referred to in the application
to an independent
expert nominated by the Commissioner, the applicant must, by a written
statement, inform the
International Bureau accordingly before completion of technical preparations
for publication
of the international application.
NORWAY
The applicant hereby requests that the application has been laid open to
public inspection (by
the Norwegian Patent Office), or has been finally decided upon by the
Norwegian Patent
Office without having been laid open inspection, the furnishing of a sample
shall only be
effected to an expert in the art. The request to this effect shall be filed by
the applicant with
the Norwegian Patent Office not later than at the time when the application is
made available
to the public under Sections 22 and 33(3) of the Norwegian Patents Act. If
such a request has
been filed by the applicant, any request made by a third party for the
furnishing of a sample
shall indicate the expert to be used. That expert may be any person entered on
the list of
recognized experts drawn up by the Norwegian Patent Office or any person
approved by the
applicant in the individual case.
AUSTRALIA
The applicant hereby gives notice that the furnishing of a sample of a
microorganism shall
only be effected prior to the grant of a patent, or prior to the lapsing,
refusal or withdrawal of
the application, to a person who is a skilled addressee without an interest in
the invention
(Regulation 3.25(3) of the Australian Patents Regulations).
FINLAND
The applicant hereby requests that, until the application has been laid open
to public
inspection (by the National Board of Patents and Regulations), or has been
finally decided
upon by the National Board of Patents and Registration without having been
laid open to
public inspection, the furnishing of a sample shall only be effected to an
expert in the art.
UNITED KINGDOM
The applicant hereby requests that the furnishing of a sample of a
microorganism shall only
be made available to an expert. The request to this effect must be filed by
the applicant with
the International Bureau before the completion of the technical preparations
for the
international publication of the application.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
226
ATCC Deposit No: PTA-1477
Page 3
DENMARK
The applicant hereby requests that, until the application has been laid open
to public
inspection (by the Danish Patent Office), or has been finally decided upon by
the Danish
Patent office without having been laid open to public inspection, the
furnishing of a sample
shall only be effected to an expert in the art. The request to this effect
shall be filed by the
applicant with the Danish Patent Office not later that at the time when the
application is made
available to the public under Sections 22 and 33(3) of the Danish Patents Act.
If such a
request has been filed by the applicant, any request made by a third party for
the furnishing of
a sample shall indicate the expert to be used. That expert may be any person
entered on a list
of recognized experts drawn up by the Danish Patent Office or any person by
the applicant in
the individual case.
SWEDEN
The applicant hereby requests that, until the application has been laid open
to public
inspection (by the Swedish Patent Office), or has been finally decided upon by
the Swedish
Patent Office without having been laid open to public inspection, the
furnishing of a sample
shall only be effected to an expert in the art. The request to this effect
shall be filed by the
applicant with the International Bureau before the expiration of 16 months
from the priority
date (preferably on the Form PCT/RO/134 reproduced in annex Z of Volume I of
the PCT
Applicant's Guide). If such a request has been filed by the applicant any
request made by a
third party for the furnishing of a sample shall indicate the expert to be
used. That expert may
be any person entered on a list of recognized experts drawn up by the Swedish
Patent Office
or any person approved by a applicant in the individual case.
NETHERLANDS
The applicant hereby requests that until the date of a grant of a Netherlands
patent or until the
date on which the application is refused or withdrawn or lapsed, the
microorganism shall be
made available as provided in the 31 F( 1 ) of the Patent Rules only by the
issue of a sample to
an expert. The request to this effect must be furnished by the applicant with
the Netherlands
Industrial Property Office before the date on which the application is made
available to the
public under Section 22C or Section 25 of the Patents Act of the Kingdom of
the Netherlands,
whichever of the two dates occurs earlier.


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
<110> Human Genome Sciences, Inc.
<120> Steroid Hormone Receptor Polynucleotides, Polypeptides, and
Antibodies
<130> PT016PCT
<140> Unassigned
<141> 2000-09-07
<150> 60/152,932
<151> 1999-09-09
<150> 60/189,032
<151> 2000-03-14
<160> 16
<170> PatentIn Ver. 2.0
<210> 1
<211> 733
<212> DNA
<213> Homo sapiens
<400>
1


gggatccggagcccaaatcttctgacaaaactcacacatgcccaccgtgcccagcacctg 60


aattcgagggtgcaccgtcagtcttcctcttccccccaaaacccaaggacaccctcatga 120


tctcccggactcctgaggtcacatgcgtggtggtggacgtaagccacgaagaccctgagg 180


tcaagttcaactggtacgtggacggcgtggaggtgcataatgccaagacaaagccgcggg 240


aggagcagtacaacagcacgtaccgtgtggtcagcgtcctcaccgtcctgcaccaggact 300


ggctgaatggcaaggagtacaagtgcaaggtctccaacaaagccctcccaacccccatcg 360


agaaaaccatctccaaagccaaagggcagccccgagaaccacaggtgtacaccctgcccc 420


catcccgggatgagctgaccaagaaccaggtcagcctgacctgcctggtcaaaggcttct 480


atccaagcgacatcgccgtggagtgggagagcaatgggcagccggagaacaactacaaga 540


ccacgcctcccgtgctggactccgacggctccttcttcctctacagcaagctcaccgtgg 600


acaagagcaggtggcagcaggggaacgtcttctcatgctccgtgatgcatgaggctctgc 660


acaaccactacacgcagaagagcctctccctgtctccgggtaaatgagtgcgacggccgc 720


gactctagaggat
733


<210>
2


<211>
1079


<212>
DNA


<213> sapiens
Homo


<400> 2
ccacgcgtccgggaaagagtctcacttgcagttgcttcagtcacaacccaggcgtctgcc 60


ttaatagcatcacctgtgccgtctgcgtttctagcgtcacctgtgccgtctgcgtttcta 120


gcgtcacctgtgccgtctgccttcctagcatgacctgtgccgtctgcctttctagcatga 180


cctgtactgtctgcctttctagcgtcacctgtgccgtctgcctttctagttaaagttgat 240


tttaaattaatttgggaaaaacggtgaacaagtggggaaaaacatcaccaaagactccaa 300


acaggtggaacgatgaacttaatgttccctgcaccctccttacagatgcctggactttgg 360


cctgtgaatgagccccggggggctgggacagtagggttttcatggaatggacatggggta 420


gcaggaagtctcagccccagagggcttggcaggagccgccctctctgcggagttcttccc 480


acgggtgtgcgttggtggctggcggggaccccccacaggaactatgtctcagctgtggaa 540


aggagctgccccagggccaggaaggccggctctgcattcccgcatgttcccagagtgaaa 600


gagaatgagaaggagacacacaagtgctgttatgacctgggttgttggttggcaaataca 660


gcattaatagaaactttcttccatcccagaataccctctaccatatgctttggtcagggt 720


gccgctcgaggatttaaaagcaacaagaagaaaaccaaacaatgctgatagactcagaaa 780


aggagtggggggcagtcaccactgctgggcgtggcatccgccttgggcctttctccgacc 840


ttctggtgtggccgccctgggggccaggccctcgccatcctccccatccttgttcctcag 900


caaatggcattagcagctgtcatcagaagctgtgagggacccacagattttggaaacgac 960


ctggacacactattgggaaggagatgtggacggcctgtctcctcctgcagggcccaccct 1020




CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
aagaatgtat ttttaaacac atgaaataag tatttttcac tgaaaaaaaa aaaaaaaaa 1079
<210> 3
<211> 1734
<212> DNA
<213> Homo Sapiens
<400> 3
ggcacgagaaatctttcaacactaaaaatcttttgaattatactggatgtagggaatttg 60


aatttttatatagagtatcttggggacttcccctatgtacgacatacctcctaagcatgg 120


tgcaggccctccgtgggagccccagcaaatccaggtggacgtggctgaaagacaaaatgc 180


tcaaaacaccactgtgcttcttcttacactctgggtcatcaacagtctcatctacatcaa 240


gcttgtgccatttgttgacactatcaaatatggcagtgtcaccaagaatctctccataaa 300


aatgtgtttagctacaccacacatgggcaacacaattgctgtctctcatacgagtgttat 360


cacattccaggacttaatttttctggttctcatgagttgagccagtggctacttggtgat 420


tttccttcacagacaccagaaaaataatccacatcttcacaacaacagctgttcctctat 480


agcctcccatgagaccggaaccatcacgactgcgctgctgcttatgatttgcttcgttgt 540


atttaatgtgagcaactcgtgccacagcatttacctaagtacggtgaagaaaagggatca 600


gttgtggaccatctcagatttgatttcctcatgttaccccattttttggtccatttttgc 660


tcattggtagagaaagtcttatcctgaattcaaaatctatgaagtagagaaagtcgtctt 720


atcccatagatatgtcaaagtaaactttcaccaatataaactttttccaaaacaacattc 780


tataaagagttggtattctgaagaaatgatgggatttaggtctgaaaaatgagatatttc 840


tcagttcactgcatgaaatataatctaatgacctttaccttcagtaaagacaatattgca 900
-


tagcttagtatttaatgtttgtatacatagatataatcatttaatagacaagtagataga 960


taatttttcatgtcagatgttataatggacttcttgcactagtcattcaatatccatatt 1020


tttcttactttaatttgtgttcagcatgaaaaatcatttcaaaaggagatcagggactcc 1080


aggaagcagagcgtaactccccacttctaaactgtgggcttcacataatgacatcctacc 1140


aagaaatcagtatagcaaaggaggaaaaagagtaacttcgcattggagaaacctaacaaa 1200


cactgtctcagccaggtgatcaaggccactgtgagcagtgatcagtcaagtcgaaagtgc 1260


atatcatatgacaggatgagaatggcattttacctcgacatcttccctcccaaaactaat 1320


aatcctactacatcagacaaacctcagctgagaaacagtttccaaaaacacctgactagt 1380


accccctaaaaccatcaaggtcatcaagaacaatgtagtcctgagaaatgatcacagccg 1440


ggaagaacctaaggacttggtgtcctttggtatcctgatatggaatcctggagcagaaaa 1500


ggacattaagtaaaaaccaaggaaatagaaataaagtataaacttaataatatatcaata 1560


ttaattcattaattgtgacaaatgtaacatagtaatgtaagacattaaaatggggaaaat 1620


tgggtatgatgtggaggagaactctgtgtattctatttgcaactcttctgtaaatgtaaa 1680


cctatttgaaaattaaaaatttattttttaaaaagaaaaaaaaaaaaaaaaaaa 1734


<210> 4
<211> 642
<212> DNA
<213> Homo Sapiens
<400>
4


ggcacgagctcgtgccgaattcggcacgaggggatactttggcagcaatctcagaagttc 60


tttatgttgatttgctagaaggggatacagaatgccatgctagatttaaaactcctgagg 120


atgctcaagcagtaataaatgcctatacagaaattaacaagaaacactgctggaaactcg 180


agatcctttctggtgatcacgaacaaaggtattggcagaagattttggttgatagacagg 240


caaaacttaatcagcctcgggaaaagaaaagaggcactgaaaagttaatcaccaaagctg 300


aaaagattagactggcaaagactcaacaagcgagtaaacatataagattttctgaatatg 360


attgaaaaaaaaaacagttcacctcttaatacttcacaagatacttgagctgttcttggg 420


agattcacttttattatggtagcactgcataattaatgtgtttttaattaaaagaaatat 480


ctttgttcctcaacttgtaaataagacttttttctagagacaaatatgatgtataccaca 540


atttttcttaaacattttatttgttgaaattatcttagatgtcagtgtcaggtgatttag 600


taaataaatgtgttttgaacattaaaaaaaaaaaaaaaaaas 642


<210> 5
<211> 1697
<212> DNA
<213> Homo Sapiens
<400> 5
ggcacgagcc accatcttgg aaaacaaaag taacagtttt ctcattttta aaagttttgt 60


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
taagatctgttaatgattcacaaagaaatagtacttgggtatatttgaaattagtattta 120


ttcatttatgttgataagtgaaaatgcaagatggttatcaagatggaagtacttaatatg 180


tatattttaatattcttagatgcagataacttcacttttgatgtatttttattttgtctc 240


agtttcatttcaacttacgtttaaggggttcttgataatctgacatgataagtggtgttg 300


gaattggcattaaaatccaccatgctctacagcacttcatccttcttcggcaggcaccaa 360


tttgatcttctactactttgcagacatctcttctgcaaacaccagacaaactgagacaat 420


gaacttccgcaggacccaaagcacctcttactgcaggaaagaagatccagtgagatagtt 480


agtccacaaatggaatgtcaatccataaacacacttaagtaacagaataaatttagtatg 540


agcgtttttatgtgggagctcttgaaatggttgctgctcatatgtcagagacatatgcag 600


ttaagaaaggtagcagttccaatcctggtttggcccaacagtcactgcatttttggtggg 660


gaaaagggatgtgggaggagatggtacatcctcatctttttctctgggttttctgtcaga 720


aagggatgttgcttactccagtggcaaaaagtgccagtgtcttctgccagagtgggttac 780


tgagggccatggtggttccaccttgtggctgatacagatagtccctttctgcttttgttt 840


ctagccaaaaaagatgtttctcgcatctcaggtatgcagatttcagcagctgttttttct 900


atatggctattttttttttctttcactctctctttctctcttttttttttttttggcttc 960


actgtgttgccatagtttcttaaatggtcccttgaaccctcccagagctattttggtttg 1020


tacataactatctatatatttttttcttggggggagtgtgtagagctaaaggctggtata 1080


tcctgctcctgctccccgaaagtgacgttattcccccaagctaatatttcaggctttcaa 1140


tttattcatgctttcatctgtttaaacataagtagaaattactttttctctccacattta 1200


gatttgatctatctactttaattgctaatagtgtcttagtcatagaatagattagttaga 1260


aaaaagtgtttttgacattataaatgattctttcaatttgtgtctaaaagggaaaatact 1320


agaaagcttaacatttattattgtattcagaccagtatttcctccagatgacctttatta 1380


caacaaagataatttaatgaagatctctctaatggtaaagctgatggctttgtgctatta 1440


caatatccttcaaataaagtgacgatttggtggaaagaacaactaaagcaaggattagga 1500


agaaaacagttaataccttcattttggtctcactctgcattaagagttttcattgtgtta 1560


aggatttattatacattaagtaatttaatgttcatttaataataaatggatcctattaaa 1620


tatgatttttaaaattataatcacattacttttattcaaaaaaaaaaaaaaaaaaaaaaa 1680


aaaaaaaaaaaaaaaaa 1697


<210> 6
<211> 1446
<212> DNA
<213> Homo Sapiens
<400>
6


ggcacgagcaaaaatagaagaaaatcaaaatctatactttagaaaataccctattgagat 60


tttcagatttatccctaattctaagtagttcattcaatataacagggccataagggttca 120


atacagaagagtcagagtttgtttttttttatattttgaatgtgtttttcatacataatt 180


gtatatcatagaatttctggcaactgcatttatccactagaaaaatgtttattgcaatct 240


aatgaactaagtctactgtatatttctgtagaaactgcccttgaacttaatgcttgcaga 300


aatatatcagaatagaagaaacagactacttctaattttaaaccaaacatctaacagaat 360


cttagtaatactaattaaaaataaacctggactaatctgattgtagtaccacatcagcaa 420


gatggctgactagagttgcctggctctcaatctctgggttttaaaacataggagacttaa 480


catcaaaaacagttactctcccaatattaggttttggataactgatcaggttgatcaaag 540


atatgaaaaaatacataataacaataccatttattgaatatttaaaagttcttggcattg 600


tggtgagtaatttatatacattgcattatttgttctcatggcaacattatgggttatatt 660


ttatctccactttataaatgaggaaatggaagcttggaggaacaaaaatatatagctaaa 720


agtcccacaggtatttagtggttgagcaaaaaattgtttctcattctgtttgactatgaa 780


accaatttttttcacaagagtatcatattttcaaactgtaagagaaattccaggtcatat 840


agtccaatatccacctgatgttagtttcttctacattacaggtacttctctgacttaaca 900


tttaaaactctctagcaaagtggaacattctgatccccaaaactaccactaaatttaact 960


gactttacctccttaaagcctctccaatctgtctactttccctctccactcgccctgcaa 1020


aaacctcaagcaaccttcagacaacagcattagccaactaactaatctacctccattcac 1080


tcttaccatttttgcatttgttttgtacacatttcagctaaatattctttttcaaatata 1140


tcactctgctcatttagtgactttttagtcttttttggatagacagcaaacatcttgcca 1200


cagcctataagaacagtaactgttaggtgctggtttgctcagatctgtcccagtttacct 1260


tgttgccaagcctaattattaattatcctctttcattctcaaaaggttcaafitttggaca1320


gtgaattttgtggttatcctatctataaagccttgcatcacctggcacaagtgattatac 1380


atttaactgttcaccactcattttgaccataaagctggacaactgttacaaacaaccatt 1440


tcagcc 1446


<210> 7
<211> 76
<212> PRT


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
4
<213> Homo Sapiens
<400> 7
Thr Arg Pro Gly Lys Ser Leu Thr Cys Ser Cys Phe Ser His Asn Pro
1 5 10 15
Gly Val Cys Leu Asn Ser Ile Thr Cys Ala Val Cys Val Ser Ser Val
20 25 30
Thr Cys Ala Val Cys Val Ser Ser Val Thr Cys Ala Val Cys Leu Pro
35 40 45
Ser Met Thr Cys Ala Val Cys Leu Ser Ser Met Thr Cys Thr Val Cys
50 55 60
Leu Ser Ser Val Thr Cys Ala Val Cys Leu Ser Ser
65 70 75
<210> 8
<211> 98
<212> PRT
<213> Homo Sapiens
<400> 8
His Glu Lys Ser Phe Asn Thr Lys Asn Leu Leu Asn Tyr Thr Gly Cys
1 5 10 15
Arg Glu Phe Glu Phe Leu Tyr Arg Val Ser Trp Gly Leu Pro Leu Cys
20 25 30
Thr Thr Tyr Leu Leu Ser Met Val Gln Ala Leu Arg Gly Ser Pro Ser
35 40 45
Lys Ser Arg Trp Thr Trp Leu Lys Asp Lys Met Leu Lys Thr Pro Leu
50 55 60
Cys Phe Phe Leu His Ser Gly Ser Ser Thr Val Ser Ser Thr Ser Ser
65 70 75 80
Leu Cys His Leu Leu Thr Leu Ser Asn Met Ala Val Ser Pro Arg Ile
85 90 95
Ser Pro
<210> 9
<211> 120
<212> PRT
<213> Homo Sapiens
<400> 9
His Glu Leu Val Pro Asn Ser Ala Arg Gly Asp Thr Leu Ala Ala Ile
1 5 10 15
Ser Glu Val Leu Tyr Val Asp Leu Leu Glu Gly Asp Thr Glu Cys His
20 25 30
Ala Arg Phe Lys Thr Pro Glu Asp Ala Gln Ala Val Ile Asn Ala Tyr
35 40 45
Thr Glu Ile Asn Lys Lys His Cys Trp Lys Leu Glu Ile Leu Ser Gly
50 55 60
Asp His Glu Gln Arg Tyr Trp Gln Lys Ile Leu Val Asp Arg Gln Ala


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
65 70 75 80
Lys Leu Asn Gln Pro Arg Glu Lys Lys Arg Gly Thr Glu Lys Leu Ile
85 90 95
Thr Lys Ala Glu Lys Ile Arg Leu Ala Lys Thr Gln Gln Ala Ser Lys
100 105 110
His Ile Arg Phe Ser Glu Tyr Asp
115 120
<210> 10
<211> 81
<212> PRT
<213> Homo Sapiens
<400> 10
Met Ser Val Phe Met Trp Glu Leu Leu Lys Trp Leu Leu Leu Ile Cys
1 5 10 15
Gln Arg His Met Gln Leu Arg Lys Val Ala Val Pro Ile Leu Val Trp
20 25 30
Pro Asn Ser His Cys Ile Phe Gly Gly Glu Lys Gly Cys Gly Arg Arg
35 40 45
Trp Tyr Ile Leu Ile Phe Phe Ser Gly Phe Ser Val Arg Lys Gly Cys
50 55 60
Cys Leu Leu Gln Trp Gln Lys Val Pro Val Ser Ser Ala Arg Val Gly
65 70 75 80
Tyr
<210> 11
<211> 51
<212> PRT
<213> Homo Sapiens
<400> 11
Met Cys Phe Ser Tyr Ile Ile Val Tyr His Arg Ile Ser Gly Asn Cys
1 5 10 15
Ile Tyr Pro Leu Glu Lys Cys Leu Leu Gln Ser Asn Glu Leu Ser Leu
20 25 30
Leu Tyr Ile Ser Val Glu Thr Ala Leu Glu Leu Asn Ala Cys Arg Asn
35 40 45
Ile Ser Glu
<210> 12
<211> 166
<212> PRT
<213> Homo Sapiens
<400> 12
Thr Ser Gly Glu Lys His His Gln Arg Leu Gln Thr Gly Gly Thr Met
1 5 10 15
Asn Leu Met Phe Pro Ala Pro Ser Leu Gln Met Pro Gly Leu Trp Pro


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
6
20 25 30
Val Asn Glu Pro Arg Gly Ala Gly Thr Val Gly Phe Ser Trp Asn Gly
35 40 45
His Gly Val Ala Gly Ser Leu Ser Pro Arg Gly Leu Gly Arg Ser Arg
50 55 60
Pro Leu Cys Gly Val Leu Pro Thr Gly Val Arg Trp Trp Leu Ala Gly
65 70 75 80
Thr Pro His Arg Asn Tyr Val Ser Ala Val Glu Arg Ser Cys Pro Arg
85 90 95
Ala Arg Lys Ala Gly Ser Ala Phe Pro His Val Pro Arg Val Lys Glu
100 105 110
Asn Glu Lys Glu Thr His Lys Cys Cys Tyr Asp Leu Gly Cys Trp Leu
115 120 125
Ala Asn Thr Ala Leu Ile Glu Thr Phe Phe His Pro Arg Ile Pro Ser
130 135 140
Thr Ile Cys Phe Gly Gln Gly Ala Ala Arg Gly Phe Lys Ser Asn Lys
145 150 155 160
Lys Lys Thr Lys Gln Cys
165
<210> 13
<211> 62
<212> PRT
<213> Homo sapiens
<400> 13
Trp Val Ile Asn Ser Leu Ile Tyr Ile Lys Leu Val Pro Phe Val Asp
1 5 10 15
Thr Ile Lys Tyr Gly Ser Val Thr Lys Asn Leu Ser Ile Lys Met Cys
20 25 30
Leu Ala Thr Pro His Met Gly Asn Thr Ile Ala Val Ser His Thr Ser
35 40 45
Val Ile Thr Phe Gln Asp Leu Ile Phe Leu Val Leu Met Ser
50 55 60
<210> 14
<211> 87
<212> PRT
<213> Homo sapiens
<400> 14
Ser Gly Tyr Leu Val Ile Phe Leu His Arg His Gln Lys Asn Asn Pro
1 5 10 15
His Leu His Asn Asn Ser Cys Ser Ser Ile Ala Ser His Glu Thr Gly
20 25 30
Thr Ile Thr Thr Ala Leu Leu Leu Met Ile Cys Phe Val Val Phe Asn
35 40 45
Val Ser Asn Ser Cys His Ser Ile Tyr Leu Ser Thr Val Lys Lys Arg
50 55 60


CA 02384672 2002-03-07
WO 01/18049 PCT/US00/24517
7
Asp Gln Leu Trp Thr Ile Ser Asp Leu Ile Ser Ser Cys Tyr Pro Ile
65 70 75 80
Phe Trp Ser Ile Phe Ala His
<210> 15
<211> 43
<212> PRT
<213> Homo sapiens
<400> 15
His Glu Pro Pro Ser Trp Lys Thr Lys Val Thr Val Phe Ser Phe Leu
1 5 10 15
Lys Val Leu Leu Arg Ser Val Asn Asp Ser Gln Arg Asn Ser Thr Trp
20 25 30
Val Tyr Leu Lys Leu Val Phe Ile His Leu Cys
35 40
<210> 16
<211> 28
<212> PRT
<213> Homo Sapiens
<400> 16
Ala Arg Ala Lys Ile Glu Glu Asn Gln Asn Leu Tyr Phe Arg Lys Tyr
1 5 10 15
Pro Ile Glu Ile Phe Arg Phe Ile Pro Asn Ser Lys
20 25

Representative Drawing

Sorry, the representative drawing for patent document number 2384672 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2000-09-07
(87) PCT Publication Date 2001-03-15
(85) National Entry 2002-03-07
Dead Application 2006-09-07

Abandonment History

Abandonment Date Reason Reinstatement Date
2002-09-09 FAILURE TO COMPLETE 2003-05-30
2005-09-07 FAILURE TO PAY APPLICATION MAINTENANCE FEE
2005-09-07 FAILURE TO REQUEST EXAMINATION

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Registration of a document - section 124 $100.00 2002-03-07
Application Fee $300.00 2002-03-07
Maintenance Fee - Application - New Act 2 2002-09-09 $100.00 2002-08-26
Maintenance Fee - Application - New Act 3 2003-09-08 $100.00 2003-08-27
Maintenance Fee - Application - New Act 4 2004-09-07 $100.00 2004-08-24
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
HUMAN GENOME SCIENCES, INC.
Past Owners on Record
NI, JIAN
RUBEN, STEVEN M.
SHI, YANGGU
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2002-03-07 1 55
Claims 2002-03-07 4 104
Cover Page 2002-08-20 1 32
Description 2002-03-07 233 13,408
PCT 2002-03-07 10 415
Assignment 2002-03-07 9 382
Correspondence 2003-04-15 1 47
Prosecution-Amendment 2003-11-20 5 141
Assignment 2009-08-10 20 998

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

No BSL files available.