Language selection

Search

Patent 2384659 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 2384659
(54) English Title: 37 HUMAN SECRETED PROTEINS
(54) French Title: 37 PROTEINES SECRETEES HUMAINES
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12N 15/12 (2006.01)
  • A61K 38/16 (2006.01)
  • C07K 7/06 (2006.01)
  • C07K 14/47 (2006.01)
  • C07K 16/18 (2006.01)
  • C12N 5/10 (2006.01)
  • C12N 15/11 (2006.01)
  • C12N 15/66 (2006.01)
  • C12P 21/00 (2006.01)
  • G01N 33/566 (2006.01)
  • A61K 38/00 (2006.01)
  • C12Q 1/68 (2006.01)
(72) Inventors :
  • ROSEN, CRAIG A. (United States of America)
  • RUBEN, STEVEN M. (United States of America)
  • KOMATSOULIS, GEORGE A. (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: 2000-09-26
(87) Open to Public Inspection: 2001-04-05
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2000/026323
(87) International Publication Number: WO2001/023546
(85) National Entry: 2002-03-11

(30) Application Priority Data:
Application No. Country/Territory Date
60/155,805 United States of America 1999-09-27

Abstracts

English Abstract




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


French Abstract

La présente invention concerne des protéines sécrétées humaines et des acides nucléiques isolés contenant les régions codantes des gènes codants pour ces protéines. Cette invention concerne aussi des vecteurs, des cellules hôtes, des anticorps et des méthodes de recombinaison permettant de produire ces protéines sécrétées humaines. Cette invention concerne encore des techniques diagnostiques et thérapeutiques qui conviennent pour diagnostiquer et traiter des maladies, des pathologies et/ou des troubles liés à ces protéines sécrétées humaines.

Claims

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



402
What Is Claimed Is:
1. An isolated nucleic acid molecule comprising a polynucleotide having
a nucleotide sequence at least 95% identical to a sequence selected from the
group
consisting of:
(a) a polynucleotide fragment of SEQ ID NO:X or a polynucleotide fragment
of the cDNA sequence included in ATCC Deposit No:Z, which is hybridizable to
SEQ ID NO:X;
(b) a polynucleotide encoding a polypeptide fragment of SEQ ID NO:Y or a
polypeptide fragment encoded by the cDNA sequence included in ATCC Deposit
No:Z, which is hybridizable to SEQ ID NO:X;
(c) a polynucleotide encoding a polypeptide domain of SEQ ID NO:Y or a
polypeptide domain encoded by the cDNA sequence included in ATCC Deposit
No:Z, which is hybridizable to SEQ ID NO:X;
(d) 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, which is hybridizable to SEQ ID NO:X;
(e) a polynucleotide encoding a polypeptide of SEQ ID NO:Y or the cDNA
sequence included in ATCC Deposit No:Z, which is hybridizable to SEQ ID NO:X,
having biological activity;
(f) a polynucleotide which is a variant of SEQ ID NO:X;
(g) a polynucleotide which is an allelic variant of SEQ ID NO:X;
(h) a polynucleotide which encodes a species homologue of the SEQ ID
NO:Y;
(i) a polynucleotide capable of hybridizing under stringent conditions to any
one of the polynucleotides specified in (a)-(h), 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.


403


2. The isolated nucleic acid molecule of claim 1, wherein the
polynucleotide fragment comprises a nucleotide sequence encoding a secreted
protein.
3. The isolated nucleic acid molecule of claim 1, wherein the
polynucleotide fragment 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, which is hybridizable to SEQ ID NO:X.
4. The isolated nucleic acid molecule of claim 1, wherein the
polynucleotide fragment comprises the entire nucleotide sequence of SEQ ID
NO:X
or the cDNA sequence included in ATCC Deposit No:Z, which is hybridizable to
SEQ ID NO:X.
5. The isolated nucleic acid molecule of claim 2, wherein the nucleotide
sequence comprises sequential nucleotide deletions from either the C-terminus
or the
N-terminus.
6. The isolated nucleic acid molecule of claim 3, wherein the nucleotide
sequence comprises sequential nucleotide deletions from either the C-terminus
or the
N-terminus.
7. A recombinant vector comprising the isolated nucleic acid molecule of
claim 1.
8. A method of making a recombinant host cell comprising the isolated
nucleic acid molecule of claim 1.
9. A recombinant host cell produced by the method of claim 8.
10. The recombinant host cell of claim 9 comprising vector sequences.


404


11. An isolated polypeptide comprising an amino acid sequence at least
95% identical to a sequence selected from the group consisting of:
(a) a polypeptide fragment of SEQ ID NO:Y or the encoded sequence
included in ATCC Deposit No:Z;
(b) a polypeptide fragment of SEQ ID NO:Y or the encoded sequence
included in ATCC Deposit No:Z, having biological activity;
(c) a polypeptide domain of SEQ ID NO:Y or the encoded sequence included
in ATCC Deposit No:Z;
(d) a polypeptide epitope of SEQ ID NO:Y or the encoded sequence included
in ATCC Deposit No:Z;
(e) a secreted form of SEQ ID NO:Y or the encoded sequence included in
ATCC Deposit No:Z;
(f) a full length protein of SEQ ID NO:Y or the encoded sequence included in
ATCC Deposit No:Z;
(g) a variant of SEQ ID NO:Y;
(h) an allelic variant of SEQ ID NO:Y; or
(i) a species homologue of the SEQ ID NO:Y.
12. The isolated polypeptide of claim 11, wherein the secreted form or the
full length protein comprises sequential amino acid deletions from either the
C-
terminus or the N-terminus.
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.
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.


405


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.
21. The gene corresponding to the cDNA sequence of SEQ ID NO:Y.
22. A method of identifying an activity in a biological assay, wherein the
method comprises:
(a) expressing SEQ ID NO:X in a cell;
(b) isolating the supernatant;


406


(c) detecting an activity in a biological assay; and
(d) identifying the protein in the supernatant having the activity.
23. The product produced by the method of claim 20.

Description

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





DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE I)E CETTE DEMANDE OU CE BREVETS
COMPRI~:ND PLUS D'UN TOME.
CECI EST ~.E TOME 1 DE 2
NOTE: Pour les tomes additionels, veillez contacter 1e Bureau Canadien des
Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.
THIS IS VOLUME 1 OF 2
NOTE: For additional vohxmes please contact the Canadian Patent Oi~ice.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
37 Human Secreted Proteins
Field of the Invention
This invention relates to newly identified polynucleotides, polypeptides
encoded by these polynucleotides, antibodies that bind these polypeptides,
uses of
such polynucleotides, polypeptides, and antibodies, and their production.
Background of the Invention
Unlike bacterium, which exist as a single compartment surrounded by a
membrane, human cells and other eucaryotes are subdivided by membranes into
many
functionally distinct compartments. Each membrane-bounded compartment, or
organelle, contains different proteins essential for the function of the
organelle. The
cell uses "sorting signals," which are amino acid motifs located within the
protein, to
target proteins to particular cellular organelles.
One type of sorting signal, called a signal sequence, a signal peptide, or a
leader sequence, directs a class of proteins to an organelle called the
endoplasmic
1 S reticulum (ER). The ER separates the membrane-bounded proteins from all
other
types of proteins. Once localized to the ER, both groups of proteins can be
further
directed to another organelle called the Golgi apparatus. Here, the Golgi
distributes
the proteins to vesicles, including secretory vesicles, the cell membrane,
lysosomes,
and the other organelles.
Proteins targeted to the ER by a signal sequence can be released into the
extracellular space as a secreted protein. For example, vesicles containing
secreted
proteins can fuse with the cell membrane and release their contents into the
extracellular space - a process called exocytosis. Exocytosis can occur
constitutively
or after receipt of a triggering signal. In the latter case, the proteins are
stored in
secretory vesicles (or secretory granules) until exocytosis is triggered.
Similarly,
proteins residing on the cell membrane can also be secreted into the
extracellular
space by proteolytic cleavage of a "linker" holding the protein to the
membrane.
Despite the great progress made in recent years, only a small number of genes
encoding human secreted proteins have been identified. These secreted proteins
include the commercially valuable human insulin, interferon, Factor VIII,
human
growth hormone, tissue plasminogen activator, and erythropoeitin. Thus, in
light of


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
the pervasive role of secreted proteins in human physiology, a need exists for
identifying and characterizing novel human secreted proteins and the genes
that
encode them. This knowledge will allow one to detect, to treat, and to prevent
medical diseases, disorders, andlor conditions by using secreted proteins or
the genes
that encode them.
Summary of the Invention
The present invention relates to novel polynucleotides and the encoded
polypeptides. Moreover, the present invention relates to vectors, host cells,
antibodies, and recombinant and synthetic methods for producing the
polypeptides
and polynucleotides. Also provided are diagnostic methods for detecting
diseases,
disorders, and/or conditions related to the polypeptides and polynucleotides,
and
therapeutic methods for treating such diseases, disorders, and/or conditions.
The
invention further relates to screening methods for identifying binding
partners of the
polypeptides.
Detailed Description
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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
In the present invention, a "secreted" protein refers to those proteins
capable
of being directed to the ER, secretory vesicles, or the extracellular space as
a result of
a signal sequence, as well as those proteins released into the extracellular
space
without necessarily containing a signal sequence. If the secreted protein is
released
into the extracellular space, the secreted protein can undergo extracellular
processing
to produce a "mature" protein. Release into the extracellular space can occur
by many
mechanisms, including exocytosis and proteolytic cleavage.
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,
50 kb, 1 S
kb, 10 kb, 7.5 kb, 5 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., S' 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).
As used herein, a "polynucleotide" refers to a molecule having a nucleic acid
sequence contained in SEQ ID NO:X or the cDNA contained within the clone
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 the signal
sequence, the
secreted 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 the translated amino acid sequence generated from the
polynucleotide as broadly defined.
In the present invention, the full length sequence identified as SEQ ID NO:X
was often generated by overlapping sequences contained in multiple clones
(contig
analysis). A representative clone containing all or most of the sequence for
SEQ ID
NO:X was deposited with the American Type Culture Collection ("ATCC"). As


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
4
shown in Table 1, each clone is identified by a cDNA Clone ID (Identifier) and
the
ATCC Deposit Number. The ATCC is located at 10801 University Boulevard,
Manassas, Virginia 20110-2209, USA. The ATCC deposit was 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, the complement thereof, or the cDNA within
the clone deposited with the ATCC. "Stringent hybridization conditions" refers
to an
overnight incubation at 42 degree C in a solution comprising 50% formamide, Sx
SSC
(750 mM NaCI, 75 mM trisodium citrate), SO mM sodium phosphate (pH 7.6), Sx
Denhardt's solution, 10% dextran sulfate, and 20 pg/ml denatured, sheared
salmon
sperm DNA, followed by washing the filters in 0.1 x SSC at about 65 degree C.
Also contemplated 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. The inclusion of specific
blocking
reagents may require modification of the hybridization conditions described
above,
due to problems with compatibility.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
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
S 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 polynucleotide 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.
The polypeptide 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
6
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 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).)
"SEQ ID NO:X" refers to a polynucleotide sequence while "SEQ ID NO:Y"
refers to a polypeptide sequence, both sequences identified by an integer
specified in
Table 1.
"A polypeptide having biological activity" refers to polypeptides exhibiting
activity similar, but not necessarily identical to, an activity of a
polypeptide of the
present invention, including mature forms, as measured in a particular
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.)
Many proteins (and translated DNA sequences) contain regions where the
amino acid composition is highly biased toward a small subset of the available


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
residues. For example, membrane spanning domains and signal peptides (which
are
also membrane spanning) typically contain long stretches where Leucine (L),
Valine
(V), Alanine (A), and Isoleucine (I) predominate. Poly-Adenosine tracts
(polyA) at
the end of cDNAs appear in forward translations as poly-Lysine (poly-K) and
poly-
Phenylalanine (poly-F) when the reverse complement is translated. These
regions are
often referred to as "low complexity" regions.
Such regions can cause database similarity search programs such as BLAST to
find high-scoring sequence matches that do not imply true homology. The
problem is
exacerbated by the fact that most weight matrices (used to score the
alignments
generated by BLAST) give a match between any of a group of hydrophobic amino
acids (L,V and I) that are commonly found in certain low complexity regions
almost
as high a score as for exact matches.
In order to compensate for this, BLASTX.2 (version 2.Oa5MP-WashU)
employs two filters ("seg" and "xnu") which "mask" the low complexity regions
in a
particular sequence. These filters parse the sequence for such regions, and
create a
new sequence in which the amino acids in the low complexity region have been
replaced with the character "X". This is then used as the input sequence
(sometimes
referred to herein as "Query" and/or "Q") to the BLASTX program. While this
regime helps to ensure that high-scoring matches represent true homology,
there is a
negative consequence in that the BLASTX program uses the query sequence that
has
been masked by the filters to draw alignments.
Thus, a stretch of "X"s in an alignment shown in the following application
does not necessarily indicate that either the underlying DNA sequence or the
translated protein sequence is unknown or uncertain. Nor is the presence of
such
stretches meant to indicate that the sequence is identical or not identical to
the
sequence disclosed in the alignment of,the present invention. Such stretches
may
simply indicate that the BLASTX program masked amino acids in that region due
to
the detection of a low complexity region, as defined above. In all cases, the
reference
sequences) (sometimes referred to herein as "Subject", "Sbjct", and/or "S")
indicated
in the specification, sequence table (Table 1), and/or the deposited clone is
(are) the
definitive embodiments) of the present invention, and should not be construed
as


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
8
limiting the present invention to the partial sequence shown in an alignment,
unless
specifically noted otherwise herein.
Polynucleotides and Polypeptides of the Invention
S
FEATURES OF PROTEIN ENCODED BY GENE NO: 1
This gene is expressed primarily in the following tissues/cDNA libraries:
Brain Frontal Cortex, re-excision; L428; Human Pancreas Tumor, Reexcision;
Human Cerebellum.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 48 as residues: Cys-3 to Gly-11. Polynucleotides
encoding said polypeptides are also provided.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
1 S related to SEQ ID NO:11 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1621 of SEQ ID
NO:11, b
is an integer of 1 S to 1635, where both a and b correspond to the positions
of
nucleotide residues shown in SEQ ID NO:11, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 2
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 85 and/or
SEQ ID
NO: 86. Polynucleotides encoding these polypeptides are also provided.
This gene is expressed primarily in the following tissues/cDNA libraries:
Human Placenta; Human Bone Marrow, treated; Soares placenta Nb2HP and to a
lesser extent in Soares retina N2b4HR; Soares melanocyte 2NbHM; Human


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
9
Osteoclastoma; CD34+cells, II, FRACTION 2; Soares testis NHT; T-Cell PHA 24
hrs; Human Chondrosarcoma; NCI CGAP_GCB1;
Soares_fetal liver spleen-1NFLS_S1; Soares fetal liver spleen 1NFLS; Soares
infant
brain 1NIB; Healing Abdomen Wound,l5 days post incision; Lung Mesothelium;
Prostate; Human Kidney; H. Striatum Depression, subt; H. Atrophic Endometrium;
Aorta endothelial cells + TNF-a; Soares NhHMPu S 1; Lung Carcinoma A549
TNFalpha activated; Human T-cell lymphoma,re-excision; Healing groin wound,
7.5
hours post incision; Soares-pregnant uterus NbHPU; Stratagene colon (#937204);
Human Osteoclastoma, re-excision; Synovial Fibroblasts (Ill/TNF), subt;
Soares NhHMPu S 1; normalized infant brain cDNA; Human Umbilical Vein,
Reexcision; TF-1 Cell Line GM-CSF Treated; Pancreas Tumor PCA4 Tu; KMH2;
L428; HUMAN JURKAT MEMBRANE BOUND POLYSOMES; Stromal cell
TF274; Human Placenta (re-excision); Soares adult brain N2b5HBS5Y; Ulcerative
Colitis; Bone Marrow Stromal Cell, untreated; CHME Cell Line,treated 5 hrs;
Smooth muscle, serum induced,re-exc; CHME Cell Line,untreated; Human T-Cell
Lymphoma; Smooth muscle, serum treated; Dendritic cells, pooled;
Soares multiple sclerosis 2NbHMSP; Human Ovarian Cancer Reexcision; Bone
marrow; 12 Week Early Stage Human II, Reexcision; NCI CGAP_CoB;
NCI CGAP_Col2; Human Microvascular Endothelial Cells, fract. A;
Soares fetal lung NbHLI9W; Soares multiple sclerosis 2NbHMSP; Spleen,
Chronic lymphocytic leukemia; Soares NFL T GBC_S1;
Soares total fetus Nb2HF8 9w; Colon Normal III; Hodgkin's Lymphoma II;
Osteoblasts; Keratinocyte and Colon Tumor II.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 49 as residues: Met-1 to Asn-8. Polynucleotides
encoding said polypeptides are also provided.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:12 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 be cumbersome. Accordingly, preferably excluded from the present
invention


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 2281 of SEQ >D
N0:12, b
is an integer of 1 S to 2295, where both a and b correspond to the positions
of
nucleotide residues shown in SEQ >D N0:12, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 3
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 87,SEQ ID
NO:
10 88,SEQ ID NO: 89,SEQ ID NO: 90,SEQ >D NO: 91,SEQ ID NO: 92 and/or SEQ >D
NO: 93. Polynucleotides encoding these polypeptides are also provided.
This gene is expressed primarily in the following tissues/cDNA libraries:
Amniotic Cells - Primary Culture; STRATAGENE Human skeletal muscle cDNA
library, cat. #936215.; Soares fetal heart NbHHI9W; Smooth muscle, serum
1 S induced,re-exc; Monocyte activated and to a lesser extent in Human
Gastrocnemius;
H. Atrophic Endometrium; HSA 172 Cells; Stratagene endothelial cell 937223;
Synovial hypoxia; Human Manic Depression Tissue; Human Thymus; Human
Pancreas Tumor; NTERA2, control; Human fetal heart, Lambda ZAP Express;
NCI CGAP HSC1; Soares testis NHT; Soares fetal liver spleen 1NFLS S1;
Stratagene fibroblast (#937212); Colon Carcinoma; Primary Dendritic cells,frac
2;
Human Fetal Heart; Human Osteoclastoma; Soares fetal heart NbHHI9W and
Human 8 Week Whole Embryo.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:13 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 2908 of SEQ 1D
N0:13, b
is an integer of 15 to 2922, where both a and b correspond to the positions of


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
11
nucleotide residues shown in SEQ ID N0:13, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 4
This gene is expressed primarily in Human Ovary.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:14 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1356 of SEQ ID
N0:14, b
is an integer of 15 to 1370, where both a and b correspond to the positions of
nucleotide residues shown in SEQ >D N0:14, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 5
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
gb~AAD29427.1 ~ (all information available through the recited accession
number is
incorporated herein by reference) which is described therein as "(AF139185)
myomegalin [Rattus norvegicus]". This protein is believed to be a novel human
homolog of myomegalin. A partial alignment demonstrating the observed homology
is shown immediately below.
>gb~AAD29427.1~ (AF139185) myomegalin [Rattus norvegicus]
>sp~AAD29427~AAD29427
Myomegalin.
Length = 2324
Plus Strand HSPS:
Score = 3504 (1233.5 bits), Expect = 0.0, Sum P(2) = 0.0
Identities = 731/936 (78~), Positives = 780/936 (83~), Frame = +3
Q: 585 RDFEKHLNDLKKENFSLKLRIYFLEERMQQKYEASREDIYKRNIELKVEVESLKRELQDK 764
R +HLNDLKKENFSLKLRIYFLEERMQQKYE SRED+YKRNIELKVEVESLKRELQD+


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
12
S: 6 RTLSQHLNDLKKENFSLKLRIYFLEERMQQKYEVSREDWKRNIELKVEVESLKRELQDR 65
Q: 765 KQHLDKTWADVENLNSQNXXXXXXXXXXXXXXXXHWELLENKIQLLQEESRLAKNEAAR 944
KQHL KTWAD E+LNSQN HWELL+N IQLLQEESR AK+EA +
S S: 66 KQHLHKTWADEEDLNSQNEAELRRQVEEPQQETEHWELLDNNIQLLQEESRFAKDEATQ 125
Q: 945 MAALVEAEKECNLELSEKLKGVTKNWEDVPGDQVKPDQYTEALAQRDKRIEELNQSLAAQ 1124
M LVEAEK CNLELSE+ K TKN ED PGDQVK DQY+ ALAQRD+RIEEL QSLAAQ
S: 126 METLVEAEKGCNLELSERWKDATKNREDAPGDQVKLDQYSAALAQRDRRIEELRQSLAAQ 185
Q: 1125 ERLVEQLSREKQQLLHLLEEPTSMEVQPMTEELLKQQKLNSHETTITQQSVSDSHLAELQ 1304
E LVEQLSREKQQLLHLLEEP MEVQPM + L QQK + +ET TQ SVSDSHLAELQ
S: 186 EGLVEQLSREKQQLLHLLEEPGGMEVQPMPKGLPTQQKPDLNETPTTQPSVSDSHLAELQ 245
IS Q: 1305 EKIQQTEATNKILQEKLNEMSYELKCAQESSQKQDGTIQNLKETLKSRERETEELYQVIE 1484
+KIQQTE TNKILQEKLN+MS EL+ AQESSQKQD TIQ+LKE LKSRE ETEELYQVIE
S: 246 DKIQQTEVTNKILQEKLNDMSCELRSAQESSQKQDTTIQSLKEMLKSRESETEELYQVIE 305
Q: 1485 GQNDTMAKLREMLHQSQLGQLHSSEGTSPAQQQVALLDLQSALFCSQLEZQKLQRWRQK 1664
2O GQNDTMAKL EMLHQSQLGQL SSEG +PAQQQVALLDLQSALFCSQLEIQKLQR++RQK
S: 306 GQNDTMAKLPEMLHQSQLGQLQSSEGIAPAQQQVALLDLQSALFCSQLEIQKLQRLLRQK 365
Q: 1665 ERQLADAKQCVQFVEAAAHESEQQKEASWKHNQELRKALQQLQEELQNKSQQLRAWEAEK 1844
ERQLAD K+C+QFVEAAA E EQQKEA+WKHNQELRKALQ LQ EL +KSQQL EAEK
ZS S: 366 ERQLADGKRCMQFVEAAAQEREQQKEAAWKHNQELRKALQHLQGELHSKSQQLHVLEAEK 425
Q: 1845 YNEIRTQEQNIQHLNHSLSHKXXXXXXXXXXXXYRDNSDKTLEANEMLLEKLRQRIHDKA 2024
YNEIRTQ QNIQHL+HSLSHK YRD +DKTL+ NE+ LEKLRQRI D+A
S: 426 YNEIRTQGQNIQHLSHSLSHKEQLIQELQELLQYRDTTDKTLDTNEVFLEKLRQRIQDRA 485
Q: 2025 VALERAIDEKFSAXXXXXXXXXXXXXAXXXXXXXXXXXXXXXSSNEATMQSMESLLRAKG 2204
VALER IDEKFSA A S+NEATMQSMESLLRA+G
S: 486 VALERVIDEKFSALEEKDKELRQLRLAVRDRDHDLERLRCVLSANEATMQSMESLLRARG 545
3S Q: 2205 LEVEQLSTTCQNLQWLKEEMETKFSRWQKEQESIIQQLQTSLHDRNKEVEDLSATLLCKL 2384
LEVEQL TCQNLQWLKEE+ETKF WQKEQESIIQQLQTSLHDRNKEVEDLSATLL KL
S: 546 LEVEQLIATCQNLQWLKEELETKFGHWQKEQESIIQQLQTSLHDRNKEVEDLSATLLHKL 605
Q: 2385 GPGQSEIAEELCQRLQRKERMLQDLLSDRNKQVLEHEMEIQGLLQSVSTREQESQAAAEK 2564
4O GPGQSE+AEELCQRLQRKER+LQDLLSDRNKQ +EHEME+QGLLQS+ TREQE QA AEK
S: 606 GPGQSEVAEELCQRLQRKERVLQDLLSDRNKQAMEHEMEVQGLLQSMGTREQERQAVAEK 665
Q: 2565 LVQALMERNSELQALRQYLGGRDSLM-SQAPISNQQAEVTPTG-RLGKQTDQGSMQIPSR 2738
+VQA MERNSELQALRQYLGG++ + SQA ISNQ A T G G+QTDQGS Q+PSR
4S S: 666 MVQAFMERNSELQALRQYLGGKELMAASQAFISNQPAGATSVGPHHGEQTDQGSTQMPSR 725
Q: 2739 DDSTSLTAKEDVSIPRSTLGDLDTVAGLEKELSNXXXXXXXXXXXXXXSQMELSALQSMM 2918
DDSTSLTA+E+ SIPRSTLGD DTVAGLEKELSN SQ+ELSALQSMM
S: 726 DDSTSLTAREEASIPRSTLGDSDTVAGLEKELSNAKEELELMAKKERESQIELSALQSMM 785
S0
Q: 2919 AVQEEELQVQAADMESLTRNIQIKEDLIKDLQMQLVDPEDIPAMERLTQEVLLLREKVAS 3098
AVQEEELQVQAAD+ESLTRNIQIKEDLIKDLQMQLVDPED+PAMERLTQEVLLLREKVAS
S: 786 AVQEEELQVQAADLESLTRNIQIKEDLIKDLQMQLWPEDMPAMERLTQEVLLLREKVAS 845
SS Q: 3099 VESQGQEISGNRRQQLLLMLEGLVDERSRLNEALQAERQLYSSLVKFHAHPETSERDRTL 3278
VE QGQE S NRRQQLLLMLEGLVDERSRLNEALQAERQLYSSLVKFHA PET ERDRTL
S: 846 VEPQGQEGSENRRQQLLLMLEGLVDERSRLNEALQAERQLYSSLVKFHAQPETFERDRTL 905
Q: 3279 QVELEGAQVLRSRLEEVLGRSLERLNRLETLAAIGG 3386
6O QVELEGAQVLRSRLEEVLGRSLERL+RLETLAAIGG
S: 906 QVELEGAQVLRSRLEEVLGRSLERLSRLETLAAIGG 941
The segment of gbf AAD29427.1 ~ that is shown as "S" above is set out in the
sequence listing as SEQ ID NO: 94. Based on the structural similarity, these
6S homologous polypeptides are expected to share at least some biological
activities.
Such activities are known in the art, some of which are described elsewhere
herein.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
13
Assays for determining such activities are also known in the art, some of
which have
been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 95 which
S corresponds to the "Q" sequence in the alignment shown above (gaps
introduced in a
sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
Human Adult Heart,re-excision; Human Amygdala,re-excision; Stratagene muscle
937209 and to a lesser extent in Soares~regnant uterus NbHPU; Smooth Muscle
Serum Treated, Norm; Human Quadriceps; H, Human adult Brain Cortex tissue;
Human Hypothalamus,schizophrenia, re-excision; Human Placenta; Smooth
muscle,control; Nine Week Old Early Stage Human; Human Cerebellum; Primary
Dendritic Cells, lib 1 and Soares infant brain 1NIB.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ >D NO: 52 as residues: Arg-33 to Ser-40, Ala-44 to Thr-
51,
Gly-63 to Gly-71. Polynucleotides encoding said polypeptides are also
provided.
The tissue distribution in adult human heart, in combination with the
homology to the rat myomegalin indicates that polynucleotides and polypeptides
of
the invention are useful in the treatment, detection, and/or prevention of
cardiovascular diseases and/or disorders, particularly degenerative and
develpomental
diseases. Moreover, the protein is useful in the detection, treatment, and/or
prevention
of a variety of vascular disorders and conditions, which include, but are not
limited to
miscrovascular disease, vascular leak syndrome, aneurysm, stroke, embolism,
thrombosis, coronary artery disease, arteriosclerosis, and/or atherosclerosis.
Based
upon The tissue distribution of this protein in adult human heart, antagonists
directed against this protein may be useful in blocking the activity of this
protein.
Accordingly, preferred are antibodies which specifically bind a portion of the
translation product of this gene. Also provided is a kit for detecting tumors
in which
expression of this protein occurs. Such a kit comprises in one embodiment an
antibody specific for the translation product of this gene bound to a solid
support.
Also provided is a method of detecting these tumors in an individual which
comprises
a step of contacting an antibody specific for the translation product of this
gene to a


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
14
bodily fluid from the individual, preferably serum, and ascertaining whether
antibody
binds to an antigen found in the bodily fluid. Preferably the antibody is
bound to a
solid support and the bodily fluid is serum. The above embodiments, as well as
other
treatments and diagnostic tests (kits and methods), are more particularly
described
elsewhere herein. Furthermore, the protein may also be used to determine
biological
activity, to raise antibodies, as tissue markers, to isolate cognate ligands
or receptors,
to identify agents that modulate their interactions, in addition to its use as
a nutritional
supplement. 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.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:15 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 3768 of SEQ ID
N0:15, b
is an integer of 15 to 3782, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:15, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 6
When tested against MVEC cell lines, supernatants removed from cells
containing this gene activated the ICAM-1 promoter element. Thus, it is likely
that
this gene activates endothelial cells, and more particularly vascular cells,
through the
ICAM-1. ICAM-1 is found on the cell surface of endothelial cells, smooth
muscle
cells, epithelial cells, and fibroblasts. It binds to its ligand, LFA-l, a
heterodimer
complex that is a member of the leukocyte integrin family of cell adhesion
receptors.
Inflammatory mediators and cytokines, such as, IL-1, TNFa and IFNg, stimulate
ICAM-1 expression on vascular cells, in addition to the aforementioned cells
and
tissue cell types. Polypeptides which increase ICAM expression are useful in
the


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
treatment of cancer, cardiovascular, autoimmune and inflammatory diseases
and/or
disorders.
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares infant brain 1NIB; normalized infant brain cDNA and to a lesser extent
in
5 Apoptotic T-cell, re-excision; Human Chronic Synovitis; Human Adipose; CD34
depleted Buffy Coat (Cord Blood), re-excision and Anergic T-cell.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:16 and may have been publicly available prior to
conception of
10 the present invention. Preferably, such related polynucleotides are
specifically
excluded from the scope of the present invention. To list every related
sequence
would be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 994 of SEQ ID
N0:16, b
15 is an integer of 15 to 1008, where both a and b correspond to the positions
of
nucleotide residues shown in SEQ ID N0:16, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 7
This gene is expressed primarily in the following tissues/cDNA libraries:
Human Thymus; L428.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:17 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1751 of SEQ ID
N0:17, b
is an integer of 15 to 1765, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:17, and where b is greater than or
equal to a
+ 14.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
16
FEATURES OF PROTEIN ENCODED BY GENE NO: 8
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ m NO: 96.
Polynucleotides encoding these polypeptides are also provided.
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares infant brain 1NIB and to a lesser extent in Human Tongue, frac 1; Human
Microvascular Endothelial Cells, fract. B; NTERA2 teratocarcinoma cell
line+retinoic
acid (14 days); KMH2; Human Fetal Dura Mater; Human Uterine Cancer; Human
Adult Testes, Large Inserts, Reexcision; Human Hypothalmus,Schizophrenia;
Human
Chondrosarcoma; Human Thymus; Macrophage (GM-CSF treated); Rejected Kidney,
lib 4; Human adult testis, large inserts; Colon Carcinoma; Adipocytes; Human
Primary Breast Cancer Reexcision; Human Microvascular Endothelial Cells,
fract. A
and Soares fetal liver spleen 1NFLS.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 55 as residues: Thr-70 to Ala-77. Polynucleotides
encoding said polypeptides are also provided.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:18 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1103 of SEQ ID
N0:18, b
is an integer of 15 to 1117, where both a and b correspond to the positions of
nucleotide residues shown in SEQ >D NO:l 8, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 9
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 97 and/or
SEQ ID
NO: 98. Polynucleotides encoding these polypeptides are also provided.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
17
This gene is expressed primarily in the following tissues/cDNA libraries:
Primary Dendritic cells,frac 2; T Cell helper I and to a lesser extent in
Human
Osteoblasts II; Primary Dendritic Cells, lib 1; Hodgkin's Lymphoma I and T
cell
helper II.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 56 as residues: Arg-63 to Phe-72, Ile-114 to Phe-
120.
Polynucleotides encoding said polypeptides are also provided.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:19 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1536 of SEQ ID
N0:19, b
is an integer of 15 to 1550, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:19, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 10
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 99.
Polynucleotides encoding these polypeptides are also provided.
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares infant brain 1NIB and to a lesser extent in Stratagene colon (#937204);
Soares
fetal liver spleen 1NFLS; Human Liver; Human Umbilical Vein, Reexcision; 12
Week Old Early Stage Human, II; Stratagene HeLa cell s3 937216; Human Ovary;
Colon Tumor; Human T-Cell Lymphoma; Early Stage Human Brain; Human Fetal
Lung III; Human Ovarian Cancer Reexcision; Nine Week Old Early Stage Human;
Soares total fetus Nb2HF8 9w; NTERA2 teratocarcinoma cell line+retinoic acid
(14 days); Soares~arathyroid tumor NbHPA; Human Pancreas Tumor, Reexcision;
Stratagene HeLa cell s3 937216; Human Adipose; Human Chondrosarcoma;


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
18
Ulcerative Colitis; Synovial Fibroblasts (control); Soares total fetus Nb2HF8
9w;
Stratagene neuroepithelium (#937231); Soares testis NHT; Soares breast
3NbHBst;
Human Placenta; NCI CGAP_GCB1; Normal colon; Soares melanocyte 2NbHM;
Soares-placenta 8to9weeks 2NbHP8to9W; 12 Week Early Stage Human II,
Reexcision; Human Amygdala; Smooth muscle,control; Colon Normal III; Hodgkin's
Lymphoma II; Osteoblasts; Human Cerebellum; Soares placenta Nb2HP; Primary
Dendritic Cells, lib 1; Kidney Pyramids; Spleen/normal; Stomach Tumour; Human
Normal Cartilage,Fraction I; Human Kidney; Human Fetal Brain, random primed;
Morton Fetal Cochlea; H. Striatum Depression, subt; Human Cardiomyopathy,
subtracted; Smooth muscle, control, re-excision; human colon cancer; Amniotic
Cells
TNF induced; Human Lung; Early Stage Human Lung, subtracted; Human
Quadriceps; Human Soleus; Human Adult Heart,re-excision;
Soares fetal liver spleen 1NFLS_Sl; Smooth Muscle- HASTE normalized; Human
Tonsils, Lib 2; Human Synovium; B-cells (unstimulated); Jurkat T-Cell, S
phase;
Gessler Wilms tumor; Soares fetal heart-NbHHI9W;
Soares fetal liver spleen 1NFLS_S1; Mo7e Cell Line GM-CSF treated (lng/ml);
Soares NFL T GBC Sl; Soares total fetus Nb2HF8 9w;
Soares-placenta 8to9weeks 2NbHP8to9W; Ovary, Cancer: Poorly differentiated
adenocarcinoma (9809C332); Human pancreatic islet; L428; Human Fetal Kidney;
human ovarian cancer; Merkel Cells; Ovary, Normal:::::::: (v>':9805C040R);
Soares testis NHT; Human Hippocampus; Human Placenta (re-excision); Human
Testes Tumor, re-excision; Bone Marrow Stromal Cell, untreated; CHME Cell
Line,treated 5 hrs; Stratagene muscle 937209; Rejected Kidney, lib 4;
NCI CGAP Panl; NCI CGAP-Brn23; NCI CGAP_Brn25; NCI CGAP_Brn35;
NCI CGAP_Kidl l; Soares~lacenta 8to9weeks 2NbHP8to9W; Macrophage-
oxLDL, re-excision; Human Gall Bladder; Pancreas Islet Cell Tumor; Fetal
Heart;
Colon Carcinoma; Human Testes Tumor; Clontech human aorta polyA+ mRNA
(#6572); Human fetal heart, Lambda ZAP Express; Human Fetal Kidney,
Reexcision;
Human Synovial Sarcoma; CD34+DIRECTIONAL;
Soares fetal liver spleen-1NFLS_S1; Human Fetal Heart; Activated T-Cell
(l2hs)/Thiouridine labelledEco; Endothelial cells-control; Human Testes; H.
Frontal
cortex,epileptic,re-excision; Human Endometrial Tumor; Human 8 Week Whole


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
19
Embryo; Keratinocyte; NCI CGAP GC 1; NCI CGAP LuS; NCI CGAP-GCB 1;
NCI CGAP Kid6; NCI CGAP_Lei2; NCI CGAP_Lip2; NCI CGAP_Prl2 and
Colon Tumor II.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:20 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 4101 of SEQ ID
N0:20, b
is an integer of 1 S to 4115, where both a and b correspond to the positions
of
nucleotide residues shown in SEQ ID N0:20, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 11
The translation product of this gene shares sequence homology to a potassium
channel protein. Based on the sequence similarity, the translation product of
this clone
is expected to share at least some biological activities with potassium
channel
proteins. Such activities are known in the art, some of which are described
elsewhere
herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 100.
Polynucleotides encoding these polypeptides are also provided.
This gene is expressed primarily in the following tissues/cDNA libraries:
Primary Dendritic Cells, lib 1 and to a lesser extent in TF-1 Cell Line GM-CSF
Treated; CD34 positive cells (Cord Blood); T cell helper II; Pharynx
carcinoma; Ku
812F Basophils Line; Human Colon, subtraction; H. Meningima, M1; Human
Rhabdomyosarcoma; Epithelial-TNFa and INF induced; breast lymph node CDNA
library; Human Placenta; Soares fetal heart NbHHI9W; Activated T-
cell(12h)/Thiouridine-re-excision; NCI CGAP_AA1; NCI CGAP-ColO;
Soares~regnant uterus NbHPU; Osteoblasts and Keratinocyte.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 58 as residues: Met-1 to Leu-10. Polynucleotides
encoding said polypeptides are also provided.
The tissue distribution in primary dendritic cells indicates the protein
product
5 of this clone is useful for the diagnosis and treatment of a variety of
immune system
disorders. Representative uses are described in the "Immune Activity" and
"Infectious
Disease" sections below, in Example 11, 13, 14, 16, 18, 19, 20, and 27, and
elsewhere
herein. Briefly, the expression of this gene product indicates a role in
regulating the
proliferation; survival; differentiation; and/or activation of hematopoietic
cell
10 lineages, including blood stem cells. This gene product is involved in the
regulation
of cytokine production, antigen presentation, or other processes suggesting a
usefulness in the treatment of cancer (e.g. by boosting immune responses).
Since the
gene is expressed in cells of lymphoid origin, the natural gene product is
involved in
immune functions. Therefore it is also useful as an agent for immunological
disorders
15 including arthritis, asthma, immunodeficiency diseases such as AIDS,
leukemia,
rheumatoid arthritis, granulomatous disease, inflammatory bowel disease,
sepsis,
acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, such as T-cell
mediated
cytotoxicity; immune reactions to transplanted organs and tissues, such as
host-
versus-graft and graft-versus-host diseases, or autoimmunity disorders, such
as
20 autoimmune infertility, tense tissue injury, demyelination, systemic lupus
erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjogren's
disease, and scleroderma. Moreover, the protein may represent a secreted
factor that
influences the differentiation or behavior of other blood cells, or that
recruits
hematopoietic cells to sites of injury. Thus, this gene product is thought to
be useful
in the expansion of stem cells and committed progenitors of various blood
lineages,
and in the differentiation and/or proliferation of various cell types.
Considering this
proteins homology to potassium channel proteins, antagonists of this protein
would be
useful therapeutically in inihibiting immune responses, signal transduction,
and other
cellular responses. Based upon the tissue distribution of this protein,
antagonists
directed against this protein may be useful in blocking the activity of this
protein.
Accordingly, preferred are antibodies which specifically bind a portion of the
translation product of this gene. Also provided is a kit for detecting tumors
in which


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
21
expression of this protein occurs. Such a kit comprises in one embodiment an
antibody specific for the translation product of this gene bound to a solid
support.
Also provided is a method of detecting these tumors in an individual which
comprises
a step of contacting an antibody specific for the translation product of this
gene to a
bodily fluid from the individual, preferably serum, and ascertaining whether
antibody
binds to an antigen found in the bodily fluid. Preferably the antibody is
bound to a
solid support and the bodily fluid is serum. The above embodiments, as well as
other
treatments and diagnostic tests (kits and methods), are more particularly
described
elsewhere herein. Furthermore, the protein may also be used to determine
biological
activity, raise antibodies, as tissue markers, to isolate cognate ligands or
receptors, to
identify agents that modulate their interactions, in addition to its use as a
nutritional
supplement. 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.
Many polynucleotide sequences, such as EST sequences, are publicly
1 S available and accessible through sequence databases. Some of these
sequences are
related to SEQ ID N0:21 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 2472 of SEQ ID
N0:21, b
is an integer of 15 to 2486, where both a and b correspond to the positions of
nucleotide residues shown in SEQ 1D N0:21, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 12
This gene is expressed primarily in the following tissues/cDNA libraries:
CD34 positive cells (cord blood),re-ex; Testes and to a lesser extent in CD34
depleted
Buffy Coat (Cord Blood), re-excision; CD34 positive cells (Cord Blood); H.
Whole
Brain #2, re-excision; Human Whole Brain, re-excision; Human Whole Brain #2 -
,
Oligo dT > I.SKb; CD34 depleted Buffy Coat (Cord Blood); Apoptotic T-cell;
Anergic T-cell; T cell helper II and Primary Dendritic Cells, lib 1.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
22
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:22 and may have been publicly available prior to
conception of
the present invention. Preferably, such related polynucleotides are
specifically
S excluded from the scope of the present invention. To list every related
sequence
would be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1323 of SEQ ID
N0:22, b
is an integer of 15 to 1337, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:22, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 13
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares infant brain 1NIB and to a lesser extent in normalized infant brain
cDNA;
Infant brain, Bento Soares; Human Fetal Brain; Human Fetal Brain; Soares
melanocyte 2NbHM; Early Stage Human Brain; NCI CGAP_LuS; Human 8 Week
Whole Embryo; Nine Week Old Early Stage Human; Tongue carcinoma; H. Striatum
Depression, subt; Frontal lobe,dementia,re-excision; Raji Cells, cyclohexamide
treated; Human Synovium; Human Hypothalamus,schizophrenia, re-excision;
Soares total fetus Nb2HF8 9w; Alzheimers, spongy change; Human Amygdala,re-
excision; Human Osteosarcoma; Stromal cell TF274; 12 Week Old Early Stage
Human; Human Substantia Nigra; Brain frontal cortex; Human Synovial Sarcoma;
Activated T-Cell (l2hs)/Thiouridine labelledEco; Human Amygdala; Smooth
muscle,control; H. Frontal cortex,epileptic,re-excision; Gessler Wilms tumor;
Human
colorectal cancer; Soares NFL T GBC_S 1; Stratagene schizo brain S 11;
Activated
T-cell(12h)/Thiouridine-re-excision; Soares NhHMPu S1 and Keratinocyte.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 60 as residues: Trp-19 to Ala-24, Gly-32 to Met-
39.
Polynucleotides encoding said polypeptides are also provided.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
23
related to SEQ ID N0:23 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1887 of SEQ ID
N0:23, b
is an integer of 15 to 1901, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:23, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 14
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares-parathyroid tumor NbHPA and to a lesser extent in T-Cell PHA 16 hrs;
Human fetal heart, Lambda ZAP Express; Soares fetal liver spleen 1NFLS_S1;
Soares senescent fibroblasts NbHSF; NCI CGAP-GCB1; Colon Tumor II;
Soares~lacenta 8to9weeks 2NbHP8to9W; Soares infant brain 1NIB; Normal
Human Trabecular Bone Cells; Soares testis NHT; NCI CGAP-Pr23; Pancreas Islet
Cell Tumor; Soares-pregnant uterus NbHPU; Soares-parathyroid tumor NbHPA;
Spleen, Chronic lymphocytic leukemia; NCI CGAP-LuS; NCI CGAP Kid3;
NCI CGAP Kids; NCI CGAP Brn25; Soares~regnant uterus NbHPU; T cell
helper II; Soares fetal liver spleen 1NFLS; Human Old Ovary; Human Leukocytes;
Lung, Cancer: Poorly-Differentiated Squamous Cell carcinoma (4005313 A3);
Ovary,
Cancer: (4004332 A2); Human Epididymus; Synovial Fibroblasts (Ill/TNF), subt;
Human Infant Brain; Soares multiple sclerosis 2NbHMSP;
Soares-placenta 8to9weeks 2NbHP8to9W; Apoptotic T-cell; Bone Marrow Stromal
Cell, untreated; Human Fetal Brain; Gessler Wilms tumor; Infant brain, Bento
Soares;
Soares NFL T GBC_S1; Soares fetal heart NbHHI9W;
Soares total fetus Nb2HF8 9w; Stratagene lung (#937210); normalized infant
brain
cDNA; Human fetal heart, Lambda ZAP Express; Human retina cDNA Tsp509I-
cleaved sublibrary; NCI CGAP_GC4; NCI CGAP_OvB; NCI CGAP_Schl; Human
Testes Tumor; Human Fetal Lung III; Human Synovial Sarcoma; B-cells
(stimulated); Bone marrow; human tonsils; Anergic T-cell; Human fetal heart,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
24
Lambda ZAP Express; Soares fetal heart_NbHHI9W;
Soares-pregnant uterus NbHPU; Soares total fetus Nb2HF8 9w;
NCI CGAP-Gas4; NCI CGAP LymS; NCI CGAP_Pr28; NCI CGAP_Brn52;
NCI CGAP-Lyml2; Soares NFL T GBC_S1 and Soares fetal lung NbHLI9W.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:24 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1085 of SEQ ID
N0:24, b
is an integer of 1 S to 1099, where both a and b correspond to the positions
of
nucleotide residues shown in SEQ ID N0:24, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 15
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 101.
Polynucleotides encoding these polypeptides are also provided.
This gene is expressed primarily in the following tissues/cDNA libraries:
HMl; Soares total fetus Nb2HF8 9w; Soares~arathyroid tumor NbHPA; Lung
Mesothelium; Supt Cells, cyclohexamide treated; STROMAL -OSTEOCLASTOMA;
Hepatocellular Tumor; Amniotic Cells - Primary Culture; H. Lymph node breast
Cancer; L428; Human umbilical vein endothelial cells, IL-4 induced;
Hepatocellular
Tumor, re-excision; Stratagene liver (#937224); Macrophage-oxLDL, re-excision;
12
Week Old Early Stage Human; Human Primary Breast Cancer Reexcision; Anergic
T-cell; Primary Dendritic Cells, lib 1.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 62 as residues: Leu-43 to Glu-48, Gly-90 to Cys-
96,
Asn-99 to Pro-113. Polynucleotides encoding said polypeptides are also
provided.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ >D N0:25 and may have been publicly available prior to
conception of
the present invention. Preferably, such related polynucleotides are
specifically
5 excluded from the scope of the present invention. To list every related
sequence
would be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 654 of SEQ ID
N0:25, b
is an integer of 1 S to 668, where both a and b correspond to the positions of
10 nucleotide residues shown in SEQ ID N0:25, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 16
This gene is expressed primarily in the following tissues/cDNA libraries:
15 Human Cerebellum; Soares NhHMPu S1; H. cerebellum, Enzyme subtracted.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ >D N0:26 and may have been publicly available prior to
conception of
the present invention. Preferably, such related polynucleotides are
specifically
20 excluded from the scope of the present invention. To list every related
sequence
would be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 982 of SEQ ID
N0:26, b
is an integer of 15 to 996, where both a and b correspond to the positions of
25 nucleotide residues shown in SEQ ID N0:26, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 17
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ~ NO: 102.
Polynucleotides encoding these polypeptides are also provided.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
26
This gene is expressed primarily in the following tissues/cDNA libraries:
NCI CGAP Kidl l; Colon Carcinoma.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
S related to SEQ ID N0:27 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 638 of SEQ >D
N0:27, b
is an integer of 15 to 652, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:27, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 18
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 103.
Polynucleotides encoding these polypeptides are also provided.
This gene is expressed primarily in the following tissues/cDNA libraries:
Human adult testis, large inserts; Soares infant brain 1NIB and to a lesser
extent in
Human Adult Testes, Large Inserts, Reexcision; breast lymph node CDNA library;
Human Primary Breast Cancer Reexcision; Stratagene fetal spleen (#937205);
NCI CGAP_Lul; Soares adult brain N2b5HB55Y; Brain frontal cortex;
NCI CGAP-Brn25; Soares_placenta 8to9weeks_2NbHP8to9W; Human
Cerebellum; Bone Cancer, re-excision; Soares ovary tumor NbHOT; Bone Cancer;
H.
Whole Brain #2, re-excision; Soares adult brain N2b4HBS5Y; Stratagene placenta
(#937225); NTERA2 + retinoic acid, 14 days; Human Frontal Cortex,
Schizophrenia;
Soares testis NHT; Soares fetal liver spleen-1NFLS S1; normalized infant brain
cDNA; Human Infant Brain; Soares fetal liver spleen 1NFLS S1; Human Fetal
Kidney; Human retina cDNA randomly primed sublibrary; NCI CGAP_KidS;
Human Umbilical Vein Endothelial Cells, uninduced; Human umbilical vein


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
27
endothelial cells, IL-4 induced; NCI CGAP_GCB1; Stratagene colon (#937204);
Rejected Kidney, lib 4; NCI CGAP_CoB; Soares senescent fibroblasts NbHSF;
Human Fetal Lung III; Human Synovial Sarcoma; Soares NhHMPu S1;
Soares fetal lung NbHLI9W; Soares fetal heart NbHHI9W; HUMAN B CELL
LYMPHOMA; NCI CGAP-GCB1; Spleen, Chronic lymphocytic leukemia; Human
Endometrial Tumor; Keratinocyte; Soares fetal liver spleen 1NFLS; Human
Uterine
Cancer, subtracted; Gessler Wilms tumor; Soares testis NHT; Colon, tumour;
Human Pituitary; Larynx Normal; Human Colon, subtraction; Human Colon; Human
Adult Retina; Smooth Muscle Serum Treated, Norm; Smooth muscle, control, re-
excision; Human Primary Breast Cancer; H. Epididiymus, cauda; Human Soleus;
Soares total fetus Nb2HF8 9w; Human Adult Heart,re-excision; Cem cells
cyclohexamide treated; Raji Cells, cyclohexamide treated; Human Normal Breast;
Human Tonsils, Lib 2; Human Lung Cancer,re-excision; B-cells (unstimulated);
Jurkat T-cell G1 phase; Jurkat T-Cell, S phase; H. Meningima, M1; Human Manic
Depression Tissue; Spleen metastic melanoma; 1-NIB; Gessler Wilms tumor; Human
epidermal keratinocyte; Infant brain, Bento Soares; Soares NFL T GBC_S1;
Soares_parathyroid tumor NbHPA; Soares senescent fibroblasts NbHSF; H.
Lymph node breast Cancer; Human Bone Marrow, re-excision; Human Brain,
Striatum; Human Fetal Dura Mater; Human heart cDNA (YNakamura);
NCI CGAP_Co2; NCI CGAP_Co3; NCI CGAP-Co4; NCI CGAP_GC4;
NCI CGAP_LuS; NCI CGAP_CoIO; NCI CGAP Prl2; Macrophage-oxLDL;
Human Adipose; Soares breast 2NbHBst; Human Fetal Brain; Stratagene liver
(#937224); Fetal Heart; NCI CGAP_KidS; NCI CGAP-Brn25; CHME Cell
Line,untreated; Human Substantia Nigra; Soares fetal lung NbHLI9W;
Soares testis NHT; Soares total fetus Nb2HF8 9w; Human Fetal Kidney,
Reexcision; Human Testes, Reexcision; Bone marrow; Human Adult Pulmonary,re-
excision; NCI CGAP_Gas4; NCI CGAP-Brn52; NCI CGAP-Lyml2;
Soares_testis NHT; Soares fetal liver spleen-1NFLS_S1; Anergic T-cell; Human
Microvascular Endothelial Cells, fract. A; Monocyte activated; Human Testes;
Human 8 Week Whole Embryo; NCI CGAP ColO; Soares testis NHT; Colon
Tumor II; Nine Week Old Early Stage Human and Infant brain, LLNL array of Dr.
M.
Soares 1NIB.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
28
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:28 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 2190 of SEQ >D
N0:28, b
is an integer of 15 to 2204, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:28, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 19
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 104,SEQ ID
NO:
105 and/or SEQ >D NO: 106. Polynucleotides encoding these polypeptides are
also
provided.
This gene is expressed primarily in the following tissues/cDNA libraries:
Salivary Gland, Lib 2; Ovary, Cancer: Grade II Papillary Carcinoma (1935AIF);
Soares NhHMPu S1; Stratagene NT2 neuronal precursor 937230;
NCI CGAP_Brn23; Soares NhHMPu S1; Keratinocyte; Primary Dendritic Cells, lib
1 and to a lesser extent in Osteoclastoma-normalized A; Ficolled Human Stromal
Cells, 5Fu treated; Human Prostate Cancer, Stage B2 fraction; H. Striatum
Depression, subt; Human colon carcinoma (HCC) cell line, remake; Fetal Heart,
re-
excision; Breast, Cancer: (4005522 A2); Human Whole Brain, re-excision; pBMC
stimulated w/ poly I/C; Human Stomach,re-excision;
Soares-pregnant uterus NbHPU; Soares fetal liver spleen-1NFLS S1; H. Ovarian
Tumor, II, OV5232; T-Cell PHA 16 hrs; KMH2; Human Osteoblasts II; Merkel
Cells;
Liver, Hepatoma; Ulcerative Colitis; Human Thymus Stromal Cells; Human Adrenal
Gland Tumor; Rejected Kidney, lib 4; NCI CGAP-Col4; NCI CGAP-Gas4;
NCI CGAP HSC2; Soares NhHMPu S1;


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
29
Soares_placenta 8to9weeks 2NbHP8to9W; Ovarian Tumor 10-3-95; NTERA2,
control; Human Ovary; Human Placenta; Human Testes Tumor; B-cells
(stimulated);
Human adult (K.Okubo); Human fetal heart, Lambda ZAP Express;
NCI CGAP_Br2; NCI CGAP_GCS; NCI CGAP-Pr22; STRATAGENE Human
skeletal muscle cDNA library, cat. #936215.; Soares_pregnant uterus NbHPU and
Soares-parathyroid tumor NbHPA.
When tested against Jurkat E and U937 myeloid cell lines, supernatants
removed from cells containing this gene activated the GAS assay. Thus, it is
likely
that this gene activates T-cells and myeloid cells through the Jak-STAT signal
transduction pathway. The gamma activating sequence (GAS) is a promoter
element
found upstream of many genes which are involved in the Jak-STAT pathway. The
Jak-STAT pathway is a large, signal transduction pathway involved in the
differentiation and proliferation of cells. Therefore, activation of the Jak-
STAT
pathway, reflected by the binding of the GAS element, can be used to indicate
proteins involved in the proliferationand differentiation of cells.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ >D NO: 66 as residues: Lys-55 to Thr-63, Thr-96 to Glu-
103,
Arg-123 to Glu-136, Ala-152 to Glu-166, Glu-226 to Arg-234, Leu-248 to Gln-
253,
Glu-273 to Glu-282. Polynucleotides encoding said polypeptides are also
provided.
The tissue distribution in immune cells indicates the polynucleotides and
polypeptides corresponding to this gene would be useful for the diagnosis and
treatment of a variety of immune system disorders. Representative uses are
described
in the "Immune Activity" and "Infectious Disease" sections below, in Example 1
l,
13, 14, 16, 18, 19, 20, and 27, and elsewhere herein. Briefly, the expression
indicates
a role in regulating the proliferation; survival; differentiation; and/or
activation of
hematopoietic cell lineages, including blood stem cells. Involvement in the
regulation
of cytokine production, antigen presentation, or other processes suggests a
usefulness
for treatment of cancer (e.g. by boosting immune responses). Expression in
cells of
lymphoid origin, indicates the natural gene product would be involved in
immune
functions. Therefore it would also be useful as an agent for immunological
disorders
including arthritis, asthma, immunodeficiency diseases such as A>DS, leukemia,
rheumatoid arthritis, granulomatous disease, inflammatory bowel disease,
sepsis,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, such as T-cell
mediated
cytotoxicity; immune reactions to transplanted organs and tissues, such as
host-
versus-graft and graft-versus-host diseases, or autoimmunity disorders, such
as
autoimmune infertility, lense tissue injury, demyelination, systemic lupus
5 erythematosis, drug induced hemolytic anemia, rheumatoid arthritis,
Sjogren's
disease, and scleroderma. Moreover, the protein may represent a secreted
factor that
influences the differentiation or behavior of other blood cells, or that
recruits
hematopoietic cells to sites of injury. Thus, this gene product is thought to
be useful
in the expansion of stem cells and committed progenitors of various blood
lineages,
10 and in the differentiation and/or proliferation of various cell types.
Furthermore, the
protein may also be used to determine biological activity, raise antibodies,
as tissue
markers, to isolate cognate ligands or receptors, to identify agents that
modulate their
interactions, in addition to its use as a nutritional supplement. Protein, as
well as,
antibodies directed against the protein may show utility as a tumor marker
and/or
15 immunotherapy targets for the above listed tissues.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:29 and may have been publicly available prior to
conception of
the present invention. Preferably, such related polynucleotides are
specifically
20 excluded from the scope of the present invention. To list every related
sequence
would be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 2913 of SEQ ID
N0:29, b
is an integer of 15 to 2927, where both a and b correspond to the positions of
25 nucleotide residues shown in SEQ ID N0:29, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 20
This gene is expressed primarily in the following tissues/cDNA libraries:
30 Soares total fetus Nb2HF8 9w and to a lesser extent in Colon Carcinoma;
Primary
Dendritic cells,frac 2; Human adult (K.Okubo); Colon Tumor II and Primary
Dendritic Cells, lib 1.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
31
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:30 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 783 of SEQ ID
N0:30, b
is an integer of 15 to 797, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:30, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 21
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 107.
Polynucleotides encoding these polypeptides are also provided.
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares fetal liver spleen 1NFLS and to a lesser extent in Human Cerebellum; H.
Frontal cortex,epileptic,re-excision; Human Adrenal Gland Tumor; Human Testes
Tumor; 12 Week Early Stage Human II, Reexcision; Human Chondrosarcoma; Nine
Week Old Early Stage Human; Human Synovial Sarcoma; 12 Week Old Early Stage
Human; Human Whole Brain #2 - Oligo dT > l.SKb; 12 Week Old Early Stage
Human, II; Human Fetal Brain; Human Placenta; Human Fetal Kidney, Reexcision;
Human Placenta; Smooth muscle,control; Soares infant brain 1NIB; Human Fetal
Liver- Enzyme subtraction; H. cerebellum, Enzyme subtracted; Soares retina
N2b4HR; H. Kidney Cortex, subtracted; Bone Marrow Stromal Cell, untreated;
Soares breast 2NbHBst; NCI CGAP_LuS; Human Substantia Nigra;
Soares fetal liver spleen-1NFLS S1; Human 8 Week Whole Embryo;
NCI CGAP_LuS; Human Fetal Liver, subtracted, neg clone; Human Fetal Liver,
subtracted; Human 8 Week Whole Embryo, subtracted; Human Umbilical Vein
Endothelial Cells, fract. A; Frontal lobe,dementia,re-excision; H. Whole Brain
#2, re-
excision; Myoloid Progenitor Cell Line; Human Infant Brain;


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
32
Soares fetal liver spleen_1NFLS S1; Olfactory epithelium,nasalcavity; Human
Hippocampus; Human Thymus; Human Whole Six Week Old Embryo; Human
Ovary; Human Gall Bladder; Fetal Heart; Human Fetal Heart; Endothelial cells-
control; Human Microvascular Endothelial Cells, fract. A; Osteoblasts;
NCI CGAP AR1; Human Pediatric Kidney; Human Infant Adrenal Gland,
subtracted; Brain, normal; Placenta; Human Fetal Heart, Differential (Fetal-
Specific);
Human Normal Cartilage Fraction IV; Ku 812F Basophils Line; Human
Fibrosarcoma; STRIATUM DEPRESSION; Human Pituitary, re-excision; Human
Cerebellum, subtracted; Human Fetal Spleen; Healing Abdomen wound,70&90 min
post incision; Human Placenta; Human Fetal Bone; Early Stage Human Lung,
subtracted; Messangial cell, frac 2; Human Lung Cancer,re-excision; Human
Whole
Brain, re-excision; Soares adult brain N2b4HB55Y; Human
Hypothalamus,schizophrenia, re-excision; Stratagene placenta (#937225);
Alzheimers, spongy change; Human Amygdala,re-excision; Synovial Fibroblasts
(Ill/TNF), subt; Pancreatic Islet; Soares_fetal lung NbHLI9W; H. Meningima,
M1;
Human Manic Depression Tissue; Human Umbilical Vein, Reexcision; TF-1 Cell
Line GM-CSF Treated; human ovarian cancer; Human Osteoblasts II; Human Fetal
Dura Mater; Human Adult Testes, Large Inserts, Reexcision; Ovary, Normal::::':
(..9805C040R); Spinal cord; Human Rhabdomyosarcoma; Soares adult brain
N2bSHB55Y; Human Thymus Stromal Cells; Rejected Kidney, lib 4; Human Liver,
normal; NCI CGAP Brn35; Smooth muscle, serum induced,re-exc; Fetal Liver,
subtraction II; Soares testis NHT; Brain frontal cortex; Early Stage Human
Brain;
Soares NFL T GBC Sl; Soares NSF F8 9W OT PA P S1;
Soares fetal liver spleen_1NFLS S1; Stratagene schizo brain 511; Human Fetal
Lung III; Endothelial-induced; Soares fetal heart NbHHI9W;
Soares fetal heart_NbHHI9W; NCI CGAP-GC6; NCI CGAP_LuS;
NCI CGAP_Brn25; Soares_fetal liver spleen-1NFLS S1; NCI CGAP_Pr4.l;
Soares placenta Nb2HP; Human adult (K.Okubo); Human fetal heart, Lambda ZAP
Express and NCI CGAP-Ewl.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:31 and may have been publicly available prior to
conception of


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
33
the present invention. Preferably, such related polynucleotides are
specifically
excluded from the scope of the present invention. To list every related
sequence
would be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 4459 of SEQ ID
N0:31, b
is an integer of 15 to 4473, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:31, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 22
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 108 and/or
SEQ
ID NO: 109. Polynucleotides encoding these polypeptides are also provided.
This gene is expressed primarily in the following tissues/cDNA libraries:
Human 8 Week Whole Embryo and to a lesser extent in Human Amygdala; Nine
Week Old Early Stage Human; Human Cerebellum; Human Ovarian Cancer
Reexcision; Soares fetal heart_NbHHI9W; Human Brain, Striatum; Human Fetal
Brain; Rejected Kidney, lib 4; Soares infant brain 1NIB; Adrenal Gland,normal;
Soares ovary tumor NbHOT; Human Hippocampus, subtracted; Breast, Cancer:
(9806C012R); H. Atrophic Endometrium; Fetal Heart, re-excision;
Soares~regnant uterus NbHPU; Stratagene corneal stroma (#937222); Stratagene
NT2 neuronal precursor 937230; Human Neutrophils, Activated, re-excision;
Amniotic Cells - TNF induced; NTERA2 teratocarcinoma cell line+retinoic acid
(14
days); Breast Cancer cell line, MDA 36; H Female Bladder, Adult; Pancreatic
Islet;
Soares fetal lung NbHLI9W; Synovial hypoxia; Breast, Normal: (4005522:::::::
B2);
L428; 12 Week Old Early Stage Human, II; Human Uterine Cancer;
NCI CGAP-LuS; Human Placenta (re-excision); Bone Marrow Stromal Cell,
untreated; Human Adrenal Gland Tumor; NTERA2, control; Macrophage-oxLDL, re-
excision; Resting T-Cell Library,II; Soares testis NHT; Soares NFL T GBC_S1;
Soares total fetus Nb2HF8 9w; normalized infant brain cDNA; Human
Eosinophils; Human Synovial Sarcoma; NCI CGAP Ut2; NCI CGAP-Lu24;
Schiller oligodendroglioma; Soares fetal lung NbHLI9W;


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
34
Soares fetal heart NbHHI9W; Stanley Frontal SN pool 2; Human retina cDNA
Tsp509I-cleaved sublibrary; NCI CGAP-Br2; NCI CGAP-Ewl; NCI CGAP-LuS;
NCI CGAP-GCBl; Human Testes, Reexcision; Bone marrow; 12 Week Early Stage
Human II, Reexcision; Endothelial-induced; Anergic T-cell; HUMAN B CELL
LYMPHOMA; Human Bone Marrow, treated; Bone Marrow Cell Line (RS4,11);
NCI CGAP_Co 14; H. Frontal cortex,epileptic,re-excision; Human Endometrial
Tumor; Hodgkin's Lymphoma II and Soares fetal liver spleen 1NFLS.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 69 as residues: Pro-46 to Ala-57, Ser-74 to Glu-
94,
Gly-104 to Ser-110. Polynucleotides encoding said polypeptides are also
provided.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:32 and may have been publicly available prior to
conception of
the present invention. Preferably, such related polynucleotides are
specifically
1 S excluded from the scope of the present invention. To list every related
sequence
would be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 2864 of SEQ ID
N0:32, b
is an integer of 15 to 2878, where both a and b correspond to the positions of
nucleotide residues shown in SEQ >D N0:32, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 23
This gene is expressed primarily in Dendritic Cells From CD34 Cells.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ 117 NO: 70 as residues: Pro-25 to Phe-31, Gln-99 to Lys-
116.
Polynucleotides encoding said polypeptides are also provided.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ B7 N0:33 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
would be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 405 of SEQ 117
N0:33, b
is an integer of 15 to 419, where both a and b correspond to the positions of
S nucleotide residues shown in SEQ ID N0:33, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 24
The computer algorithm BLASTX has been used to determine that the
10 translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
emb~CAB46879.1 ~ (all information available through the recited accession
number is
incorporated herein by reference) which is described therein as "(AL022240)
dJ328E19.2 (based on ESTs/predictions) [Homo sapiensJ". A partial alignment
1 S demonstrating the observed homology is shown immediately below.
>emb~CAH46879.1~ (AL022240) dJ328E19.2 (based on ESTs/predictions) [Homo
sapiens]
Length = 997
Plus Strand HSPS:
Score = 188 (66.2 bits), Expect = 3.3e-16, Sum P(2) = 3.3e-16
Identities = 37/39 (94$), Positives = 39/39 (100$), Frame = +1
2S
Q: 337 ENRSQEPLAGDSMSPRTLPVQNMNNAMFLQKTLSLSFIG 453
ENRSQEPLAGDSMSPRTLPVQNM+NAMFLQKTL+LSFIG
S: 850 ENRSQEPLAGDSMSPRTLPVQNMSNAMFLQKTLNLSFIG 888
3 0 Score = 113 (39.8 bits), Expect = 3.3e-16, Sum P(2) = 3.3e-16
Identities = 22/26 (84$), Positives = 22/26 (84$), Frame = +1
Q: 1663 EPKHIPVTFCDYHLAPTLV--FGLDP 1734
EPKHIPVTFCDYHLAPTLV FG P
3S S: 891 EPKHIPVTFCDYHLAPTLVLVFGAVP 916
The segments of emb~CAB46879.1 ~ that are shown as "S" above are set out in
the sequence listing as SEQ ID NO: 110 and SEQ ID NO: 112. Based on the
structural similarity, these homologous polypeptides are expected to share at
least
40 some biological activities. Such activities are known in the art, some of
which are
described elsewhere herein. Assays for determining such activities are also
known in
the art, some of which have been described elsewhere herein.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
36
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 111 and/or
SEQ
ID NO: 113 which correspond to the "Q" sequences in the alignment shown above
(gaps introduced in a sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
Human 8 Week Whole Embryo and to a lesser extent in Soares fetal liver spleen
1NFLS; Human Eosinophils; Soares infant brain 1NIB; Human Bone Marrow,
treated; Soares fetal heart-NbHHI9W; Soares testis NHT; Nine Week Old Early
Stage Human; Colon Normal III; Smooth muscle,control; 12 Week Old Early Stage
Human, II; NCI CGAP Panl; Fetal Heart; Human Placenta; T Cell helper I; Human
Testes; Colon Tumor II; Human Cerebellum; Monocyte activated, re-excision;
Human Adipose; Human Testes Tumor, re-excision; Gessler Wilms tumor; Dendritic
cells, pooled; Human Fetal Heart; Monocyte activated; Hodgkin's Lymphoma II; T
cell helper II; Synovial hypoxia; Synovial Fibroblasts (Ill/TNF), subt; T-Cell
PHA 16
hrs; human ovarian cancer; Human Uterine Cancer; Human Placenta (re-excision);
Human Chondrosarcoma; NTERA2, control; Human Ovary; Human Gall Bladder;
Clontech human aorta polyA+ mRNA (#6572); PC3 Prostate cell line; Human Testes
Tumor; Human Fetal Lung III; Human Adult Pulinonary,re-excision; Human
Amygdala; NCI CGAP-Co3; Osteoblasts; Breast, Cancer: (4004943 A5); Healing
groin wound, 6.5 hours post incision; H. Meningima, M1; Human Neutrophil;
Human
Prostate; Soares NhHMPu_S1; KMH2; B-Cells; Soares total fetus Nb2HF8 9w;
Human Fetal Dura Mater; T-Cell PHA 24 hrs; Human fetal heart, Lambda ZAP
Express; Rejected Kidney, lib 4; Human fetal heart, Lambda ZAP Express; Human
T-
Cell Lymphoma; Soares total fetus Nb2HF8 9w; Stratagene schizo brain 511;
Soares breast 3NbHBst; 12 Week Early Stage Human II, Reexcision; Endothelial
cells-control; Human Microvascular Endothelial Cells, fract. A; NCI CGAP Kid6;
Human Fetal Brain; HUMAN STOMACH; NCI CGAP-Gas4; Apoptotic T-cell, re-
excision; Human Synovium; STROMAL -OSTEOCLASTOMA; Human
Hypothalamus,schizophrenia, re-excision; Hepatocellular Tumor,re-excision;
Stratagene placenta (#937225); Synovial hypoxia-RSF subtracted; Glioblastoma;
Salivary Gland, Lib 2; Human T-cell lymphoma,re-excision; Healing groin wound,
7.5 hours post incision; LNCAP prostate cell line; Human endometrial stromal
cells;


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
37
Soares-pregnant uterus NbHPU; Prostate BPH; Human Infant Brain; Hippocampus,
Alzheimer Subtracted; H. Kidney Medulla, re-excision; Gessler Wilms tumor;
Soares fetal heart NbHHI9W; L428; Human Pancreas Tumor; Human
Hypothalmus,Schizophrenia; Human Rhabdomyosarcoma; Bone Marrow Stromal
Cell, untreated; Human Adrenal Gland Tumor; Pancreatic Islet; Fetal Liver,
subtraction II; Colon Tumor; Resting T-Cell Library,II; 12 Week Old Early
Stage
Human; Soares multiple sclerosis 2NbHMSP;
Soares fetal liver spleen_1NFLS S1; Stratagene HeLa cell s3 937216; Early
Stage
Human Brain; Adipocytes; Human Testes, Reexcision; Human Neutrophil,
Activated;
NCI CGAP-Br2; NCI CGAP Prl; Clontech human aorta polyA+ mRNA (#6572);
Human adult (K.Okubo); Human fetal heart, Lambda ZAP Express; Human Pancreas;
H. Adipose, subtracted; Human Testes; Human Eosinophils; PCR, pBMC I/C
treated;
Aryepiglottis Normal; Testis, normal; brain stem; Human Umbilical Vein
Endothelial
cells, frac B, re-excision; Salivary Gland, Lib 3; Human Pre-Differentiated
Adipocytes; CD34+cells, II, FRACTION 2; Human Leukocytes; LNCAP + 30nM
81881; H. Meniingima, M6; Dermatofibrosarcoma Protuberance; Human Adult
Liver, subtracted; Human Fetal Brain, random primed; Morton Fetal Cochlea;
Human
Gall Bladder, fraction II; Human OB HOS control fraction I; Human (HCC) cell
line
liver (mouse) metastasis, remake; Human Cerebellum, subtracted; Human
Umbilical
Vein Endothelial Cells, fract. A; H. Atrophic Endometrium; Human Placenta;
NCI CGAP_Brn35; Fetal Heart, re-excision; Lung, Cancer: Poorly-Differentiated
Squamous Cell carcinoma (4005313 A3); Human Primary Breast Cancer;
NCI CGAP Utl; NCI CGAP_Panl; Ovary, Cancer: Papillary Serous Cystic, Low
Malignant Potential (4004562 B6); H. cerebellum, Enzyme subtracted; Breast
Lymph
node cDNA library; Early Stage Human Lung, subtracted; Lung Carcinoma A549
TNFalpha activated; Soares NhHMPu Sl; Raji Cells, cyclohexamide treated;
NTERA2 teratocarcinoma cell line+retinoic acid (14 days); CD34 positive cells
(cord
blood),re-ex; Human Tonsils, Lib 2; Breast, Cancer: (4005522 A2); Healing
groin
wound - zero hr post-incision (control); Human Epididymus; B Cell lymphoma;
NCI CGAP-Ov23; Soares fetal lung NbHLI9W; LPS activated derived dendritic
cells; Gessler Wilms tumor; NCI CGAP GC4; NCI CGAP LuS; NCI CGAP Kids;
normalized infant brain cDNA; NTERA2 + retinoic acid, 14 days; H. Kidney
Cortex,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
38
subtracted; Human Stomach,re-excision; Human Adipose Tissue, re-excision;
Human
Osteosarcoma; Myoloid Progenitor Cell Line; Jurkat T-Cell, S phase; wilm's
tumor;
H. Lymph node breast Cancer; Brain Frontal Cortex, re-excision; Human Chronic
Synovitis; Snares-placenta 8to9weeks 2NbHP8to9W; Human pancreatic islet;
Snares total fetus Nb2HF8 9w; Snares fetal liver spleen-1NFLS_S1; Mo7e Cell
Line GM-CSF treated (lng/ml); TF-1 Cell Line GM-CSF Treated; Human Thymus;
NCI CGAP-Br7; NCI CGAP-GC4; NCI CGAP-LuS; NCI CGAP_GCB1;
NCI CGAP-PNSl; WATM1; Apoptotic T-cell; Human Fetal Kidney; Stratagene
fetal spleen (#937205); Human Osteoblasts II; Human Activated T-Cells; Stromal
cell
TF274; Macrophage-oxLDL; Gessler Wilms tumor; NCI CGAP-LymS;
NCI CGAP-Lyml2; Snares NhHMPu S1; Snares testis NHT;
Snares fetal heart NbHHI9W; Snares NSF F8 9W_OT PA P S1; Ovary,
Normaf:a: (9805C040R); Olfactory epithelium,nasalcavity; Human Pancreas
Tumor, Reexcision; Human Hippocampus; Spinal cord; Stratagene neuroepithelium
NT2RAMI 937234; Ulcerative Colitis; PERM TF274; Hemangiopericytoma;
Snares testis NHT; Snares total fetus Nb2HF8 9w; Human Thymus Stromal Cells;
Human Whole Six Week Old Embryo; Stratagene liver (#937224);
Snares fetal liver spleen_1NFLS S1; Macrophage-oxLDL, re-excision; Pancreas
Islet Cell Tumor; NCI CGAP AA1; NCI CGAP-GC3; NCI CGAP-GC4;
NCI CGAP Pr3; NCI CGAP_Brl.l; Human Substantia Nigra; Human fetal lung;
Snares NhHMPu Sl; Snares fetal heart NbHHI9W; Stratagene hNT neuron
(#937233); Stratagene lung carcinoma 937218; normalized infant brain cDNA;
Colon
Normal II; Normal colon; B-cells (stimulated); human tonsils; Endothelial-
induced;
Human Osteoclastoma; Anergic T-cell; Snares-pregnant uterus NbHPU;
Snares fetal lung NbHLI9W; Snares fetal heart NbHHI9W; CD34 positive cells
(Cord Blood); NCI CGAP_GC4; NCI CGAP-CLL1; NCI CGAP_KidS;
NCI CGAP-Lei2; Snares testis NHT; Snares total fetus Nb2HF8 9w;
NCI CGAP-CoB; NCI CGAP-LuS; NCI CGAP Ut2; NCI CGAP-Ut3;
NCI CGAP_Col4; NCI CGAP-Gas4; NCI CGAP-Brn35; Bone Marrow Cell Line
(RS4,11 ); Human Endometrial Tumor; NCI CGAP_Alv 1; NCI CGAP_GCB 1;
NCI CGAP-HSC1 and Snares fetal lung NbHLI9W.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
39
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:34 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 3165 of SEQ ID
N0:34, b
is an integer of 15 to 3179, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:34, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 25
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
gb~AAD39763.1 ~AF144899_1 (all information available through the recited
accession
number is incorporated herein by reference) which is described therein as
"(AF144899) immunoglobulin heavy chain variable region [Mus musculus]". A
partial alignment demonstrating the observed homology is shown immediately
below.
>gb~AAD39763.1~AF144899 1 (AF144899) immunoglobulin heavy chain variable
region
[MUS musculus]
Length = 91
Plus Strand HSPs:
Score = 66 (23.2 bits), Expect = 3.8, P = 0.98
Identities = 13/31 (41~), Positives = 22/31 (70~), Frame = +2
Q: 113 SLEEPPLVFAESPQKGVEAVKTVNSTGSFSW 205
S+ P L F E+P+K +E V T++S GS+++
S: 3 SVASPCLGFTETPEKRLEWVATISSGGSYTY 33
The segment of gb~AAD39763.1 ~AF144899_1 that is shown as "S" above is
set out in the sequence listing as SEQ >D NO: 114. Based on the structural
similarity,
these homologous polypeptides are expected to share at least some biological
activities. Such activities are known in the art, some of which are described
elsewhere


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
herein. Assays for determining such activities are also known in the art, some
of
which have been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 115 which
5 corresponds to the "Q" sequence in the alignment shown above (gaps
introduced in a
sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
Human Frontal Cortex, Schizophrenia; Human Whole Six Week Old Embryo.
Many polynucleotide sequences, such as EST sequences, are publicly
10 available and accessible through sequence databases. Some of these
sequences are
related to SEQ ID N0:35 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 be cumbersome. Accordingly, preferably excluded from the present
invention
15 are one or more polynucleotides comprising a nucleotide sequence described
by the
general formula of a-b, where a is any integer between 1 to 1269 of SEQ ID
N0:35, b
is an integer of 1 S to 1283, where both a and b correspond to the positions
of
nucleotide residues shown in SEQ ID N0:35, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 26
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
pir~JC5238~JC5238 (all information available through the recited accession
number is
incorporated herein by reference) which is described therein as
"galactosylceramide-
like protein, GCP - human". A partial alignment demonstrating the observed
homology is shown immediately below.
>pir I JC5238 ~ JC5238 galactosylceramide-like protein, GCP - human
Length = 114
Minus Strand HSPs:
Score = 93 (32.7 bits), Expect = 3.6e-05, Sum P(2) = 3.6e-05


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
41
Identities = 17/24 (70~), Positives = 19/24 (79~), Frame = -2
Q: 89 GRLRWADHLRPGVRDQPGKHDETP 18
GR R DHLR GVRDQPG+H +TP
S S: 76 GRPRQVDHLRSGVRDQPGQHGKTP 99
Score = 45 (15.8 bits), Expect = 3.6e-05, Sum P(2) = 3.6e-05
Identities = 9/13 (69~), Positives = 9/13 (69~), Frame = -3
Q: 127 ARRGARACNPSTL 89
A GA CNPSTL
S: 63 AGHGALTCNPSTL 75
The segments of pir~JCS238~JCS238 that are shown as "S" above are set out in
1 S the sequence listing as SEQ ID NO: 116 and SEQ ID NO: 118. Based on the
structural similarity, these homologous polypeptides are expected to share at
least
some biological activities. Such activities are known in the art, some of
which are
described elsewhere herein. Assays for determining such activities are also
known in
the art, some of which have been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 117 and/or
SEQ
ID NO: 119 which correspond to the "Q" sequences in the alignment shown above
(gaps introduced in a sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
2S NCI CGAP-GCB1; Soares infant brain 1NIB and to a lesser extent in Soares
ovary
tumor NbHOT; Soares fetal liver spleen 1NFLS; Soares melanocyte 2NbHM;
NCI CGAP_LuS; Spleen, Chronic lymphocytic leukemia;
Soares fetal liver spleen-1NFLS S1; Human Osteosarcoma; Jurkat T-cell G1
phase; Stratagene fetal spleen (#937205); Soares fetal lung NbHLI9W; Human
Whole Six Week Old Embryo; Rejected Kidney, lib 4; Stratagene lung (#937210);
Stratagene HeLa cell s3 937216; NCI CGAP Kids; NCI CGAP-Kid6;
NCI CGAP Prl2; Human Testes, Reexcision;
Soares fetal liver spleen 1NFLS_S1; Colon Normal III; Human 8 Week Whole
Embryo; Nine Week Old Early Stage Human; STRATAGENE Human skeletal
3S muscle cDNA library, cat. #936215.; Soares NhHMPu S1;
Soares_pineal_gland N3HPG; Soares~regnant uterus NbHPU;
Soares~arathyroid tumor NbHPA; Stratagene schizo brain S11; Bone Cancer; H.
Adipose Tissue; Frontal lobe,dementia,re-excision; Human Lung; Soares retina
N2b4HR; Human Pineal Gland; H Female Bladder, Adult; NTERA2 + retinoic acid,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
42
14 days; Stratagene NT2 neuronal precursor 937230; Human Adipose Tissue, re-
excision; T-Cell PHA 16 hrs; TF-1 Cell Line GM-CSF Treated; Human Fetal
Kidney;
Stratagene pancreas (#937208); Merkel Cells; NCI CGAP_GC6; NCI CGAP Ut2;
NCI CGAP_Lul9; NCI CGAP-Pr28; NCI CGAP Brn25; NCI CGAP Kidl l;
Soares NhHMPu_S1; Soares testis NHT; Soares NFL T GBC_Sl;
Soares fetal liver spleen-1NFLS_S1; Soares senescent fibroblasts NbHSF; Ovary,
Normal:::::::: (9805C040R); Hemangiopericytoma; NCI CGAP-Kid3;
NCI CGAP-Kids; NCI CGAP_Pr28; Human Ovary; PC3 Prostate cell line; Human
Testes Tumor; Human immortalized fibroblasts (H.L.Ozer); Soares NhHMPu_S1;
Human Fetal Kidney, Reexcision; Primary Dendritic cells,frac 2; BATM2; Human
retina cDNA Tsp509I-cleaved sublibrary; NCI CGAP-Br2; NCI CGAP-GC4;
NCI CGAP_Pr2; NCI CGAP-Pr2l; Soares fetal heart NbHHI9W; Human Fetal
Heart; Soares fetal lung NbHLI9W; Soares fetal heart NbHHI9W; Human
Testes; Activated T-cell(12h)/Thiouridine-re-excision and Primary Dendritic
Cells,
lib 1.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:36 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 2194 of SEQ ID
N0:36, b
is an integer of 15 to 2208, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:36, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 27
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
dbj ~BAA74863.1 ~ (all information available through the recited accession
number is


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
43
incorporated herein by reference) which is described therein as "(AB020647)
KIAA0840 protein [Homo Sapiens]". A partial alignment demonstrating the
observed
homology is shown immediately below.
>dbj~BAA74863.1~ (AB020647) KIAA0840 protein [Homo Sapiens] >dbj~BAA74863.1~
(AB020647) KIAA0840 protein [Homo Sapiens] >sp~094926~094926
KIAA0840 PROTEIN (FRAGMENT). >sp~BAA74863~BAA74863 KIAA0840 protein
(fragment).
Length = 483
Plus Strand HSPs:
Score = 109 (38.4 bits), Expect = 0.057, P = 0.055
Identities = 26/62 (41~), Positives = 37/62 (59~), Frame = +3
Q: 1056 QETGRVLLFL---SLSGCYQITDHGLRVLTLGGGLPYLEHLNLSGCLTITGAGLQDLVSA 1226
Q+T V L L ++SGC ++TD GL T+ P L L +SGC I+ + D+VS
S: 171 QDTPNVCLMLETVTVSGCRRLTDRGL--YTIAQCCPELRRLEVSGCYNISNEAVFDWSL 228
Q: 1227 CPSL 1238
CP+L
S: 229 CPNL 232
The segment of dbj ~BAA74863.1 ~ that is shown as "S" above is set out in the
sequence listing as SEQ ID NO: 120. Based on the structural similarity, these
homologous polypeptides are expected to share at least some biological
activities.
Such activities are known in the art, some of which are described elsewhere
herein.
Assays for determining such activities are also known in the art, some of
which have
been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 121 which
corresponds to the "Q" sequence in the alignment shown above (gaps introduced
in a
sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
normalized infant brain cDNA and to a lesser extent in Soares infant brain
1NIB;
NCI CGAP-GCB1; Human Ovarian Cancer Reexcision; Human Bone Marrow,
treated; Soares fetal liver spleen 1NFLS; Soares retina N2b4HR; Human T-Cell
Lymphoma; Soares NFL T GBC_S1; Human Fetal Heart; Human Osteoclastoma;
NCI CGAP_GCB1; Soares placenta Nb2HP; Hepatocellular Tumor, re-excision; 12
Week Old Early Stage Human; Human Placenta; Soares melanocyte 2NbHM;
Osteoblasts; Primary Dendritic Cells, lib 1; Soares-pregnant uterus NbHPU;
Human
Amygdala,re-excision; Stromal cell TF274; Human Activated T-Cells, re-
excision;


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
44
Fetal Heart; Soares-parathyroid tumor NbHPA; Soares fetal lung NbHLI9W;
Smooth muscle,control; Human Testes; Human Endometrial Tumor;
NCI CGAP_KidS; Human Cerebellum; H. Atrophic Endometrium; Human Thyroid;
H. Kidney Cortex, subtracted; Human T-cell lymphoma,re-excision; Breast,
Cancer:
S (4004943 AS); H. Kidney Medulla, re-excision; TF-1 Cell Line GM-CSF Treated;
Stratagene fetal spleen (#937205); 12 Week Old Early Stage Human, II; Human
Pancreas Tumor, Reexcision; Liver, Hepatoma; Human Adipose; Human Placenta
(re-excision); Stratagene HeLa cell s3 937216; Stratagene hNT neuron
(#937233);
Ulcerative Colitis; Bone Marrow Stromal Cell, untreated; Colon Tumor; Human
Fetal
Lung III; Normal Human Trabecular Bone Cells; Soares~ineal-gland N3HPG;
Bone marrow; Human Adult Pulmonary,re-excision; NCI CGAP_Co9; Monocyte
activated; T Cell helper I; Human 8 Week Whole Embryo; Nine Week Old Early
Stage Human; T cell helper II; Human Macrophage, subtracted; Human Adult
Skeletal Muscle; Activated T-Cells, 8 hrs.; Human Microvascular Endothelial
Cells,
fract. B; LNCAP + o.3nM 81881; Human 8 Week Whole Embryo, subtracted; HL-
60, RA 4h, Subtracted; Morton Fetal Cochlea; Human Gall Bladder, fraction II;
Human colon carcinoma (HCC) cell line, remake; Human Cerebellum, subtracted;
Human Cardiomyopathy, subtracted; Human Adult Pulmonary; Human Fetal Spleen;
Fetal Heart, re-excision; NCI CGAP Kidl l; Soares testis NHT;
Soares~regnant uterus NbHPU; Soares-placenta 8to9weeks 2NbHP8to9W; Lung,
Cancer: Poorly-Differentiated Squamous Cell carcinoma (4005313 A3); Human
Liver; Soares fetal lung NbHLI9W; Soares fetal heart NbHHI9W; Human Pineal
Gland; Smooth Muscle- HASTE normalized; Ovary, Cancer: Poorly differentiated
carcinoma (***); Messangial cell, frac 2; Human adult small intestine,re-
excision;
Human Tonsils, Lib 2; Stomach cancer (human),re-excision; Human
Hypothalamus,schizophrenia, re-excision; pBMC stimulated w/ poly I/C; Human
Osteoclastoma Stromal Cells - unamplified; Human Stomach,re-excision; Human
Osteosarcoma; Jurkat T-Cell, S phase; Healing groin wound, 6.5 hours post
incision;
Human Manic Depression Tissue; Soares total fetus Nb2HF8 9w; H. Lymph node
breast Cancer; Breast, Normal: (4005522:::::::: B2); Human Infant Brain; Human
Neutrophil; Human Chronic Synovitis; Soares multiple sclerosis 2NbHMSP; T-Cell
PHA 16 hrs; Human Umbilical Vein, Reexcision; Atrium cDNA library Human heart;


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
Human promyelocyte; Human pancreatic islet; Soares NhHMPu S 1;
Soares fetal liver spleen 1NFLS_Sl; Soares senescent fibroblasts NbHSF; Testis
1; Human Osteoblasts II; Human Activated T-Cells; Human Heart; Macrophage-
oxLDL; Human Hypothalmus,Schizophrenia; Human Hippocampus; PERM TF274;
S Synovial Fibroblasts (control); Human Testes Tumor, re-excision; Human
Thymus
Stromal Cells; Rejected Kidney, lib 4; Human fetal heart, Lambda ZAP Express;
Resting T-Cell Library,II; Colon Carcinoma; H Macrophage (GM-CSF treated), re-
excision; Normal colon; Dendritic cells, pooled; Soares fetal lung NbHLI9W;
Neutrophils control, re-excision; Primary Dendritic cells,frac 2; Gessler
Wilms tumor;
10 Human pancreatic islet; Human fetal brain S. Meier-Ewert; NCI CGAP_Co3;
NCI CGAP_Pr6; NCI CGAP Pr22; Soares testis NHT;
Soares total fetus Nb2HF8 9w; Stratagene schizo brain S 11; Human Placenta;
Human Testes, Reexcision; Soares-parathyroid tumor NbHPA; 12 Week Early
Stage Human II, Reexcision; human tonsils; Endothelial-induced; Endothelial
cells-
15 control; NCI CGAP-Co3; NCI CGAP-GCBl; NCI CGAP-Brl.l; WATM1;
Spleen, Chronic lymphocytic leukemia; Colon Normal III; Hodgkin's Lymphoma II;
Activated T-cell(12h)/Thiouridine-re-excision; neutrophils control; Human
pancreatic
islet; Human retina cDNA randomly primed sublibrary; NCI CGAP_GC4;
NCI CGAP_LuS; NCI CGAP_CoIO; NCI CGAP_Kid6; Stratagene NT2 neuronal
20 precursor 937230; Colon Tumor II; NCI CGAP_GC4; NCI CGAP_LuS;
NCI CGAP Ut3; NCI CGAP_Col4; NCI CGAP_Kid3; NCI CGAP-Kids;
Soares multiple sclerosis 2NbHMSP and Infant brain, LLNL array of Dr. M.
Soares
1NIB.
Many polynucleotide sequences, such as EST sequences, are publicly
25 available and accessible through sequence databases. Some of these
sequences are
related to SEQ ID N0:37 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 be cumbersome. Accordingly, preferably excluded from the present
invention
30 are one or more polynucleotides comprising a nucleotide sequence described
by the
general formula of a-b, where a is any integer between 1 to 2065 of SEQ ID
N0:37, b
is an integer of 15 to 2079, where both a and b correspond to the positions of


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
46
nucleotide residues shown in SEQ >D N0:37, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 28
This gene is expressed primarily in NTER.A2, control.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:38 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1287 of SEQ ID
N0:38, b
is an integer of 15 to 1301, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:38, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 29
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
dbj~BAA11748.1 ~ (all information available through the recited accession
number is
incorporated herein by reference) which is described therein as "nuclear
protein,
NP220 [Homo Sapiens]". A partial alignment demonstrating the observed homology
is shown immediately below.
>dbj~BAA11748.1~ nuclear protein, NP220 [Homo Sapiens] >sp~Q14966~Q14966
NUCLEAR PROTEIN, NP220. >pir~JU0239~JU0239 nuclear matrix protein
N/MAX-92 - human (fragment) (SUB 670-761} >pir~JU0240~JU0240
nuclear matrix protein N/MAX-74 - human (fragment) {SUB 899-972)
Length = 1978
Plus Strand HSPs:
Score = 99 (34.8 bits), Expect = 2.7, P = 0.93
Identities = 22/22 (1000 , Positives = 22/22 (1000 , Frame = +3
Q: 1155 ASTLKRDADASKAVEIVTSTSA 1220
ASTLKRDADASKAVEIVTSTSA


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
47
S: 771 ASTLKRDADASKAVEIVTSTSA 792
The segment of dbj ~BAA11748.1 ~ that is shown as "S" above is set out in the
sequence listing as SEQ ID NO: 122. Based on the structural similarity, these
S homologous polypeptides are expected to share at least some biological
activities.
Such activities are known in the art, some of which are described elsewhere
herein.
Assays for determining such activities are also known in the art, some of
which have
been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 123 which
corresponds to the "Q" sequence in the alignment shown above (gaps introduced
in a
sequence by the computer are, of course, removed).
This gene is expressed primarily in prostate-edited.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:39 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between ~to 1-5-83 of SEQ ID
N0:39, b
is an integer of 15 to 1597, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:39, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 30
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares infant brain 1NIB and to a lesser extent in Smooth muscle, serum
treated;
Human Fetal Heart; Human Adult Pulmonary,re-excision; Human Microvascular
Endothelial Cells, fract. A; Smooth muscle,control; Soares NFL T GBC_S1; Human
Rhabdomyosarcoma; Smooth muscle, serum induced,re-exc; Soares NhHMPu S1;
Stratagene fetal spleen (#937205); Jia bone marrow stroma; Pancreatic Islet;
Synovial


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
48
Fibroblasts (control); Stratagene lung (#937210); NCI CGAP-GC6; Soares
melanocyte 2NbHM; Soares fetal heart NbHHI9W; H. Frontal cortex,epileptic,re-
excision; Soares-pregnant uterus NbHPU; Osteoblasts; Soares placenta Nb2HP;
Human OB HOS control fraction I; Human OB MG63 treated (10 nM E2) fraction I;
Human White Adipose; Smooth muscle, control, re-excision; Early Stage Human
Lung, subtracted; Ovary, Cancer: Poorly differentiated carcinoma (***);
Synovial
hypoxia-RSF subtracted; Human Whole Brain #2 - Oligo dT > 1.SKb; Human Manic
Depression Tissue; Stratagene HeLa cell s3 937216; Brain Frontal Cortex, re-
excision; Human Umbilical Vein, Reexcision; Human Osteoblasts II; Human
umbilical vein endothelial cells, IL-4 induced; Normal Human Trabecular Bone
Cells;
Soares testis NHT; Human Pancreas Tumor, Reexcision;
Soares senescent fibroblasts NbHSF; Spinal cord; Human retina cDNA randomly
primed sublibrary; Human Testes Tumor, re-excision; Bone Marrow Stromal Cell,
untreated; Pancreas Islet Cell Tumor; NCI CGAP-Gas4; NCI CGAP-Panl;
Soares testis NHT; Soares-pregnant uterus NbHPU; CHME Cell Line,untreated;
NCI CGAP_GC2; NCI CGAP-GCB1; NCI CGAP PNS1; Spleen, Chronic
lymphocytic leukemia; T Cell helper I; Hodgkin's Lymphoma II; Colon Tumor II;
Nine Week Old Early Stage Human and Primary Dendritic Cells, lib 1.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ >D NO: 77 as residues: Pro-21 to Ser-26. Polynucleotides
encoding said polypeptides are also provided.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:40 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 3102 of SEQ ID
N0:40, b
is an integer of 15 to 3116, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:40, and where b is greater than or
equal to a
+ 14.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
49
FEATURES OF PROTEIN ENCODED BY GENE NO: 31
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
emb~CAA06588.1 ~ (all information available through the recited accession
number is
incorporated herein by reference) which is described therein as "(AJ005559)
SPR2A
protein [Mus musculus]". A partial alignment demonstrating the observed
homology
is shown immediately below.
>emb~CAA06588.1~ (AJ0055590 SPR2A protein [MUS musculus] >sp~070553~070553
SPR2A PROTEIN.
Length = 83
Minus Strand HSPs:
Score = 82 (28.9 bits), Expect = 0.040, P = 0.039
Identities = 21/55 (38~), Positives = 27/55 (49~), Frame = -3
2O Q: 323 PSSCPLRAWAAP--P---FEVC-PMAPYGPAPP---FEVCPMAPYRPSCLPRVWP 186
P CP +AW P P FE C P GP P +E CP P++ +C P +P
S: 24 PEPCPPQAWPGPCRPVMCFEPCLPSVWPGPCRPWCYEQCPPQPWQSTCPPVQFP 78
The segment of emb~CAA06588.1 ( that is shown as "S" above is set out in the
sequence listing as SEQ ID NO: 124. Based on the structural similarity, these
homologous polypeptides are expected to share at least some biological
activities.
Such activities are known in the art, some of which are described elsewhere
herein.
Assays for determining such activities are also known in the art, some of
which have
been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 125 which
corresponds to the "Q" sequence in the alignment shown above (gaps introduced
in a
sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
Human Testes, Reexcision; Human Testes and to a lesser extent in Human Adult
Testes, Large Inserts, Reexcision; Soares testis NHT; Human adult testis,
large
inserts; NCI CGAP-GC4; H Female Bladder, Adult; Soares NFL T GBC_S1;
Testis, normal; Human testis (C. De Smet) and Testis 1.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 78 as residues: Phe-30 to Lys-37, Pro-43 to Lys-
7S.
Polynucleotides encoding said polypeptides are also provided.
Many polynucleotide sequences, such as EST sequences, are publicly
S available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:41 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 be cumbersome. Accordingly, preferably excluded from the present
invention
10 are one or more polynucleotides comprising a nucleotide sequence described
by the
general formula of a-b, where a is any integer between 1 to 1067 of SEQ ID
N0:41, b
is an integer of 1S to 1081, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:41, and where b is greater than or
equal to a
+ 14.
1S
FEATURES OF PROTEIN ENCODED BY GENE NO: 32
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
20 pir~PL0126~PL0126 (all information available through the recited accession
number is
incorporated herein by reference) which is described therein as "H-2 class II
histocompatibility antigen I-A beta chain - mouse (fragment)". A partial
alignment
demonstrating the observed homology is shown immediately below.
2S >pir~PL0126~PL0126 H-2 class II histocompatibility antigen I-A beta chain -
mouse (fragment)
Length = 89
Plus Strand HSPs:
Score = 68 (23.9 bits), Expect = 1.9, P = 0.85
Identities = 15/45 (33~), Positives = 29/45 (64~), Frame = +2
Q: 20 ILDIGSPIAKHFTFIFQYLLLSRILLDKLCHFSEKETELLSVLRQ 154
3S + ++G P A+++ QYL +R LD +C ++ +ETE+ + LR+
S: 45 VTELGRPDAEYYNK--QYLERTRAELDTVCRYNYEETEVPTSLRR 87
The segment of pir~PL0126~PL0126 that is shown as "S" above is set out in the
sequence listing as SEQ ID NO: 126. Based on the structural similarity, these


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
51
homologous polypeptides are expected to share at least some biological
activities.
Such activities are known in the art, some of which are described elsewhere
herein.
Assays for determining such activities are also known in the art, some of
which have
been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 127 which
corresponds to the "Q" sequence in the alignment shown above (gaps introduced
in a
sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares infant brain 1NIB and to a lesser extent in Human Adrenal Gland Tumor;
Human Cerebellum; Soares retina N2b4HR; Soares testis NHT; normalized infant
brain cDNA; Human Testes; Human Brain, Striatum;
Soares-parathyroid tumor NbHPA; Human Testes, Reexcision;
Soares multiple sclerosis 2NbHMSP; Human Amygdala; CD34 positive cells (Cord
Blood); Neutrophils IL-1 and LPS induced and H. Frontal cortex,epileptic,re-
excision.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 79 as residues: Glu-34 to Ser-41. Polynucleotides
encoding said polypeptides are also provided.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:42 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1131 of SEQ ID
N0:42, b
is an integer of 15 to 1145, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:42, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 33


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
52
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
emb~CAA06752.1 ~ (all information available through the recited accession
number is
incorporated herein by reference) which is described therein as "(AJ005895)
protein
translocase [Homo Sapiens]". A partial alignment demonstrating the observed
homology is shown immediately below.
>emb~CAA06752.1~ (AJ005895) protein translocase [Homo Sapiens]
>gb~AAD27772.1~AF077039_1 (AF077039) inner mitochondrial membrane
translocase TIM17 homolog [Homo Sapiens] >gb~AAC24694.1~ (AF034790)
inner mitochondrial membrane translocase Timl7b [Homo Sapiens]
>sp~060830~060830 PROTEIN TRANSLOCASE. >sp~AAD27772~AAD27772 Inner
mitochondrial membrane translocase TIM17 homolog.
Length = 172
Minus Strand HSPS:
Score = 347 (122.2 bits), Expect = 6.4e-31, P = 6.4e-31
Identities = 68/70 (97~), Positives = 68/70 (97~), Frame = -2
Q: 571 AAXGAVLAARSGPLAMVGSAMMGGILLALIEGVGILLTRYTAQQFRNAPPFLEDPSQLPP 392
A GAVLAARSGPLAMVGSAMMGGILLALIEGVGILLTRYTAQQFRNAPPFLEDPSQLPP
S: 97 ALTGAVLAARSGPLAMVGSAMMGGILLALIEGVGILLTRYTAQQFRNAPPFLEDPSQLPP 156
Q: 391 KDGTPAPGYP 362
KDGTPAPGYP
S: 157 KDGTPAPGYP 166
The segment of emb~CAA06752.1 ~ that is shown as "S" above is set out in the
sequence listing as SEQ ID NO: 128. Based on the structural similarity, these
homologous polypeptides are expected to share at least some biological
activities.
Such activities are known in the art, some of which are described elsewhere
herein.
Assays for determining such activities are also known in the art, some of
which have
been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ a7 NO: 129 which
corresponds to the "Q" sequence in the alignment shown above (gaps introduced
in a
sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
Activated T-Cells,l2 hrs,re-excision and to a lesser extent in Human Primary
Breast
Cancer Reexcision; Soares testis NHT; Amniotic Cells - Primary Culture;
NCI CGAP_LuS; Human Normal Breast; Human endometrial stromal cells-treated


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
53
with estradiol; Soares-pineal-gland N3HPG;
Soares_placenta 8to9weeks_2NbHP8to9W; Mo7e Cell Line GM-CSF treated
(lng/ml); Soares fetal heart-NbHHI9W; Human Chondrosarcoma; Soares
melanocyte 2NbHM; Soares fetal liver spleen 1NFLS; Soares infant brain 1NIB;
S Human Fetal Brain, normalized c5-11-26; Human Primary Breast Cancer;
Salivary
gland, re-excision; Activated T-Cells, 12 hrs.; Human OB HOS treated (10 nM
E2)
fraction I; Human Primary Breast Cancer,re-excision; Human Pituitary,
subtracted;
Human Primary Breast Cancer; Breast Lymph node cDNA library; Raji Cells,
cyclohexamide treated; Human Synovium; Breast, Cancer: (4004943 AS); H. Lymph
node breast Cancer; NCI CGAP_LuS; Breast Cancer Cell line, angiogenic; Human
Brain, Striatum; Apoptotic T-cell; Human Uterine Cancer;
Soares fetal lung NbHLI9W; Human umbilical vein endothelial cells, IL-4
induced;
Soares-placenta 8to9weeks 2NbHP8to9W; Human Thymus; Soares breast
2NbHBst; Human Whole Six Week Old Embryo; Rejected Kidney, lib 4; Pancreas
Islet Cell Tumor; Human Fetal Lung III; Activated T-Cell (l2hs)/Thiouridine
labelledEco; HTCDL1; NCI CGAP_Br2; NCI CGAP_Co3; NCI CGAP_GC2;
NCI CGAP_GC4; NCI CGAP_Pr2; NCI CGAP Pr3; NCI CGAP Kid3;
NCI CGAP_KidS; NCI CGAP Kid6; NCI CGAP_Pr22; NCI CGAP Brn23;
Stratagene fetal retina 937202; Human Microvascular Endothelial Cells, fract.
A;
Hodgkin's Lymphoma II; Keratinocyte and T cell helper II.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 80 as residues: Tyr-19 to Ala-27, Asp-33 to Thr-
43,
Ala-45 to His-55. Polynucleotides encoding said polypeptides are also
provided.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:43 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 654 of SEQ ID
N0:43, b
is an integer of 15 to 668, where both a and b correspond to the positions of


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
54
nucleotide residues shown in SEQ ID N0:43, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 34
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
gb~AAB31223.1 ~ (all information available through the recited accession
number is
incorporated herein by reference) which is described therein as "influenza
virus
hemagglutinin 5' epitope tag=fusion protein {N-terminal, frame 1 }
[Saccharomyces
cerevisiae=yeast, YCpIF15,16,17 cloning vector, Peptide Plasmid Synthetic
Partial,
45 as]". A partial alignment demonstrating the observed homology is shown
immediately below.
>gb~AAB31223.1~ influenza virus hemagglutinin 5' epitope tag=fusion protein
{N-terminal, frame 1} [Saccharomyces cerevisiae=yeast,
YCpIF15,16,17 cloning vector, Peptide Plasmid Synthetic Partial, 45
aa]
Length = 45
Plus Strand HSPS:
Score = 124 (43.7 bits), Expect = 1.9e-06, P = 1.9e-06
Identities = 22/35 (62~), Positives = 24/35 (68~), Frame = +1
Q: 1144 PDGVEHQALCGLVPNSCSPGDPLVLERPPPRWXSS 1248
PD L+ NSCSPGDPLVLERPPPRW 5+
S: 9 PDYASSTVSISLISNSCSPGDPLVLERPPPRWSSN 43
The segment of gb~AAB31223.1 ~ that is shown as "S" above is set out in the
sequence listing as SEQ ID NO: 130. Based on the structural similarity, these
homologous polypeptides are expected to share at least some biological
activities.
Such activities are known in the art, some of which are described elsewhere
herein.
Assays for determining such activities are also known in the art, some of
which have
been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ~ NO: 131 which
corresponds to the "Q" sequence in the alignment shown above (gaps introduced
in a
sequence by the computer are, of course, removed).


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares ovary tumor NbHOT and to a lesser extent in Soares breast 3NbHBst;
normalized infant brain cDNA; Human Fetal Brain; Endothelial-induced; Soares
placenta Nb2HP; Soares infant brain 1NIB; NCI CGAP-Pr28; Soares NhHMPu S1;
5 Soares fetal liver spleen 1NFLS_S1; Stratagene fetal spleen (#937205); Human
Chondrosarcoma; Soares NhHMPu Sl; Soares~regnant uterus NbHPU;
Osteoblasts; Barstead spleen HPLRB2; Infant brain, Bento Soares; NCI CGAP_Utl;
NCI CGAP Ut4; NCI CGAP-Kidl l; Soares fetal heart NbHHI9W;
Soares senescent fibroblasts NbHSF; Stratagene schizo brain 511;
10 Soares fetal heart NbHHI9W; Soares senescent fibroblasts NbHSF; Prostate
BPH,Lib 2, subtracted; Rectum tumour; Larynx tumor; Human Kidney Cortex, re-
rescue; Human Adult Retina; Amniotic Cells - TNF induced; H. Epididiymus,
cauda;
Human Hypothalamus,schizophrenia, re-excision; Amniotic Cells - Primary
Culture;
Jurkat T-Cell, S phase; Human Brain, Striatum; L428; Human Uterine Cancer;
15 Human umbilical vein endothelial cells, IL-4 induced; Human Hippocampus;
Human
Whole Six Week Old Embryo; Hepatocellular Tumor, re-excision; 12 Week Old
Early Stage Human; Bone marrow; Endothelial cells-control; Human Amygdala;
NCI CGAP Co3; NCI CGAP GC4; NCI CGAP LuS; NCI CGAP Pr3;
NCI CGAP Kid3; Colon Normal III; Soares total fetus Nb2HF8 9w; Stratagene
20 pancreas (#937208); Soares fetal liver spleen 1NFLS and Primary Dendritic
Cells, lib
1.
Preferred polypeptides of the present invention comprise immunogenic
epitopes shown in SEQ ID NO: 81 as residues: Pro-59 to Ala-64. Polynucleotides
encoding said polypeptides are also provided.
25 Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ )D N0:44 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
30 would be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1245 of SEQ ID
N0:44, b


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
56
is an integer of 1 S to 1259, where both a and b correspond to the positions
of
nucleotide residues shown in SEQ ID N0:44, and where b is greater than or
equal to a
+ 14.
S FEATURES OF PROTEIN ENCODED BY GENE NO: 35
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
emb~CAAS9990.1 ~ (all information available through the recited accession
number is
incorporated herein by reference) which is described therein as "elastin like
protein
[Drosophila melanogaster]". A partial alignment demonstrating the observed
homology is shown immediately below.
>emb~CAA59990.1~ elastin like protein [Drosophila melanogaster]
1S >Sp~Q24333~Q24333 ELASTIN LIKE PROTEIN (FRAGMENT).
Length = 110
Plus Strand HSPs:
Score = 104 (36.6 bits), Expect = 0.00013, P = 0.00013
Identities = 21/26 (80~), Positives = 21/26 (80~), Frame = +3
Q: 18 WSXTAVAXALELVDXPGCRNSARADQ 95
WS TAVA ALELVD PGCRNSAR Q
2S S: 1 WSSTAVAAALELVDPPGCRNSARDRQ 26
The segment of emb~CAAS9990.1 ~ that is shown as "S" above is set out in the
sequence listing as SEQ ID NO: 132. Based on the structural similarity, these
homologous polypeptides are expected to share at least some biological
activities.
Such activities are known in the art, some of which are described elsewhere
herein.
Assays for determining such activities are also known in the art, some of
which have
been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ >D NO: 133 which
3S corresponds to the "Q" sequence in the alignment shown above (gaps
introduced in a
sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
NCI CGAP-Co3 and to a lesser extent in Human Chondrosarcoma;
Soares NhHMPu S1; Human Adult Testes, Large Inserts, Reexcision; Human


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
57
Pituitary, subtracted; Breast, Normal: (4005522,:;:<: B2); HM1; NCI CGAP-ColO;
NCI CGAP_KidS; NCI CGAP_Panl; Soares-pregnant uterus NbHPU;
Soares-pineal_gland N3HPG; Human adult testis, large inserts; B-cells
(stimulated);
Soares NhHMPu S1; Soares fetal lung NbHLI9W and CD34 depleted Buffy Coat
(Cord Blood), re-excision.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ )D N0:45 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 1001 of SEQ )D
N0:45, b
is an integer of 15 to 1015, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:45, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED SY GENE NO: 36
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.
gb~AAA68426.1 ~ (all information available through the recited accession
number is
incorporated herein by reference) which is described therein as "Gcapl gene
product
[Mus musculus]". A partial alignment demonstrating the observed homology is
shown
immediately below.
>gb~AAA68426.1~ Gcapl gene product [Mus musculus] >sp~Q61402~Q61402 GRANULE
CELL ANTISERUM POSITIVE 8 (GCAP1) (FRAGMENT).
Length = 85
Plus Strand HSPs:
Score = 63 (22.2 bits), Expect = 4.0, P = 0.98
Identities = 13/56 (23~), Positives = 30/56 (53~), Frame = +3
Q: '171 YYWKCSCTCWQQIKIFECLNA-LCLDL-VGIFAKLVMLLELYDYIFF*LFSWTSLYVH 338
Y C C C + ++ C++ +C+ + + I+ + + L +Y YI + T +Y++
S: 4 YMCLCVCLC---VYVYACVSLYICISIYLSIYLSISIYLSIYTYIHTHTHTHTYIYIY 58


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
58
The segment of gb~AAA68426.1 ~ that is shown as "S" above is set out in the
sequence listing as SEQ ID NO: 134. Based on the structural similarity, these
homologous polypeptides are expected to share at least some biological
activities.
Such activities are known in the art, some of which are described elsewhere
herein.
Assays for determining such activities are also known in the art, some of
which have
been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 135 which
corresponds to the "Q" sequence in the alignment shown above (gaps introduced
in a
sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
NCI CGAP_GCBl and to a lesser extent in NCI CGAP_KidS; TF-1 Cell Line GM-
CSF Treated; T-Cell PHA 24 hrs; Ulcerative Colitis; Hepatocellular Tumor, re-
excision; Colon Tumor; Colon Normal II; NCI CGAP AA1; Normalized infant
1 S brain, Bento Soares; Soares testis NHT; Soares NFL T GBC S 1; Normal
colon;
Hodgkin's Lymphoma II and Soares infant brain 1NIB.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:46 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 be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 815 of SEQ ID
N0:46, b
is an integer of 15 to 829, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:46, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 37
The computer algorithm BLASTX has been used to determine that the
translation product of this gene shares sequence homology with, as a non-
limiting
example, the sequence accessible through the following database accession no.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
59
gb~AAC48138.1 ~ (all information available through the recited accession
number is
incorporated herein by reference) which is described therein as "(AF003740)
weak
similarity to rat mitochondrial capsule selenoprotein (GB:X87883) and integrin
beta
oligodendroglia chain precursor (N>D:g520566) [Caenorhabditis elegans]". A
partial
alignment demonstrating the observed homology is shown immediately below.
>gb~AAC48138.1~ (AF003740) weak similarity to rat mitochondrial capsule
selenoprotein (GB:X87883) and integrin beta oligodendroglia chain
precursor (NID:g520566) [Caenorhabditis elegans] >sp~001972~001972
1O SIMILARITY TO RAT MITOCHONDRIAL CAPSULE SELENOPROTEIN.
Length = 532
Plus Strand HSPS:
Score = 160 (56.3 bits), Expect = 1.5e-11, Sum P(2) = 1.5e-11
Identities = 33/94 (35~), Positives = 51/94 (54~), Frame = +1
Q: 529 SGVRRIDREEXRELQALRQSREDCGCHCDR-ICDPETCSCXLAGIKCQMDHTAFP---CG 696
SGV+ +++ + +LR +R +CGC C+ +C PETC C + GI CQ+D +P C
2O S: 277 SGVK-VEKNTDA-IDSLRNTRVECGCSCENGVCLPETCQCAIDGIHCQVDGGEWPTQPCA 334
Q: 697 CCREGCENPMGRVEFNQARVQTHFIHTLTRLQLEQE 804
C E C NP GR ++ V T+ + Q+
S: 335 CFAETCTNPEGRTFYDPEAVHEFRQRTIMNWRASQK 370
The segment of gb~AAC48138.1 ~ that is shown as "S" above is set out in the
sequence listing as SEQ >D NO: 136. Based on the structural similarity, these
homologous polypeptides are expected to share at least some biological
activities.
Such activities are known in the art, some of which are described elsewhere
herein.
Assays for determining such activities are also known in the art, some of
which have
been described elsewhere herein.
Preferred polypeptides of the invention comprise a polypeptide having the
amino acid sequence set out in the sequence listing as SEQ ID NO: 137 which
corresponds to the "Q" sequence in the alignment shown above (gaps introduced
in a
sequence by the computer are, of course, removed).
The translation product of this gene shares sequence homology with Rat
neuronal immediate early gene L100 protein (see, e.g., Genseq accession
Y42774)
which is thought to be important in neural function.
Based on the sequence similarity, the translation product of this clone is
expected to
share at least some biological activities with neuronal immediate early gene
L100
protein. Such activities are known in the art, some of which are described
elsewhere
herein.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
This gene is expressed primarily in the following tissues/cDNA libraries:
Human Neutrophil, Activated and to a lesser extent in Activated T-
cell(12h)/Thiouridine-re-excision; Human Neutrophils, Activated, re-excision;
Colon
Carcinoma; Human Adult Pulmonary,re-excision; Human adult (K.Okubo);
5 Stratagene endothelial cell 937223; NCI CGAP_Panl;
Soares-placenta 8to9weeks 2NbHP8to9W; Stratagene colon (#937204); H Female
Bladder, Adult; Human Neutrophil; Human Activated T-Cells, re-excision;
NCI CGAP_GCB1; Soares-placenta 8to9weeks 2NbHP8to9W; Activated T-Cell
(l2hs)/Thiouridine labelledEco; Soares fetal liver spleen 1NFLS; Soares infant
brain
10 1NIB; Soares fetal heart NbHHI9W; H. hypothalamus, frac A,re-excision; K562
+
PMA (36 hrs); Human Pre-Differentiated Adipocytes; H. Adipose Tissue; K562 +
PMA (36 hrs),re-excision; Hodgkin's Lymphoma I; Human Pituitary, subtracted;
Human Normal Breast; Soares fetal liver spleen 1NFLS S1; Dendritic Cells From
CD34 Cells; Ovary, Cancer: Well-Differentiated Micropapillary Serous
Carcinoma,
1 S Metastatic; Human endometrial stromal cells-treated with estradiol;
Stratagene
placenta (#937225); Breast Cancer cell line, MDA 36; Salivary Gland, Lib 2;
Human
Adipose Tissue, re-excision; Human endometrial stromal cells; Human Prostate;
B-
Cells; Human Activated T-Cells; Human Adult Testes, Large Inserts, Reexcision;
Merkel Cells; Olfactory epithelium,nasalcavity; Human fetal lung; Normalized
infant
20 brain, Bento Soares; Soares NSF F8 9W OT PA P S1;
Soares fetal liver spleen_1NFLS S1; Human Testes Tumor, re-excision; Soares
breast 2NbHBst; CHME Cell Line,treated 5 hrs; Human Liver, normal; CHME Cell
Line,untreated; NCI CGAP_GC4; NCI CGAP_Pr3; NCI CGAP_Pr4.l;
Soares senescent fibroblasts NbHSF; Gessler Wilms tumor; Soares testis NHT;
25 Soares NFL T GBC_S1; Human Fetal Kidney, Reexcision; human tonsils;
Endothelial-induced; Human Bone Marrow, treated; Neutrophils IL-1 and LPS
induced; T cell helper II and Primary Dendritic Cells, lib 1.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
30 and for diagnosis of diseases and conditions which include but are not
limited to:
treatment and/or detection of immune disorders including autoimmune disease,
asthma, eosinophilia immundeficiencies, transplant rejection, GVHD, leukemia
and


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
61
microbial infection. 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.,
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 immune cells indicates the polynucleotides and
polypeptides corresponding to this gene would be useful for the diagnosis and
treatment of a variety of immune system disorders. Representative uses are
described
in the "Immune Activity" and "Infectious Disease" sections below, in Example
11,
1 S 13, 14, 16, 18, 19, 20, and 27, and elsewhere herein. Briefly, the
expression indicates
a role in regulating the proliferation; survival; differentiation; and/or
activation of
hematopoietic cell lineages, including blood stem cells. Involvement in the
regulation
of cytokine production, antigen presentation, or other processes suggests a
usefulness
for treatment of cancer (e.g. by boosting immune responses). Expression in
cells of
lymphoid origin, indicates the natural gene product would be involved in
immune
functions. Therefore it would also be useful as an agent for immunological
disorders
including arthritis, asthma, immunodeficiency diseases such as AIDS, leukemia,
rheumatoid arthritis, granulomatous disease, inflammatory bowel disease,
sepsis,
acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, such as T-cell
mediated
cytotoxicity; immune reactions to transplanted organs and tissues, such as
host-
versus-graft and graft-versus-host diseases, or autoimmunity disorders, such
as
autoimmune infertility, Tense tissue injury, demyelination, systemic lupus
erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjogren's
disease, and scleroderma. Moreover, the protein may represent a secreted
factor that
influences the differentiation or behavior of other blood cells, or that
recruits
hematopoietic cells to sites of injury. Thus, this gene product is thought to
be useful
in the expansion of stem cells and committed progenitors of various blood
lineages,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
62
and in the differentiation and/or proliferation of various cell types.
Expression of this
gene primarily in immune cells also suggests a role for the encoded protein in
the
treatment and/or detection of immune disorders including autoimmune disease,
asthma, eosinophilia immundeficiencies, transplant rejection, GVHD, leukemia
and
microbial infection.
The tissue distribution and sequence homology with Rat neuronal immediate
early gene L100 protein indicates that polynucleotides and polypeptides
corresponding to this gene would be useful for the detection, treatment,
and/or
prevention of neurodegenerative disease states, behavioral disorders, or
inflammatory
conditions. Representative uses are described in the "Regeneration" and
"Hyperproliferative Disorders" sections below, in Example 11, 15, and 18, and
elsewhere herein. Briefly, the uses include, but are not limited to the
detection,
treatment, and/or prevention of Alzheimer's Disease, Parkinson's Disease,
Huntington's Disease, Tourette Syndrome, meningitis, encephalitis,
demyelinating
diseases, peripheral neuropathies, neoplasia, trauma, congenital
malformations,
spinal cord injuries, ischemia and infarction, aneurysms, hemorrhages,
schizophrenia,
mania, dementia, paranoia, obsessive compulsive disorder, depression, panic
disorder, learning disabilities, ALS, psychoses, autism, and altered
behaviors,
including disorders in feeding, sleep patterns, balance, and perception. In
addition,
elevated expression of this gene product in regions of the brain indicates it
plays a
role in normal neural function. Potentially, this gene product is involved in
synapse
formation, neurotransmission, learning, cognition, homeostasis, or neuronal
differentiation or survival. Furthermore, the protein may also be used to
determine
biological activity, to raise antibodies, as tissue markers, to isolate
cognate ligands or
receptors, to identify agents that modulate their interactions, in addition to
its use as a
nutritional supplement. 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.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:47 and may have been publicly available prior to
conception of
the present invention. Preferably, such related polynucleotides are
specifically


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
63
excluded from the scope of the present invention. To list every related
sequence
would be cumbersome. Accordingly, preferably excluded from the present
invention
are one or more polynucleotides comprising a nucleotide sequence described by
the
general formula of a-b, where a is any integer between 1 to 2904 of SEQ ID
N0:47, b
is an integer of 1 S to 2918, where both a and b correspond to the positions
of
nucleotide residues shown in SEQ >D N0:47, and where b is greater than or
equal to a
+ 14.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
64
'+-W' 01 O v0 M ~ N
O O .--~ .~ ,~ ,~ 00 M ~' N
4r ~ .T",
y N O 01
N
4., d0
O~ 00
by
N .~ .--n .~ ,~ ~ ,~ .-r
° v~ a.
zoo°~~°' ~ ~ °
(s., ~ C/~ ~ N N ~-i N ~ N N
H
cue,, ~ ,~ ~ 41 O N ~D
00 ~D N ~n M
N ~ N N
~n ~n N O N oo v~ I~
M O~ N I~ o0 O
N a, M ~ o
N N ~~ M .~ ,-.
N
y z o_p ~
wn ~n N O N oo ~n l~
M O~ N l~ oo O vp
4 ''~j p ~p N 01 M l~ O l~ .-.
(-i ~ ~ N N ,~ M .--r
N M W v0 I~ 00
p
,~ b ~ O~ ~ O~ ~ a1 ~t a1 ~ Q\ ~ a1 ~ Q\ ~t Q\
U ~~ ~ a, a, a1 a, a, a\ a, a\ o\ o, a\ a, a, a, a, a,
~_° ~~~~~~~~~~~~~~~~
Q.N ~ ~'N~'NQ~'NdNQi'NQ~'N~'NQ~'N
d~zr~~o~o~o~o~o~o~o~o
00 V1 M N
M ~ CO
A _O ~C E"i C/~ ,~ a (~ ~ A
UU x x ~ ~ x x x x
N M vt v) ~p l~ 00
~z


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
w c,..., O O ,-, 01 01 ~' ~ ,~ N l~ v'1
O


O y p N M M ~ ~. N N


4.~


~ 00 ~O O o0 N ~ 01 ~ V


M M ~ N N ~ N M



O ~ ~ ~ M N -~ N N ON M



~ N ,-r ,~ '-. .--...-m-, '--i



~O l~ 00 01 O ~ N M ~ v~ ~O



H
z~~~ M ~ ~ ~ o


C1 N ~ N N M M M N ~ ~' 00


.~


z M ~ O ~ ~ ~'
~


b N ~ N ~t M N M 00 d~ O~O
p ~ N M



E.., ~ ~" O ~n ~O a1 0o v> o0 ~ N M
~+-~ V7 .~ 00 O M ~ M N


O ~ ~ ~ N O o0 O \p Q1 V7 N
O ~p Q\ ~O
~


U N


z O O
~'"' N
~


p _O ~ ,~ ,~ ~ M .~ ,~ ,-, ~
v~ N


U


o ~n ~ ~ .- o~ ~ ~ N


oz~ ~ ~ ~ M o ~ ~ ~
~


E-i C ,~ ~ N -~ .-y ,~ N N


01 O ~ N M ~ V7 l0 I~ 00 01


N N N N N N N N N N


O O O O


C/~ C/~ C/~ C/~
U O O O O N O N N O N O
~M ,'M ~M ,'M


~ U U U U a ~ Q
~ ~ ~ ~


~ . ~ ~ .. ~ .,


~. . ~. u.


b ~ ~ ~ ~ ~ ~ wt ~ ~ ~ ~
a1 a1 ov av ov o a, a, o~ a\ a1


a, o~ Q, o, a, a, o~ o~ a, o, o~
~ o, o, o~ o~ ~ ~ a, ~ o, o, o,


O . . . . . . . . . . .
~ c~ c~ ~ c~ ~ ~ ~ ~ ~ ~ c~


N ~ Q~' ~ d d d d Q; Q~' Q~' d
N N N N N N N N N N N


~
A


z


N ~ ~ l~ M ~ "' 00


a
~


o ~ ~ ' ~ ~


A ~ ~ ~ A ~ v


~ x ~ ~


x x x x x x x x x



O O .~ N M <t N ~O l~ 00 01





CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
66
~ M M O ~O <Y l~
O O ~ ~ ~ ~ ~ ~ N ~~ M M N
~ 4~ ~ ~'
N ~ N N N N
N
4, b4
O~~ NON ONNN
f~ ~ ~r
r
_w~
w ~ O ~ 00 M Q\ ~ ~ M N_ l~ O O
O \O ~ N .-M~ N M ~ 01
.b ~ 00 M D1 ~ ~ M N O O
O ~ N r, ~.,~ ,-, 01
p_o~~~~~~oMO~~o
N l~ ~ ~~ N ~ O M v~
M U ~ ~ ~ M ~ N N .-a .~ M
H
in U ~ M ~-
~ ~ M 00 ~ 01 M 00 O~ ,--r I~
oz~~ ~ ~ ~ ~ N o o ° °'
N M ~ N N .-. .~ M
E'' a ~ O ~~ N M own ~O t~ oo Q\ O
M M M M M M M M M M
-.-i .--n .-r Q.,
p'O~ O O ~ ~~/ ~ ~ ~OM ~~.,~~
Q. Q, ~~ ~ ~~ x Q. ~~ ~ U ~ ~~ x Pa
,r ~ ~ a, ~ oW a, ~r a\ ~ a, ~ a, ~ oW aW o, ~ a, ~ o,
(~ ~~ ~ o, a, a, ~ a, o, a~ a1 a\ a, o, a\ a1 a1 a1 a, a\ o, a1 a, o, a1
~ w ~ w ~ w ~ w ~ w ~ w ~ w ~ w ~ w ~ w ~ w
O ,'' W~ ~W~ W~ W~ W~ W~ W~ W~ W~ W~ W~
N cd ~NQ~'NQ~'N d'N Q~'N d'NdN dN d'N d'N Q,'N
ate., o aE''., o p.E''., o ate.., o ate., o p.~., o per.., o ~ o QE''., o
ate.., o ate., o
M N ~ N ,~ l~ ~ l~ l~ N
z~o x ~ ~ ~w ~' ~ ~ W d
~ W U U W ~ ~ a ~ pV-, ran
U x x ~ ~ x x x x
O O ~ N M ~t ~ ~O l~ 00 01 O
,UzNNNNNNNNNNM


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
67
O o
o


O l~ ~ m I~ ~ M N


.b
~"..
4-r


~ 0 ~0 01 ~ I~ ~ Q1
~
~'~


y. N N ~ M N N N
.,~


N



d~
~ ~ O N N


O~~ N M N



bA
4~ ~ ~ ~ --. -
_ O ~ N


w P~


00 41 O ~ N M



O


z p ~ ~ ~ ~D ~ N
~ O


p~ ~ ,~ .~ .-. ~. ,~


~ N


H
.~ I~ N ~


_
O ~ \D ~ ~


C/~ O ' N
U -'


H O M V7 O~ I~
z
o


o o
~ ~ ~ ~


cn U .~ ,.~ ,~ N


O N
zo
o~


_


U v~ '-' '''


-~ v~ ~ pm n ~ 00
H oo ~ ~n


Hz~ o



a~
H ~ N M ~ V~ ~O l~
~


W Z
z


o ~ ~ ~ ~ ~


~ ~ ~ ~ ~ ~ ~
. .


. .
x _ _


~


.



~ a~
a\ a\ ~ ~ a\ o, a\
ov o, ov a, ~ a, a\


U O


~
H d ~' d' d ~ ~' ~'
r~ N N N N N N N


dr~z ~o ~o ~o ~o ~o ~o ~o



M ~ O ~ V7 V7
W N f~
W


~ ~


H ~ ~


d U U U
x ~


x x x x x


N M ~ vW p l~


M M M M M M M




CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
68
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 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.
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 "Gene No." The deposited clone may contain all or most of these
sequences, reflected by the nucleotide position indicated as "S' 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 start codon (methionine) is identified as "5' NT of Start
Codon."
Similarly , the nucleotide position of SEQ ID NO:X of the predicted signal
sequence
is identified as "5' NT of First AA of Signal Pep."
The translated amino acid sequence, beginning with the methionine, 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.
The first and last amino acid position of SEQ ID NO:Y of the predicted signal
peptide is identified as "First AA of Sig Pep" and "Last AA of Sig Pep." The
predicted first amino acid position of SEQ ID NO:Y of the secreted portion is
identified as "Predicted First AA of Secreted Portion." Finally, the amino
acid
position of SEQ ID NO:Y of the last amino acid in the open reading frame is
identified as "Last AA of ORF."
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
69
accurate and otherwise suitable for a variety of uses well known in the art
and
described further below. For instance, SEQ >D NO:X is useful for designing
nucleic
acid hybridization probes that will detect nucleic acid sequences contained in
SEQ >D
NO:X or the cDNA contained in the deposited clone. 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 may be used, for example, to generate antibodies
which bind specifically to proteins containing the polypeptides and 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 ID 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 each deposited clone can
readily
be determined by sequencing the deposited clone 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 clone
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.
The present invention also relates to the genes corresponding to SEQ ID
NO:X, SEQ ID NO:Y, or the deposited clone. The corresponding gene can be
isolated in accordance with known methods using the sequence information
disclosed


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
herein. Such methods include 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
S 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, or a
deposited clone, using information from the sequences disclosed herein or the
clones
deposited with the ATCC. For example, allelic variants and/or species homologs
may
10 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.
Table 2 summarizes the expression profile of polynucleotides corresponding to
the
15 clones disclosed in Table 1. The first column provides a unique clone
identifier,
"Clone ID", for a cDNA clone related to each contig sequence disclosed in
Table 1.
Column 2, "Library Codes" 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
20 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
invention
or to identify polynucleotides which show predominant and/or specific tissue
25 expression.
Table 3, column 1, provides a nucleotide sequence identifier, "SEQ ID
NO:X," that matches a nucleotide SEQ ID NO:X disclosed in Table 1, column 5.
Table 3, column 2, provides the chromosomal location, "Cytologic Band or
Chromosome," of polynucleotides corresponding to SEQ ID NO:X. Chromosomal
30 location was determined by finding exact matches to EST and cDNA sequences
contained in the NCBI (National Center for Biotechnology Information) UniGene
database. Given a presumptive chromosomal location, disease locus association
was


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
71
determined by comparison with the Morbid Map, derived from Online Mendelian
Inheritance in Man (Online Mendelian Inheritance in Man, OMIMTM. McKusick-
Nathans Institute for Genetic Medicine, Johns Hopkins University (Baltimore,
MD)
and National Center for Biotechnology Information, National Library of
Medicine
(Bethesda, MD) 2000. World Wide Web URL: http://www.ncbi.nlm.nih.gov/omim/).
If the putative chromosomal location of the Query overlapped with the
chromosomal
location of a Morbid Map entry, the OMIM reference identification number of
the
morbid map entry is provided in Table 3, column 3, labelled "OMIM ID." A key
to
the OMIM reference identification numbers is provided in Table 5.
Table 4 provides a key to the Library Code disclosed in Table 2. Column 1
provides the Library Code disclosed in Table 2, column 2. Column 2 provides a
description of the tissue or cell source from which the corresponding library
was
derived.
Table 5 provides a key to the OMIM reference identification numbers
disclosed in Table 3, column 3. OMIM reference identification numbers (Column
1)
were derived from Online Mendelian Inheritance in Man (Online Mendelian
Inheritance in Man, OMIM. McKusick-Nathans Institute for Genetic Medicine,
Johns
Hopkins University (Baltimore, MD) and National Center for Biotechnology
Information, National Library of Medicine, (Bethesda, MD) 2000. World Wide Web
URL: http://www.ncbi.nlm.nih.gov/omim/). Column 2 provides diseases associated
with the cytologic band disclosed in Table 3, column 2, as determined using
the
Morbid Map database.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
72
Table 2
... ., .,


HFXLH94 H0052 H0561 H0622 50282


HLTGE14 H0031 H0083 H0251 H0266 H0356 H0369 H0422 H0423
H0445 H0455


H0459 H0486 H0506 H0560 H0580 H0581 H0586 H0587
H0591 H0601


H0623 H0624 H0633 H0644 H0660 H0690 L1290 S0003
S0013 S0026


50027 S0116 50126 S0212 50214 S0242 S0358 50360
50380 S0418


S0420 S0424 S0472 S3014 T0049


HMSHT78 H0063 L1290 S0002 S0362


HOVAW46 H0135 H0428


HSLCR68 H0014 H0036 H0046 H0050 H0052 H0136 H0144 H0150
H0178 H0341


H0365 H0519 H0521 H0542 H0553 H0556 H0591 H0598
H0599 H0638


H0646 H0648 H0663 H0667 L1290 S0002 S0026 S0028
50114 50132


S0134 50250 S0330 50346 50358 S0360 50364 50388
53012 56028


HTGER35 H0309 H0402 H0427 L1290 50114 S0218


HAJAA13 H0561


HCDAS02 H0014 H0059 H0087 H0123 H0251 H0253 H0266 H0267
H0379 H0547


H0555 H0560 H0580 H0617 H0618 H0638 H0656 H0688
L1290 50040


S0051 S0144 50222 S0356


HDPDI72 H0521 S0358 50374


HDPIE44 H0012 H0013 H0014 H0024 H0038 H0040 H0046 H0050
H0051 H0052


H0090 H0135 H0144 H0163 H0171 H0172 H0196 H0208
H0251 H0263


H0265 H0294 H0328 H0355 H0356 H0375 H0423 H0427
H0428 H0455


H0486 H0488 H0494 H0506 H0521 H0538 H0539 H0547
H0551 H0553


H0555 H0561 H0591 H0599 H0615 H0619 H0620 H0622
H0623 H0624


H0625 H0634 H0644 H0651 H0656 H0658 H0670 H0672
H0683 H0687


H0688 H0689 L1290 50007 S0010 50016 S0028 S0045
50126 S0192


50206 S0212 50222 50308 S0316 50330 50344 S0356
50360 S0364


S0366 50374 S0376 50386 50390 50418 50422 S0436
T0042


HDPKD16 H0124 H0231 H0392 H0494 H0521 H0543 H0553 H0556
H0625 H0628


H0674 L1290 S0126 S0216 S0332 S0424


HDPSZ07 H0521 H0543 H0589 S0114 50134


HFCBL53 H0009 H0013 H0134 H0135 H0144 H0163 H0170 H0178
H0252 H0265


H0455 H0494 H0556 H0677 L1290 50001 S0007 50010
50026 50028


50036 S0222 50300 50346 S0384 S0388 S6024


HAAAT06 H0009 H0013 H0024 H0028 H0040 H0050 H0135 H0144
H0150 H0244


H0264 H0265 H0271 H0284 H0286 H0292 H0341 H0354
H0393 H0413


H0422 H0423 H0436 H0445 H0486 H0519 H0520 H0539
H0542 H0543


H0545 H0553 H0556 H0581 H0599 H0615 H0616 H0619
H0620 H0622


H0631 H0646 H0656 H0657 H0658 H0666 H0672 H0684
H0689 L1290


50046 S0114 S0116 50134 S0208 50212 50220 S0242
50260 S0360


S0378 50400 S0420 T0010 T0023


HAQBKO1 H0030 H0050 H0136 H0144 H0170 H0246 H0252 H0295
H0331 H0370


H0412 H0521 H0544 H0561 H0590 H0620 H0622 L1290
50011 S0344


50472 T0010


HCEEG48 H0052 H0257 H0261 L1290


HCRMA24 L1290 S0356


HTLHN86 H0009 H0012 H0013 H0024 H0033 H0038 H0046 H0052
H0085 H0117


H0123 H0125 H0134 H0135 H0144 H0181 H0188 H0231
H0253 H0255


H0266 H0318 H0370 H0381 H0392 H0412 H0413 H0419
H0421 H0423


H0427 H0436 H0438 H0444 H044S H0488 H0494 HOS20
H0529 H0547




CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
73
H0550H0555H0556H0575H0594H0599H0616H0617 H0618
H0619


H0620H0656H0687H0690H0694L129050001S0002 50036
S0049


S0050S0114S011650142
50330
S0360
S0364
S0366
50378
50390


S0410S0418S042050448
53012
56028
T0010
T0041
T0042
T0082


HCFCG91 H0040H0052H0156H0166H0329H0415H0422H0428 H0435
H0455


H0478H0494H0506H0509H0518H0520H0521H0538 H0551
H0553


H0555H0560H0598H0639H0645H0657H0659H0663 H0687
L1290


S00315012250358S0386
S0406


HDPOD73 H0521H0522H0581L129050356S0360


HE9HE13 H0024H0031H0144H0156H0623L129050001S0038 50222
50300


S6024S6028


HBCJL35 H0009H0013H0046H0052H0059H0135H0144H0156 H0171
H0201


H0294H0318H0341H0369H0411H0416H0423H0436 H0457
H0483


H0486H0519H0539H0547H0555H0561H0581H0615 H0616
H0624


H0644H0645H0658H0660H0662H0664H0688L1290 S0010
S0046


50049S0114S0116
S0194
S0212
S0222
50344
50360
50402


HDCBM09 H0637


HFRBF41 HO1005005050374


HKGBJ47 H0012H0013H0024H0038H0040H0050HO100H0144 H0252
H0333


H0375H0381H0411H0422H0428H0445H0520H0521 H0522
H0538


H0555H0556H0616H0620H0651L1290S015250280 50300
50358


50424S6022T0006T0041


HLTCV76 H0013H0014H0015H0024H0031H0038H0040H0042 H0046
H0050


H0051H0052H0057H0069H0090H0109H0122H0131 H0140
H0144


H0151H0170H0171H0179H0194H0196H0244H0252 H0264
H0266


H0267H0309H0355H0369H0370H0373H0422H0427 H0431
H0436


H0441H0445H0486H0488H0492H0506H0509H0520 H0521
H0522


H0529H0540H0542H0543H0547H0553H0555H0556 H0574
H0575


H0580H0581H0586H0590H0591H0595H0598H0599 H0615
H0616


H0619H0622H0623H0624H0634H0635H0644H0645 H0646
H0648


H0656H0659H0660H0661H0662H0667H0672H0674 H0684
L1290


50002S0003S0022
S0026
S0028
50045
S0046
S0051
S0052
S0116


S01265013450136
50142
S0152
S0192
50206
50210
50212
S0250


S02785034650356
S0358
S0360
S0374
50376
50378
S0380
50388


S0408S0418S0424
S0428
S6028
T0006
T0010
T0041
T0067


HNTEF67 H0519


HPCAL37 H0255S0146


HSKGN92 H0050H0124H0144H0208H0266H0286H0288H0412 H0445
H0486


H0521H0539H0542H0555H0575H0599H0622H0623 H0634
H0660


H0682L1290S00275002850031
50038
S0045
S0126
50192
50212


5022250250S0276
S0282
S0360
50390
50420
S3014
S6028
T0060


HTLIF12 H0038H0253H0411H0616H0618L1290S0398


HATCX03 H0038H0052H0156H0305H0616L1290S001050049 S0053
S0222


HBGBE75 H0012H0087HO100H0163H0181H0182H0188H0213 H0251
H0254


H0265H0266H0295H0412H0539H0546H0556H0585 H0598
H0606


H0617H0620H0661H0673H0684L129050134S0330 S0366
S0380


S0422


HBIBB20 H0009H0051H0052H0059HO100H0135H0170H0251 H0266
H0271


H0294H0295H0356H0370H0399H0412H0427H0483 H0519
H0520


H0521H0544H0550H0561H0570H0574H0586H0592 H0599
H0619


H0622H0623L129050010S004550046 0116 50126
S0049 S0152
5


S0190S0294S0328
S0358
S0378
S0388
S0406
50414
50418
T0042


T0082




CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
74
HCDAA24 H0052 H0213 H0251 H0253 H0255 H0402 H0445 H0478
H0546 H0617


H0618 H0638 H0657 H0661 H0662 H0696 L1290 50330
50354 S0358


50378 S6028


HCFMR75 H0423 H0486 H0506 H0574 L1290 50330 50354 S0374
S0376 S0424



HCRMR35 H0032 H0069 H0179 H0188 H0213 H0264 H0265 H0271
H0280 H0411


H0416 H0429 H0436 H0478 H0483 H0485 H0510 H0521
H0538 H0543


H0544 H0546 H0556 H0575 H0581 H0593 H0618 H0620
H0628 H0634


H0635 H0637 H0641 H0648 H0650 H0656 H0670 L1290
S0046 S0053


S0220 S0356 S0418 S0420 S6022




Image


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
76
Table 4
". , , ,
H0009 Human Fetal Brain


H0012 Human Fetal Kidne


H0013 Human 8 Week Whole Emb o


H0014 Human Gall Bladder


H0015 Human Gall Bladder, fraction II


H0024 Human Fetal Lung III


H0028 Human Old Ovary


H0030 Human Placenta


H0031 Human Placenta


H0032 Human Prostate


H0033 Human Pituitar


H0036 Human Adult Small Intestine


H0038 Human Testes


H0040 Human Testes Tumor


H0042 Human Adult Pulmonar


H0046 Human Endometrial Tumor


H0050 Human Fetal Heart


H0051 Human Hi ocam us


H0052 Human Cerebellum


H0057 Human Fetal Spleen


H0059 Human Uterine Cancer


H0063 Human Thymus


H0069 Human Activated T-Cells


H0083 HUMAN JURKAT MEMBRANE BOUND POLYSOMES


H0085 Human Colon


H0087 Human Th us


H0090 Human T-Cell L m homa


HO100 Human Whole Six Week Old Emb o


H0109 Human Macro ha e, subtracted


H0117 Human Uterine Cancer, subtracted


H0122 Human Adult Skeletal Muscle


H0123 Human Fetal Dura Mater


H0124 Human Rhabdom osarcoma


H0125 Cem cells cyclohexamide treated


H0131 LNCAP + o.3nM 81881


H0134 Raji Cells, cyclohexamide treated


H0135 Human S ovial Sarcoma


H0136 Su t Cells, c clohexamide treated


H0140 Activated T-Cells, 8 hrs.


H0144 Nine Week Old Early Sta a Human


HO150 Human E idid us


H0151 Earl Sta a Human Liver


H0156 Human Adrenal Gland Tumor


H0163 Human S ovium


H0166 Human Prostate Cancer, Sta a B2 fraction


H0170 12 Week Old Earl Sta a Human


H0171 12 Week Old Early Stage Human, II


H0172 Human Fetal Brain, random rimed




CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
77
H0178 Human Fetal Brain


H0179 Human Neutro hil


H0181 Human Primar Breast Cancer


H0182 Human Primar Breast Cancer


H0188 Human Normal Breast


H0194 Human Cerebellum, subtracted


H0196 Human Cardiom o athy, subtracted


H0201 Human Hi ocam us, subtracted


H0208 Early Stage Human Lung, subtracted


H0213 Human Pituitar , subtracted


H0231 Human Colon, subtraction


H0244 Human 8 Week Whole Embr o, subtracted


H0246 Human Fetal Liver- Enz me subtraction


H0251 Human Chondrosarcoma


H0252 Human Osteosarcoma


H0253 Human adult testis, lar a inserts


H0254 Breast L h node cDNA librar


H0255 breast I h node CDNA Libra


H0257 HL-60, PMA 4H


H0261 H. cerebellum, Enzyme subtracted


H0263 human colon cancer


H0264 human tonsils


H0265 Activated T-Cell (l2hs)/Thiouridine IabelledEco


H0266 Human Microvascular Endothelial Cells, fract.
A


H0267 Human Microvascular Endothelial Cells, fract.
B


H0271 Human Neutro hil, Activated


H0280 K562 + PMA 36 hrs)


H0284 Human OB MG63 control fraction I


H0286 Human OB MG63 treated 10 nM E2) fraction I


H0288 Human OB HOS control fraction I


H0292 Human OB HOS treated 10 nM E2) fraction I


H0294 Amniotic Cells - TNF induced


H0295 Amniotic Cells - Prima Culture


H0305 CD34 ositive cells (Cord Blood)


H0309 Human Chronic Synovitis


H0318 HUMAN B CELL LYMPHOMA


H0328 human ovarian cancer


H0329 Dermatofibrosarcoma Protuberance


H0331 He atocellular Tumor


H0333 Hemangio eric oma


H0341 Bone Marrow Cell Line (RS4,11


H0354 Human Leukoc es


H0355 Human Liver


H0356 Human Kidne


H0365 Osteoclastoma-normalized B


H0369 H. Atro hic Endometrium


H0370 H. Lym h node breast Cancer


H0373 Human Heart


H0375 Human Lung


H0379 Human Ton ue, frac 1


H0381 Bone Cancer


H0392 H. Menin ima, M1


H0393 ~ Fetal Liver, subtraction II




CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
78
H0399 Human Kidne Cortex, re-rescue


H0402 CD34 de leted Buff Coat Cord Blood , re-excision


H0411 H Female Bladder, Adult


H0412 Human umbilical vein endothelial cells, IL-4
induced


H0413 Human Umbilical Vein Endothelial Cells, uninduced


H0415 H. Ovarian Tumor, II, OV5232


H0416 Human Neutro hils, Activated, re-excision


H0419 Bone Cancer, re-excision


H0421 Human Bone Marrow, re-excision


H0422 T-Cell PHA 16 hrs


H0423 T-Cell PHA 24 hrs


H0427 Human Adi ose


H0428 Human Ova


H0429 K562 + PMA 36 hrs ,re-excision


H0431 H. Kidne Medulla, re-excision


H0435 Ovarian Tumor 10-3-95


H0436 Resting T-Cell Librar ,II


H0438 H. Whole Brain #2, re-excision


H0441 H. Kidney Cortex, subtracted


H0444 Spleen metastic melanoma


H0445 Spleen, Chronic 1 hocytic leukemia


H0455 H. Striatum Depression, subt


H0457 Human Eosino hils


H0459 CD34+cells, II, FRACTION 2


H0478 Salivary Gland, Lib 2


H0483 Breast Cancer cell line, MDA 36


H0485 Hodgkin's L homy I


H0486 Hod kin's L m homy II


H0488 Human Tonsils, Lib 2


H0492 HL-60, RA 4h, Subtracted


H0494 Keratinoc a


H0506 Ulcerative Colitis


H0509 Liver, He atoma


H0510 Human Liver, normal


H0518 BMC stimulated w/ oly I/C


H0519 NTERA2, control


H0520 NTERA2 + retinoic acid, 14 da s


H0521 Prima Dendritic Cells, lib 1


H0522 Primar Dendritic cells,frac 2


H0529 M oloid Pro enitor Cell Line


H0538 Merkel Cells


H0539 Pancreas Islet Cell Tumor


H0540 Skin, burned


H0542 T Cell hel er I


H0543 T cell hel er II


H0544 Human endometrial stromal cells


H0545 Human endometrial stromal cells-treated with
rogesterone


H0546 Human endometrial stromal cells-treated with
estradiol


H0547 NTERA2 teratocarcinoma cell line+retinoic acid
( 14 days)


H0550 H. E ididi us, cauda


H0551 Human Th us Stromal Cells


H0553 Human Placenta


H0555 Rejected Kidney, lib 4




CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
79
H0556 Activated T-cell 12h /Thiouridine-re-excision


H0560 KMH2


H0561 L428


H0570 Human Fetal Brain, normalized C500H


H0574 He atocellular Tumor, re-excision


H0575 Human Adult Pulmonar ,re-excision


H0580 Dendritic cells, ooled


H0581 Human Bone Marrow, treated


H0585 Activated T-Cells,l2 hrs,re-excision


H0586 Healin roin wound, 6.5 hours ost incision


H0587 Healin roin wound, 7.5 hours ost incision


H0589 CD34 ositive cells cord blood ,re-ex


H0590 Human adult small intestine,re-excision


H0591 Human T-cell 1 m homa,re-excision


H0592 Healing roin wound - zero hr ost-incision control


H0593 Olfacto a ithelium,nasalcavit


H0594 Human Lun Cancer,re-excision


H0595 Stomach cancer human ,re-excision


H0598 Human Stomach,re-excision


H0599 Human Adult Heart,re-excision


H0601 Healin Abdomen Wound,l5 da s ost incision


H0606 Human Primar Breast Cancer,re-excision


H0615 Human Ovarian Cancer Reexcision


H0616 Human Testes, Reexcision


H0617 Human Primar Breast Cancer Reexcision


H0618 Human Adult Testes, Lar a Inserts, Reexcision


H0619 Fetal Heart


H0620 Human Fetal Kidne , Reexcision


H0622 Human Pancreas Tumor, Reexcision


H0623 Human Umbilical Vein, Reexcision


H0624 12 Week Earl Sta a Human II, Reexcision


H0625 Ku 812F Baso hils Line


H0628 Human Pre-Differentiated Adi oc es


H0631 Saos2, Dexamethosome Treated


H0633 Lung Carcinoma A549 TNFaI ha activated


H0634 Human Testes Tumor, re-excision


H0635 Human Activated T-Cells, re-excision


H0637 Dendritic Cells From CD34 Cells


H0638 CD40 activated monoc a dendridic cells


H0639 Ficolled Human Stromal Cells, 5Fu treated


H0641 LPS activated derived dendritic cells


H0644 Human Placenta re-excision


H0645 Fetal Heart, re-excision


H0646 Lung, Cancer (4005313 A3): Invasive Poorly Differentiated
Lung
Adenocarcinoma,


H0648 Ovary, Cancer: (4004562 B6) Papillary Serous
Cystic Neoplasm, Low
Mali nant Pot


H0650 B-Cells


H0651 Ovar , Normal: (9805C040R


H0656 B-cells unstimulated


H0657 B-cells stimulated)


H0658 Ova , Cancer 9809C332 : Poorl differentiated
adenocarcinoma


H0659 Ova , Cancer 15395A1F : Grade II Pa illa Carcinoma


H0660 Ovary, Cancer: (15799A1F) Poorly differentiated
carcinoma




CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
H0661 Breast, Cancer: 4004943 A5)


H0662 Breast, Norial: 400552282


H0663 Breast, Cancer: 4005522 A2


H0664 Breast, Cancer: 9806C012R


H0666 Ova , Cancer: 4004332 A2


H0667 Stromal cells HBM3.18


H0670 Ovary, Cancer(4004650 A3): Well-Differentiated
Micropapillary Serous
Carcinoma


H0672 Ovar , Cancer: 4004576 A8


H0673 Human Prostate Cancer, Stage B2, re-excision


H0674 Human Prostate Cancer, Stage C, re-excission


H0677 TNFR degenerate oligo


H0682 Ovarian cancer, Serous Pa illary Adenocarcinoma


H0683 Ovarian cancer, Serous Pa illar Adenocarcinoma


H0684 Ovarian cancer, Serous Pa illar Adenocarcinoma


H0687 Human normal ova (#96106215


H0688 Human Ovarian Cancer(#98076017


H0689 Ovarian Cancer


H0690 Ovarian Cancer, # 97026001


H0694 Prostate cancer adenocarcinoma)


H0696 Prostate Adenocarcinoma


L1290 Strata ene lacenta #937225)


50001 Brain frontal cortex


50002 Monocyte activated


50003 Human Osteoclastoma


50007 Early Stage Human Brain


50010 Human Am dala


50011 STROMAL -OSTEOCLASTOMA


50013 Prostate


50016 Kidne P amids


50022 Human Osteoclastoma Stromal Cells - unam lified


50026 Stromal cell TF274


S0027 Smooth muscle, serum treated


50028 Smooth muscle,control


50031 S final cord


50036 Human Substantia Ni a


S0038 Human Whole Brain #2 - Oligo dT > l.SKb


S0040 Adi oc es


S0045 Endothelial cells-control


S0046 Endothelial-induced


S0049 Human Brain, Striatum


S0050 Human Frontal Cortex, Schizo hrenia


50051 Human H othalmus,Schizo hrenia


50052 neutro hils control


S0053 Neutro hils IL-1 and LPS induced


S0114 Aner is T-cell


S0116 Bone marrow


S0122 Osteoclastoma-normalized A


S0126 Osteoblasts


50132 E ithelial-TNFa and INF induced


50134 A o totic T-cell


S0136 PERM TF274


50142 Macro hage-oxLDL


_ Macrophage (GM-CSF treated)
50144 ~




CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
81
S0146 rostate-edited


50152 PC3 Prostate cell line


50190 Prostate BPH,Lib 2, subtracted


50192 S novial Fibroblasts control


S0194 S novial h oxia


S0206 Smooth Muscle- HASTE normalized


S0208 Messangial cell, frac 1


S0210 Messangial cell, frac 2


S0212 Bone Marrow Stromal Cell, untreated


S0214 Human Osteoclastoma, re-excision


S0216 Neutro hils IL-1 and LPS induced


50218 A o totic T-cell, re-excision


50220 H. h othalamus, frac A,re-excision


50222 H. Frontal cortex,e file tic,re-excision


50242 S ovial Fibroblasts (Ill/TNF , subt


50250 Human Osteoblasts II


S0260 S final Cord, re-excision


S0276 S ovial h oxia-RSF subtracted


S0278 H Macro hage (GM-CSF treated , re-excision


S0280 Human Adi ose Tissue, re-excision


S0282 Brain Frontal Cortex, re-excision


50294 Larynx tumor


50300 Frontallobe,dementia,re-excision


S0308 S leen/normal


S0316 Human Normal Cartilage,Fraction I


S0328 Palate carcinoma


S0330 Palate normal


S0332 Pha carcinoma


S0344 Macro hage-oxLDL, re-excision


S0346 Human Am gdala,re-excision


50354 Colon Normal II


S0356 Colon Carcinoma


S0358 Colon Normal III


50360 Colon Tumor II


50362 Human Gastrocnemius


50364 Human Quadrice s


50366 Human Soleus


S0374 Normal colon


S0376 Colon Tumor


S0378 Pancreas normal PCA4 No


S0380 Pancreas Tumor PCA4 Tu


50384 Ton ue carcinoma


S0386 Human Whole Brain, re-excision


S0388 Human H othalamus,schizo hrenia, re-excision


S0390 Smooth muscle, control, re-excision


S0398 Testis, normal


S0400 Brain, normal


S0402 Adrenal Gland,normal


S0406 Rectum tumour


S0408 Colon, normal


S0410 Colon, tumour


S0414 Hi ocam us, Alzheimer Subtracted


S0418 CHME Cell Line,treated 5 hrs




CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
82
50420 CHME Cell Line,untreated


50422 Mo7e Cell Line GM-CSF treated In /ml


50424 TF-1 Cell Line GM-CSF Treated


S0428 Neutro hils control, re-excision


S0436 Stomach Tumour


50448 Lar x Normal


S0472 Lung Mesothelium


53012 Smooth Muscle Serum Treated, Norm


S3014 Smooth muscle, serum induced,re-exc


56022 H. Adi ose Tissue


S6024 Alzheimers, s on chan a


S6028 Human Manic De ression Tissue


T0006 Human Pineal Gland


T0010 Human Infant Brain


T0023 Human Pancreatic Carcinoma


T0041 Jurkat T-cell G1 hase


T0042 Jurkat T-Cell, S hase


T0049 Aorta endothelial cells + TNF-a


T0060 Human White Adi ose


T0067 Human Thyroid


T0082 Human Adult Retina



Image


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
84
The polypeptides of the invention can be prepared in any suitable manner.
Such polypeptides include isolated naturally occurnng 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 secreted
protein.
The present invention provides a polynucleotide comprising, or alternatively
consisting of, the nucleic acid sequence of SEQ ID NO:X, and/or a cDNA
contained
in ATCC deposit Z. The present invention also provides a polypeptide
comprising, or
alternatively, consisting of, the polypeptide sequence of SEQ ID NO:Y and/or a
polypeptide encoded by the cDNA contained in ATCC deposit Z. Polynucleotides
encoding a polypeptide comprising, or alternatively consisting of the
polypeptide
sequence of SEQ ID NO:Y and/or a polypeptide sequence encoded by the cDNA
contained in ATCC deposit Z are also encompassed by the invention.
Signal Sequences
The present invention also encompasses mature forms of the polypeptide
having the polypeptide sequence of SEQ m NO:Y and/or the polypeptide sequence
encoded by the cDNA in a deposited clone. Polynucleotides encoding the mature
forms (such as, for example, the polynucleotide sequence in SEQ m NO:X and/or
the


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
polynucleotide sequence contained in the cDNA of a deposited clone) are also
encompassed by the invention. According to the signal hypothesis, proteins
secreted
by mammalian cells have a signal or secretary leader sequence which is cleaved
from
the mature protein once export of the growing protein chain across the rough
5 endoplasmic reticulum has been initiated. Most mammalian cells and even
insect
cells cleave secreted proteins with the same specificity. However, in some
cases,
cleavage of a secreted protein is not entirely uniform, which results in two
or more
mature species of the protein. Further, it has long been known that cleavage
specificity of a secreted protein is ultimately determined by the primary
structure of
10 the complete protein, that is, it is inherent in the amino acid sequence of
the
polypeptide.
Methods for predicting whether a protein has a signal sequence, as well as the
cleavage point for that sequence, are available. For instance, the method of
McGeoch, Virus Res. 3:271-286 (1985), uses the information from a short N-
terminal
15 charged region and a subsequent uncharged region of the complete
(uncleaved)
protein. The method of von Heinje, Nucleic Acids Res. 14:4683-4690 (1986) uses
the
information from the residues surrounding the cleavage site, typically
residues -13 to
+2, where +1 indicates the amino terminus of the secreted protein. The
accuracy of
predicting the cleavage points of known mammalian secretory proteins for each
of
20 these methods is in the range of 75-80%. (von Heinje, supra.) However, the
two
methods do not always produce the same predicted cleavage points) for a given
protein.
In the present case, the deduced amino acid sequence of the secreted
polypeptide was analyzed by a computer program called SignalP (Henrik Nielsen
et
25 al., Protein Engineering 10:1-6 (1997)), which predicts the cellular
location of a
protein based on the amino acid sequence. As part of this computational
prediction of
localization, the methods of McGeoch and von Heinje are incorporated. The
analysis
of the amino acid sequences of the secreted proteins described herein by this
program
provided the results shown in Table 1.
30 As one of ordinary skill would appreciate, however, cleavage sites
sometimes
vary from organism to organism and cannot be predicted with absolute
certainty.
Accordingly, the present invention provides secreted polypeptides having a
sequence


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
86
shown in SEQ >D NO:Y which have an N-terminus beginning within 5 residues
(i.e.,
+ or - 5 residues) of the predicted cleavage point. Similarly, it is also
recognized that
in some cases, cleavage of the signal sequence from a secreted protein is not
entirely
uniform, resulting in more than one secreted species. These polypeptides, and
the
polynucleotides encoding such polypeptides, are contemplated by the present
invention.
Moreover, the signal sequence identified by the above analysis may not
necessarily predict the naturally occurring signal sequence. For example, the
naturally occurnng signal sequence may be further upstream from the predicted
signal
sequence. However, it is likely that the predicted signal sequence will be
capable of
directing the secreted protein to the ER. Nonetheless, the present invention
provides
the mature protein produced by expression of the polynucleotide sequence of
SEQ >D
NO:X and/or the polynucleotide sequence contained in the cDNA of a deposited
clone, in a mammalian cell (e.g., COS cells, as desribed below). These
polypeptides,
and the polynucleotides encoding such polypeptides, are contemplated by the
present
invention.
Polynucleotide and Polypeptide Variants
The present invention is directed to variants of the polynucleotide sequence
disclosed in SEQ m NO:X, the complementary strand thereto, and/or the cDNA
sequence contained in a deposited clone.
The present invention also encompasses variants of the polypeptide sequence
disclosed in SEQ ID NO:Y and/or encoded by a deposited clone.
"Variant" refers to a polynucleotide or polypeptide differing from the
polynucleotide or polypeptide of the present invention, but retaining
essential
properties thereof. Generally, variants are overall closely similar, and, in
many
regions, identical to the polynucleotide or polypeptide of the present
invention.
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% or 99% identical to, for example, the nucleotide
coding sequence in SEQ ID NO:X or the complementary strand thereto, the
nucleotide coding sequence contained in a deposited cDNA clone or the


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
87
complementary strand thereto, a nucleotide sequence encoding the polypeptide
of
SEQ m NO:Y, a nucleotide sequence encoding the polypeptide encoded by the
cDNA contained in a deposited clone, and/or polynucleotide fragments of any of
these nucleic acid molecules (e.g., those fragments described herein).
Polynucleotides which hybridize to these nucleic acid molecules under
stringent
hybridization conditions or 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% identical to, for example, the polypeptide sequence
shown in SEQ m NO:Y, the polypeptide sequence encoded by the cDNA contained
in a deposited clone, andlor polypeptide fragments of any of these
polypeptides (e.g.,
those fragments described herein).
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 5% of the nucleotides in
the
reference sequence may be deleted or substituted with another nucleotide, or a
number of nucleotides up to 5% of the total nucleotides in the reference
sequence may
be inserted into the reference sequence. The query sequence may be an entire
sequence shown inTable 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 presence 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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
88
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=S, Gap Size Penalty 0.05, Window Size=500 or the lenght of the subject
nucleotide sequence, whichever is shorter.
If the subject sequence is shorter than the query sequence because of S' 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
5' 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
5' 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 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 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
89
deletions are internal deletions so that there are no bases on the 5' or 3' of
the subject
sequence which are not matched/aligned with the query. In this case the
percent
identity calculated by 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 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 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%, 95%, 96%, 97%, 98% or 99% identical to, for instance, an amino acid
sequences shown in Table 1 (SEQ >D NO:Y) or to the amino acid sequence encoded
by cDNA contained in a deposited clone can be determined conventionally using
known computer programs. A preferred method for determing 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 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
Size=S00 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-

5 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
10 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
15 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
20 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
25 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
30 with 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 subject sequence, as displayed in the FASTDB alignment, which are
not


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
91
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 5-10, 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 occurring
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, one or more amino acids
can be
deleted from the N-terminus or C-terminus of the secreted protein 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 a$er deleting 8-10 amino acid
residues from
the carboxy terminus of this protein. (Dobeli et al., J. Biotechnology 7:199-
216
(1988).)
Moreover, ample evidence demonstrates that variants o$en retain a biological
activity similar to that of the naturally occurring protein. For example,
Gayle and
coworkers (J. Biol. Chem 268:22105-22111 (1993)) conducted extensive
mutational
analysis of human cytokine IL-1 a. They used random mutagenesis to generate
over


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
92
3,500 individual IL-1 a 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, 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 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
substantial biological activity. 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. For example, guidance concerning how
to make
phenotypically silent amino acid substitutions is provided in Bowie et al.,
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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
93
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 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.
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, 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, 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 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 or (v) fusion of the
polypeptide with
another compound, such as albumin (including, but not limited to, recombinant
albumin (see, e.g., U.S. Patent No. 5,876,969, issued March 2, 1999, EP Patent
0 413
622, and U.S. Patent No. 5,766,883, issued June 16, 1998, herein incorporated
by
reference in their entirety)). Such variant polypeptides are deemed to be
within the
scope of those skilled in the art from the teachings herein.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
94
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 (1987); Cleland et al., Crit.
Rev.
Therapeutic Drug Carrier Systems 10:307-377 (1993).)
A further embodiment of the invention relates to a polypeptide which
comprises the amino acid sequence of the present invention having an amino
acid
sequence which contains at least 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, in
order of ever-increasing preference, it is highly preferable for a peptide or
polypeptide
to have an amino acid sequence which comprises the amino acid sequence of the
present invention, which contains 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 the present
invention or
fragments thereof (e.g., the mature form and/or other fragments described
herein), is
1-5, 5-10, 5-25, 5-50, 10-50 or 50-150, conservative amino acid substitutions
are
preferable.
Polynucleotide and Polypeptide Fragments
The present invention is also directed to polynucleotide fragments of the
polynucleotides of the invention.
In the present invention, a "polynucleotide fragment" refers to a short
polynucleotide having a nucleic acid sequence which: is a portion of that
contained in
a deposited clone, or encoding the polypeptide encoded by the cDNA in a
deposited
clone; is a portion of that shown in SEQ ID NO:X or the complementary strand
thereto, or is a portion of a polynucleotide sequence encoding the polypeptide
of SEQ
ID NO:Y. 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,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
and even more preferably, at least about 40 nt, at least about 50 nt, at least
about 75
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 the cDNA
sequence contained in a deposited clone or the nucleotide sequence shown in
SEQ ID
5 NO:X. In this context "about" includes the particularly recited value, a
value larger
or smaller by several (5, 4, 3, 2, or 1) nucleotides, at either terminus or at
both
termini. 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.,
50, 150, 500, 600, 2000 nucleotides) are preferred.
10 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,
15 1151-1200, 1201-1250, 1251-1300, 1301-1350, 1351-1400, 1401-1450, 1451-
1500,
1501-1550, 1551-1600, 1601-1650, 1651-1700, 1701-1750, 1751-1800, 1801-1850,
1851-1900, 1901-1950, 1951-2000, or 2001 to the end of SEQ ID NO:X, or the
complementary strand thereto, or the cDNA contained in a deposited clone. In
this
context "about" includes the particularly recited ranges, and ranges larger or
smaller
20 by several (5, 4, 3, 2, or 1 ) nucleotides, at either terminus or at both
termini.
Preferably, these fragments encode a polypeptide which has biological
activity. More
preferably, these polynucleotides can be used as probes or primers as
discussed
herein. Polynucleotides which hybridize to these nucleic acid molecules under
stringent hybridization conditions or lower stringency conditions are also
25 encompassed by the invention, as are polypeptides encoded by these
polynucleotides.
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 or encoded by the
cDNA contained in a deposited clone. Protein (polypeptide) fragments may be
"free-
standing," or comprised within a larger polypeptide of which the fragment
forms a
30 part or region, most preferably as a single continuous region.
Representative
examples of polypeptide fragments of the invention, include, for example,
fragments
comprising, or alternatively consisting of, from about amino acid number 1-20,
21-40,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
96
41-60, 61-80, 81-100, 102-120, 121-140, 141-160, or 161 to the end of the
coding
region. Moreover, polypeptide fragments can be about 20, 30, 40, 50, 60, 70,
80, 90,
100, 110, 120, 130, 140, or 150 amino acids in length. In this context "about"
includes the particularly recited ranges or values, and ranges or values
larger or
smaller by several (5, 4, 3, 2, or 1) amino acids, at either extreme or at
both extremes.
Polynucleotides encoding these polypeptides are also encompassed by the
invention.
Preferred polypeptide fragments 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
1 S preferred. Similarly, polynucleotides encoding these polypeptide fragments
are also
preferred.
Also preferred are polypeptide and polynucleotide fragments characterized by
structural or functional domains, such as fragments that comprise alpha-helix
and
alpha-helix forming regions, beta-sheet and beta-sheet-forming regions, turn
and turn-
forming regions, coil and coil-forming regions, hydrophilic regions,
hydrophobic
regions, alpha amphipathic regions, beta amphipathic regions, flexible
regions,
surface-forming regions, substrate binding region, and high antigenic index
regions.
Polypeptide fragments of SEQ ID NO:Y falling within conserved domains are
specifically contemplated by the present invention. Moreover, polynucleotides
encoding these domains are also contemplated.
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. Polynucleotides encoding these polypeptide fragments are
also
encompassed by the invention.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
97
Preferably, the polynucleotide fragments of the invention encode a
polypeptide which demonstrates a functional activity. 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)
polypeptide of invention protein. Such functional activities include, but are
not
limited to, biological activity, antigenicity [ability to bind (or compete
with a
polypeptide of the invention for binding) to an antibody to the polypeptide of
the
invention], immunogenicity (ability to generate antibody which binds to a
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 of the invention.
The functional activity of polypeptides of the invention, 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 invention for binding to an
antibody of
the polypeptide of the invention, 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 for a polypeptide of the invention
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
98
chromatography, protein affinity chromatography, and affinity blotting. See
generally, Phizicky, E., et al., 1995, Microbiol. Rev. 59:94-123. In another
embodiment, physiological correlates of binding of a polypeptide of the
invention to
its substrates (signal transduction) can be assayed.
In addition, assays described herein (see Examples) and otherwise known in
the art may routinely be applied to measure the ability of polypeptides of the
invention and fragments, variants derivatives and analogs thereof to elicit
related
biological activity related to that of the polypeptide of the invention
(either in vitro or
in vivo). Other methods will be known to the skilled artisan and are within
the scope
of the invention.
Epitopes and Antibodies
The present invention encompasses polypeptides comprising, or alternatively
consisting of, an epitope of the polypeptide having an amino acid sequence of
SEQ ID
NO:Y, or an epitope of the polypeptide sequence encoded by a polynucleotide
sequence contained in ATCC deposit No. Z or encoded by a polynucleotide that
hybridizes to the complement of the sequence of SEQ ID NO:X or contained in
ATCC deposit No. Z under stringent hybridization conditions or lower
stringency
hybridization conditions 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 the complementary strand under stringent hybridization conditions or lower
stringency hybridization conditions 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
99
described infra. (See, for example, Geysen et al., Proc. Natl. Acad. Sci. USA
81:3998- 4002 (1983)). The term "antigenic epitope," as used 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, 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 1 l, 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 carrier protein, such as an
albumin, to


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
100
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 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 Garner, 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 ~g 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 comprising an immunogenic or antigenic
epitope can be fused to other polypeptide sequences. For example, the
polypeptides
30. 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), or albumin (including but not limited to
recombinant


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
101
albumin (see, e.g., U.S. Patent No. 5,876,969, issued March 2, 1999, EP Patent
0 413
622, and U.S. Patent No. 5,766,883, issued June 16, 1998, herein incorporated
by
reference in their entirety)), 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 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). 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.,
1991,
Proc. Natl. Acad. Sci. USA 88:8972- 897). In 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 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 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;


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
102
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, 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
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 components, motifs, sections,
parts,
1 S domains, fragments, etc. of one or more heterologous molecules.
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 ID 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., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2) or
subclass
of immunoglobulin molecule. In preferred embodiments, the immunoglobulin


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
103
molecules of the invention are IgGI. In other preferred embodiments, the
immunoglobulin molecules of the invention are IgG4.
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
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 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,
by size in
contiguous amino acid residues, or listed in the Tables and Figures.
Antibodies which
specifically bind any epitope or polypeptide of the present invention may also
be


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
104
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 55%, and less than 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 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-Z M, 10-Z M, 5 X 10-3 M, 10-3 M, 5 X 10-4 M,
10-4 M,
5 X 10-5 M, 10-5 M, 5 X 10-6 M, 10-6M, 5 X 10-~ M, 10' M, 5 X 10-g M, 10-g M,
5 X
10-9 M, 10-9 M, 5 X 10~~° M, 10-x° M, 5 X 10-11 M, 10-~~ M, 5 X
10-~Z M,'°-12 M, 5 X
10-'3 M, 10-13 M, 5 X 10-~4 M, 10-4 M, 5 X 10-5 M, or 10-~5 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
105
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
1 S 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.
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 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 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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
106
5,811,097; Deng et al., Blood 92(6):1981-1988 (1998); Chen et al., Cancer Res.
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 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 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 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 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 tv a cellular ligand or other protein, etc. Any
of


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
107
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 "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 (e.g., Example 16). In a non-limiting example, mice can be immunized
with a polypeptide of the invention or a cell expressing such peptide. Once an


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
108
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
CHl 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 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 M 13 binding domains expressed from phage with Fab, Fv or
disulfide stabilized Fv antibody domains recombinantly fused to either the
phage


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
109
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, Science 229:1202
(1985); Oi et al., BioTechniques 4:214 (1986); Gillies et al., (1989) J.
Immunol.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
110
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 immunoglobulin 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
111
mouse embryonic stem cells in addition to the human 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 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 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,
1 S 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
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 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 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
112
techniques 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 polypeptide of the invention to a ligand
can be
S 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.
Polynucleotides 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
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
113
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
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
114
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 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.5. 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
115
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, 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
116
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 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 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. 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, such


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
117
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 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. 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 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
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 Enzymol. 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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
118
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.
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,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
119
which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl.
Acad.
Sci. USA 78:2072 (1981)); neo, which confers resistance to the aminoglycoside
6-
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 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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
120
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, 50, 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 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
121
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 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
122
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 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 "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 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
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 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 luciferase,
luciferin,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
123
and aequorin; and examples of suitable radioactive material include 125I,
131I, 111In
or 99Tc.
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
S 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 (CCNU),
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, 13-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"),


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
124
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 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 Garners 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,
1 S 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
125
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)).
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. 1, 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
126
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 modif ed 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
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. l, 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
127
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 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 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.
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, 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, including, but not limited
to, any


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
128
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 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.
1 S 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.
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,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
129
including fragments thereof. Preferred binding affinities include those with a
dissociation constant or Kd less than 5 X 10-2 M, 10-2 M, 5 X 10-3 M, 10-3 M,
5 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-~ M, 10-~ M, S 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-' Z M,
10-' 2 M, 5 X 10-' 3 M, 10-' 3 M, S 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.
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):155-215 (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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
130
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
S 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 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
131
(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 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
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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
132
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 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
carried 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.
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 Tlymphocytes, Blymphocytes, monocytes, macrophages,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
133
neutrophils, eosinophils, 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
S patient.
In an embodiment in which recombinant cells are used in gene therapy,
nucleic acid 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 94/08598; Stemple and
Anderson, Cell 71:973-985 (1992); Rheinwald, Meth. Cell Bio. 21A: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 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 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 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 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
134
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 an antibody of the invention. In a
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;


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
135
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 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; SeBon, 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
136
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 (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
1 S 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 carriers are described in "Remington's Pharmaceutical Sciences"
by
E.W. Martin. Such compositions will contain a therapeutically effective amount
of


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
137
the compound, preferably in purified form, together with a suitable amount of
Garner
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 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 cations 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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
138
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.
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 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 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 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) 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,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
139
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 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.
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. 105:3087-3096 (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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
140
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 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 carried out by
repeating 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
141
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, 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
andlor 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
142
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 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
143
recombinant antigens, and a reporter-labeled anti-human antibody for detecting
surface-bound anti-antigen antibody.
Fusion Proteins
Any polypeptide of the present invention can be used to generate fusion
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, the polypeptides of the
present
invention 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.
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.
Moreover, polypeptides of the present invention, including fragments, and
specifically epitopes, can be combined with parts of the constant domain of
immunoglobulins (IgA, IgE, IgG, IgM) or portions thereof (CH1, CH2, CH3, and
any
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).)


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
144
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
secreted protein or protein fragment alone. (Fountoulakis et al., J. Biochem.
270:3958-3964 (1995).) Polynucleotides comprising or alternatively consisting
of
nucleic acids which encode these fusion proteins are also encompassed by the
invention.
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,
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 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.
Vectors, Host Cells, and Protein Production


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
145
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 may be joined to a vector containing a selectable marker
for propagation in a host. Generally, a plasmid vector is introduced in a
precipitate,
such as a calcium phosphate precipitate, or in a complex with a charged lipid.
If the
vector is a virus, it 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
1 S 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 S~ 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,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
146
pNHl6a, pNHlBA, 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
S 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.
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 (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 canon 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, and preferably the secreted form, can
also be recovered from: products purified from natural sources, including
bodily
fluids, tissues and cells, whether directly isolated or 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-


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
147
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 the
polypeptide of the present 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, Pichia pastoris must
generate
high levels of alcohol oxidase due, in part, to the relatively low affinity of
alcohol
oxidase for O2. 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
AOXI
regulatory sequence 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 secretion of a protein 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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
148
Many other yeast vectors could be used in place of pPIC9K, such as, pYES2,
pYDI, pTEFl/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalpha, pPIC9, pPIC3.5,
pHIL-D2, pHIL-S1, 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 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 replace endogenous genetic material (e.g., coding
sequence), and/or to include genetic material (e.g., heterologous
polynucleotide
sequences) that is operably associated with the 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, resulting in the formation of a new
transcription unit
(see, e.g., U.S. Patent No. 5,641,670, issued June 24, 1997; U.S. Patent No.
5,733,761, issued March 31, 1998; 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
149
polypeptide sequence of the invention 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 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 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
150
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). For example, the polyethylene glycol may have
an
average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000,
3500,
4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500,
10,000,
10,500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500,
15,000,
15,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500,
20,000,
25,000, 30,000, 35,000, 40,000, 50,000, 55,000, 60,000, 65,000, 70,000,
75,000,
80,000, 85,000, 90,000, 95,000, or 100,000 kDa.
As noted above, the polyethylene glycol may have a branched structure.
Branched polyethylene glycols are described, for example, in U.S. Patent No.
5,643,575; Morpurgo et al., Appl. Biochem. Biotechnol. 56:59-72 (1996);
Vorobjev et
al., Nucleosides Nucleotides 18:2745-2750 (1999); and Caliceti et al.,
Bioconjug.
Chem. 10:638-646 (1999), the disclosures of each of which are incorporated
herein by
reference.
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
151
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 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.
As suggested above, polyethylene glycol may be attached to proteins via
linkage to any of a number of amino acid residues. For example, polyethylene
glycol
can be linked to a proteins via covalent bonds to lysine, histidine, aspartic
acid,
glutamic acid, or cysteine residues. One or more reaction chemistries may be
employed to attach polyethylene glycol to specific amino acid residues (e.g.,
lysine,
histidine, aspartic acid, glutamic acid, or cysteine) of the protein or to
more than one
type of amino acid residue (e.g., lysine, histidine, aspartic acid, glutamic
acid,
cysteine and combinations thereof) of the protein.
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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
152
As indicated above, pegylation of the proteins of the invention may be
accomplished by any number of means. For example, polyethylene glycol may be
attached to the protein either directly or by an intervening linker.
Linkerless systems
for attaching polyethylene glycol to proteins are described in Delgado et al.,
Crit. Rev.
Thera. Drug Carrier Sys. 9:249-304 (1992); Francis et al., Intern. .l.
ofHematol.
68:1-18 (1998); U.S. Patent No. 4,002,531; U.S. Patent No. 5,349,052; WO
95/06058;
and WO 98/32466, the disclosures of each of which are incorporated herein by
reference.
One system for attaching polyethylene glycol directly to amino acid residues
of proteins without an intervening linker employs tresylated MPEG, which is
produced by the modification of monmethoxy polyethylene glycol (MPEG) using
tresylchloride (C1SOZCHZCF3). Upon reaction of protein with tresylated MPEG,
polyethylene glycol is directly attached to amine groups of the protein. Thus,
the
invention includes protein-polyethylene glycol conjugates produced by reacting
proteins of the invention with a polyethylene glycol molecule having a
2,2,2-trifluoreothane sulphonyl group.
Polyethylene glycol can also be attached to proteins using a number of
different intervening linkers. For example, U.S. Patent No. 5,612,460, the
entire
disclosure of which is incorporated herein by reference, discloses urethane
linkers for
connecting polyethylene glycol to proteins. Protein-polyethylene glycol
conjugates
wherein the polyethylene glycol is attached to the protein by a linker can
also be
produced by reaction of proteins with compounds such as MPEG-
succinimidylsuccinate, MPEG activated with 1,1'-carbonyldiimidazole, MPEG-
2,4,5-trichloropenylcarbonate, MPEG-p-nitrophenolcarbonate, and various MPEG-
succinate derivatives. A number additional polyethylene glycol derivatives and
reaction chemistries for attaching polyethylene glycol to proteins are
described in
WO 98/32466, the entire disclosure of which is incorporated herein by
reference.
Pegylated protein products produced using the reaction chemistries set out
herein are
included within the scope of the invention.
The number of polyethylene glycol moieties attached to each protein of the
invention (i.e., the degree of substitution) may also vary. For example, the
pegylated
proteins of the invention may be linked, on average, to 1, 2, 3, 4, 5, 6, 7,
8, 9, 10, 12,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
153
15, 17, 20, or more polyethylene glycol molecules. Similarly, the average
degree of
substitution within ranges such as 1-3, 2-4, 3-5, 4-6, 5-7, 6-8, 7-9, 8-10, 9-
11, 10-12,
11-13, 12-14, 13-15, 14-16, 15-17, 16-18, 17-19, or 18-20 polyethylene glycol
moieties per protein molecule. Methods for determining the degree of
substitution are
discussed, for example, in Delgado et al., Crit. Rev. Thera. Drug Carrier Sys.
9:249-
304 (1992).
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 ID NO:Y or encoded by the cDNA
contained in a deposited clone (including fragments, variants, splice
variants, and
fusion proteins, corresponding to these polypeptides 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 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
154
embodiments, the heteromeric 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 the
sequence
listing, or contained in the polypeptide encoded by a deposited clone). 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
occurring) polypeptide. In another instance, the covalent associations are the
consequence of chemical or recombinant manipulation. Alternatively, such
covalent
associations may involve one or more amino acid residues contained in the
heterologous polypeptide sequence in a fusion protein of the invention.
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 sequence contained in an 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, oseteoprotegerin (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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
155
(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
S 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 derived from naturally occurring 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
156
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
S 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
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 incorporated by membrane reconstitution techniques
into


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
157
liposomes (see, e.g., US Patent Number 5,478,925, which is herein incorporated
by
reference in its entirety).
Uses of the Polynucleotides
S 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 polynucleotide of the present
invention can be used as a chromosome marker.
Briefly, sequences can be mapped to chromosomes by preparing PCR primers
(preferably 15-25 bp) from the sequences shown in SEQ ID NO:X. Primers can 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
hybrids
containing the human gene corresponding to the 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 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., Shiner,
Trends
Biotechnol 16:456-459 (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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
158
Verma et al., "Human 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).
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 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); Herrick 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.
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 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 SO-500 potential causative genes.
Thus, once coinheritance is established, differences in the polynucleotide 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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
159
Furthermore, increased or decreased expression of the gene in affected
individuals as compared to unaffected individuals can be assessed using
polynucleotides of the present 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 present invention and a suitable container. In a
specific
embodiment, the kit includes two polynucleotide probes defining an internal
region of
the polynucleotide of the present 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 polymerise chain reaction amplification.
Where a diagnosis of a disorder, 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
present
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 polynucleotide of the present
invention" is intended qualitatively or quantitatively measuring or estimating
the level
of the polypeptide of the present invention or the level of the mRNA encoding
the
polypeptide 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
160
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 disorder or being determined by averaging levels from a population of
individuals
not having a 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 the
polypeptide of the present invention or 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
other 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 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 present invention
attached may
be used to identify polymorphisms between the polynucleotide sequences, with
polynucleotides isolated from a test subject. The knowledge of such
polymorphisms
(i.e. their location, as well as, their existence) would be beneficial in
identifying
disease loci for many disorders, including 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 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,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
161
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. Egholm, O. Buchardt,
S 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 DNAlDNA 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 1 S-mer
lowers the melting point (Tm) by 8°-20° C, vs. 4°-
16° C for the DNA/DNA 1 S-
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 is useful for 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 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 diseases, disorders, and/or conditions 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
162
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 would
not be
limited to treatment of proliferative diseases, disorders, and/or conditions
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 can be used to control gene
expression through triple helix formation or antisense DNA or RNA. Antisense
techniques are discussed, for example, in Okano, J. Neurochem. 56: 560 (1991);
"Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression,CRCPress,
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
163
(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. Both 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 or prevent disease.
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.
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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
164
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 specific to particular tissue 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.
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.
A polypeptide of the present invention can be used to assay protein levels in
a
biological sample using antibody-based techniques. For example, protein
expression


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
165
in tissues can be studied with classical immunohistological methods.
(Jalkanen, M.,
et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, M., 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, and
radioisotopes, such
as iodine (125I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium
(112In), and
technetium (99mTc), and fluorescent labels, such as fluorescein and rhodamine,
and
biotin.
In addition to assaying secreted protein levels 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,
131I,
1 l2In, 99mTc), a radio-opaque substance, or a material detectable by nuclear
magnetic resonance, is introduced (for example, parenterally, subcutaneously,
or
intraperitoneally) into the mammal. 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).)


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
166
Thus, the invention provides a diagnostic method of a disorder, which
involves (a) assaying the expression of a polypeptide of the present invention
in cells
or body fluid of an individual; (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 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, prevent,
and/or diagnose disease. 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, prevent, and/or diagnose disease. For example,
administration of
an antibody directed to a polypeptide of the present invention can bind and
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
167
be used to raise antibodies, which in turn are used to measure protein
expression from
a recombinant cell, as a way of assessing transformation of the host cell.
Moreover,
the polypeptides of the present invention can be used to test the following
biological
activities.
S
Gene Therapy Methods
Another aspect of the present invention is to gene therapy methods for
treatingor 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 a polypeptide of the present
invention. This method requires a polynucleotide which codes for a polypeptide
of the
invention that 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 invention ex vivo, with the engineered cells then being
provided
to a patient to be treated with the polypeptide. Such methods are well-known
in the
art. For example, see Belldegrun et al., J. Natl. Cancer Inst., 85:207-216
(1993);
Ferrantini et al., Cancer Research, 53:107-1112 (1993); Ferrantini et al., J.
Immunology 153: 4604-4615 (1994); Kaido, T., et al., Int. J. Cancer 60: 221-
229
(1995); Ogura et al., Cancer Research 50: 5102-5106 (1990); Santodonato, et
al.,
Human Gene Therapy 7:1-10 (1996); Santodonato, et al., Gene Therapy 4:1246-
1255
(1997); and Zhang, 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,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
168
and the like). The polynucleotide constructs may be delivered in a
pharmaceutically
acceptable liquid or aqueous Garner.
In one embodiment, the polynucleotide of the invention is delivered as a naked
polynucleotide. 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 polynucleotides
of the
invention can also be delivered in liposome formulations and lipofectin
formulations
and the like can be 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 of the invention 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, pXTI and pSG available from Stratagene;
pSVK3, pBPV, pMSG and pSVL available from Pharmacia; and pEFl/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 expression of polynucleotide sequence of the invention. 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-
actin
promoter; and human growth hormone promoters. The promoter also may be the
native promoter for the polynucleotides of the 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
169
sequences can be introduced into cells to provide production of the desired
polypeptide for periods of up to six months.
The polynucleotide construct of the invention 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 nakednucleic 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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
170
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 of the invention 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 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 ,
84:7413-7416 (1987), which is herein incorporated by reference); mRNA (Malone
et
al., Proc. Natl. Acad. Sci. USA , 86:6077-6081 (1989), which is herein
incorporated
by reference); and purified transcription factors (Debs et al., J. Biol.
Chem.,
265:10189-10192 (1990), 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
,
84:7413-7416 (1987), 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., Felgner et al.,
Proc.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
171
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
1 S 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 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 , 101:512-527
(1983),
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
172
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 prefonned 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,
394:483 (1975); Wilson et al., Cell , 17:77 (1979)); ether injection (Deamer
et al.,
Biochim. Biophys. Acta, 443:629 ( 1976); Ostro et al., Biochem. Biophys. Res.
Commun., 76:836 (1977); Fraley et al., Proc. Natl. Acad. Sci. USA, 76:3348
(1979));
detergent dialysis (Enoch et al., Proc. Natl. Acad. Sci. USA , 76:145 (1979));
and
reverse-phase evaporation (REV) (Fraley et al., J. Biol. Chem., 255:10431
(1980);
Szoka et al., Proc. Natl. Acad. Sci. USA , 75:145 (1978); Schaefer-Ridder et
al.,
Science, 215:166 (1982)), 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.
U.5. 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.5. 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.5. 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
polypeptides of the 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
173
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 polypeptides of the 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 polypeptides of the
invention.
In certain other embodiments, cells are engineered, ex vivo or in vivo, with
polynucleotides of the invention contained in an adenovirus vector. Adenovirus
can
be manipulated such that it encodes and expresses polypeptides of the
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, thereby alleviating concerns about insertional
mutagenesis. Furthermore, adenoviruses have been used as live enteric vaccines
for
many years with an excellent safety profile (Schwartzet al., Am. Rev. Respir.
Dis.,
109:233-238 (1974)). 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 et al.,Science , 252:431-434
(1991);
Rosenfeld et al., Cell, 68:143-155 (1992)). Furthermore, extensive studies to
attempt
to establish adenovirus as a causative agent in human cancer were uniformly
negative
(Green et al. Proc. Natl. Acad. Sci. USA , 76:6606 (1979)).
Suitable adenoviral vectors useful in the present invention are described, for
example, in Kozarsky and Wilson, Curr. Opin. Genet. Devel., 3:499-503 (1993);


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
174
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
S 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 LS.
In certain other embodiments, the cells are engineered, ex vivo or in vivo,
using an adeno-associated virus (AAV). AAVs are naturally occurring defective
viruses that require helper viruses to produce infectious particles (Muzyczka,
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.
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 containing polynucleotides of the
invention
is inserted into the AAV vector using standard cloning methods, such as those
found
in Sambrook et al., Molecular Cloning: A Laboratory Manual, 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
175
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 of the
invention.
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 the desired gene product.
Another method of gene therapy involves operably associating heterologous
control regions and endogenous polynucleotide sequences (e.g. encoding the
polypeptide sequence of interest) via homologous recombination (see, e.g.,
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
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 5' end
of the
second targeting sequence contains the same restriction site as the 3' end of
the
amplified promoter. The amplified promoter and targeting sequences are
digested
and ligated together.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
176
The promoter-targeting sequence construct is delivered to the cells, either as
naked polynucleotide, or in conjunction with transfection-facilitating agents,
such as
liposomes, 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.
The polynucleotides encoding polypeptides of the present invention may be
administered along with other polynucleotides encoding other angiongenic
proteins.
Angiogenic proteins include, but are not limited to, acidic and basic
fibroblast growth
factors, VEGF-1, VEGF-2 (VEGF-C), VEGF-3 (VEGF-B), 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.
Preferably, the polynucleotide encoding a polypeptide of the 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
177
(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 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 carrier capable of withstanding degradation by digestive
enzymes in the
gut of an animal. Examples of such carriers, 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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
178
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
Biological Activities
The 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 and polypeptides 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 or polypeptides, or agonists or
antagonists could
be used to treat the associated disease.
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 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
179
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
scarring.
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
180
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
181
not limited to, 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., 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,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
182
prevented, and/or diagnosed using for example, antagonists or agonists,
polypeptides
or polynucleotides, or antibodies of the present invention.
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
(SC>D)-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-
lymphocytopenia,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
183
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.
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 SC>D, 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
184
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, ischemic brain injury
and/or
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, 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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
185
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.).
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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
186
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 adjuvant 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 o~ A)DS, 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 immune response to a
virus,
disease, or symptom selected from the group consisting o~ HN/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.
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: Yibrio cholerae, Mycobacterium
leprae, Salmonella typhi, Salmonella paratyphi, Meisseria meningitidis,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
187
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
S 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.
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
188
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.
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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
189
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, or ribozymes. 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.
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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
190
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).
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 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.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
191
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 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),


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
192
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 and/or
conditions associated with the diseases and disorders recited above are
treated,
prognosed, prevented, and/or diagnosed using antibodies against the
polypeptide of
1 S the invention.
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, and/or
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 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
193
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,
S 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, lymphangioendotheliosarcoma, 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.
Diseases associated with increased apoptosis that could be treated or detected
by polynucleotides, polypeptides, and/or antagonists of the invention, include
A>DS;
neurodegenerative disorders (such as Alzheimer's disease, Parkinson's disease,
Amyotrophic lateral sclerosis, Retinitis pigmentosa, Cerebellar degeneration
and


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
194
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
A polynucleotides or polypeptides, or agonists or antagonists of the invention
can be used to treat, prevent, and/or diagnose hyperproliferative diseases,
disorders,
and/or conditions, including neoplasms. A polynucleotides or polypeptides, or
agonists or antagonists of the present invention may inhibit the proliferation
of the
disorder through direct or indirect interactions. Alternatively, a
polynucleotides or
polypeptides, or agonists or antagonists of the present invention may
proliferate other
cells which can inhibit the hyperproliferative disorder.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
195
For example, by increasing an immune response, particularly increasing
antigenic qualities of the hyperproliferative disorder or by proliferating,
differentiating, or mobilizing T-cells, hyperproliferative diseases,
disorders, and/or
conditions can be treated, prevented, and/or diagnosed. 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, preventing, and/or diagnosing hyperproliferative diseases,
disorders, and/or conditions, such as a chemotherapeutic agent.
Examples of hyperproliferative diseases, disorders, and/or conditions that can
be treated, prevented, and/or diagnosed 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 diseases, disorders, and/or conditions can
also be treated, prevented, and/or diagnosed by a polynucleotides or
polypeptides, or
agonists or antagonists of the present invention. Examples of such
hyperproliferative
diseases, disorders, andlor conditions include, but are not limited to:
hypergammaglobulinemia, lymphoproliferative diseases, disorders, and/or
conditions,
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 or preventing cell
proliferative diseases, disorders, and/or conditions 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 or
preventing cell-proliferative diseases, disorders, and/or conditions in
individuals


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
196
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
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
1 S 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.
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 delivered by known gene delivery systems such as, but
not
limited to, retroviral vectors (Gilboa, J. Virology 44:845 (1982); Hocke,
Nature


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
197
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 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 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
198
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, preventing, and/or
diagnosing one
or more of the described diseases, disorders, and/or conditions. 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, preventing, and/or diagnosing a subject having or
developing
cell proliferative and/or differentiation diseases, disorders, and/or
conditions 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 diseases, disorders, and/or conditions 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
199
with a dissociation constant or Kd less than 5X10-6M, 10-6M, 5X10-~M, 10-~M,
SX10-
gM, 10~8M, SX10-9M, 10-9M, 5X10-'°M, 10-'°M, 5X10-"M, 10-"M,
5X10-'ZM, 10-
'ZM, 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
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).


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
200
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
S 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.
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 invention
may be used to treat, prevent, and/or diagnose cardiovascular diseases,
disorders,
and/or conditions, including peripheral artery disease, such as limb ischemia.
Cardiovascular diseases, disorders, and/or conditions 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
201
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
S Syndrome, trilogy of Fallot, ventricular heart septal defects.
Cardiovascular diseases, disorders, and/or conditions 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 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, mural valve stenosis, pulmonary atresia, pulmonary
valve
insufficiency, pulmonary valve stenosis, tricuspid atresia, tricuspid valve
insufficiency, and tricuspid valve stenosis.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
202
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
diseases, disorders,
and/or conditions, diabetic angiopathies, diabetic retinopathy, embolisms,
thrombosis,
erythromelalgia, hemorrhoids, hepatic veno-occlusive disease, hypertension,
hypotension, ischemia, peripheral vascular diseases, phlebitis, 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 diseases, disorders, and/or conditions 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
203
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 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 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 of the invention may be administered as part of a
Therapeutic,
described in more detail below. Methods of delivering polynucleotides of the
invention are described in more detail herein.
Anti-An~io~enesis Activity
The naturally occurring 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
204
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
diseases,
disorders, and/or conditions, 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, disorders, and/or
conditions 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
disorders, see
Fishman et al., Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia
(1985)).Thus, the
present invention provides a method of treating, preventing, and/or diagnosing
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 treator prevent a cancer or
tumor.
Cancers which may be treated, prevented, and/or diagnosed with
polynucleotides,
polypeptides, antagonists and/or agonists include, but are not limited to
solid tumors,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
205
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 or prevent cancers such as skin cancer, head and neck
tumors, breast
tumors, 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, preventing, andlor diagnosing other diseases, disorders, andlor
conditions,
besides cancers, which involve angiogenesis. These diseases, disorders, and/or
conditions 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 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, preventing, and/or diagnosing hypertrophic scars and keloids,
comprising


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
206
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, preventing,
and/or
diagnosing neovascular diseases of the eye, including for example, corneal
neovascularization, neovascular glaucoma, proliferative diabetic retinopathy,
retrolental fibroplasia and macular degeneration.
Moreover, Ocular diseases, disorders, and/or conditions associated with
neovascularization which can be treated, prevented, and/or diagnosed 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., SuYV. Ophthal. 22:291-312 (1978).
Thus, within one aspect of the present invention methods are provided for
treating or preventing 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 become complete if the cornea completely opacitates. A wide variety of


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
207
diseases, disorders, and/or conditions 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),
S 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-


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
208
3 times per year. A steroid could also be added to the injection solution to
reduce
inflammation resulting from the injection itself.
Within another aspect of the present invention, methods are provided for
treating or preventing 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 or prevent 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 or preventing 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 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 or preventing 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 and/or via
intraocular implants.
Additionally, diseases, disorders, and/or conditions which can be treated,
prevented, and/or diagnosed 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,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
209
pyogenic granuloma, scleroderma, trachoma, and vascular adhesions.
Moreover, diseases, disorders, and/or conditions and/or states, which can be
treated, prevented, and/or diagnosed 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
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 after" 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
210
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.
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
211
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 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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
212
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;
S 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.
Diseases at the Cellular Level
Diseases associated with increased cell survival or the inhibition of
apoptosis
that could be treated, prevented, and/or diagnosed by the polynucleotides or
polypeptides and/or antagonists or agonists 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 cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma,
osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast
cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer); autoimmune
diseases,
disorders, and/or conditions (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, the polynucleotides or
polypeptides, and/or


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
213
agonists 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, prevented or diagnosed by the polynucleotides or
polypeptides, or
S agonists 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, lymphangioendotheliosarcoma, 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.
Diseases associated with increased apoptosis that could be treated, prevented,
and/or diagnosed by the polynucleotides or polypeptides, and/or agonists or
antagonists of the invention, include AIDS; neurodegenerative diseases,
disorders,
and/or conditions (such as Alzheimer's disease, Parkinson's disease,
Amyotrophic
lateral sclerosis, Retinitis pigmentosa, Cerebellar degeneration and brain
tumor or
prior associated disease); autoimmune diseases, disorders, and/or conditions
(such as,
multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary
cirrhosis,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
214
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 the polynucleotides or polypeptides, and/or
agonists
or antagonists of the 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
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
antimetabolites. Polynucleotides or polypeptides, and/or agonists or
antagonists of
the invention, could be used to promote dermal reestablishment subsequent to
dermal
loss
The polynucleotides or polypeptides, and/or agonists or antagonists of the
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 a non-
exhaustive list of grafts that polynucleotides or polypeptides, agonists or
antagonists
of the 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,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
215
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. The polynucleotides or polypeptides, and/or agonists
or
S antagonists of the invention, can be used to promote skin strength and to
improve the
appearance of aged skin.
It is believed that the polynucleotides or polypeptides, and/or agonists or
antagonists of the invention, will also produce changes in hepatocyte
proliferation,
and epithelial cell proliferation in the lung, breast, pancreas, stomach,
small intesting,
and large intestine. The polynucleotides or polypeptides, and/or agonists or
antagonists of the 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. The polynucleotides or polypeptides, and/or
agonists
or antagonists of the invention, may promote proliferation of endothelial
cells,
keratinocytes, and basal keratinocytes.
The polynucleotides or polypeptides, andlor agonists or antagonists of the
invention, could also be used to reduce the side effects of gut toxicity that
result from
radiation, chemotherapy treatments or viral infections. The polynucleotides or
polypeptides, and/or agonists or antagonists of the invention, may have a
cytoprotective effect on the small intestine mucosa. The polynucleotides or
polypeptides, and/or agonists or antagonists of the invention, may also
stimulate
healing of mucositis (mouth ulcers) that result from chemotherapy and viral
infections.
The polynucleotides or polypeptides, and/or agonists or antagonists of the
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. The
polynucleotides or polypeptides, and/or agonists or antagonists of the
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. The polynucleotides or polypeptides,
and/or


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
216
agonists or antagonists of the 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, the polynucleotides or polypeptides, and/or
agonists or
antagonists of the 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 the polynucleotides or polypeptides, and/or agonists
or
antagonists of the 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.
The polynucleotides or polypeptides, and/or agonists or antagonists of the
invention,
could be used to treat diseases associate with the under expression of the
polynucleotides of the invention.
Moreover, the polynucleotides or polypeptides, and/or agonists or antagonists
of
the invention, could be used to prevent and heal damage to the lungs due to
various
pathological states. A growth factor such as the polynucleotides or
polypeptides,
andlor agonists or antagonists of the 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, prevented, and/or diagnosed using the
polynucleotides or polypeptides, and/or agonists or antagonists of the
invention.
Also, the polynucleotides or polypeptides, and/or agonists or antagonists of
the
invention, could be used to stimulate the proliferation of and differentiation
of type II
pneumocytes, which may help treat or prevent disease such as hyaline membrane
diseases, such as infant respiratory distress syndrome and bronchopulmonary
displasia, in premature infants.
The polynucleotides or polypeptides, and/or agonists or antagonists of the
invention, could stimulate the proliferation and differentiation of
hepatocytes and,


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
217
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, the polynucleotides or polypeptides, and/or agonists or
antagonists
of the 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, the polynucleotides or polypeptides, and/or agonists or antagonists
of the
invention, could be used to maintain the islet function so as to alleviate,
delay or
prevent permanent manifestation of the disease. Also, the polynucleotides or
polypeptides, and/or agonists or antagonists of the invention, could be used
as an
auxiliary in islet cell transplantation to improve or promote islet cell
function.
Neurological Diseases
Nervous system diseases, disorders, and/or conditions, which can be treated,
prevented, and/or diagnosed with the compositions of the invention (e.g.,
polypeptides, polynucleotides, and/or agonists or antagonists), include, but
are not
limited to, nervous system injuries, and diseases, disorders, and/or
conditions which
result in either a disconnection of axons, a diminution or degeneration of
neurons, or
demyelination. Nervous system lesions which may be treated, prevented, and/or
diagnosed in a patient (including human and non-human mammalian patients)
according to the invention, include but are not limited to, the following
lesions of
either the central (including spinal cord, brain) or peripheral nervous
systems: (1)
ischemic lesions, in which a lack of oxygen in a portion of the nervous system
results
in neuronal injury or death, including cerebral infarction or ischemia, or
spinal cord
infarction or ischemia; (2) traumatic lesions, including lesions caused by
physical
injury or associated with surgery, for example, lesions which sever a portion
of the
nervous system, or compression injuries; (3) malignant lesions, in which a
portion of
the nervous system is destroyed or injured by malignant tissue which is either
a
nervous system associated malignancy or a malignancy derived from non-nervous
system tissue; (4) infectious lesions, in which a portion of the nervous
system is


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
218
destroyed or injured as a result of infection, for example, by an abscess or
associated
with infection by human immunodeficiency virus, herpes zoster, or herpes
simplex
virus or with Lyme disease, tuberculosis, syphilis; (5) degenerative lesions,
in which
a portion of the nervous system is destroyed or injured as a result of a
degenerative
process including but not limited to degeneration associated with Parkinson's
disease,
Alzheimer's disease, Huntington's chorea, or amyotrophic lateral sclerosis
(ALS); (6)
lesions associated with nutritional diseases, disorders, and/or conditions, in
which a
portion of the nervous system is destroyed or injured by a nutritional
disorder or
disorder of metabolism including but not limited to, vitamin B 12 deficiency,
folic
acid deficiency, Wernicke disease, tobacco-alcohol amblyopia, Marchiafava-
Bignami
disease (primary degeneration of the corpus callosum), and alcoholic
cerebellar
degeneration; (7) neurological lesions associated with systemic diseases
including,
but not limited to, diabetes (diabetic neuropathy, Bell's palsy), systemic
lupus
erythematosus, carcinoma, or sarcoidosis; (8) lesions caused by toxic
substances
including alcohol, lead, or particular neurotoxins; and (9) demyelinated
lesions in
which a portion of the nervous system is destroyed or injured by a
demyelinating
disease including, but not limited to, multiple sclerosis, human
immunodeficiency
virus-associated myelopathy, transverse myelopathy or various etiologies,
progressive
multifocal leukoencephalopathy, and central pontine myelinolysis.
In a preferred embodiment, the polypeptides, polynucleotides, or agonists or
antagonists of the invention are used to protect neural cells from the
damaging effects
of cerebral hypoxia. According to this embodiment, the compositions of the
invention are used to treat, prevent, and/or diagnose neural cell injury
associated with
cerebral hypoxia. In one aspect of this embodiment, the polypeptides,
polynucleotides, or agonists or antagonists of the invention are used to
treat, prevent,
and/or diagnose neural cell injury associated with cerebral ischemia. In
another
aspect of this embodiment, the polypeptides, polynucleotides, or agonists or
antagonists of the invention are used to treat, prevent, and/or diagnose
neural cell
injury associated with cerebral infarction. In another aspect of this
embodiment, the
polypeptides, polynucleotides, or agonists or antagonists of the invention are
used to
treat, prevent, and/or diagnose or prevent neural cell injury associated with
a stroke.
In a further aspect of this embodiment, the polypeptides, polynucleotides, or
agonists


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
219
or antagonists of the invention are used to treat, prevent, and/or diagnose
neural cell
injury associated with a heart attack.
The compositions of the invention which are useful for treating or preventing
a nervous system disorder may be selected by testing for biological activity
in
promoting the survival or differentiation of neurons. For example, and not by
way of
limitation, compositions of the invention which elicit any of the following
effects may
be useful according to the invention: (1) increased survival time of neurons
in culture;
(2) increased sprouting of neurons in culture or in vivo; (3) increased
production of a
neuron-associated molecule in culture or in vivo, e.g., choline
acetyltransferase or
acetylcholinesterase with respect to motor neurons; or (4) decreased symptoms
of
neuron dysfunction in vivo. Such effects may be measured by any method known
in
the art. In preferred, non-limiting embodiments, increased survival of neurons
may
routinely be measured using a method set forth herein or otherwise known in
the art,
such as, for example, the method set forth in Arakawa et al. (J. Neurosci.
10:3507-3515 (1990)); increased sprouting of neurons may be detected by
methods
known in the art, such as, for example, the methods set forth in Pestronk et
al. (Exp.
Neurol. 70:65-82 (1980)) or Brown et al. (Ann. Rev. Neurosci. 4:17-42 (1981));
increased production of neuron-associated molecules may be measured by
bioassay,
enzymatic assay, antibody binding, Northern blot assay, etc., using techniques
known
in the art and depending on the molecule to be measured; and motor neuron
dysfunction may be measured by assessing the physical manifestation of motor
neuron disorder, e.g., weakness, motor neuron conduction velocity, or
functional
disability.
In specific embodiments, motor neuron diseases, disorders, and/or conditions
that may be treated, prevented, andlor diagnosed according to the invention
include,
but are not limited to, diseases, disorders, and/or conditions such as
infarction,
infection, exposure to toxin, trauma, surgical damage, degenerative disease or
malignancy that may affect motor neurons as well as other components of the
nervous
system, as well as diseases, disorders, and/or conditions that selectively
affect neurons
such as amyotrophic lateral sclerosis, and including, but not limited to,
progressive
spinal muscular atrophy, progressive bulbar palsy, primary lateral sclerosis,
infantile
and juvenile muscular atrophy, progressive bulbar paralysis of childhood
(Fazio-


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
220
Londe syndrome), poliomyelitis and the post polio syndrome, and Hereditary
Motorsensory Neuropathy (Charcot-Marie-Tooth Disease).
Further, polypeptides or polynucleotides of the invention may play a role in
neuronal survival; synapse formation; conductance; neural differentiation,
etc. Thus,
compositions of the invention (including polynucleotides, polypeptides, and
agonists
or antagonists) may be used to diagnose and/or treat or prevent diseases or
disorders
associated with these roles, including, but not limited to, learning and/or
cognition
disorders. The compositions of the invention may also be useful in the
treatment or
prevention of neurodegenerative disease states and/or behavioural disorders.
Such
neurodegenerative disease states and/or behavioral disorders include, but are
not
limited to, Alzheimers Disease, Parkinsons Disease, Huntingtons 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,
compositions of the invention may also play a role in the treatment,
prevention and/or
detection of developmental disorders associated with the developing embryo, or
sexually-linked disorders.
Additionally, polypeptides, polynucleotides and/or agonists or antagonists of
the invention, may be useful in protecting neural cells from diseases, damage,
disorders, or injury, associated with cerebrovascular disorders including, but
not
limited to, carotid artery diseases (e.g., carotid artery thrombosis, carotid
stenosis, or
Moyamoya Disease), cerebral amyloid angiopathy, cerebral aneurysm, cerebral
anoxia, cerebral arteriosclerosis, cerebral arteriovenous malformations,
cerebral artery
diseases, cerebral embolism and thrombosis (e.g., carotid artery thrombosis,
sinus
thrombosis, or Wallenberg's Syndrome), cerebral hemorrhage (e.g., epidural or
subdural hematoma, or subarachnoid hemorrhage), cerebral infarction, cerebral
ischemia (e.g., transient cerebral ischemia, Subclavian Steal Syndrome, or
vertebrobasilar insufficiency), vascular dementia (e.g., mufti-infarct),
leukomalacia,
periventricular, and vascular headache (e.g., cluster headache or migraines).
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


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
221
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 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.
Additional neurologic diseases which can be treated or detected with
polynucleotides, polypeptides, agonists, and/or antagonists of the present
invention
include 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 multi-infarct dementia, periventricular
leukomalacia,
vascular headache such as cluster headache and migraine.


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
222
Additional neurologic diseases which can be treated or detected with
polynucleotides, polypeptides, agonists, and/or antagonists of the present
invention
include 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
1 S epilepsy, status epilepticus such as Epilepsia Partialis Continua, and
Hallervorden-
Spatz Syndrome.
Additional neurologic diseases which can be treated or detected with
polynucleotides, polypeptides, agonists, and/or antagonists of the present
invention
include 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, and
cerebral
malaria.
Additional neurologic diseases which can be treated or detected with
polynucleotides, polypeptides, agonists, and/or antagonists of the present
invention
include meningitis such as arachnoiditis, aseptic meningtitis such as viral
meningtitis
which includes lymphocytic choriomeningitis, Bacterial meningtitis which
includes
Haemophilus Meningtitis, Listeria Meningtitis, Meningococcal Meningtitis such
as


CA 02384659 2002-03-11
WO 01/23546 PCT/US00/26323
223
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
S bulbar poliomyelitis and postpoliomyelitis syndrome, prion diseases (such as
Creutzfeldt-Jakob Syndrome, Bovine Spongiform Encephalopathy, Gerstmann-
Straussler Syndrome, Kuru, Scrapie), and cerebral toxoplasmosis.
Additional neurologic diseases which can be treated or detected with
polynucleotides, polypeptides, agonists, and/or antagonists of the present
invention
include central nervous system neoplasms such as brain neoplasms that include
cerebellar 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 optica, 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-Hoffmann 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,




DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPRI~:ND PLUS D'UN TOME.
CECI EST L,E TOME 1 DE 2
NOTE: Pour les tomes additionels, veillez contacter 1e Bureau Canadien des
Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.
THIS IS VOLUME 1 OF 2
NOTE: For additional valumes please contact the Canadian Patent Office.

Representative Drawing

Sorry, the representative drawing for patent document number 2384659 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 2000-09-26
(87) PCT Publication Date 2001-04-05
(85) National Entry 2002-03-11
Dead Application 2005-09-26

Abandonment History

Abandonment Date Reason Reinstatement Date
2002-09-27 FAILURE TO COMPLETE 2003-04-30
2004-09-27 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $300.00 2002-03-11
Maintenance Fee - Application - New Act 2 2002-09-26 $100.00 2002-09-05
Registration of a document - section 124 $100.00 2003-03-04
Maintenance Fee - Application - New Act 3 2003-09-26 $100.00 2003-09-18
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
HUMAN GENOME SCIENCES, INC.
Past Owners on Record
KOMATSOULIS, GEORGE A.
ROSEN, CRAIG A.
RUBEN, STEVEN M.
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 2002-03-11 1 53
Claims 2002-03-11 5 142
Description 2002-03-11 225 11,786
Description 2002-03-11 259 12,164
Cover Page 2002-09-27 1 31
PCT 2002-03-11 5 213
Assignment 2002-03-11 3 91
Correspondence 2002-09-25 1 23
Correspondence 2002-11-07 1 28
PCT 2002-03-12 5 199
Assignment 2003-03-04 3 173
Correspondence 2003-04-15 1 41
Prosecution-Amendment 2003-10-17 5 173
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.

BSL Files

To view selected files, please enter reCAPTCHA code :