Sélection de la langue

Search

Sommaire du brevet 2423559 

Énoncé de désistement de responsabilité concernant l'information provenant de tiers

Une partie des informations de ce site Web a été fournie par des sources externes. Le gouvernement du Canada n'assume aucune responsabilité concernant la précision, l'actualité ou la fiabilité des informations fournies par les sources externes. Les utilisateurs qui désirent employer cette information devraient consulter directement la source des informations. Le contenu fourni par les sources externes n'est pas assujetti aux exigences sur les langues officielles, la protection des renseignements personnels et l'accessibilité.

Disponibilité de l'Abrégé et des Revendications

L'apparition de différences dans le texte et l'image des Revendications et de l'Abrégé dépend du moment auquel le document est publié. Les textes des Revendications et de l'Abrégé sont affichés :

  • lorsque la demande peut être examinée par le public;
  • lorsque le brevet est émis (délivrance).
(12) Demande de brevet: (11) CA 2423559
(54) Titre français: PROTEINES SECRETEES HUMAINES 22
(54) Titre anglais: 22 HUMAN SECRETED PROTEINS
Statut: Réputée abandonnée et au-delà du délai pour le rétablissement - en attente de la réponse à l’avis de communication rejetée
Données bibliographiques
(51) Classification internationale des brevets (CIB):
  • C12N 15/12 (2006.01)
  • A61K 38/00 (2006.01)
  • A61K 38/17 (2006.01)
  • C07H 21/02 (2006.01)
  • C07H 21/04 (2006.01)
  • C07K 05/00 (2006.01)
  • C07K 14/47 (2006.01)
  • C12N 01/21 (2006.01)
  • C12N 15/63 (2006.01)
  • C12N 15/66 (2006.01)
  • C12N 15/85 (2006.01)
  • C12N 15/86 (2006.01)
  • C12P 21/00 (2006.01)
(72) Inventeurs :
  • ROSEN, CRAIG A. (Etats-Unis d'Amérique)
  • KOMATSOULIS, GEORGE A. (Etats-Unis d'Amérique)
  • BAKER, KEVIN P. (Etats-Unis d'Amérique)
  • BIRSE, CHARLES E. (Etats-Unis d'Amérique)
  • SOPPET, DANIEL R. (Etats-Unis d'Amérique)
  • OLSEN, HENRIK S. (Etats-Unis d'Amérique)
  • MOORE, PAUL A. (Etats-Unis d'Amérique)
  • WEI, PING (Etats-Unis d'Amérique)
  • EBNER, REINHARD (Etats-Unis d'Amérique)
  • DUAN, D. ROXANNE (Etats-Unis d'Amérique)
  • SHI, YANGGU (Etats-Unis d'Amérique)
  • CHOI, GIL H. (Etats-Unis d'Amérique)
  • FISCELLA, MICHELE (Etats-Unis d'Amérique)
  • NI, JIAN (Etats-Unis d'Amérique)
(73) Titulaires :
  • HUMAN GENOME SCIENCES, INC.
(71) Demandeurs :
  • HUMAN GENOME SCIENCES, INC. (Etats-Unis d'Amérique)
(74) Agent: MBM INTELLECTUAL PROPERTY AGENCY
(74) Co-agent:
(45) Délivré:
(86) Date de dépôt PCT: 2001-01-17
(87) Mise à la disponibilité du public: 2002-03-21
Requête d'examen: 2006-01-17
Licence disponible: S.O.
Cédé au domaine public: S.O.
(25) Langue des documents déposés: Anglais

Traité de coopération en matière de brevets (PCT): Oui
(86) Numéro de la demande PCT: PCT/US2001/001386
(87) Numéro de publication internationale PCT: US2001001386
(85) Entrée nationale: 2003-03-04

(30) Données de priorité de la demande:
Numéro de la demande Pays / territoire Date
60/232,104 (Etats-Unis d'Amérique) 2000-09-12

Abrégés

Abrégé français

La présente invention concerne de nouvelles protéines sécrétées humaines et des acides nucléiques isolés contenant les zones de codage des gènes codant ces protéines. L'invention traite aussi de vecteurs, de cellules hôtes, d'anticorps et de procédés de recombinaison pour produire des protéines sécrétées humaines. L'invention traite également de procédés de diagnostic et de thérapeutique permettant de diagnostiquer et de traiter des maladies, des troubles et/ou états liés à ces nouvelles protéines sécrétées humaines.


Abrégé anglais


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.

Revendications

Note : Les revendications sont présentées dans la langue officielle dans laquelle elles ont été soumises.


430
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 m 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.

431
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.

432
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.

433
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
II 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;

434
(b) isolating the supernatant;
(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 : Les descriptions sont présentées dans la langue officielle dans laquelle elles ont été soumises.


DEMANDE OU BREVET VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.
CECI EST LE TOME 1 DE 2
CONTENANT LES PAGES 1 A 254
NOTE : Pour les tomes additionels, veuillez 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
CONTAINING PAGES 1 TO 254
NOTE: For additional volumes, please contact the Canadian Patent Office
NOM DU FICHIER / FILE NAME
NOTE POUR LE TOME / VOLUME NOTE:

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
22 Human Secreted Proteins
Field of the Iuventiotz
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.
Backgroutzd of the hzveutioh
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
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
extracellu~ar 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
include the commercially valuable human insulin, interferon, Factor VIII,
human
growth hormone, tissue plasminogen activator, and erythropoeitin. Thus, in
light of
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, and/or conditions by using secreted proteins or
the genes
that encode them.
Summary of the I~ventioh
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 occurring), 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
preparations or other compositions where the art demonstrates no
distinguishing
features of the polynucleotide/sequences of the present invention.
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, 15
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., 5' or 3' to the gene of interest
in the
genome). In other embodiments, the polynucleotides of the invention do not
contain
the coding sequence of more than 1000, 500, 250, 100, 50, 25, 20, 15, 10, 5,
4, 3, 2, or
1 genomic flanking gene(s).
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 m NO:X
was often generated by overlapping sequences contained in multiple clones
(contig

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
4
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
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), 50 mM sodium phosphate (pH 7.6), Sx
Denhardt's solution, 10% dextran sulfate, and 20 ~.g/ml denatured, sheared
salmon
sperm DNA, followed by washing the filters in O.lx 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 NaH2P04; 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
reagents may require modification of the hybridization conditions described
above,
due to problems with compatibility.
Of course, a polynucleotide which hybridizes only to polyA+ sequences (such
as any 3' terminal polyA+ tract of a cDNA shown in the sequence listing), or
to a
complementary stretch of T (or ~ residues, would not be included in the
definition of
"polynucleotide," since such a polynucleotide would hybridize to any nucleic
acid
molecule containing a poly (A) stretch or the complement thereof (e.g.,
practically
any double-stranded cDNA clone generated using oligo dT as a primer).
The 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, enzylnatically, 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
6
a given polypeptide. Also, a given polypeptide may contain many types of
modifications. Polypeptides may be branched , for example, as a result of
ubiquitination, and they may be cyclic, with or without branching. Cyclic,
branched,
and branched cyclic polypeptides may result from posttranslation natural
processes or
may be made by synthetic methods. Modifications include acetylation,
acylation,
ADP-ribosylation, amidation, covalent attachment of flavin, covalent
attachment of a
heme moiety, covalent attachment of a nucleotide or nucleotide derivative,
covalent
attachment of a lipid or lipid derivative, covalent attachment of
phosphotidylinositol,
cross-linking, cyclization, disulfide bond formation, demethylation, formation
of
covalent cross-links, formation of cysteine, formation of pyroglutamate,
formylation,
gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation,
iodination, methylation, myristoylation, oxidation, pegylation, proteolytic
processing,
phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-
RNA
mediated addition of amino acids to proteins such as arginylation, and
ubiquitination.
(See, for instance, PROTEINS - STRUCTURE AND MOLECULAR PROPERTIES,
2nd 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., Aml 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.)

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
7
Polynucleotides and Polypeptides of the Invention
FEATURES OF PROTEIN ENCODED BY GENE NO: 1
The translation product of this gene shares sequence homology with human
alpha-1-B-glycoprotein (see, e.g. Genbank accession No. P04217; all references
available through this accession are hereby incorporated in their entirety by
reference
herein) which is thought to be important in immune modulation. "Amino acid
sequence of human plasma alpha 1 B-glycoprotein: homology to the
immunoglobulin
supergene family". Proc. Natl. Acad. Sci. U.S.A. 83 (8), 2363-2367 (1986) This
protein contains 5 immunoglobulin-like V-type domains.
This gene is expressed primarily in human liver and to a lesser extent in
lung,
brain and colon.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to liver
diseases. 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 hepatic system, expression of this gene at significantly
higher or
lower levels may be routinely detected in certain tissues or cell types (e.g.,
liver,
cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine,
bile,
lymph, 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.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ m NO: 61 as
residues: Gly-85 to Gly-96, Pro-162 to Val-168, Pro-177 to Gly-188, Arg-254 to
Asp-
260, Leu-283 to Asp-293, Glu-310 to Gly-318, Arg-338 to Arg-345, Ala-387 to
Leu-
392, Gly-398 to Gln-404, His-464 to Trp-475. Polynucleotides encoding said
polypeptides are also encompassed by the invention.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
The tissue distribution almost exclusively in liver and homology to alpha-1-B-
glycoprotein indicates that polynucleotides and polypeptides corresponding to
this
gene would be useful for the detection and treatment of liver disorders and
cancers
(e.g. hepatoblastoma, jaundice, hepatitis, liver metabolic diseases and
conditions that
are attributable to the differentiation of hepatocyte progenitor cells). In
addition the
expression in fetus would suggest a useful role for the protein product in
developmental abnormalities, fetal deficiencies, pre-natal disorders and
various
would-healing models and/or tissue trauma. 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 N0:11 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 1684 of SEQ ID
NO:11, b
is an integer of 15 to 1698, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID NO:l 1, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 2
The translation product of this gene shares homology with the marine G1RP
(see, e.g., Genbank Accession No. gb~AAF05310.1~AF171875_1 (AF171875); all
references available through this accession are hereby incorporated in their
entirety by
reference herein). Northern blot analysis using RNAs derived from 32Dc13 cells
that
have been grown in the absence of IL-3 demonstrated that the marine G1RP
message

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
is upregulated in these cells following the removal of IL-3, suggesting that
this gene
may regulate growth factor withdrawal-induced apoptosis of myeloid precursor
cells
(see, e.g., Baker and Reddy Gene 248:33-40 (2000), hereby incorporated in its
entirety by reference herein). Based on the sequence similarity, the
translation product
of this gene is expected to share at least some biological activities with the
murine
G1RP.
Moreover, the polypeptide encoded by this gene has been determined to have
transmembrarie domains at about amino acid position 195 to about 211 and at
about
388 to about 404 of the amino acid sequence referenced in Table 1 for this
gene.
Based upon these characteristics, it is believed that the protein product of
this gene
shares structural features to type IIIa membrane proteins.
This gene is expressed primarily in immune, reproductive and developing
tissues.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
disorders of the immune or reproductive system, or developmental disorders.
Similarly, polypeptides and antibodies directed to these polypeptides are
useful in
providing immunological probes for differential identification of the tissues)
or cell
type(s). For a number of disorders of the above tissues or cells, particularly
of the
immune and/or reproductive systems) or developing fetus, expression of this
gene at
significantly higher or lower levels may be routinely detected in certain
tissues or cell
types (e.g., reproductive, immune, fetal, cancerous and wounded tissues) or
bodily
fluids (e.g., serum, plasma, urine, lymph, amniotic fluid, 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.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 62 as
residues: Ala-27 to Glu-33, Asp-57 to Glu-69, Asn-134 to Thr-143, Met-185 to
Arg
192, Ile-221 to Leu-234, Thr-249 to Asp-260, Ser-270 to Asp-275, Pro-366 to
Gly-

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
376. Polynucleotides encoding said polypeptides are also encompassed by the
invention.
The tissue distribution indicates and homology to G1RP that polynucleotides
and polypeptides corresponding to this gene would be useful for the diagnosis,
5 detection, prevention and/or 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. Involvement in the regulation of
cytokine
production, antigen presentation, or other processes indicates a usefulness in
the
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 may be also used as an agent for immunological
disorders
including arthritis, asthma, immunodeficiency diseases such as AIDS, leukemia,
rheumatoid artlmitis, 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, Hodgkin's rheumatoid arthritis,
Sjogren's disease, scleroderma and tissues. 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. In addition, this gene
product may
have commercial utility in the expansion of stem cells and committed
progenitors of
various blood lineages, and in the differentiation and/or proliferation of
various cell
types. Moreover, the tissue distribution in reproductive and developing
tissues and
homology to murine G1RP indicates that polynucleotides and/or polypeptides
corresponding to this gene would be useful for the treatment, prevention,
detection,
and/or diagnosis of disorders of reproductive system organs, including
cancers,
disorders affecting fertility, and/or developmental disorders. Specifically,
expression
in prostate indicates that polynucleotides and/or polypeptides of the
invention would

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
11
be useful fox diagnosis, treatment and/or prevention of the disorders of the
prostate,
including inflammatory disorders, such as chronic prostatitis, granulomatous
prostatitis and malacoplakia, prostatic hyperplasia and prostate neoplastic
disorders,
including adenocarcinoma, transitional cell carcinomas, ductal carcinomas,
squamous
cell carcinomas, or as hormones or factors with systemic or reproductive
functions.
Likewise, expression in breast tissue indicates that polynucleotides and/or
polypeptides of the invention would be useful for diagnosis, treatment and/or
prevention of breast neoplasia and breast cancers, such as fibroadenoma,
pipillary
carcinoma, ductal carcinoma, Paget's disease, medullary carcinoma, mucinous
carcinoma, tubular carcinoma, secretory carcinoma and apocrine carcinoma, as
well
as juvenile hypertrophy and gynecomastia, mastitis and abscess, duct ectasia,
fat
necrosis and fibrocystic diseases. Similarly, expression in ovarian tissue,
indicates
that polynucleotides and polypeptides corresponding to this gene would be
useful for
the treatment, prevention, detection and diagnosis of conditions concerning
proper
ovarian function (e.g., egg maturation, endocrine function), as well as
cancer. The
expression in ovarian tissue may indicate the gene or its products can be used
to treat,
prevent, detect and/or diagnose disorders of the ovary, including inflammatory
disorders, such as oophoritis (e.g., caused by viral or bacterial infection),
ovarian
cysts, amenorrhea, infertility, hirsutism, and ovarian cancer (including, but
not limited
to, primary and secondary cancerous growth, endometrioid carcinoma of the
ovary,
ovarian papillary serous adenocarcinoma, ovarian mucinous adenocarcinoma,
Ovarian Krukenberg tumor).
Furthermore, expression in testicular tissue indicates that polynucleotides
and
polypeptides corresponding to this gene would be useful for the treatment,
prevention,
detection and/or diagnosis of conditions concerning proper testicular function
(e.g.
endocrine function, sperm maturation), as well as cancer. Therefore,
polynucleotides
and/or polypeptides of the invention would be useful in the treatment of male
infertility and/or impotence. Polynucleotides and/or polypeptides of the
invention
would be also useful in assays designed to identify binding agents, as such
agents
(antagonists) would be useful as male contraceptive agents. Similarly,
polynucleotides and/or polypeptides of the invention are believed to be useful
in the
treatment and/or diagnosis of testicular cancer. The testes are also a site of
active gene

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
12
expression of transcripts that may be expressed, particularly at low levels,
in other
tissues of the body. Therefore, this gene product may be expressed in other
specific
tissues or organs where it may play related functional roles in other
processes, such as
hematopoiesis, inflammation, bone formation, and kichley function, to name a
few
possible target indications.
Moreover, the expression within embryonic tissue and other cellular sources
marked by proliferating cells indicates that polypeptides and/or
polynucleotides
corresponding to this gene may play a role in the regulation of cellular
division, and
may show utility in the diagnosis, treatment, and/or prevention of
developmental
diseases and disorders, cancer, and other proliferative conditions.
Representative uses
are described in the "Hyperproliferative Disorders" and "Regeneration"
sections
below and elsewhere herein. Briefly, developmental tissues rely on decisions
involving cell differentiation and/or apoptosis in pattern formation.
Dysregulation of
apoptosis can result in inappropriate suppression of cell death, as occurs in
the
development of some cancers, or in failure to control the extent of cell
death, as is
believed to occur in acquired immunodeficiency and certain neurodegenerative
disorders, such as spinal muscular atrophy (SMA). Because of potential roles
in
proliferation and differentiation, this gene product may have applications in
the adult
for tissue regeneration and the treatment of cancers. It may also act as a
morphogen to
control cell and tissue type specification. Therefore, the polynucleotides and
polypeptides of the present invention would be useful in treating, detecting,
and/or
preventing said disorders and conditions, in addition to other types of
degenerative
conditions. Thus this protein may modulate apoptosis or tissue differentiation
and
would be useful in the detection, treatment, and/or prevention of degenerative
or
proliferative conditions and diseases. The protein is useful in modulating the
immune
response to aberrant polypeptides, as may exist in proliferating and cancerous
cells
and tissues. The protein can also be used to gain new insight into the
regulation of
cellular growth and proliferation. 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
13
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 m 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
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 1475 of SEQ m
N0:12, b
is an integer of 15 to 1489, where both a and b correspond to the positions of
nucleotide residues shown in SEQ m NO:12, and where b is greater than or equal
to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 3
The translation product of this gene shares sequence homology with a family
of secreted cyteine rich proteins including a Drosophila protein called
sprouty which
acts as a FGF antagonist (see, e.g., GeneSeq Accession No. W48794; all
references
available through this accession are hereby incorporated in their entirety by
reference
herein). The polypeptide encoded by this gene has been determined to have a
transmembrane domain at about amino acid position 1 to about 23 of the amino
acid
sequence referenced in Table 1 for this gene. Moreover, a cytoplasmic tail
encompassing about amino acids 24 to about 159 of this protein has also been
determined. Based upon these characteristics, it is believed that the protein
product of
this gene shares structural features to type Ib membrane proteins.
This gene is expressed primarily in testes and epidiymus.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
male reproductive disorders, vascular disorders, and wound healing. Similarly,
polypeptides and antibodies directed to these polypeptides are useful in
providing

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
14
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 male
reproductive system, the vascular and nervous systems, expression of this gene
at
significantly higher or lower levels may be routinely detected in certain
tissues or cell
types (e.g., reproductive, cancerous and wounded tissues) or bodily fluids
(e.g.,
serum, plasma, urine, semen, lymph, 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.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the ixnmunogenic epitopes shown in SEQ m NO: 63 as
residues: Gly-25 to Leu-30, Pro-40 to Ser-49, Pro-74 to Ser-91, Asn-97 to Cys-
104,
Pro-115 to Phe-123, Ser-125 to Ser-132. Polynucleotides encoding said
polypeptides
are also encompassed by the invention.
The tissue distribution in testes and homology to an fibroblast growth factor
antagonist indicates that polynucleotides and polypeptides corresponding to
this gene
would be useful for the treatment of testicular cancer, male sterility,
impotence and
potentially in the regulation of testoserone production and the induction and
maintenance of male characteristics; in addition there is potential for this
molecule in
would healing, anti-angiogenesis and tumor treatment, agiogenesis and the
treatment
of cardiac arrest, stroke and other vascular disorders. In addition,
expression in
testicular tissue indicates that polynucleotides and polypeptides
corresponding to this
gene would be useful for the treatment, prevention, detection and/or diagnosis
of
conditions concerning proper testicular function (e.g. endocrine function,
sperm
maturation), as well as cancer. Therefore, this gene product is useful in the
treatment
of male infertility and/or impotence. This gene product is also useful in
assays
designed to identify binding agents, as such agents (antagonists) would be
useful as
male contraceptive agents. Similarly, the protein is believed to be useful in
the
treatment and/or diagnosis of testicular cancer. The testes are also a site of
active gene
expression of transcripts that may be expressed, particularly at low levels,
in other
tissues of the body. Therefore, this gene product may be expressed in other
specific
tissues or organs where it may play related functional roles in other
processes, such as

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
hematopoiesis, inflammation, bone formation, and kidney function, to name a
few
possible target indications. 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
5 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
10 related to SEQ m 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
axe one or more polynucleotides comprising a nucleotide sequence described by
the
15 general formula of a-b, where a is any integer between 1 to 965 of SEQ >D
N0:13, b
is an integer of 15 to 979, where both a and b correspond to the positions of
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
The gene encoding the disclosed cDNA is believed to reside on chromosome
22, specifically at interval D22S420-D22S 1144. Accordingly, polynucleotides
related
to this invention would be useful as a marker in linkage analysis for
chromosome 22.
Moreover, the polypeptide encoded by this gene has been determined to have
a transmembrane domain at about amino acid position 26 to about 42 of the
amino
acid sequence referenced in Table 1 for this gene. Moreover, a cytoplasmic
tail
encompassing about amino acids 1 to about 25 of this protein has also been
determined. Based upon these characteristics, it is believed that the protein
product of
this gene shares structural features to type II membrane proteins.
This gene is expressed primarily in monocytes and dendritic cells and to a
lesser extent in other immune cell types.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
16
Polynucleotides and polypeptides of the invention would be useful as reagents
for differential identification of the tissues) or cell types) present in a
biological
sample and for diagnosis of diseases and conditions which include but are not
limited
to: immune disorders. Similarly, polypeptides and antibodies directed to these
polypeptides are useful in providing immunological probes fox 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, dendritic, cancerous and wounded tissues) or bodily
fluids (e.g.,
lymph, serum, plasma, urine, synovial fluid and spinal fluid) or another
tissue or
sample taken from an individual having such a disorder, relative to the
standard gene
expression level, i.e., the expression level in healthy tissue or bodily fluid
from an
individual not having the disorder.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 64 as
residues: Gly-100 to Pro-105, Pro-111 to Lys-116. Polynucleotides encoding
said
polypeptides are also encompassed by the invention.
The tissue distribution indicates that polynucleotides and polypeptides
corresponding to this gene would be useful for the diagnosis, detection,
prevention
and/or 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 lineages,
including
blood stem cells. Involvement in the regulation of cytokine production,
antigen
presentation, or other processes indicates a usefulness in the 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 may
be
also used as an agent for immunological disorders including arthritis,
astluna,
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;

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
17
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, Hodgkin's rheumatoid arthritis, Sjogren's disease,
scleroderma and
tissues. 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. W addition, this gene product may have commercial utility in
the
expansion of stem cells and committed progenitors of various blood lineages,
and in
the differentiation and/or proliferation of various cell types. Moreover, the
predicted
membrane localization indicates that polynucleotides and/or polypeptides
corresponding to this gene would be a good target for antagonists,
particularly small
molecules or antibodies, which block functional activity (such as, for
example,
binding of the receptor by its cognate ligand(s); transport function;
signalling
function). Accordingly, preferred are antibodies and or small molecules which
specifically bind an extracellular portion of the translation product of this
gene. The
extracellular regions can be ascertained from the information regarding the
transmembrane domains as set out above. Also provided is a kit for detecting
immune
disorders such as cancer. 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 immune disorders, including but not limited
to,
cancer, 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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
18
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ m 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 1079 of SEQ m
N0:14, b
is an integer of 15 to 1093, where both a and b correspond to the positions of
nucleotide residues shown in SEQ m N0:14, and where b is greater than or equal
to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 5
This gene is expressed primarily in brain and during early development.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
developmental disorders and neurological diseases. 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 nervous system,
expression
of this gene at significantly higher or lower levels may be routinely detected
in certain
tissues or cell types (e.g., nervous, neural, neuronal, cancerous and wounded
tissues)
or bodily fluids (e.g., lymph, serum, plasma, urine, synovial fluid and spinal
fluid) or
another tissue or sample taken from an individual having such a disorder,
relative to
the standard gene expression level, i.e., the expression level in healthy
tissue or bodily
fluid from an individual not having the disorder.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ m NO: 65 as
residues: Ser-53 to Thr-59, Lys-65 to Glu-70, Pro-80 to Gly-91, Ser-94 to Pro-
100,
Ala-116 to Asn-124, Pro-135 to Trp-151, Asn-172 to Phe-189, Asn-191 to.Trp-
196,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
19
Pro-201 to Trp-223, Gly-234 to Gly-245, Thr-251 to Asn-260, Arg-340 to Lys-
357,
Gly-428 to Tyr-454, Phe-458 to Tyr-464. Polynucleotides encoding said
polypeptides
are also encompassed by the invention.
The tissue distribution indicates that polynucleotides and/or polypeptides
corresponding to this gene would be useful for the detection, diagnosis,
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, toxic neuropathies induced by neurotoxins, peripheral
neuropathies,
multiple sclerosis, neoplasia of neuroectodermal origin, 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 )D 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
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 2628 of SEQ ID
N0:15, b
is an integer of 15 to 2642, 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 SY GENE NO: 6
This gene is expressed primarily in the salivary gland and in the palate.
10 Polynucleotides and polypeptides of the invention are useful as reagents
for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
digestive and salivary gland disorders. Similarly, polypeptides and antibodies
directed
to these polypeptides are useful in providing immunological probes for
differential
15 identification of the tissues) or cell type(s). For a number of disorders
of the above
tissues or cells, particularly of the endocrine systems, expression of this
gene at
significantly higher or lower levels may be routinely detected in certain
tissues or cell
types (e.g., digestive, cancerous and wounded tissues) or bodily fluids (e.g.,
serum,
plasma, urine, saliva, synovial fluid and spinal fluid) or another tissue or
sample taken
20 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.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ m NO: 66 as
residues: Pro-28 to Asp-41, Thr-107 to Tyr-114, Glu-158 to Pro-164.
Polynucleotides
encoding said polypeptides are also encompassed by the invention.
The tissue distribution indicates that polynucleotides and polypeptides
corresponding to this gene are useful for the treatment and/or detection of
digestive
disorders and salivary gland disorders including slivary gland carcinoma,
dental
caries, oral candidiasis, Sjorgens syndrome and xerostomia. 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
21
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 N0:16 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 681 of SEQ ID
N0:16, b
is an integer of 15 to 695, 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 fetal spleen and immune cell populations
and to a lesser extent in testes.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
immune disorders and male reproductive disorders. Similarly, polypeptides and
antibodies directed to these polypeptides are useful in providing
immunological
probes for differential identification of the tissues) or cell type(s). For a
number of
- disorders of the above tissues or cells, particularly of the male
reproductive system
and immune systems, expression of this gene at significantly higher or lower
levels
may be routinely detected in certain tissues or cell types (e.g.,
reproductive, immune,
cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine,
lymph,
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
22
expression level in healthy tissue or bodily fluid from an individual not
having the
disorder.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 67 as
residues: Met-1 to Ala-11, Leu-20 to Gly-26, Leu-51 to Ser-61, Phe-82 to Leu-
87,
Gln-94 to Pro-99. Polynucleotides encoding said polypeptides are also
encompassed
by the invention.
The tissue distribution indicates that polynucleotides and polypeptides
corresponding to this gene would be useful for the diagnosis, detection,
prevention
and/or 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 lineages,
including
blood stem cells. Involvement in the regulation of cytokine production,
antigen
presentation, or other processes indicates a usefulness in the 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 may
be
also used 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, Hodgkin's rheumatoid arthritis, Sjogren's disease,
scleroderma and
tissues. 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. In addition, this gene product may have commercial utility in
the
expansion of stem cells and committed progenitors of various blood lineages,
and in
the differentiation and/or proliferation of various cell types.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
23
In addition, expression in testicular tissue indicates that polynucleotides
and
polypeptides corresponding to this gene would be useful for the treatment,
prevention,
detection and/or diagnosis of conditions concerning proper testicular function
(e.g.
endocrine function, sperm maturation), as well as cancer, male sterility,
impotence
and potentially in the regulation of testoserone.production and the induction
and
maintenance of male characteristics. Therefore, polynucleotides and/or
polypeptides
of the invention would be useful in the treatment of male infertility and/or
impotence.
Polynucleotides and/or polypeptides of the invention would also be useful in
assays
designed to identify binding agents, as such agents (antagonists) would be
useful as
male contraceptive agents. Similarly, polynucleotides and/or polypeptides of
the
invention are believed to be useful in the treatment and/or diagnosis of
testicular
cancer. The testes are also a site of active gene expression of transcripts
that may be
expressed, particularly at low levels, in other tissues of the body.
Therefore, this gene
product may be expressed in other specific tissues or organs where it may play
related
functional roles in other processes, such as hematopoiesis, inflammation, bone
formation, and kidney function, to name a few possible target indications.
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
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 2507 of SEQ ID
N0:17, b
is an integer of 15 to 2521, 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 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
24
FEATURES OF PROTEIN ENCODED BY GENE NO: 8
The translation product of this gene shares sequence homology with
polyadenylate binding proteins of several organisms (see, e.g., Genbank
Accession
No. gb~AAB88449.1 ~ (AF032896) and/or emb~CAA88401.1 ~; all references
available
through these accessions are hereby incorporated in their entirety by
reference herein)
and may represent a fragment of a novel protein with similar function.
In specific embodiments, polypeptides of the invention comprise, or
alternatively consist of, the following amino acid sequence:
PGS THANXGRV GPXVHIPGQEPLTASMLAAAPLHEQKQMIGTCYLVLKRW S
DWMVLSFLPLLLSCDFEGSVSTPLSMMSTPSWLARSRACCWRLTTXSCCSCW
SLQNPSMPR (SEQ ID NO: 111). Moreover, fragments and variants of these
polypeptides (such as, for example, fragments as described herein,
polypeptides at
least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to these
polypeptides, or polypeptides encoded by a polynucleotide which hybridizes,
under
stringent conditions, to the polynucleotide encoding these polypeptides) are
encompassed by the invention. Antibodies that bind polypeptides of the
invention and
polynucleotides encoding these polypeptides are also encompassed by the
invention.
This gene is expressed primarily in kidney and to a lesser extent in the
developing embryo.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
renal disorders and/or developmental disorders. Similarly, polypeptides and
antibodies directed to these polypeptides are useful in providing
immunological
probes for differential identification of the tissues) or cell type(s). For a
number of
disorders of the above tissues or cells, particularly of the developing fetus,
and/or
renal system, expression of this gene at significantly higher or lower levels
may be
routinely detected in certain tissues or cell types (e.g., fetal, renal,
reproductive,
cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine,
lymph,
amniotic fluid, synovial fluid and spinal fluid) or another tissue or sample
taken from

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
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.
Preferred polypeptides of the present invention comprise, or alternatively
5 consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 68
as
residues: His-8 to Met-13. Polynucleotides encoding said polypeptides are also
encompassed by the invention.
The tissue distribution in kidney indicates that polynucleotides and/or
polypeptides corresponding to this gene would be useful in the treatment,
prevention,
10 diagnosis and/or detection of kidney diseases including renal failure,
nephritus, renal
colic, renal diabetes, renal hypertension, renal osteodystrophy, renal tubular
acidosis
proteinuria, pyuria, edema, pyelonephritis, hydronephritis, nephrotic
syndrome, crush
syndrome, glomerulonephritis, hematuria, renal colic and kidney stones, in
addition to
Wilm's Tumor Disease, and congenital kidney abnormalities such as horseshoe
15 kidney, polycystic kidney, and Falconi's syndrome. Moreover, the expression
within
embryonic tissue and homology to polyadenylate binding proteins indicates that
polypeptides and/or polynucleotides corresponding to this gene may play a role
in the
regulation of cellular division, and may show utility in the diagnosis,
treatment,
and/or prevention of developmental diseases and disorders, cancer, and other
20 proliferative conditions. Representative uses are described in the
"Hyperproliferative
Disorders" and "Regeneration" sections below and elsewhere herein. Briefly,
developmental tissues rely on decisions involving cell differentiation and/or
apoptosis
in pattern formation. Dysregulation of apoptosis can result in inappropriate
suppression of cell death, as occurs in the development of some cancers, or in
failure
25 to control the extent of cell death, as is believed to occur in acquired
immunodeficiency and certain neurodegenerative disorders, such as spinal
muscular
atrophy (SMA). Because of potential roles in proliferation and
differentiation, this
gene product may have applications in the adult for tissue regeneration and
the
treatment of cancers. It may also act as a morphogen to control cell and
tissue type
specification. Therefore, the polynucleotides and polypeptides of the present
invention would be useful in treating, detecting, and/or preventing said
disorders and
conditions, in addition to other types of degenerative conditions. Thus this
protein

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
26
may modulate apoptosis or tissue differentiation and would be useful in the
detection,
treatment, and/or prevention of degenerative or proliferative conditions and
diseases.
The protein is useful in modulating the immune response to aberrant
polypeptides, as
may exist in proliferating and cancerous cells and tissues. The protein can
also be
used to gain new insight into the regulation of cellular growth and
proliferation.
Furthermore, the protein may also be used to determine biological activity, to
raise
antibodies, as tissue markers, to isolate cognate ligaaids 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 m NO: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 1262 of SEQ ID
N0:18, b
is an integer of 15 to 1276, where both a and b correspond to the positions of
nucleotide residues shown in SEQ m N0:18, and where b is greater than or equal
to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 9
The translation product of this gene shares sequence homology with
DOCK180 protein (see, e.g., Genbank Accession No. dbj ~BAA09454.1 ~, all
references
available through this accession are hereby incorporated in their entirety by
reference
herein), a new effector molecule which transducer signals from tyrosine
kinases
through the CRK adaptor protein, which is thought to be important in detection
and/or
treatment of tumors.
In specific embodiments, polypeptides of the invention comprise, or
alternatively consist of, the following amino acid sequence:

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
27
IHHPIPSALLEVMLTAVKMFRLSAVTLCAFSLTLHSGVQLCEQLVLRIALFQN
CRAEDGFGLRVCWRRLMRSFCRSAKFWGSNDLRTWGSRFLWKDCT (SEQ
m NO: 112). Moreover, fragments and variants of these polypeptides (such as,
for
example, fragments as described herein, polypeptides at least 80%, 85%, 90%,
95%,
96%, 97%, 98%, 99%, or 100% identical to these polypeptides, or polypeptides
encoded by a polynucleotide which hybridizes, under stringent conditions, to
the
polynucleotide encoding these polypeptides) are encompassed by the invention.
Antibodies that bind polypeptides of the invention and polynucleotides
encoding
these polypeptides are also encompassed by the invention.
This gene is expressed primarily in colon and to a lesser extent in chronic
lymphotic leukemia.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of a variety of diseases and conditions which include but
are not
limited to: disorders of the digestive system, particularly diseases of the
colon,
including but not limited to, cancer; and/or disorders of the immune system
Similarly,
polypeptides and antibodies directed to these polypeptides are useful in
providing
immunological probes for differential identification of the tissues) or cell
type(s). For
a number of disorders of the above tissues or cells, particularly of the
digestive tract
and immune system expression of this gene at significantly higher or lower
levels
may be routinely detected in certain tissues or cell types (e.g., digestive,
immune,
cancerous and wounded tissues) or bodily fluids (e.g., lymph, serum, plasma,
urine,
synovial fluid and spinal fluid) or another tissue or sample taken from an
individual
having 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.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 69 as
residues: Asn-71 to Trp-76. Polynucleotides encoding said polypeptides are
also
encompassed by the invention.
The tissue distribution in colon and immune tissues and homology to
DOCK180 indicates that polynucleotides and polypeptides corresponding to this
gene

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
28
would be useful for diagnosis, detection, prevention and/or treatment of a
variety of
conditions including cancer. The tissue distribution in colon indicates that
polynucleotides and polypeptides corresponding to this gene would be useful
for
diagnosis, treatment, prevention and/or detection of tumors, especially of the
intestine, such as, carcinoid tumors, lymphomas, non-neoplastic polyps,
adenomas,
familial syndromes, colorectal carcinogenesis, colorectal carcinoma, cancer of
the
colon, cancer of the rectum and carcinoid tumors, as well as cancers in other
tissues
where expression has been indicated. The expression in the colon tissue may
indicate
the gene or its products can be used to treat, detect, prevent and/or diagnose
disorders
of the colon, including inflammatory disorders such as, congenital
abnormalities, such
as atresia and stenosis, Meckel diverticulum, congenital aganglionic megacolon-
Hirschsprung disease; enterocolitis, such as diarrhea and dysentary,
infectious
enterocolitis, including viral gastroenteritis, bacterial enterocolitis,
necrotizing
enterocolitis, antiboitic-associated colitis (pseudomembranous colitis), and
collagenous and lymphocytic colitis, miscellaneous intestinal inflammatory
disorders,
including parasites and protozoa, amoebic colitis, acquired irmnunodeficiency
syndrome, transplantation, drug-induced intestinal injury, radiation
enterocolitis,
neutropenic colitis, diverticular colon disease (DCD), inflammatory colonic
disease,
idiopathic inflammatory bowel disease, such as Crohn's disease (CD), non-
inflammatory bowel disease (non-IBD) colonic inflammation; ulcerative
disorders
such as, ulcerative colitis (UC); eosinophilic colitis; noncancerous tumors,
such as,
polyps in the colon, adenomas, leiomyomas, lipomas, and angiomas.
Similarly, the tissue distribution in B cells indicates that polynucleotides
and
polypeptides corresponding to this gene would be useful for the diagnosis,
detection,
prevention and/or 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 lineages,
including
blood stem cells. Involvement in the regulation of cytokine production,
antigen
presentation, or other processes indicates a usefulness in the treatment of
cancer (e.g.,
by boosting immune responses). Expression in cells of lymphoid origin,
indicates the

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
29
natural gene product would be involved in immune functions. Therefore it may
be
also used as an agent for immunological disorders including arthritis, asthma,
immunodeficiency diseases such as AIDS, leulcemia, 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,
lense tissue injury, demyelination, systemic lupus erythematosis, drug induced
hemolytic anemia, Hodgkin's rheumatoid arthritis, Sjogren's disease,
scleroderma and
tissues. 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. In addition, this gene product may have commercial utility in
the
expansion of stem cells and committed progenitors of various blood lineages,
and in
the differentiation and/or proliferation of various cell types. 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 N0:19 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 1201 of SEQ ID
NO:19, b
is an integer of 15 to 1215, where both a and b correspond to the positions of
nucleotide residues shown in SEQ m N0:19, and where b is greater than or equal
to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 10

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
The polypeptide encoded by this gene has been determined to have a
transmembrane domain at about amino acid position 12 to about 28 of the amino
acid
sequence referenced in Table 1 for this gene. Moreover, a cytoplasmic tail
5 encompassing about amino acids 29 to about 122 of this protein has also been
determined. Based upon these characteristics, it is believed that the protein
product of
this gene shares structural features to type Ib membrane proteins.
This gene is expressed primarily in immune system.
Polynucleotides and polypeptides of the invention are useful as reagents for
10 differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
disorders of the immune system. Similarly, polypeptides and antibodies
directed to
these polypeptides are useful in providing immunological probes for
differential
identification of the tissues) or cell type(s). For a number of disorders of
the above
15 immune system, expression of this gene at significantly higher or lower
levels may be
routinely detected in certain tissues or cell types (e.g., immune, cancerous
and
wounded tissues) or bodily fluids (e.g., lymph, serum, plasma, 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
20 healthy tissue or bodily fluid from an individual not having the disorder.
Expression of this gene primarily in immune cells suggests a role for the
encoded protein in the treatment, prevention, diagnosis and/or detection of
immune
disorders including autoimmune disease, asthma, and cancer. Moreover, the
tissue
distribution indicates that polynucleotides and polypeptides corresponding to
this
25 gene would be useful for the diagnosis, detection, prevention andlor
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
30 activation of hematopoietic cell lineages, including blood stem cells.
Involvement in
the regulation of cytokine production, antigen presentation, or other
processes
indicates a usefulness in the treatment of cancer (e.g., by boosting immune

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
31
responses). Expression in cells of lymphoid origin, indicates the natural gene
product
would be involved in immune functions. Therefore it may be also used as an
agent for
immunological disorders including arthritis, asthma, irnmunodeficiency
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, lense tissue injury, demyelination, systemic
lupus
erythematosis, drug induced hemolytic anemia, Hodgkin's rheumatoid arthritis,
Sjogren's disease, scleroderma and tissues. 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. In addition, this gene
product may
have commercial utility in the expansion of stem cells and committed
progenitors of
various blood lineages, and in the differentiation and/or proliferation of
various cell
types. 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
available and accessible through sequence databases. Some of these sequences
are
related to SEQ 1D NO: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 1615 of SEQ ID
N0:20, b
is an integer of 15 to 1629, 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
32
The polypeptide encoded by this gene has been determined to have a
transmembrane domain at about amino acid position 38 to about 54 of the amino
acid
sequence referenced in Table 1 for this gene. Moreover, a cytoplasmic tail
encompassing about amino acids 55 to about 184 of this protein has also been
determined. Based upon these characteristics, it is believed that the protein
product of
this gene shares structural features to type Ib membrane proteins.
This gene is expressed specifically expressed in normal and diseased pancreas.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to
digestive diseases, particularly, disorders of the pancreas. 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 pancreas,
expression of this
gene at significantly higher or lower levels may be routinely detected in
certain
tissues or cell types (e.g., digestive, cancerous and wounded tissues) or
bodily fluids
(e.g., bile, lymph, serum, plasma, urine, synovial fluid and spinal fluid) or
another
tissue or sample taken from an individual having such a disorder, relative to
the
standard gene expression level, i.e., the expression level in healthy tissue
or bodily
fluid from an individual not having the disorder.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ m NO: 71 as
residues: Arg-128 to Tyr-134. Polynucleotides encoding said polypeptides are
also
encompassed by the invention.
The tissue distribution in pancreas indicates that polynucleotides and
polypeptides corresponding to this gene would be useful for the treatment,
prevention
and/or diagnosis of disorders of the pancreas, including inflammatory
disorders, such
as chronic or acute pancreatitis; diabetes mellitus; pancreatic cancer.
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,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
33
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 m 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 1001 of SEQ m
N0:21, b
is an integer of 15 to 1015, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:21, 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 brain and to a lesser extent in liver and
immune cell types.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
neurological and/or immune system disorders. Similarly, polypeptides and
antibodies
directed to these polypeptides are useful in providing immunological probes
for
differential identification of the tissues) or cell type(s). For a number of
disorders of
the above tissues or cells, particularly of the immune, nervous and hepatic
systems,
expression of this gene at significantly higher or lower levels may be
routinely
detected in certain tissues or cell types (e.g., immune, neural, neuronal,
nervous,
cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine,
lymph,
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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
34
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 72 as
residues: Gly-4 to Glu-9, Asp-22 to Cys-28, Glu-39 to Leu-44, Phe-88 to Phe-
94.
Polynucleotides encoding said polypeptides are also encompassed by the
invention.
S The tissue distribution in brain indicates that polynucleotides andlor
polypeptides corresponding to this gene would be useful for the detection,
diagnosis,
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 1
l,
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, toxic neuropathies induced by neurotoxins, peripheral
neuropathies,
multiple sclerosis, neoplasia of neuroectodermal origin, 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. In
addition,
the tissue distribution in immune tissues indicates that polynucleotides and
polypeptides corresponding to this gene would be useful for the diagnosis,
detection,
prevention and/or 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; andlor activation of hematopoietic cell lineages,
including
blood stem cells. Involvement in the regulation of cytokine production,
antigen
presentation, or other processes indicates a usefulness in the treatment of
cancer (e.g.,
by boosting immune responses). Expression in cells of lymphoid origin,
indicates the

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
natural gene product would be involved in immune functions. Therefore it may
be
also used as an agent for immunological disorders including arthritis, asthma,
immunodeficiency diseases such as ADDS, leulcemia, rheumatoid arthritis,
granulomatous disease, inflammatory bowel disease, sepsis, acne, neutropenia,
5 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, Hodgkin's rheumatoid arthritis, Sjogren's disease,
scleroderma and
10 tissues. 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. In addition, this gene product may have commercial utility in
the
expansion of stem cells and committed progenitors of various blood lineages,
and in
the differentiation and/or proliferation of various cell types. Furthermore,
the protein
15 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.
20 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
excluded from the scope of the present invention. To list every related
sequence
25 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 1226 of SEQ ID
N0:22, b
is an integer of 15 to 1240, 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
30 + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 13

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
36
The polypeptide encoded by this gene has been determined to have a
transmembrane domain at about amino acid position 75 to about 91 of the amino
acid
sequence referenced in Table 1 for this gene. Moreover, a cytoplasmic tail
encompassing about amino acids 1 to about 74 of this protein has also been
determined. Based upon these characteristics, it is believed that the protein
product of
this gene shares structural features to type II membrane proteins.
This gene is expressed primarily in immune cell types.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
immune disorders. Similarly, polypeptides and antibodies directed to these
polypeptides are useful in providing immunological probes for differential
identification of the tissues) or cell type(s). For a number of disorders of
the above
tissues or cells, particularly of the 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, lymph, 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.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 73 as
residues: Ser-34 to Ile-39. Polynucleotides encoding said polypeptides are
also
encompassed by the invention.
The tissue distribution indicates that polynucleotides and polypeptides
corresponding to this gene would be useful for the diagnosis, detection,
prevention
and/or 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 of this gene product indicates a role in regulating the
proliferation;
survival; differentiation; and/or activation of hematopoietic cell lineages,
including

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
37
blood stem cells. W volvement in the regulation of cytokine production,
antigen
presentation, or other processes indicates a usefulness in the 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 may
be
also used as an agent for immunological disorders including arthritis, asthma,
immunodeficiency diseases such as A>DS, 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,
lense tissue injury, demyelination, systemic lupus erythematosis, drug induced
hemolytic anemia, Hodgkin's rheumatoid arthritis, Sjogren's disease,
scleroderma and
tissues. 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. In addition, this gene product may have commercial utility in
the
expansion of stem cells and committed progenitors of various blood lineages,
and in
the differentiation and/or proliferation of various cell types. 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
available and accessible through sequence databases. Some of these sequences
are
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 555 of SEQ ID
N0:23, b
is an integer of 15 to 569, where both a and b correspond to the positions of

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
38
nucleotide residues shown in SEQ m N0:23, and where b is greater than or equal
to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 14
The translation product of this gene shares sequence homology with
dishevelled gene products which are involved in signal transduction during
development. The gene encoding the disclosed cDNA is believed to reside on
chromosome 17. Accordingly, polynucleotides related to this invention would be
useful as a marker in linkage analysis for chromosome 17.
In specific embodiments, polypeptides of the invention comprise, or
alternatively consists of, the following amino acid sequence:
RPESYLVNLSLNDNDGSSGASDQDTLAPLPGATPWPLLPTFSYQYPAPHPYSP
QPPPYHELSSYTYGGGSASSQHSEGSRSSGSTRSDGGAGRTGRPEERAPESKS
GSGSESEPSSRGGSLRRGGEASGTSDGGPPPSRGSTGGAPNLRAHPGLHPYGP
PPGMALPYNPMMVVMMPPPPPPVPPAVQPPGAPPVRDLGSVPPELTASRQSF
HGHGQSQRVLCGCYVAHCGARLGRALLVCDWVSWPSCACSYSAWAQPTSC
CHTGDCGHCDSHQQCLVPPPSLRGRQGTFDYF (SEQ ID NO: 113). Moreover,
fragments and variants of these polypeptides (such as, for example, fragments
as
described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or
99% identical to these polypeptides and polypeptides encoded by the
polynucleotide
which hybridizes, under stringent conditions, to the polynucleotide encoding
these
polypeptides , or the complement there of are encompassed by the invention.
Antibodies that bind polypeptides of the invention are also encompassed by the
invention. Polynucleotides encoding these polypeptides are also encompassed by
the
invention.
This gene is expressed primarily in reproductive tissues, such as placenta and
to a lesser extent in the brain.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
reproductive and/or immune systems) disorders, and/or neurological disorders.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
39
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
nervous systems and/or the reproductive system, expression of this gene at
significantly higher or lower levels may be routinely detected in certain
tissues or cell
types (e.g., reproductive, nervous, neural, neuronal, cancerous and wounded
tissues)
or bodily fluids (e.g., serum, plasma, urine, vaginal pool, amniotic fluid,
lymph,
semen, 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.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ m NO: 74 as
residues: Pro-39 to Leu-44, Gln-80 to Pro-93, Pro-153 to Pro-158.
Polynucleotides
encoding said polypeptides are also encompassed by the invention.
The tissue distribution in reproductive and developing tissues indicates that
polynucleotides and/or polypeptides corresponding to this gene would be useful
for
the treatment, prevention, detection, and/or diagnosis of disorders of
reproductive
system organs, including cancers, disorders affecting fertility, and/or
developmental
disorders. Specifically, expression in prostate indicates that polynucleotides
and/or
polypeptides of the invention would be useful for diagnosis, treatment and/or
prevention of the disorders of the prostate, including inflammatory disorders,
such as
chronic prostatitis, granulomatous prostatitis and malacoplakia, prostatic
hyperplasia
and prostate neoplastic disorders, including adenocarcinoma, transitional cell
carcinomas, ductal carcinomas, squamous cell carcinomas, or as hormones or
factors
with systemic or reproductive functions. Likewise, expression in breast tissue
indicates that polynucleotides and/or polypeptides of the invention would be
useful
for diagnosis, treatment and/or prevention of breast neoplasia and breast
cancers, such
as fibroadenoma, pipillary carcinoma, ductal carcinoma, Paget's disease,
medullary
carcinoma, mucinous carcinoma, tubular carcinoma, secretory carcinoma and
apocrine carcinoma, as well as juvenile hypertrophy and gynecomastia, mastitis
and
abscess, duct ectasia, fat necrosis and fibrocystic diseases.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
Similarly, expression in ovarian tissue, indicates that polynucleotides and
polypeptides corresponding to this gene would be useful for the treatment,
prevention,
detection and diagnosis of conditions concerning proper ovarian function
(e.g., egg
maturation, endocrine function), as well as cancer.
5 The expression in ovarian tissue may indicate the gene or its products can
be
used to treat, prevent, detect and/or diagnose disorders of the ovary,
including
inflammatory disorders, such as oophoritis (e.g., caused by viral or bacterial
infection), ovarian cysts, amenorrhea, infertility, hirsutism, and ovarian
cancer
(including, but not limited to, primary and secondary cancerous growth,
endometrioid
10 carcinoma of the ovary, ovarian papillary serous adenocarcinoma, ovarian
mutinous
adenocarcinoma, Ovarian Krukenberg tumor). Furthermore, expression in
testicular
tissue indicates that polynucleotides and polypeptides corresponding to this
gene
would be useful for the treatment, prevention, detection and/or diagnosis of
conditions Concerning proper testicular function (e.g. endocrine function,
sperm
15 maturation), as well as cancer. Therefore, this gene product is useful in
the treatment
of male infertility and/or impotence. This gene product is also useful in
assays
designed to identify binding agents, as such agents (antagonists) would be
useful as
male Contraceptive agents. Similarly, the protein is believed to be useful in
the
treatment and/or diagnosis of testicular cancer. The testes are also a site of
active gene
20 expression of transcripts that may be expressed, particularly at low
levels, in other
tissues of the body. Therefore, this gene product may be expressed in other
specific
tissues or organs where it may play related functional roles in other
processes, such as
hematopoiesis, inflammation, bone formation, and kidney function, to name a
few
possible target indications.
25 In addition, the tissue distribution in nervous system tissues and homology
to
the dishevelled family of signaling proteins indicates that polynucleotides
and/or
polypeptides corresponding to this gene would be useful for the detection,
diagnosis,
treatment, and/or prevention of neurodegenerative disease states, behavioral
disorders, or inflammatory conditions. Representative uses are described in
the
30 "Regeneration" and "Hyperproliferative Disorders" sections below, in
Example 1 l,
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,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
41
Huntington's Disease, Tourette Syndrome, meningitis, encephalitis,
demyelinating
diseases, peripheral neuropathies, neoplasia, trauma, congenital
malformations, spinal
cord injuries, toxic neuropathies induced by neurotoxins, peripheral
neuropathies,
multiple sclerosis, neoplasia of neuroectodermal origin, 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 m 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 1240 of SEQ ~
N0:24, b
is an integer of 15 to 1254, where both a and b correspond to the positions of
nucleotide residues shown in SEQ m N0:24, and where b is greater than or equal
to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 15
The translation product of this gene shares sequence homology with type-2
phosphatidic acid phosphatase-gamma (see, e.g., Genbank Accession Nos.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
42
gb~AAC15968.1~ (AF035959) and gb~AAD24061.1~AF123611_1; all references
available through these accession are hereby incorporated in their entirety by
reference herein).
In specific embodiments, polypeptides of the invention comprise, or
alternatively consist of, an amino acid sequence selected from the group:
YAVTYTAMYVTLVFRVI~GSRLVKPSLCLALLCPAFLVGVVRVAEYRNHWSD
VLAGFLTGAAIATFLVTCVVH (SEQ ID NO: 114);
AALCAYAVTYTAMYVTLVFRVKGSRLVKPSLCLALLCPAFLVGVVRVAEYR
NHWSDVLAGFLTGAAIATFLVTCVVHNFQXR (SEQ ID NO: 115) and/or
AALCAYAVTYTAMYVTLVFRVKGSRLVKPSLCLALLCPAFLVGVVRVAEYR
NHWSDVLAGFLTGAAIATFLVTCVVHNFQXRPPSGRXLSPQSAYPRLPGPXFP
HLHNGGDHPCPAGCRXGCESSAWMQPGGSHRAAFTGLALPWAGGRPHPKR
(SEQ ID NO: 116). Moreover, fragments and variants of these polypeptides (such
as,
for example, fragments as described herein, polypeptides at least 80%, 85%,
90%,
95%, 96%, 97%, 98%, 99%, or 100% identical to these polypeptides, or
polypeptides
encoded by a polynucleotide which hybridizes, under stringent conditions, to
the
polynucleotide encoding these polypeptides) are encompassed by the invention.
Antibodies that bind polypeptides of the invention and polynucleotides
encoding
these polypeptides are also encompassed by the invention.
Contact of human T cells with supernatant expressing the product of this gene
was shown to increase the expression of a the surface molecule, CD152. Thus it
is
likely that the product of this gene is involved in the activation of T cells,
in addition
to other cell-lines or tissue cell types. Thus, polynucleotides and
polypeptides related
to this gene have uses which include, but are not limited to, activating
immune cells,
such as during an inflammatory response.
This gene is expressed primarily in brain and to a lesser extent in
reproductive
and immune tissues.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
neurological disorders, immune system disorders, and disorders of the
reproductive
system. Similarly, polypeptides and antibodies directed to these polypeptides
are

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
43
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 nervous system, reproductive system, and immune system,
expression of this gene at significantly higher or lower levels may be
routinely
detected in certain tissues or cell types (e.g., nervous, neural, neuronal,
immune,
reproductive, 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.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ m NO: 75 as
residues: Glu-37 to Trp-42, Phe-67 to Gly-88, Pro-101 to Leu-110.
Polynucleotides
encoding said polypeptides are also encompassed by the invention.
The tissue distribution in brain tissue indicates that polynucleotides and
polypeptides corresponding to this gene would be useful for the detection,
diagnosis,
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, aald 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, toxic neuropathies induced by neurotoxins, peripheral
neuropathies,
multiple sclerosis, neoplasia of neuroectodermal origin, 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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
44
Similarly, the tissue distribution in immune tissues indicates that
polynucleotides and polypeptides corresponding to this gene would be useful
for the
diagnosis, detection, prevention and/or 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
lineages, including blood stem cells. Involvement in the regulation of
cytokine
production, antigen presentation, or other processes indicates a usefulness in
the
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 may be also used 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, Hodgkin's rheumatoid arthritis,
Sjogren's disease, scleroderma and tissues. 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. In addition, this gene
product may
have commercial utility in the expansion of stem cells and committed
progenitors of
various blood lineages, and in the differentiation and/or proliferation of
various cell
types. Moreover, the tissue distribution in reproductive and developing
tissues
indicates that polynucleotides and/or polypeptides corresponding to this gene
would
be useful for the treatment, prevention, detection, and/or diagnosis of
disorders of
reproductive system organs, including cancers, disorders affecting fertility,
and/or
developmental disorders. Specifically, expression in prostate indicates that
polynucleotides and/or polypeptides of the invention would be useful for
diagnosis,
treatment and/or prevention of the disorders of the prostate, including
inflarmnatory
disorders, such as chronic prostatitis, granulomatous prostatitis and
malacoplakia,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
prostatic hyperplasia and prostate neoplastic disorders, including
adenocarcinoma,
transitional cell carcinomas, ductal carcinomas, squamous cell carcinomas, or
as
hormones or factors with systemic or reproductive functions. Lilcewise,
expression in
breast tissue indicates that polynucleotides and/or polypeptides of the
invention would
5 be useful for diagnosis, treatment and/or prevention of breast neoplasia and
breast
cancers, such as fibroadenoma, pipillary carcinoma, ductal carcinoma, Paget's
disease, medullary carcinoma, mucinous carcinoma, tubular carcinoma, secretory
carcinoma and apocrine carcinoma, as well as juvenile hypertrophy and
gynecomastia, mastitis and abscess, duct ectasia, fat necrosis and fibrocystic
diseases.
10 Similarly, expression in ovarian tissue, indicates that polynucleotides and
polypeptides corresponding to this gene would be useful for the treatment,
prevention,
detection and diagnosis of conditions concerning proper ovarian function
(e.g., egg
maturation, endocrine function), as well as cancer. The expression in ovarian
tissue
may indicate the gene or its products can be used to treat, prevent, detect
and/or
15 diagnose disorders of the ovary, including inflammatory disorders, such as
oophoritis
(e.g., caused by viral or bacterial infection), ovarian cysts, amenorrhea,
infertility,
hirsutism, and ovarian cancer (including, but not limited to, primary and
secondary
cancerous growth, endometrioid carcinoma of the ovary, ovarian papillary
serous
adenocarcinoma, ovarian mucinous adenocarcinoma, Ovarian Krukenberg tumor).
20 Furthermore, expression in testicular tissue indicates that polynucleotides
and
polypeptides corresponding to this gene would be useful for the treatment,
prevention,
detection and/or diagnosis of conditions concerning proper testicular function
(e.g.
endocrine function, sperm maturation), as well as cancer. Therefore, this gene
product
is useful in the treatment of male infertility and/or impotence. This gene
product is
25 also useful in assays designed to identify binding agents, as such agents
(antagonists)
would be useful as male contraceptive agents. Similarly, the protein is
believed to be
useful in the treatment andlor diagnosis of testicular cancer. The testes are
also a site
of active gene expression of transcripts that may be expressed, particularly
at low
levels, in other tissues of the body. Therefore, this gene product may be
expressed in
30 other specific tissues or organs where it may play related functional roles
in other
processes, such as hematopoiesis, inflammation, bone formation, and kidney
function,
to name a few possible target indications. Furthermore, the protein may also
be used

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
46
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 N0:25 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 1970 of SEQ ID
N0:25, b
is an integer of 15 to 1984, where both a and b correspond to the positions of
nucleotide residues shown in SEQ DJ N0:25, and where b is greater than or
equal to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 16
The translation product of this gene shares sequence homology with a portion
of renal specific transporter proteins from several different species (see,
e.g., Genbank
AccessionNos. dbj~BAA23875.1~ (AB005451) and gb~AAC70004.1~ (AF057039); all
references available through these accession numbers are hereby incorporated
in their
entirety by reference herein) which are thought to be involved in the cellular
transport
of anionic and cationic compounds (see, e.g., Mori, et al., FEBS Lett. 417:371-
374
(1997), hereby incorporated in its entirety by reference herein).
In specific embodiments, polypeptides of the invention comprise, or
alternatively consist of, an amino acid sequence selected from the group:
MAFSKLLEQAGGVGLFQTLQVLTFILPCLMIPSQMLLENFSAAIPGHRCWTH
MLDNGSAVSTNMTPKALLTISIPPGPNQGPHQCRRFRQPQWQLLDPNATATS
WSEADTEPCVDGWVYD (SEQ ID NO: 117) and
MAFSI~LLEQAGGVGLFQTLQVLTFILPCLMIPSQMLLENFSAAIPGHRCWTH

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
47
MLDNGSAVSTNMTPI~ALLTISIPPGPNQGPHQCRRFRQPQWQLLDPNATATS
WSEADTEPCVDGWVYXRRSSPPPSWPSGTWCAAPRLEXP (SEQ ID NO: ).
Moreover, fragments and variants of these polypeptides (such as, for example,
fragments as described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%,
97%, 98%, 99%, or 100% identical to these polypeptides, or polypeptides
encoded by
a polynucleotide which hybridizes, under stringent conditions, to the
polynucleotide
encoding these polypeptides) are encompassed by the invention. Antibodies that
bind
polypeptides of the invention and polynucleotides encoding these polypeptides
are
also encompassed by the invention.
The polypeptide encoded by this gene has been determined to have a
transmembrane domain at about amino acid position 21 to about 37 of the amino
acid
sequence referenced in Table 1 for this gene. Moreover, a cytoplasmic tail
encompassing about amino acids 1 to about 20 of this protein has also been
determined. Based upon these characteristics, it is believed that the protein
product of
this gene shares structural features to type II membrane proteins.
This gene is expressed primarily in placenta.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to
developmental disorders and reproductive disorders. Similarly, polypeptides
and
antibodies directed to these polypeptides are useful in providing
immunological
probes for differential identification of the tissues) or cell type(s). For a
number of
disorders of the above tissues or cells, particularly of developing or
proliferating
tissues, expression of this gene at significantly higher or lower levels may
be
routinely detected in certain tissues or cell types (e.g., fetal, cancerous
and wounded
tissues) or bodily fluids (e.g., serum, plasma, urine, amniotic fluid,
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.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ff~ NO: 76 as

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
48
residues: Pro-45 to Trp-50, Pro-76 to Trp-93. Polynucleotides encoding said
polypeptides are also encompassed by the invention.
The tissue distribution in placenta and homology to a transporter protein
indicates that polynucleotides and/or polypeptides corresponding to this gene
would
be useful for the treatment, prevention, detection, and/or diagnosis of
disorders of
reproductive system organs, including cancers, disorders affecting fertility,
developmental disorders, placenta abruptio, placenta previa, placental
failure, and
placental insufficiency.
In addition, the specific expression in placental tissue and predicted
membrane
localization indicates that polynucleotides and/or polypeptides corresponding
to this
gene would be a good target for antagonists, particularly small molecules or
antibodies, which block functional activity (e.g., transport function).
Accordingly,
preferred are antibodies and or small molecules which specifically bind an
extracellular portion of the translation product of this gene. The
extracellular regions
can be ascertained from the information regarding the transmembrane domains as
set
out above. Also provided is a kit for detecting developmental disorders or
cancer.
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
developmental disorders or cancer 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
'~5 elsewhere herein.
Many polynucleotide sequences, such as EST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ ID N0:26 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
49
general formula of a-b, where a is any integer between 1 to 2086 of SEQ ID
N0:26, b
is an integer of 15 to 2100, where both a and b correspond to the positions of
nucleotide residues shown in SEQ m N0:26, and where b is greater than or equal
to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 17
In specific embodiments, polypeptides of the invention comprise, or
alternatively consist of, the following amino acid sequence:
MRQEVHGCFAQMDRSLALPKIRARVLLQQFQTAWREAEFVKLDQAVAAPEL
QQQSKVRKSRSKSKSKGELLKKCIEDKIHLCEEQASEDLVEKVRGELLRERVQ
RMEAQEGGFAQSLVALQFQKASRVTETLSAYTALLSIQDLLLEELSASEMLTK
SACTQILESHSRELQELERKLEDQLVQQEAAQQQQALASWQQWVADGPGIL
NEPGEVDSERQVSTVLHQALSKSQTLLEQHQQCLREEQQNSVVLEDLLENME
ADTFATLCSQELRLASYLARMAMVPGATLRRLLSVVLPTASQPQLLALLDSA
TERHVDHAAESDGGAEQADVGRRRKHQSWWQALDGKLRGDLISRGLEKML
WARKRKQSILKKTCLPLRERMIFS GKGS WPHLS LEPIGELXPVPIV GAETIDLL
NTGEKLFIFRNPKEPEISLHVPPRKKKNFLNAKI~Al~GMD (SEQ ID NO:
118) and
MAMVPGATLRRLLSVVLPTASQPQLLALLDSATERHVDHAAESDGGAEQAD
VGRR
RKHQSWWQALDGKLRGDLISRGLEKMLWARKRKQSILKKTCLPLRERMIFS
GKGSWPHLSLEPIGELXPVPIVGAETIDLLNTGEKLFIFRNPKEPEISLHVPPRK
KKNFLNAKKAMRAXGMD (SEQ ID NO: ). Moreover, fragments and variants of
these polypeptides (such as, for example, fragments as described herein,
polypeptides
at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to these
polypeptides, or polypeptides encoded by a polynucleotide which hybridizes,
under
stringent conditions, to the polynucleotide encoding these polypeptides) are
encompassed by the invention. Antibodies that bind polypeptides of the
invention and
polynucleotides encoding these polypeptides are also encompassed by the
invention.
This gene is expressed primarily in immune and reproductive tissues and to a
lesser extent in epithelial cells and connective tissues.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
Polynucleotides and polypeptides of the invention axe useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to
immune or reproductive system disorders. Similarly, polypeptides and
antibodies
5 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 and/or reproductive
system(s),
expression of this gene at significantly higher or lower levels may be
routinely
detected in certain tissues or cell types (e.g., immune, reproductive,
cancerous and
10 wounded tissues) or bodily fluids (e.g., serum, plasma, urine, lymph,
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.
Preferred polypeptides of the present invention comprise, or alternatively
15 consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 77
as
residues: Val-52 to Trp-61, Leu-65 to Gly-71, Lys-86 to Leu-92.
Polynucleotides
encoding said polypeptides are also encompassed by the invention.
The tissue distribution indicates that polynucleotides and polypeptides
corresponding to this gene would be useful for the diagnosis, detection,
prevention
20 and/or 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 lineages,
including
25 blood stem cells. Involvement in the regulation of cytokine production,
antigen
presentation, or other processes indicates a usefulness in the 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 may
be
also used as an agent for immunological disorders including arthritis, asthma,
30 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;

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
51
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 erythematosis, drug induced
hemolytic anemia, Hodgkin's rheumatoid arthritis, Sjogren's disease,
scleroderma and
tissues. 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. In addition, this gene product may have commercial utility in
the
expansion of stem cells and committed progenitors of various blood lineages,
and in
the differentiation and/or proliferation of various cell types.
Moreover, the tissue distribution in reproductive tissues indicates that
polynucleotides and/or polypeptides corresponding to this gene would be useful
for
the treatment, prevention, detection, and/or diagnosis of disorders of
reproductive
system organs, including cancers, disorders affecting fertility, and/or
developmental
disorders. Specifically, expression in prostate indicates that polynucleotides
and/or
polypeptides of the invention would be useful for diagnosis, treatment and/or
prevention of the disorders of the prostate, including inflarninatory
disorders, such as
chronic prostatitis, granulomatous prostatitis and malacoplakia, prostatic
hyperplasia
and prostate neoplastic disorders, including adenocarcinoma, transitional cell
carcinomas, ductal carcinomas, squamous cell carcinomas, or as hormones or
factors
with systemic or reproductive functions. Likewise, expression in breast tissue
indicates that polynucleotides and/or polypeptides of the invention would be
useful
for diagnosis, treatment and/or prevention of breast neoplasia and breast
cancers, such
as fibroadenoma, pipillary carcinoma, ductal carcinoma, Paget's disease,
medullary
carcinoma, mucinous carcinoma, tubular carcinoma, secretory carcinoma and
apocrine carcinoma, as well as juvenile hypertrophy and gynecomastia, mastitis
and
abscess, duct ectasia, fat necrosis and fibrocystic diseases. Furthermore,
expression in
testicular tissue indicates that polynucleotides and polypeptides
corresponding to this
gene would be useful for the treatment, prevention, detection and/or diagnosis
of
conditions concerning proper testicular function (e.g. endocrine function,
sperm
maturation), as well as cancer. Therefore, polynucleotides and/or polypeptides
of the
invention would be useful in the treatment of male infertility and/or
impotence.
Polynucleotides and/or polypeptides of the invention would be also useful in
assays

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
52
designed to identify binding agents, as such agents (antagonists) would be
useful as
male contraceptive agents. Similarly, polynucleotides and/or polypeptides of
the
invention are believed to be useful in the treatment and/or diagnosis of
testicular
cancer. The testes are also a site of active gene expression of transcripts
that may be
expressed, particularly at low levels, in other tissues of the body.
Therefore, this gene
product may be expressed in other specific tissues or organs where it may play
related
functional roles in other processes, such as hematopoiesis, inflammation, bone
formation, and kidney function, to name a few possible target indications.
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
available and accessible through sequence databases. Some of these sequences
are
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 1122 of SEQ ID
N0:27, b
is an integer of 15 to 1136, 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
The translation product of this gene shares sequence homology with human
heat shock protein hsp40 and homologs thereof (see, e.g., Genbank accession
Nos.
gb~AAB07346.1~ and gb~AA.C14483.2~; all references available through these
accessions are hereby incorporated in their entirety by reference herein)
which are

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
53
thought to be important in maintaining the structure and activity of labile
proteins
through direct interactions.
The polypeptide encoded by this gene has been determined to have a
transmembrane domain at about amino acid position 36 to about 52 of the amino
acid
sequence referenced in Table 1 for this gene. Moreover, a cytoplasmic tail
encompassing about amino acids 1 to about 35 of this protein has also been
determined. Based upon these characteristics, it is believed that the protein
product of
this gene shares structural features to type II membrane proteins.
The gene encoding the disclosed cDNA is believed to reside on chromosome
12, specifically at interval D12S325-D12S1691. Accordingly, polynucleotides
related
to this invention would be useful as a marker in linkage analysis for
chromosome 12.
The polynucleotide of this clone encodes a 412aa protein with at least one
transmembrane domain at the 5'. It shows strong homology to a fragment of the
Bovine viral diarrhea virus-2 viral nonstructural protein NS2-3 and to
pestivirus type
1 viral nonstructural protein p125. '
This gene is expressed primarily in immune/hematopoetic tissues and to a
lesser extent in lung and ovarian cancers. The tissue distribution also shows
some
enrichment in digestive, reproductive, and immune/hematopoetic organs.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to
immune disorders and cancers. 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, lymph, 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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
54
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 78 as
residues: Leu-66 to Lys-71, Arg-92 to Trp-97, Lys-115 to Gly-122, Pro-182 to
Glu-
192, Ala-207 to Glu-212, Asn-221 to Val-227, Leu-234 to Lys-239, Cys-250 to
Arg-
255, Asp-260 to Ala-266, Pro-336 to Pro-349, Ser-387 to Arg-412.
Polynucleotides
encoding said polypeptides are also encompassed by the invention.
The tissue distribution and homology to human heat shock proteins hsp40
indicates that polynucleotides and polypeptides corresponding to this gene
would be
useful for the diagnosis, detection, prevention, and/or treatment of
conditions or
disorders associated with heat shock protein expression. They are also useful
as
antagonists for prevention or treatment of cancer (e.g. lung cancer, ovarian
cancer,
adenocarcinoma, breast and skin cancers). They are also useful for drug
screening
using libraries of compounds.
In addition, the tissue distribution in immune tissues indicates that
polynucleotides and polypeptides corresponding to this gene would be useful
for the
diagnosis, detection, prevention and/or 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
lineages, including blood stem cells. Involvement in the regulation of
cytokine
production, antigen presentation, or other processes indicates a usefulness in
the
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 may be also used 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, Hodgkin's rheumatoid arthritis,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
Sjogren's disease, scleroderma and tissues. 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. In addition, this gene
product may
have commercial utility in the expansion of stem cells and committed
progenitors of
5 various blood lineages, 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
10 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 N0:28 and may have been publicly available prior to
conception of
the present invention. Preferably, such related polynucleotides are
specifically
15 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 2265 of SEQ ID
N0:28, b
is an integer of 15 to 2279, where both a and b correspond to the positions of
20 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
25 The translation product of this gene shares sequence homology with
mitochondria) transcription factor A (see, e.g., Genbank accession Nos.
gb~AAC52815.1 ~ and gb~AAA59849.1 ~; all references available through these
accessions are hereby incorporated in their entirety by reference herein).
This gene is expressed primarily in ovarian cancer tissue.
30 Polynucleotides and polypeptides of the invention are useful as reagents
for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
56
cancer and disorders of the reproductive system, particularly diseases of the
ovary
(for example, ovarian cancer). 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 reproductive system, expression of this
gene at
significantly higher or lower levels may be routinely detected in certain
tissues or cell
types (e.g., reproductive, ovarian, cancerous and wounded tissues) or bodily
fluids
(e.g., serum, plasma, urine, lymph, vaginal pool, 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.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 79 as
residues: Thr-45 to Leu-54, Gln-71 to Asn-77, Glu-90 to Ile-98, Lys-111 to Thr-
122,
Lys-131 to Lys-140, Gly-153 to AIa-161, Arg-169 to Ser-178, Thr-184 to Gln-
198,
Glu-214 to Glu-220, Thr-234 to Tyr-246. Polynucleotides encoding said
polypeptides
are also encompassed by the invention.
The tissue distribution in ovarian cancer tissue, indicates that
polynucleotides
and polypeptides corresponding to this gene would be useful for the treatment,
prevention, detection and diagnosis of tumors, especially ovarian cancer, as
well as
cancers of other tissues where expression has been indicated.
The expression in ovarian cancer tissue may indicate that polynucleotides
and/or polypeptides of the invention can be used to treat, prevent, detect
and/or
diagnose disorders of the ovary, including inflammatory disorders, such as
oophoritis
(e.g., caused by viral or bacterial infection), ovarian cysts, amenorrhea,
infertility,
hirsutism, and ovarian cancer (including, but not limited to, primary and
secondary
cancerous growth, endometrioid carcinoma of the ovary, ovarian papillary
serous
adenocarcinoma, ovarian mucinous adenocarcinoma, Ovarian Krukenberg tumor).
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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
57
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 SST sequences, are publicly
available and accessible through sequence databases. Some of these sequences
are
related to SEQ m N0:29 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 1579 of SEQ m
N0:29, b
is an integer of 15 to 1593, where both a and b correspond to the positions of
nucleotide residues shown in SEQ m N0:29, and where b is greater than or equal
to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 20
The gene encoding the disclosed cDNA is believed to reside on chromosome
13, specifically at interval D13S153-D13S163. Accordingly, polynucleotides
related
to this invention would be useful as a marker in linkage analysis for
chromosome 13.
The polypeptide encoded by this gene has been determined to have a
transmembrane domain at about amino acid position 55 to about 71 of the amino
acid
sequence referenced in Table 1 for this gene. Moreover, a cytoplasmic tail
encompassing about amino acids72 to about 490 of this protein has also been
determined. Based upon these characteristics, it is believed that the protein
product of
this gene shares structural features to type Ib membrane proteins.
The translated gene has homology to neurofilament protein NF-220
(gi~161292~gb~AAA29991.1 ~).
In specific embodiments, polypeptides of the invention comprise, or
alternatively consist of, the following amino acid sequence:
MLKDFSNLLLVVLCDYVLGEAEYLLLREPGHVALSNDTVYVDFQYFDGANG
TLRNV S V LLLEANTNQTV TTKYLLTNQS QGTLKFECFYFKEAGDYWFTMTPE
ATDNSTPFPWWEKSAFLKVEWPVFHVDLNRSAKAAEGTFQVGLFTSQPLCPF

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
58
PVDKPNIVVDVIFTNSLPEARRNSRQPLEIRTSKRTELAQGQWVEFGCAPLGPE
AYVTVVLKLLGRDSVITSTGPIDLAQKFGYKLVMVPELTCESGVEVTVLPPPC
TFVQGVVTVFKEAPRYPGKRTIHLAENSLPWERGGQFLTVLCLTWGRISTALT
LAFQAEAIFLQRRSAC (SEQ lD NO: I 19). Moreover, fragments and variants of
this polypeptide (such as, for example, fragments as described herein,
polypeptides at
least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to these
polypeptides
and polypeptides encoded by the polynucleotide which hybridize, under
stringent
conditions, to the polynucleotide encoding this polypeptide are encompassed by
the
invention. Antibodies that bind polypeptides of the invention are also
encompassed by
the invention. Polynucleotides encoding this polypeptide are also encompassed
by the
invention.
This gene is expressed primarily in lung and fetal tissues and to a less
extent
in colon and pancreas tumors.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to
developmental disorders and diseases or conditions associated with
proliferating cells.
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
developing fetus, and profliferating tissues or cells, expression of this gene
at
significantly higher or lower levels may be routinely detected in certain
tissues or cell
types (e.g., fetal, cancerous and wounded tissues) or bodily fluids (e.g.,
amniotic
fluid, 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.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 80 as
residues: Pro-35 to Ser-49, Thr-85 to Ile-92, Pro-95 to Glu-103, Ser-110 to
Thr-130,
Val-142 to Gly-150, Glu-152 to Ala-158, Gln-173 to Leu-183, Gly-238 to Leu-
245,
Gly-272 to His-282, Arg-298 to Met-308, Ala-318 to Gly-339, Ala-353 to Leu-
360,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
59
Pro-362 to Tyr-367, Pro-372 to Asp-380, Glu-390 to Ser-414, Ile-416 to Arg-
424,
Ser-429 to Gln-439, Gly-472 to Thr-480. Polynucleotides encoding said
polypeptides
are also encompassed by the invention.
The tissue distribution in lung and fetal tissues indicates that
polynucleotides
and polypeptides corresponding to this gene would be useful for the diagnosis
and
treatment of lung diseases and developmental disorders. Moreover, the
expression
within embryonic tissue and other cellular sources marked by proliferating
cells
indicates that polypeptides and/or polynucleotides corresponding to this gene
may
play a role in the regulation of cellular division, and may show utility in
the diagnosis,
treatment, and/or prevention of developmental diseases and disorders, cancer,
and
other proliferative conditions. Representative uses are described in the
"Hyperproliferative Disorders" and "Regeneration" sections below and elsewhere
herein. Briefly, developmental tissues rely on decisions involving cell
differentiation
and/or apoptosis in pattern formation. Dysregulation of apoptosis can result
in
inappropriate suppression of cell death, as occurs in the development of some
cancers,
or in failure to control the extent of cell death, as is believed to occur in
acquired
immunodeficiency and certain neurodegenerative disorders, such as spinal
muscular
atrophy (SMA). Because of potential roles in proliferation and
differentiation, this
gene product may have applications in the adult for tissue regeneration and
the
treatment of cancers. It may also act as a morphogen to control cell and
tissue type
specification. Therefore, the polynucleotides and polypeptides of the present
invention would be useful in treating, detecting, and/or preventing said
disorders and
conditions, in addition to other types of degenerative conditions. Thus this
protein
may modulate apoptosis or tissue differentiation and would be useful in the
detection,
treatment, and/or prevention of degenerative or proliferative conditions and
diseases.
The protein is useful in modulating the immune response to aberrant
polypeptides, as
may exist in proliferating and cancerous cells and tissues. The protein can
also be
used to gain new insight into the regulation of cellular growth and
proliferation.
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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
Protein, as well as, antibodies directed against the protein may show utility
as a tumor
marlcer 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
5 related to SEQ m 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
10 general formula of a-b, where a is any integer between 1 to 2652 of SEQ m
N0:30, b
is an integer of 15 to 2666, where both a and b correspond to the positions of
nucleotide residues shown in SEQ m N0:30, and where b is greater than or equal
to a
+ 14.
15 FEATURES OF PROTEIN ENCODED BY GENE NO: 21
The translation product of this gene shares sequence homology with three
isoforms of rat tomosyn (see, e.g., Genbank Accession Nos.
gb~AAD27818.1~AF118889_1 (AF118889), gb~AAD27819.1~AF118890-1
20 (AF118890), and gb~AAD04756.1 ~; all references available through these
accessions
are hereby incorporated in their entirety by reference herein). Tomosyn is a
neural
tissue-specific syntaxin-1-binding proteinwhich is capable of dissociating
MunclBln-
Secl/rbSecl from syntaxin-1 to form a lOS tomosyn complex, an intermediate
complex converted to the 7S SNARE complex.
25 The three isoforms (m-tomosyn, b-tomosyn, and s-tomosyn) bind to syntaxin-
1, but not to syntaxin-2, -3, or -4, and have a region highly homologous to
VAMP,
another syntaxin-binding protein. This region is necessary but not sufficient
for high-
affinity binding of tomosyn to syntaxin-1. See, e.g., Yokoyama, et al.,
Biochem.
Biophys. Res. Commun. 256 (1), 218-222 (1999), incorporated herein by
reference.
30 Based on the sequence similarity, the translation product of this gene is
expected to
share at least some biological activities with any one of the three isoforms
of the
tomosyn proteins.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
61
In specific embodiments, polypeptides of the invention comprise, or
alternatively consist of, the following amino acid sequence:
MDWWFLAIAMALLWLTTSRKQCCSTWALLNYMALMILIGENPDLLVNLDSL
QEPVCVILVKGLLFQRIAANLQPLQRCQGS (SEQ ID NO: 120) and
MVDYLQKAVLLNLGTIELYGSNDPYRREPRSPRKSRQPSGAGLCDISEGTVVP
EDRCKSPTSAKMSRKLSLPTDLKPDLDVKDNSFSRSRSSSVTSIDKESREAISA
LHFCETFTRKTDSSPSPCLWVGTTLGTVLVIALNLPPGGEXXLLQPVIVSPSGTI
LRLKGAILRMAFLDTTGCLIPPAYEPWREHNVPEEKDEKEKXKI~RRPVSVSPS
SSQEISENQYAVICSEKQAKVISLPTQNCAYKQNITETSFVLRGDIVALSNSICL
ACFCANGHIMTFSLPSLRPLLXVYYLPLTNMRXARTFCFTNNGQALYLVSPTE
IQRLTYSQETCENLQEMLGELFTPVETPEAPNRGFFKGLFGGGAQSLDREELF
GE S S S GKASRSLAQHIP GP GGIEGVKGAAS GV V GELAR.ARLALDERGQI~LGD
LEERTAAMLSSAESFSKHAHEIMLKYKDKKWYQF (SEQ ID NO: 12I).
Moreover, fragments and variants of this polypeptide (such as, for example,
fragments as described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%,
97%, 98%, or 99% identical to these polypeptides and polypeptides encoded by
the
polynucleotide which hybridize, under stringent conditions, to the
polynucleotide
encoding this polypeptide are encompassed by the invention. Antibodies that
bind
polypeptides of the invention are also encompassed by the invention.
Polynucleotides
encoding this polypeptide are also encompassed by the invention.
This gene is expressed primarily in immune tissues.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to
immune system disorders. Similarly, polypeptides and antibodies directed to
these
polypeptides are useful in providing immunological probes for differential
identification of the tissues) or cell type(s). For a number of disorders of
the above
tissues or cells, particularly of the immune system, expression of this gene
at
signif cantly 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, lymph, synovial fluid and spinal fluid) or another tissue or
sample
taken from an individual having such a disorder, relative to the standard gene

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
62
expression level, i.e., the expression Ievel in healthy tissue or bodily fluid
from an
individual not having the disorder.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 81 as
residues: Thr-17 to Cys-23, Gln-76 to Ser-81. Polynucleotides encoding said
polypeptides are also encompassed by the invention.
The tissue distribution in immune tissues and homology to the rat tomosyn
protein indicates that polynucleotides and polypeptides corresponding to this
gene
would be useful for the diagnosis, detection, prevention and/or treatment of a
variety
of immune system disorders. Representative uses axe 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 lineages, including blood stem cells.
Involvement in
the regulation of cytokine production, antigen presentation, or other
processes
indicates a usefulness in the 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 may be also used 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, lense tissue injury, demyelination, systemic
lupus
erythematosis, drug induced hemolytic anemia, Hodgkin's rheumatoid arthritis,
Sjogren's disease, scleroderma and tissues. 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. In addition, this gene
product may
have commercial utility in the expansion of stem cells and committed
progenitors of
various blood lineages, and in the differentiation and/or proliferation of
various cell
types. Furthermore, the protein may also be used to determine biological
activity,
raise antibodies, as tissue markers, to isolate cognate ligands or receptors,
to identify

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
63
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 m N0:31 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 2687 of SEQ m
N0:31, b
is an integer of 15 to 2701, where both a and b correspond to the positions of
nucleotide residues shown in SEQ m N0:31, and where b is greater than or equal
to a
+ 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 22
This gene is expressed primarily in B-cell leukemia and to a lesser extent in
ovarian cancer.
Polynucleotides and polypeptides of the invention are useful as reagents for
differential identification of the tissues) or cell types) present in a
biological sample
and for diagnosis of diseases and conditions which include but are not limited
to:
immune disorders and disorders of the reproductive system, including, but not
limited
to ovarian cancer. 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 and the female
reproductive
system, expression of this gene at significantly higher or lower levels may be
routinely detected in certain tissues or cell types (e.g., reproductive,
immune,
cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine,
lymph,
vaginal pool, 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,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
64
i.e., the expression level in healthy tissue or bodily fluid from an
individual not
having the disorder.
Preferred polypeptides of the present invention comprise, or alternatively
consist of, one or more of the immunogenic epitopes shown in SEQ ID NO: 82 as
residues: Thr-35 to Gly-48. Polynucleotides encoding said polypeptides are
also
encompassed by the invention.
The tissue distribution indicates that polynucleotides and polypeptides
corresponding to this gene would be useful for the diagnosis, detection,
prevention
and/or 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 lineages,
including
blood stem cells. Involvement in the regulation of cytokine production,
antigen
presentation, or other processes indicates a usefulness in the 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 may
be
also used 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,
lense tissue injury, demyelination, systemic lupus erythematosis, drug induced
hemolytic anemia, Hodgkin's rheumatoid arthritis, Sjogren's disease,
scleroderma and
tissues. 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. In addition, this gene product may have commercial utility in
the
expansion of stem cells and committed progenitors of various blood lineages,
and in
the differentiation and/or proliferation of various cell types. Similarly, the
tissue
distribution in ovarian cancer tissue, indicates that polynucleotides and
polypeptides
corresponding to this gene would be useful for the treatment, prevention,
detection

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
and diagnosis of tumors, especially ovarian cancer, as well as cancers of
other tissues
where expression has been indicated. The expression in ovarian cancer tissue
may
indicate the gene or its products can be used to treat, prevent, detect and/or
diagnose
disorders of the ovary, including inflammatory disorders, such as oophoritis
(e.g.,
5 caused by viral or bacterial infection), ovarian cysts, amenorrhea,
infertility,
hirsutism, and ovarian cancer (including, but not limited to, primary and
secondary
cancerous growth, endometrioid carcinoma of the ovary, ovarian papillary
serous
adenocarcinoma, ovarian mutinous adenocarcinoma, Ovarian Krukenberg tumor).
Furthermore, the protein may also be used to determine biological activity,
raise
10 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
15 available and accessible through sequence databases. Some of these
sequences are
related to SEQ B7 N0:32 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
20 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 514 of SEQ m
NO:32, b
is an integer of 15 to 528, where both a and b correspond to the positions of
nucleotide residues shown in SEQ ID N0:32, and where b is greater than or
equal to a
+ 14.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
''"' ~ ~n Ov ~ 01 N N d-
~ ~ ' o
j ~ d i i ~ ~ ~ n N
p Wit'
N
~ ~ N N o0 0o M .-a l~ O O
' '
V C~l N N N N d ~ M M
~
N P~
4~
N N N N N do' ~ N N
~
+~
a -.
~
H
O
~ M M ~ ~ d' O N
~ N
M M o ~ N
~ M N ~ ~
z~o~ ~
' ~ ~ ' ov o
~
~ ,
H
-.~ ,~ ,-.
' ~ ~ U
cO O_ 01 v0 ~ M ~t N
H ue
O ~ t~ d ~ ~ O
~ ' O> --~ -, N
, ,
r~ ~ M N ~ M d' ~n V7
F-~ r'~ .~ M ~ M ~ ~ M ~ M
i--i5.-Wr S.-~
O O O O
O ~ ~ ~
M O O O
~ M ~
M M
~ ~ ~ ~
~
~ U U U ~
~ ~ ~ ~ ~ ~ ~ ~ ~
o o o o o o o o o
~ ~ o o o o o o o 0
0 0
~
O +' N N N N N N N N N
~ ~ p~ ~ p~ p~ p~ p~ ~ p~
~~ ~ H~ H~ H~ H~ H~ H~ ~ H~
z ~ ~ ~ o ~ ~ ~ ~ o
o o o o o o o
~ ~
N N O N N
N
er
O ~ (~ p
..,
N N M d- d- ~n ~n

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
0o O O N ~O
O l~ ~ ~ ~ 00 i ~ ~ ~
, p o 0 0
M p 0 0 0 M
N
N N ' ' N ' d0' ' N
N d d d d ~h
' P-a
+~
_'' N N ~ ~ .-r 01 ~ ~ due,N
~ a
~
~ lp ~ O M ~ O O
~ M N ~ ~ ~ ~ 00 0o r, r, N
c%~
l0 ~ O M O O
b N ,.~ lw 0 00 00 ~ ~ N
U
~+-~ O ~' ~ ~ N ~ ~ 1 01 ~ ~ N
o vo '~ N ~ ~ 00 00
y '
U ,
~
a~
zo~~
~ 00 ~ l~ ~ M V7 M d1
+-~ H ~ ~ N 01 l~ N ~ N
~' ~ ~ ~ N ~ ~ ~ ~ N r., ~O
oo
V~ ,.
..,
O r [~ ~D o0 I~ 01 O o0 .-W 1 N O
.
~ M ~ M ~ M d' ~ d' ~ Wit'N
N o o N N N 0 0 0 0 0
~ ~ ~ ~
~ r~ '~ '~ '~ ~ ~ ~ ~ r~
p, u.~ ~ ~.
.~, d ~ ~ ~ ' ~ ' ~ ~ ' ~ ~
o o o o o o o o o o o
w
U ~, ~ ~ o o o o o o o o o o 0
0
~
N O ~ N N N N N N N N N N N
z~,N ~ p~ ~ ~ p~ p~ p~ 01 p~ p~ p~ p~
' ' ' ' ' ' ' ' ' ' '
o o o o o o o o o o o
~azr~ ~o ~o ~o ~o ~~ ~o ~o ~o ~o ~o ~o
d' O ~ ~ ~ N N M M
U
. ~ ~ ~ ~ ~ , ,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
-. ~p ~t al ~D t~ 00 00
b
N N M M N N N M N N
N P~
O ~ ~ N '-' M c~r1 N N '~ M M N N
OM1 0~1 t~ ~ ~ 0~1 0~1 ~ 0~1
4-i .--a
z p ~ O ~ ~ ~ ~ ~ ~ N N ~ N ~ O
U1 ~ N v~ ,--~ ~ N N ~ ~ N M d'
O o0 ,--WO ~O ~t
U N ,~-~ ~ N N ~ N d0
N
z p O ~ ~ ~ p o o ~ ~ ~ N
M ~ ~ 00 \O r, .-1 ,-i ~O ~ V~ l~ ~, 00
z p O ~ ~ ,-~ ,--~ ~ ,-i .-~ ~ due'
L-~ ~' ~ ~ ~ O d0' ~ N
z ~j 00 l~ o ~ ~ l0 ~ tn I~ ,N-, 00
dM' due' N ~ N due' d~ N d N due'
z
0 0 0 o N ~ N ~ N ~ N ~ N ~ 0 0
~ ~, ~ ~,
b ~ o ~ o ~ o ~ o ~ o ~. o ~ o ~ o ~ o ~ o ~ o
0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
U O ~ N ~p~ N 01 N 01 N \~ N ~p~ N ~p~ N p1 N p~ N ~p~ N ~p~ N p~
[~ Q.. N ~ ~ O ~ O ~ O ~ O ~ O ~ O ~ O ~ O ~ O ~ O ~ O
o ~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o
-a o 0
~ d- d' d- ,~ ,-~ 00 00
a1 a1 a1
U U O O
a W W ~ O
a~
O O O ~ .-i N N N M M d' ~Y

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
v cue, y0 O N v~ M O oo N d- ~ N
~ ~n ~ N ~ d' N N
+~ 4-r N ~',
N ~ Q1 Ov o0 00 N N d' 01 01
N M M M M N N N d'
O
o ~ ~ ,~ oMO M M M N N ~"'' ~ ono °°
~aa~z~ o ~ o ~ o ~ o ~ o
_~~
0
w
z 4-~ ~ O ~ o0 d' d' ~n oo ~O N r' 00
O ~ ~ ~ M V7 ~ ~ ~ M
zoo ~ ~ ~ ° ~ ~ ~ N
M U ~ M '-' ~ N ~ '~ .-a N
p O ~ 0~0 ~ ~ ~ ,~ ~ 0~1 ~ O ~ ,--i
~O N
C~ a" N oho ~ p ~ m ~ ~ ~ OM1 N
d~ N ~ ~ ~ N oho
N V7 N ~ N ~ ~ N
s~ t-r s~ . ~, s~ ~,
O O O O ~ ~ O O
'~'., ~ ~ V1 O ~!1 O L/1 O C!~ O ~ ~ ~ O C/~~ O
O ~ ~~ ~~~M~ ~ ~N C~ ~ -~-~N~N
~ ~~v~~Y~~~
;~, 'o ~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o h o ~ o ~ o
U ~ ~ ~ 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 o O o 0 0 0 0
U O +' N ~ N ~p~ N ~O~ N ~p~ N ~ N 01 N ~ N ~O~ N p~ N ~~ N ~p~
i ~ ~ i i ~ ~ i i i i
~ N ~ ~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o
~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o ~ o
a, a,
o ~ ~ H H a a
~~x x x x x ~ ~ x x

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
00
V y ~,,~O N O N N O O
~ P-r
OC
O ~ N 01 r., 01 01 ~ N N
~
+~
~ o ~ o o
~ ~
H _~~
M V7 O~'vV7 O M d' M
M
H o
M V7 ~ O M d' M
01 ~ I~ M
p O ~ ~ 0 O ~ O
~ ~ '~' 0 oo M o, N ~n
0 ~n
~
cn U N r,
H
~
N l ~ ~O M
pH~ ~ ~ N ~ N ~ N O
H Z ~ N ~ ~' N r p
o p
~ N
O r~ O \O t~ .~ 00 01 N O
M ~ ~ M ~ ~ M \O
H
O O O O O O
_ _
o ~ ~ ~ ~ ~ ~ '~
o o o o o o
N ~ ~ ~ ~ ~ O O
N N M M M
U U U U U U
~, u.~ ~, u.~ ~, ~,
N 0 N ~ 0 N O O
\
N ~ O 0 O 0 0 O O O
O1
~pzr~ ~o ~o ~o ~o ~o ~o ~o ~~
v z z z
U U U
o
O
O O O O .--~,--~,-i N N
N N N N N N N N

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
71
Table 1 summarizes the information corresponding to each "Gene No."
described above. The nucleotide sequence identified as "NT SEQ m NO:X" was
assembled from partially homologous ("overlapping") sequences obtained from
the
"cDNA clone m" 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 m 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
identif ed by "Gene No." The deposited clone may contain all or most of these
sequences, reflected by the nucleotide position indicated as "5' NT of Clone
Seq."
and the "3' NT of Clone Seq." of SEQ ID NO:X. The nucleotide position of SEQ m
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 m 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 m 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 m NO:Y of the secreted portion is
identified as "Predicted First AA of Secreted Portion." Finally, the amino
acid
position of SEQ m NO:Y of the last amino acid in the open reading frame is
identified as "Last AA of ORF."
SEQ >D NO:X (where X may be any of the polynucleotide sequences
disclosed in the sequence listing) and the translated SEQ m NO:Y (where Y may
be
any of the polypeptide sequences disclosed in the sequence listing) are
sufficiently

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
72
accurate and otherwise suitable for a variety of uses well known in the art
and
described further below. For instance, SEQ ID NO:X is useful for designing
nucleic
acid hybridization probes that will detect nucleic acid sequences contained in
SEQ ID
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 ff~ 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 l~nown methods using the sequence information
disclosed

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
73
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
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
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 provides predicted epitopes contained in certain embodiments of the
invention and polynucleotide sequences that may be disclaimed according to
certain
embodiments of the invention. The first column refers to each "Gene #"
described
above in Table 1. The second column provides the sequence identifier, "NT SEQ
ID
NO:X", for polynucleotide sequences disclosed in Table 1. The third column
provides
the sequence identifier, "AA SEQ III NO:Y", for polypeptide sequences
disclosed in
Table 1. The fourth column provides a unique integer "ntA" where "ntA" is any
integer between 1 and the final nucleotide minus 15 of SEQ ID NO:X, and the
fifth
column provides a unique integer "ntB" where "ntB" is any integer between 15
and
the final nucleotide of SEQ 117 NO:X, where both ntA and ntB correspond to the
positions of nucleotide residues shown in SEQ ID NO:X, and where ntB is
greater
than or equal to a + 14. For each of the polynucleotides shown as SEQ ID NO:X,
the
uniquely defined integers can be substituted into the general formula of a-b,
and used
to describe polynucleotides which may be preferably excluded from the
invention.
Column 6 lists residues comprising predicted epitopes contained in the
polypeptides
encoded by each of the preferred ORFs (SEQ ID NO:Y). Identification of
potential
immunogenic regions was performed according to the method of Jameson and Wolf
((1988) CABIOS, 4; 181-186); specifically, the Genetics Computer Group (GCG)
implementation of this algorithm, embodied in the program PEPTIDESTRUCTURE
(Wisconsin Package v10.0, Genetics Computer Group (GCG), Madison, Wisc.). This

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
74
method returns a measure of the probability that a given residue is found on
the
surface of the protein. Regions where the antigenic index score is greater
than 0.9
over at least 6 amino acids are indicated in Table 2 as "Predicted epitopes".
Polypeptides of the invention may possess one, two, three, four, five or more
antigenic epitopes comprising residues described in Table 2. It will be
appreciated
that depending on the analytical criteria used to predict antigenic
determinants, the
exact address of the determinant may vary slightly.
Table 3 summarizes the expression profile of polynucleotides corresponding
to the clones disclosed in Table 1. The first column provides a unique clone
identifier, "Clone DJ", for a cDNA clone related to each contig sequence
disclosed in
Table 1. Column 2, "Library Code(s)" shows the expression profile of tissue
and/or
cell line libraries which express the polynucleotides of the invention. Each
Library
Code in column 2 represents a tissue/cell source identifier code corresponding
to the
Library Code and Library description provided in Table 5. 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
expression.
Table 4, column l, provides a nucleotide sequence identifier, "SEQ ID
NO:X," that matches a nucleotide SEQ ID NO:X disclosed in Table 1, column 5.
Table 4, column 2, provides the chromosomal location, "Cytologic Band or
Chromosome," of polynucleotides corresponding to SEQ ID NO:X. Chromosomal
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
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
location of a Morbid Map entry, the OMIM reference identification number of
the
morbid map entry is provided in Table 4, column 3, labelled "OMIM
Reference(s)."
A key to the OMIM reference identification numbers is provided in Table 6.
Table 5 provides a key to the Library Code disclosed in Table 3. Column 1
5 provides the Library Code disclosed in Table 3, column 2. Column 2 provides
a
description of the tissue or cell source from which the corresponding library
was
derived. Library codes corresponding to diseased Tissues are indicated in
column 3
with the word "disease".
Table 6 provides a key to the OMIM reference identification numbers
10 disclosed in Table 4, 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
15 URL: http://www.ncbi.nlm.nih.gov/omim~. Column 2 provides diseases
associated
with the cytologic band disclosed in Table 4, column 2, as determined using
the
Morbid Map database.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
76
Table 2
. . . . : , ., .; , ".
.
1 11 61 1 - 15 - Gly-85 to Gly-96
1684 1698
Pro-162 to Val-168
Pro-177to Gly-188
Arg-254 to Asp-260
Leu-283 to Asp-293
Glu-310 to Gly-318
Arg-338 to Arg-345
Ala-387 to Leu-392
Gly-398 to Gln-404
His-464 to T -475.
1 33 83 1 - 15 - Gly-85 to Gly-96
1696 1710
Glu-163 to Val-168
Pro-177 to Gly-188
Arg-254 to Asp-260
Leu-283 to Asp-293
Glu-310 to Gly-318.
2 12 62 1 - 15 - Ala-27 to Glu-33
1475 1489
Asp-57 to Glu-69
Asn-134 to Thr-143
Met-185 to Arg-192
Ile-221 to Leu-234
Thr-249 to Asp-260
Ser-270 to Asp-275
Pro-366 to Gly-376.
2 34 84 1 - 15 - Ala-27 to Glu-33
1532 1546
Asp-57 to Glu-69
Asn-134 to Thr-143
Met-185 to Arg-192
Ile-221 to Leu-234
Thr-249 to Asp-260
Ser-270 to Asp-275
Pro-366 to Gl -376.
3 13 63 1 - 15 - Gly-25 to Leu-30
965 979
Pro-40 to Ser-49
Pro-74 to Ser-91
Asn-97 to Cys-104
Pro-115 to Phe-123
Ser-125 to Ser-132.
4 14 64 1 - 15 - Gly-100 to Pro-105
1079 1093
Pro-111 to Lys-116.
4 35 85 1 - 15 -
1590 1604
15 65 1 - 15 - Ser-53 to Thr-59
2628 2642
Lys-65 to Glu-70
Pro-80 to Gly-91
Ser-94 to Pro-100
Ala-116 to Asn-124
Pro-135 to Trp-151
Asn-172 to Phe-189
Asn-191 to Trp-196
Pro-201 to Trp-223

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
77
Gly-234 to Gly-24S
Thr-2S1 to Asn-260
Arg-340 to Lys-3S7
Gly-428 to Tyr-4S4
Phe-458 to Tyr-464.
S 36 86 1 - 1S - Ser-S3 to Thr-59
743 7S7
Lys-6S to Glu-70
Pro-80 to Gly-91
Ser-94 to Pro-100
Ala-116 to Asn-124
Pro-l3Sto Trp-151
Asn-172 to Phe-189
Asn-191 to T -196.
S 37 87 1 - 1 S His-1 to His-18
793 - 807
Pro-36 to Gly-44.
6 16 66 1 - 1S - Pro-28 to Asp-41
681 69S
Thr-107 to Tyr-114
Glu-1S8 to Pro-164.
6 38 88 1 - 1S - Pro-28 to As -41.
1504 1518
7 17 67 1 - 1 S Met-1 to Ala-11
2507 - 2521
Leu-20 to Gly-26
Leu-S 1 to Ser-61
Phe-82 to Leu-87
Gln-94 to Pro-99.
7 39 89 1 - 1S - Met-1 to Ala-11
682 696
Leu-20 to Gly-26
Leu-S 1 to Ser-61
Phe-82 to Leu-87
Gln-94 to Pro-99.
7 40 90 1 - 1S - Gly-1 to Asn-14
1383 1397
Lys-80 to Phe-88
Gly-139 to Lys-147
Gly-174 to Thr-179
Ala-198 to Ser-208
Asn-210 to Leu-223
Gln-226 to Arg-2S3
Ser-260 to Asn-270
Ser-289 to Asp-296
Met-303 to Pro-310
Arg-332 to Thr-340
Asn-3S3 to Asn-361.
8 18 68 1 - 1S - His-8 to Met-13.
1262 1276
8 41 91 1 - 1S - His-8 to Met-13.
1309 1323
9 19 69 1 - 1 S Asn-71 to Trp-76.
1201 - 1215
9 42 92 1 - 1 S Asn-71 to Trp-76.
799 - 813
20 70 1 - 1 S
161 - 1629
S
10 43 93 1 - 15 - Ser-42 to Asn-S0.
803 817
10 44 94 1 - 1 S Ser-8 to Glu-14
629 - 643
Tyr-23 to Ala-28
Glu-42 to Trp-S
1
Glu-107 to Glu-121.
11 21 71 1 - 1S - Ar -128 to Tyr-134.
1001 lOlS
11 4S 95 1 - 1S -
992 1006
12 22 72 1 - 1 S Gly-4 to Glu-9
1226 - 1240
Asp-22 to Cys-28
Glu-39 to Leu-44

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
78
Phe-88 to Phe-94.
12 46 96 1 - 15 - Gly-4 to Glu-9.
633 647
12 47 97 1 - 15 - Gly-1 to Glu-8
1307 1321
Gly-37 to Gly-61
Gln-71 to Phe-81
Asp-95 to Gly-103
Leu-126 to Ile-131
Val-166 to Glu-171.
13 23 73 1 - 15 - Ser-34 to Ile-39.
555 569
13 48 98 1 - 15 - Ser-34 to Ile-39
711 725
Leu-120 to Glu-128.
14 24 74 1 - 15 - Pro-39 to Leu-44
1240 1254
Ghl-80 to Pro-93
Pro-153 to Pro-158.
14 49 99 1 - 15 - Pro-39 to Leu-44
. 855 869
Gln-80 to Pro-93
Pro-153 to Pro-158.
14 50 100 1 - 15 - Pro-12 to His-25.
678 692
15 25 75 1 - 15 - Glu-37 to Trp-42
1970 1984
Phe-67 to Gly-88
Pro-101 to Leu-110.
15 51 101 1 - 15 - Glu-37 to Trp-42.
451 465
16 26 76 1 - 15 - Pro-45 to Trp-50
2086 2100
Pro-76 to Trp-93.
16 52 102 1 - 15 - Pro-45 to Trp-50
532 546
Pro-76 to T -93.
17 27 77 1 - 15 - Val-52 to Trp-61
1122 1136
Leu-65 to Gly-71
Lys-86 to Leu-92.
17 53 103 1 - 15 - Val-52 to Trp-61
1847 1861
Leu-65 to Gly-71
Lys-86 to Leu-92.
18 28 78 1 - 15 - Leu-66 to Lys-71
2265 2279
Arg-92 to Trp-97
Lys-115 to Gly-122
Pro-182 to Glu-192
Ala-207 to Glu-212
Asn-221 to Val-227
Leu-234 to Lys-239
Cys-250 to Arg-255
Asp-260 to Ala-266
Pro-336 to Pro-349
Ser-387 to Arg-412.
18 54 104 1 - 15 - Leu-52 to Lys-57
870 884
Arg-78 to Trp-83
Lys-101 to Gly-108
Pro-168 to Glu-178.
19 29 79 1 - 15 - Thr-45 to Leu-54
1579 1593
Gln-71 to Asn-77
Glu-90 to Ile-98
Lys-111 to Thr-122
Lys-131 to Lys-140
Gly-153 to Ala-161
Arg-169 to Ser-178
Thr-184 to Gln-198
Glu-214 to Glu-220

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
79
Thr-234 to Tyr-246.
19 55 105 1 - 15 - Thr-45 to Leu-54
558 572
Gln-71 to Asn-77
Glu-90 to Ile-98.
20 30 80 1 - 15 - Pro-35 to Ser-49
2652 2666
Thr-85 to Ile-92
Pro-95 to Glu-I03
Ser-110 to Thr-130
Val-142 to Gly-150
Glu-152 to Ala-158
Gln-173 to Leu-183
Gly-238 to Leu-245
Gly-272 to His-282
Arg-298 to Met-308
Ala-318 to Gly-339
Ala-353 to Leu-360
Pro-362 to Tyr-367
Pro-372 to Asp-380
Glu-390 to Ser-414
Ile-416 to Arg-424
Ser-429 to Gln-439
Gly-472 to Thr-480.
20 56 106 1 - 15 - Asn-64 to Thr-70
450 464
Glu-103 to Pro-110.
20 57 107 1 - 15 - Ser-1 to Arg-12
715 729
Ser-17 to Gln-27
Gly-60 to Thr-68.
21 31 81 1 - 15 -
2687 2701
21 58 108 1 - 15 - Thr-17 to Cys-23.
1311 1325
21 59 109 1 - 15 - Thr-17 to Gys-23
2864 2878
Gln-76 to Ser-81.
22 32 82 1 - 15 - Thr-35 to Gly-48.
514 528
22 60 ~ 110 1 - 15 - Thr-35 to Gly-48.
~ 496 S I
O

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
Table 3
Clone Librar Code s
ID
HLDAB75 H0509
HDPGT25 H0038 H0042 H0051 H0124 H0163 H0170 H0318
H0328 H0341
H0355 H0376 H0400 H0416 H0427 H0428 H0438
H0441 H0445
H0484 H0486 H0510 H0521 H0522 H0539 H0546
H0551 H0552
H0553 H0555 H0574 H0586 H0591 H0615 H0616
H0619 H0624
H0638 H0639 H0641 H0644 H0648 H0659 H0662
H0663 H0676
H0683 H0689 H0696 H0711 L1290 S0002 50010
50031 50051 50052
50142 50144 50242 50276 S0278 50300 50328
S0330 50344 S0346
50358 S0360 S0388 S0418 50420 50422 50426
50444 50460 50474
T0003 T0004 T0006 T0041 T0049
HEEBI05 H0150 H0549 L1290
HLTIP27 H0040 H0050 H0087 H0090 H0255 H0318 H0506
H0521 H0522
H0581 H0591 H0597 H0599 H0640 H0650 H0672
H0689 H0710
L1290 50031 S0116 50142 S0358 50360 50378
50426 50458 T0023
HAHFU44 H0333 H0520 H0539 H0599 H0622 L1290 S0312
50330
HNKC080 H0032 H0096 H0263 H0402 H0478 H0542 H0593
H0696 L1290
50150 50328 50330 50380 S0392
HEEBB55 H0038 H0090 H0120 H0135 H0144 H0166 H0170
H0310 H0331
H0423 H0494 H0506 H0519 H0520 H0538 H0543
H0545 H0549
H0616 H0632 H0657 H0661 H0673 L1290 50016
50051 50310 S0358
S0364 S0420 S0448 56024
HOCPM23 H0660 L1290
HWMGN33 L1290 S0358
HWMLN52 H0393 H0551 H0638 L1290 S0358
HVARW53 50378 S0380
HLTIP94 H0170 H0591 56026
HEGCLll H0267 H0457 H0497 H0550 H0586 L1290 50028
S0152 50344
HCOOS80 H0014 H0033 H0052 HO100 H0290 H0333 H0352
H0402 H0436
H0521 H0543 H0545 H0546 H0550 H0574 H0580
H0618 H0643
H0660 L1290 50280 50354 50404 50406 50418
HLCND09 H0009 HO100 H0123 H0144 H0179 H0266 H0271
H0327 H0435
H0438 H0545 H0551 H0575 H0594 H0615 H0628
H0696 L1290
S0007 S0028 S0038 S0210 50212 50300 S0376
50418 50420 50464
HLWBT09 H0031 H0553 H0620 L1290
HKAEL28 H0255 H0494 H0556 H0587 H0618 L1290 S0132
HNTPB82 H0003 H0059 H0081 H0090 H0144 H0265 H0428
H0543 H0547
H0555 H0594 H0599 H0600 H0662 H0670 H0684
L1290 50046 50050
50344 50358 50406 50408 50444 S0462
HOFMM69 H0415
HI~ACC80 H0013 H0039 H0494 H0586 H0644 L1290 50037
S0346 S0354 S0356
50420
HHEDN80 H0036 H0038 H0042 HO100 H0130 H0267 H0272
H0486 H0542
H0616 H0625 H0634 H0637 H0666 L1290 S0051
50212 50214 50222
50276 50346 50358 50430 53012
HPDWP28 H0658 L1290
~

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
81
Table 4
SEQ ID Cytologic Band OMIM
or Reference(s):
NO: X Chromosome:
11 l9cen- 13.2
23 6p21.3 106300108800120290120810120820142857
142858150270167250170261177900179450
201910217000222100233100235200248611
256550600202600261601868602280602475
24 17

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
82
Table 5
Library Library Description Disease
Code
H0003 Human Adult Liver
H0009 Human Fetal Brain
H0013 Human 8 Week Whole Embryo
H0014 Human Gall Bladder
H0031 Human Placenta
H0032 Human Prostate
H0033 Human Pituitary
H0036 Human Adult Small Intestine
H0038 Human Testes
H0039 Human Pancreas Tumor disease
H0040 Human Testes Tumor disease
H0042 Human Adult Pulmonary
H0050 Human Fetal Heart
H0051 Human Hi ocam us
H0052 Human Cerebellum
H0059 Human Uterine Cancer disease
H0081 Human Fetal Epithelium (Skin)
H0087 Human Thymus
H0090 Human T-Cell Lymphoma disease
H0096 Human Parotid Cancer disease
HO100 Human Whole Six Week Old Embryo
H0120 Human Adult S Teen, subtracted
H0123 Human Fetal Dura Mater
H0124 Human Rhabdomyosarcoma disease
H0130 LNCAP untreated
H0135 Human Synovial Sarcoma
HOI44 Nine Week Old Early Stage Human
HO150 Human E ididymus
H0163 Human Synovium
H0166 Human Prostate Cancer, Stage B2 fractiondisease
H0170 12 Week Old Early Stage Human
H0179 Human Neutrophil
H0255 breast lymph node CDNA library
H0263 human colon cancer disease
H0265 Activated T-Cell (l2hs)/Thiouridine
labelledEco
H0266 Human Microvascular Endothelial Cells,
fract. A
H0267 Human Microvascular Endothelial Cells,
fract. B
H0271 Human Neutrophil, Activated
H0272 HUMAN TONSILS, FRACTION 2
H0290 Human OB HOS txeated (1 nM E2) fraction
I
H0310 human caudate nucleus
H0318 HUMAN B CELL LYMPHOMA disease
H0327 human co us colosum
H0328 human ovarian cancer disease
H0331 Hepatocellular Tumor disease
H0333 Hemangiopericytoma disease
H034I Bone Marrow Cell Line (RS4,11)

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
83
H0352 wilm's tumor disease
H0355 Human Liver
H0376 Human S leen
H0393 Fetal Liver, subtraction II
H0400 Human Striatum De ression, re-rescue
H0402 CD34 de feted Buffy Coat Cord Blood),
re-excision
H0415 H. Ovarian Tumor, II, OV5232 disease
H0416 Human Neutro hits, Activated, re-excision
H0423 T-Cell PHA 24 hrs
H0427 Human Adipose
H0428 Human Ovary
H0435 Ovarian Tumor 10-3-95
H0436 Resting T-Cell Library,II
H0438 H. Whole Brain#2, re-excision
H0441 H. Kidney Cortex, subtracted
H0445 S Teen, Chronic lym hocytic leukemiadisease
H0457 Human Eosino hits
H0478 Salivary Gland, Lib 2
H0484 Breast Cancer Cell line, an iogenic
H0486 Hodgkin's Lymphoma II ~ disease
H0494 Keratinocyte
H0497 HEL cell line
H0506 Ulcerative Colitis
H0509 Liver, He atoma disease
H0510 Human Liver, normal
H0519 NTERA2, control
H0520 NTERA2 + retinoic acid, 14 days
H0521 Primary Dendritic Cells, lib 1
H0522 Primary Dendritic cells,frac 2
H0538 Merkel Cells
H0539 Pancreas Islet Cell Tumor disease
H0542 T Cell hel er I
H0543 T cell hel er II
H0545 Human endometrial stromal cells-treated
with progesterone
H0546 Human endometrial stromal cells-treated
with estradiol
H0547 NTERA2 teratocarcinoma cell line+retinoic
acid (14 days)
H0549 H. Epididiymus, caput & corpus
H0550 H. E ididi us, cauda
H0551 Human Thymus Stromal Cells
H0552 Si nal tra ,Femur Bone Marrow,pooled
H0553 Human Placenta
H0555 Rejected Kidney, lib 4 ~ disease
H0556 Activated T-cell(12h)/Thiouridine-re-excision
H0574 He atocellular Tumor, re-excision disease
H0575 Human Adult Pulmonary,re-excision
H0580 Dendritic cells, poled
H0581 Human Bone Marrow, treated
H0586 Healing groin wound, 6.5 hours post disease
incision
H0587 Healing groin wound, 7.5 hours post disease
incision
H0591 Human T-cell lym homa,re-excision disease
H0593 Olfactory a ithelium,nasalcavity
H0594 Human Lung Cancer,re-excision disease
H0597 Human Colon, re-excision

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
84
H0599 Human Adult Heart,re-excision
H0600 Healing Abdomen wound,70&90 min ost disease
incision
H06I5 Human Ovarian Cancer Reexcision disease
H0616 Human Testes, Reexcision
H0618 Human Adult Testes, Large Inserts,
Reexcision
H0619 Fetal Heart
H0620 Human Fetal Kidney, Reexcision
H0622 Human Pancreas Tumor, Reexcision disease
H0624 12 Week Early Stage Human II, Reexcision
H0625 Ku 812F Baso hits Line
H0628 Human Pre-Differentiated Adi ocytes
H0632 He atocellular Tumor,re-excision
H0634 Human Testes Tumor, re-excision disease
H0637 Dendritic Cells From CD34 Cells
H0638 CD40 activated monocyte dendridic
cells
H0639 Ficolled Human Stromal Cells, 5Fu
treated
H0640 Ficolled Human Stromal Cells, Untreated
H0641 LPS activated derived dendritic cells
H0643 He G2 Cells, PCR library
H0644 Human Placenta (re-excision)
H0648 Ovary, Cancer: (4004562 B6) Papillarydisease
Serous Cystic
Neo lasm, Low Malignant Pot
H0650 B-Cells
H0657 B-cells (stimulated)
H0658 Ovary, Cancer (9809C332): Poorly differentiateddisease
adenocarcinoma
H0659 Ovary, Cancer (15395A1F): Grade II disease
Papillary Carcinoma
H0660 Ovary, Cancer: (15799A1F) Poorly differentiateddisease
carcinoma
H0661 Breast, Cancer: (4004943 AS) disease
H0662 Breast, Normal: (400S522B2)
H0663 Breast, Cancer: (4005522 A2) disease
H0666 Ovary, Cancer: (4004332 A2) disease
H0670 Ovary, Cancer(4004650 A3): Well-Differentiated
Micro a illary Serous Carcinoma
H0672 Ovary, Cancer: (4004576 A8)
H0673 Human Prostate Cancer, Sta a B2, re-excision
H0676 Colon, Cancer: (9808C064R)-total RNA
H0683 Ovarian cancer, Serous Papillary Adenocarcinoma
H0684 Ovarian cancer, Serous Pa illary Adenocarcinoma
H0689 Ovarian Cancer
H0696 Prostate Adenocarcinoma
H0710 Patient #6 Acute Myeloid Leukemia/SGAH
H0711 Ovarian Cancer Cell Line(Xenograft)
ES-2
L1290 NCI CGAP Sub4
S0002 Monocyte activated
S0007 Early Stage Human Brain
S0010 Human Am gdala
50016 Kidney Pyramids
S0028 Smooth muscle,control
S0031 S final cord
50037 Smooth muscle, ILlb induced
50038 Human Whole Brain #2 - Oligo dT >
l.SKb
50046 Endothelial-induced
SOOSO Human Frontal Cortex, Schizophrenia disease

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
50051 Human H othalmus,Schizo hrenia disease
50052 neutro hits control
50116 Bone marrow
S0132 E ithelial-TNFa and INF induced
50142 Macro hage-oxLDL
50144 Macro ha a (GM-CSF treated)
SO150 LNCAP prostate cell line
50152 PC3 Prostate cell line
S0210 Messangial cell, frac 2
50212 Bone Marrow Stromal Cell, untreated
50214 Human Osteoclastoma, re-excision disease
50222 H. Frontal cortex,e ile tic,re-excisiondisease
50242 Synovial Fibroblasts (Ill/TNF), subt
S0276 Synovial h oxia-RSF subtracted
S0278 H Macro huge (GM-CSF treated), re-excision
50280 Human Adi ose Tissue, re-excision
50300 Frontallobe,dementia,re-excision
50310 Normal trachea
50312 Human osteoarthritic,fraction II disease
S0328 Palate carcinoma disease
S0330 Palate normal
S0344 Macrophage-oxLDL, re-excision
50346 Human Amygdala,re-excision
S0354 Colon Normal II
50356 Colon Carcinoma disease
50358 Colon Normal III
50360 Colon Tumor II disease
50364 Human Quadrice s
50376 Colon Tumor disease
50378 Pancreas normal PCA4 No
S0380 Pancreas Tumor PCA4 Tu disease
50388 Human H othalamus,schizophrenia, re-excisiondisease
S0392 Salivary Gland
50404 Rectum normal
50406 Rectum tumour
50408 Colon, normal
50418 CHME Cell Line,treated 5 hrs
50420 CHME Cell Line,untreated
50422 Mo7e Cell Line GM-CSF treated (lng/ml)
50426 Monocyte activated, re-excision
50430 Aryepiglottis Normal
50444 Colon Tumor disease
S0448 Larynx Normal
50458 Thyroid Normal (SDCA2 No)
S0460 Thyroid Tumour
50462 Thyroid Thyroiditis
50464 Larynx Normal
50474 Human blood latelets _
53012 Smooth Muscle Serum Treated, Norm
56024 Alzheimers, spongy chan a disease
56026 Frontal Lobe, Dementia
T0003 Human Fetal Lun
T0004 Human White Fat

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
86
T0006 Human Pineal Gland
T0023 Human Pancreatic Carcinoma disease
T0041 Jurkat T-cell G1 hase
T0049 Aorta endothelial cells + TNF-a

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
87
Table 6
OMIM Description
Reference
106300 Ankylosin s ondylitis
108800 Atrial se tal defect, secundum a
120290 OSMED syndrome, 215150
Stickler syndrome, t a II, 184840
120810 C4 deficiency
120820 C4 deficiency
142857 Pem hi oid, susce tibility to
142858 Beryllium disease, chronic, susceptibility
to
150270 Laryngeal adductor paralysis
167250 Paget disease of bone
170261 Bare lym hocyte syndrome, a I, due to TAP2
deficiency
177900 Psoriasis susce tibility-1
179450 Ragweed sensitivity
201910 Adrenal h erplasia, con enital, due to 21-hydroxylase
deficiency
217000 C2 deficiency
222100 Diabetes mellitus, insulin-dependent-1
233100 [Renal lucosuria]
235200 Hemochromatosis
248611 Maple syru urine disease, a Ib
256550 Sialidosis, type I
Sialidosis, a II
600202 Dyslexia, specific, 2
600261 Ehlers-Danlos-like s drome
601868 Deafness, autosomal dominant 13
602280 Retinitis pigmentosa-14, 600132
602475 Ossification of posterior longitudinal ligament
of spine

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
88
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 Seguences
The present invention also encompasses mature forms of the polypeptide
having the polypeptide sequence of SEQ ID NO:Y and/or the polypeptide sequence
encoded by the cDNA in a deposited clone. Polynucleotides encoding the mature

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
89
forms (such as, for example, the polynucleotide sequence in SEQ m NO:X and/or
the
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
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 unifoim, 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
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
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
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
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.
As one of ordinary skill would appreciate, however, cleavage sites sometimes
vary from organism to organism and cannot be predicted with absolute
certainty.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
Accordingly, the present invention provides secreted polypeptides having a
sequence
shown in SEQ m 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
5 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
10 naturally occurring 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
15 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
20 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 m NO:Y andlor encoded by a deposited clone.
25 "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
30 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 m NO:X or the complementary strand thereto, the

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
91
nucleotide coding sequence contained in a deposited cDNA clone or the
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°l0, 90%,
95%, 96%, 97%, 98%, 99% identical to, for example, the polypeptide sequence
shown in SEQ ff~ NO:Y, the polypeptide sequence encoded by the cDNA contained
in a deposited clone, and/or 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 l, 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
92
using the FASTDB computer program based on the algorithm of Brutlag et al.
(Comp.
App. Biosci. 6:237-245(1990)). In a sequence alignment the query and subject
sequences are both DNA sequences. An RNA sequence can be compared by
converting U's to T's. The result of said global sequence alignment is in
percent
identity. Preferred parameters used in a FASTDB alignment of DNA sequences to
calculate percent identiy are: Matrix=Unitary, k-tuple=4, Mismatch Penalty=1,
Joining Penalty=30, Randomization Group Length=0, Cutoff Score=1, Gap
Penalty=5, Gap Size Penalty 0.05, Window Size=500 or the lenght of the subject
nucleotide sequence, whichever is shorter.
If the subject sequence is shorter than the query sequence because of 5' or 3'
deletions, not because of internal deletions, a manual correction must be made
to the
results. This is because the FASTDB program does not account for 5' and 3'
truncations of the subject sequence when calculating percent identity. For
subject
sequences truncated at the 5' or 3' ends, relative to the query sequence, the
percent
identity is corrected by calculating the number of bases of the query sequence
that are
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 arnve 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 subj ect 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
93
subject sequence is compared with a 100 base query sequence. This time the
deletions axe 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 subj ect sequence which are not matchedlaligned 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 m 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,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
94
Window Size=sequence length, Gap Penalty=5, Gap Size Penalty=0.05, Window
Size=500 or the length of the subject amino acid sequence, whichever is
shorter.
If the subject sequence is shorter than the query sequence due to N- or C-
terminal deletions, not because of internal deletions, a manual correction
must be
made to the results. This is because the FASTDB program does not account for N-
and C-terminal truncations of the subject sequence when calculating global
percent
identity. For subj ect 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 subj ect residue, as a percent of the
total
bases of the query sequence. Whether a residue is matched/aligned is
determined by
results of the FASTDB sequence alignment. This percentage is then subtracted
from
the percent identity, calculated by the above FASTDB program using the
specified
parameters, to arrive at a final percent identity score. This final percent
identity score
is what is used for the purposes of the present invention. Only residues to
the N- and
C-termini of the subject sequence, which are not matched/aligned with the
query
sequence, are considered for the purposes of manually adjusting the percent
identity
score. That is, only query residue positions outside the farthest N- and C-
terminal
residues of the subject sequence.
For example, a 90 amino acid residue subj ect sequence is aligned with a 100
residue query sequence to determine percent identity. The deletion occurs at
the N-
terminus of the subject sequence and therefore, the FASTDB alignment does not
show a matching/alignment of the first 10 residues at the N-terminus. The 10
unpaired residues represent 10% of the sequence (number of residues at the N-
and C-
termini not matched/total number of residues in the query sequence) so 10% is
subtracted from the percent identity score calculated by the FASTDB program.
If the
remaining 90 residues were perfectly matched the final percent identity would
be
90%. In another example, a 90 residue subject sequence is compared with a 100
residue query sequence. This time the deletions are internal deletions so
there are no
residues at the N- or C-termini of the subject sequence which are not
matched/aligned
with 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
ends of the subject sequence, as displayed in the FASTDB alignment, which are
not
matched/aligned with the query sequnce are manually corrected for. No other
manual
corrections are to made for the purposes of the present invention.
The variants may contain alterations in the coding regions, non-coding
S 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
10 added in any combination are also preferred. Polynucleotide variants can be
produced
for a variety of reasons, e.g., to optimize colon expression for a particular
host
(change colons in the human mRNA to those preferred by a bacterial host such
as E.
coli).
Naturally occurnng variants are called "allelic variants," and refer to one of
15 several alternate forms of a gene occupying a given locus on a chromosome
of an
organism. (Genes II, Lewin, B., el., 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.
20 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
25 (1993), reported variant KGF proteins having heparin binding activity even
after
deleting 3, 8, or 27 amino-terminal amino acid residues. Similarly, Interferon
gamma
exhibited up to ten times higher activity after deleting 8-10 amino acid
residues from
the carboxy terminus of this protein. (Dobeli et al., J. Biotechnology 7:199-
216
(1988).)
30 Moreover, ample evidence demonstrates that vaxiants often 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
96
analysis of human cytokine IL-la. They used random mutagenesis to generate
over
3,500 individual IL-la mutants that averaged 2.5 amino acid changes per
variant over
the entire length of the molecule. Multiple mutations were examined at every
possible amino acid position. The investigators found that "[m]ost of the
molecule
could be altered with little effect on either [binding or biological
activity]." (See,
Abstract.) In fact, only 23 unique amino acid sequences, out of more than
3,500
nucleotide sequences examined, produced a protein that significantly differed
in
activity from wild-type.
Furthermore, 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 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
97
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 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
98
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 Carner 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 Polyue~tide 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 m NO:X or the complementary strand
thereto, or is a portion of a polynucleotide sequence encoding the polypeptide
of SEQ
m 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 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
99
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
NO:X. hi 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.
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, 70I-
750,
751-800, 800-850, 851-900, 901-950, 951-1000, 1001-1050, 1051-1100, 1101-1150,
1151-1200, 1201-1250, 1251-1300, 1301-1350, 1351-1400, 1401-1450, 1451-1500,
1501-1550, 1551-1600, 1601-1650, 1651-1700, 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
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
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
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 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
100
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
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 m NO:Y falling within conserved domains are
specifically contemplated by the present invention. Moreover, polynucleotides
encoding these domains are also contemplated.
Qther 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 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
101
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 diffusionw
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 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
102
chromatography, protein affinity chromatography, and affinity blotting. See
generally, Phiziclcy, 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 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
103
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 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
104
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 ~ 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 carrier, such as keyhole limpet hemacyanin (KL,H) 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 earner- coupled peptides, for instance, by intraperitoneal
and/or
intradermal injection of emulsions containing about 100 ~,g ofpeptide or
carrier
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 (e.g., those comprising an immunogenic
or
antigenic epitope) can be fused to heterologous polypeptide sequences. For
example,
polypeptides of the present invention (including fragments or variants
thereof), 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,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
105
resulting in chimeric polypeptides. By way of another non-limiting example,
polypeptides and/or antibodies of the present invention (including fragments
or
variants thereof] may be fused with albumin (including but not limited to
recombinant
human serum albumin or fragments or variants thereof (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)).
In a preferred embodiment, polypeptides and/or antibodies of the present
invention
(including fragments or variants thereof) are fused with the mature form of
human
serum albumin (i.e., amino acids 1- 585 of human serum albumin as shown in
Figures 1 and 2 of EP Patent 0 322 094) which is herein incorporated by
reference in
its entirety. In another preferred embodiment, polypeptides and/or antibodies
of the
present invention (including fragments or variants thereof) are fused with
polypeptide
fragments comprising, or alternatively consisting of, amino acid residues 1-z
of
human serum albumin, where z is an integer from 369 to 419, as described in
U.S.
Patent 5,766,883 herein incorporated by reference in its entirety.
Polypeptides and/or
antibodies of the present invention (including fragments or variants thereof)
may be
fused to either the N- or C-terminal end of the heterologous protein (e.g:,
immunoglobulin Fc polypeptide or human serum albumin polypeptide).
Polynucleotides encoding fusion proteins of the invention are also encompassed
by
the invention.
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
106
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;
5,834,252; and 5,837,458, and Patten et al., Curr. Opinion Biotechnol. 8:724-
33 1
(1997); Harayama, Trends Biotechnol. 16(2):76-82 (1998); Hansson, et al., J.
Mol.
Biol. 287:265-76 (1999); and Lorenzo and Blasco, Bioteclmiques 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 subj ected 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,
domains, fragments, etc. of one or more heterologous molecules.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
107
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
molecules of the invention are IgGl . 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,
CHl, CH2, and CH3 domains. The antibodies of the invention may be from any
animal origin including birds and mammals. Preferably, the antibodies are
human,
marine (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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
108
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 I~ucherlapati 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
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
109
invention axe 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
I O constant or Kd less than S X 10-z M, 10-z M, S X 10-3 M, I O-3 M, S X 10-4
M, 10-4 M,
S X 10-s M, I O-s M, S X 10-G M, 10-6M, 5 X 10-' M, 10' M, S X 10-8 M, 10-g M,
S X
10-9 M, 10-9 M, S X 10-1° M, IO-r° M, S X 10-11 M, 10-11 M, S X
10-lz M, io-iz M, S X
10-13 M, 10-is M, S X 10-14 M, I0-14 M, 5 X 10-is M, or 10-is M.
The invention also provides antibodies that competitively inhibit binding of
an
1 S antibody to an epitope of the invention as determined by any method known
in the art
for determining competitive binding, for example, the immunoassays described
herein. In preferred embodiments, the antibody competitively inhibits binding
to the
epitope by at least 9S%, at least 90%, at least 8S %, at least 80%, at least
75%, at least
70%, at least 60%, or at least SO%.
20 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
25 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
30 (e.g., tyrosine or serine/threonine) of the receptor or its substrate by
immunoprecipitation followed by western blot analysis (for example, as
described
supra). In specific embodiments, antibodies are provided that inhibit ligand
activity

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
110
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 axe
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.
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. Tm_m__unol. 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.,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
111
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 to a cellular ligand or other protein, etc. Any
of
numerous chemical modifications may be carried out by known techniques,
including, but not limited to specific chemical cleavage, acetylation,
formylation,
metabolic synthesis of tunicamycin, etc. Additionally, the derivative may
contain
one or more non-classical amino acids.
The antibodies of the present invention may be generated by any suitable
method known in the art. Polyclonal antibodies to an antigen-of interest can
be
produced by various procedures well known in the art. For example, a
polypeptide of
the invention can be administered to various host animals including, but not
limited
to, rabbits, mice, rats, etc. to induce the production of sera containing
polyclonal
antibodies specific for the antigen. Various adjuvants may be used to increase
the
immunological response, depending on the host species, and include but are not
limited to, Freund's (complete and incomplete), mineral gels such as aluminum
hydroxide, surface active substances such as lysolecithin, pluronic polyols,
polyanions, peptides, oil emulsions, keyhole limpet hemocyanins,
dinitrophenol, and

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
112
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
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
113
hybridomas resulting from the fusion for hybridoma clones that secrete an
antibody
able to bind a polypeptide of the invention.
Antibody fragments which recognize specific epitopes may be generated by
known techniques. For example, Fab and F(ab')2 fragments of the invention may
be
produced by proteolytic cleavage of immunoglobulin molecules, using enzymes
such
as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
F(ab')2 fragments contain the variable region, the light chain constant region
and the
CH1 domain of the heavy chain.
For example, the antibodies of the present invention can also be generated
using various phage display methods known in the art. In phage display
methods,
functional antibody domains are displayed on the surface of phage particles
which
carry the polynucleotide sequences encoding them. In a particular embodiment,
such
phage can be utilized to display antigen binding domains expressed from a
repertoire
or combinatorial antibody library (e.g., human or murine). Phage expressing an
antigen binding domain that binds the antigen of interest can be selected or
identified
with antigen, e.g., using labeled antigen or antigen bound or captured to a
solid
surface or bead. Phage used in these methods are typically filamentous phage
including fd and M13 binding domains expressed from phage with Fab, Fv or
disulfide stabilized Fv antibody domains recombinantly fused to either the
phage
gene III or gene VIII protein. Examples of phage display methods that can be
used to
make the antibodies of the present invention include those disclosed in
Brinkman et
al., J. Immunol. Methods 182:41-50 (1995); Ames et al., J. Immunol. Methods
184:177-186 (1995); Kettleborough et al., Eur. J. Imnnunol. 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,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
114
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 Enzyrnology 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.
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
115
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
98124893, WO 98116654, WO 96134096, WO 96133735, and WO 91/10741; each of
which is incorporated herein by reference in its entirety.
Human antibodies can also be produced using transgenic mice which are
incapable of expressing functional endogenous immunoglobulins, but which can
express human immunoglobulin genes. For example, the human heavy and light
chain immunoglobulin gene complexes may be introduced randomly or by
homologous recombination into mouse embryonic stem cells. Alternatively, the
human variable region, constant region, and diversity region may be introduced
into
mouse embryonic stem cells in addition to the human 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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
116
Thus, using such a technique, it is possible to produce therapeutically useful
IgG, IgA,
IgM and IgE antibodies. For an overview of this technology for producing human
antibodies, see Lonberg and Huszar, Int. Rev. Immunol. 13:65-93 (1995). For a
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
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
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.
Polyraucleotides En.codirag Antibodies

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
117
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 ID NO:Y.
The polynucleotides may be obtained, and the nucleotide sequence of the
polynucleotides determined, by any method known in the art. For example, if
the
nucleotide sequence of the antibody is known, a polynucleotide encoding the
antibody
may be assembled from chemically synthesized oligonucleotides (e.g., as
described .
in Kutmeier et al., BioTechniques 17:242 (1994)), which, briefly, involves the
synthesis of overlapping oligonucleotides containing portions of the sequence
encoding the antibody, annealing and ligating of those oligonucleotides, and
then
amplification of the ligated oligonucleotides by PCR.
Alternatively, a polynucleotide encoding an antibody may be generated from
nucleic acid from a suitable source. If a clone containing a nucleic acid
encoding a
particular antibody is not available, but the sequence of the antibody
molecule is
known, a nucleic acid encoding the immunoglobulin may be chemically
synthesized
or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA
library
generated from, or nucleic acid, preferably poly A+ RNA, isolated from, any
tissue
or cells expressing the antibody, such as hybridoma cells selected to express
an
antibody of the invention) by PCR amplification using synthetic primers
hybridizable
to the 3' and 5' ends of the sequence or by cloning using an oligonucleotide
probe
specific for the particular gene sequence to identify, e.g., a cDNA clone from
a
cDNA library that encodes the antibody. Amplified nucleic acids generated by
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
118
example, the techniques described in Sambrook et al., 1990, Molecular Cloning,
A
Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor,
NY and Ausubel et al., eds., 1998, Current Protocols in Molecular Biology,
John
Wiley & Sons, NY, which are both incorporated by reference herein in their
entireties ), to generate antibodies having a different amino acid sequence,
for
example to create amino acid substitutions, deletions, and/or insertions.
In a specific embodiment, the amino acid sequence of the heavy and/or light
chain variable domains may be inspected to identify the sequences of the
complementarity determining regions (CDRs) by methods that are well know in
the
art, e.g., by comparison to known amino acid sequences of other heavy and
light
chain variable regions to determine the regions of sequence hypervariability.
Using
routine recombinant DNA techniques, one or more of the CDRs may be inserted
within framework regions, e.g., into human framework regions to humanize a non-
human antibody, as described supra. The framework regions may be naturally
occurring or consensus framework regions, and preferably human framework
regions
(see, e.g., Chothia et al., J. Mol. Biol. 278: 457-479 (1998) for a listing of
human
framework regions). Preferably, the polynucleotide generated by the
combination of
the framework regions and CDRs encodes an antibody that specifically binds a
polypeptide of the invention. Preferably, as discussed supra, one or more
amino acid
substitutions may be made within the framework regions, and, preferably, the
amino
acid substitutions improve binding of the antibody to its antigen.
Additionally, such
methods may be used to make amino acid substitutions or deletions of one or
more
variable region cysteine residues participating in an intrachain disulfide
bond to
generate antibody molecules lacking one or more intrachain disulfide bonds.
Other
alterations to the polynucleotide are encompassed by the present invention and
within
the skill of the art.
In addition, techniques developed for the production of "chimeric antibodies"
(Mornson 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
119
derived from different animal species, such as those having a variable region
derived
from a marine mAb and a human immunoglobulin constant region, e.g., humanized
antibodies.
Alternatively, techniques described for the production of single chain
antibodies (U.S. Patent No. 4,946,778; Bird, Science 242:423- 42 (1988);
Huston et
al., Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988); and Ward et al., Nature
334:544-54 (1989)) can be adapted to produce single chain antibodies. Single
chain
antibodies are formed by linking the heavy and light chain fragments of the Fv
region
via an amino acid bridge, resulting in a single chain polypeptide. Techniques
for the
assembly of functional Fv fragments in E. coli may also be used (Skerra et
al.,
Science 242:1038- 1041 (1988)).
Methods of Producing Antibodies
The antibodies of the invention can be produced by any method known in the
art for the synthesis of antibodies, in particular, by chemical synthesis or
preferably,
by recombinant expression techniques.
Recombinant expression of an antibody of the invention, or fragment,
derivative or analog thereof, (e.g., a heavy or light chain of an antibody of
the
invention or a single chain antibody of the invention), requires construction
of an
expression vector containing a polynucleotide that encodes the antibody. Once
a
polynucleotide encoding an antibody molecule or a heavy or light chain of an
antibody, or portion thereof (preferably containing the heavy or light chain
variable
domain), of the invention has been obtained, the vector for the production of
the
antibody molecule may be produced by recombinant DNA technology using
techniques well known in the art. Thus, methods for preparing a protein by
expressing a polynucleotide containing an antibody encoding nucleotide
sequence are
described herein. Methods which are well known to those skilled in the art can
be
used to construct expression vectors containing antibody coding sequences and
appropriate transcriptional and translational control signals. These methods
include,
for example, in vitro recombinant DNA techniques, synthetic techniques, and in
vivo
genetic recombination. The invention, thus, provides replicable vectors
comprising a
nucleotide sequence encoding an antibody molecule of the invention, or a heavy
or

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
120
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. I11 preferred embodiments for the expression of double-chained
antibodies,
vectors encoding both the heavy and light chains may be co-expressed in the
host cell
for expression of the entire immunoglobulin molecule, as detailed below.
A variety of host-expression vector systems may be utilized to express the
antibody molecules of the invention. Such host-expression systems represent
vehicles by which the coding sequences of interest may be produced and
subsequently
purified, but also represent cells which may, when transformed or transfected
with
the appropriate nucleotide coding sequences, express an antibody molecule of
the
invention in situ. These include but are not limited to microorganisms such as
bacteria (e.g., E. coli, B. subtilis) transformed with recombinant
bacteriophage DNA,
plasmid DNA or cosmid DNA expression vectors containing antibody coding
sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant
yeast
expression vectors containing antibody coding sequences; insect cell systems
infected with recombinant virus expression vectors (e.g., baculovirus)
containing
antibody coding sequences; plant cell systems infected with recombinant virus
expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic
virus,
TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti
plasmid)
containing antibody coding sequences; or mammalian cell systems (e.g., COS,
CHO,
BHI~, 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
121
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 ~z 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). W
general, such
fusion proteins are soluble and can easily be purified from lysed cells by
adsorption
and binding to matrix glutathione-agarose beads followed by elution in the
presence
of free glutathione. The pGEX vectors are designed to include thrombin or
factor Xa
protease cleavage sites so that the cloned target gene product can be released
from the
GST moiety.
In an insect system, Autographa californica nuclear polyhedrosis virus
(AcNPV) is used as a vector to express foreign genes. The virus grows in
Spodoptera f-ugiperda 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).

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
122
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.
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, BHI~, 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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
123
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,
which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl.
Acad.
Sci. USA 78:2072 (1981)); neo, which confers resistance to the aminoglycoside
G-
418 Clinical Pharmacy 12:488-505; Wu and Wu, Biotherapy 3:87-95 (1991);
Tolstoshev, Ann. Rev. Pharmacol. Toxicol. 32:573-596 (1993); Mulligan, Science
260:926-932 (1993); and Morgan and Anderson, Ann. Rev. Biochem. 62:191-217
(1993); May, 1993, TIB TECH 11(5):155-215); and hygro, which confers
resistance
to hygromycin (Santerre et al., Gene 30:147 (1984)). Methods commonly known in
the art of recombinant DNA technology may be routinely applied to select the
desired
recombinant clone, and such methods are described, for example, in Ausubel et
al.
(eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY (1993);

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
124
I~riegler, Gene Transfer and Expression, A Laboratory Manual, Stoclcton 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); I~ohler, Proc. Natl. Acad.
Sci.
USA 77:2197 (1980)). The coding sequences for the heavy and light chains may
comprise cDNA or genomic DNA.
Once an antibody molecule of the invention has been produced by an animal,
chemically synthesized, or recombinantly expressed, it may be purified by any
method known in the art for purification of an immunoglobulin molecule, for
example, by chromatography (e.g., ion exchange, affinity, particularly by
affinity for
the specific antigen after Protein A, and sizing column chromatography),
centrifugation, differential solubility, or by any other standard technique
for the
purification of proteins. In addition, the antibodies of the present invention
or
fragments thereof can be fused to heterologous polypeptide sequences described
herein or otherwise known in the art, to facilitate purification.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
125
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; Dillies 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
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);

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
126
Zheng et al., J. linmunol. 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
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
127
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
urnbelliferone,
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,
and aequorin; and examples of suitable radioactive material include 125I,
131I, 11 lIn
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
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-

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
128
dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine,
propranolol, and
puromycin and analogs or homologs thereof. Therapeutic agents include, but are
not
limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-
thioguanine,
cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g.,
mechlorethamine,
thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNL~,
cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and
cis-
dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g.,
daunorubicin
(formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin
(formerly
actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic
agents (e.g., vincristine and vinblastine).
The conjugates of the invention can be used for modifying a given biological
response, the therapeutic agent or drug moiety is not to be construed as
limited to
classical chemical therapeutic agents. For example, the drug moiety may be a
protein
or polypeptide possessing a desired biological activity. Such proteins may
include,
for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or
diphtheria
toxin; a protein such as tumor necrosis factor, a-interferon,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., Iut. l~cynunol., 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"), iriterleukin-6
("IL-6"),
granulocyte macrophage colony stimulating factor ("GM-CSF"), granulocyte
colony
stimulating factor ("G-CSF"), or other growth factors.
Antibodies may also be attached to solid supports, which are particularly
useful for immunoassays or purification of the target antigen. Such solid
supports
include, but are not 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
129
et al. (eds.), pp. 243-56 (Alan R. Liss, Inc. 1985); Hellstrom et al.,
"Antibodies For
Drug Delivery", in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.),
pp.
623-53 (Marcel Dekker, Inc. 1987); Thorpe, "Antibody Carriers Of Cytotoxic
Agents
In Cancer Therapy: A Review", in Monoclonal Antibodies '84: Biological And
Clinical Applications, Pinchera et aI. (eds.), pp. 475-506 (1985); "Analysis,
Results,
And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In
Cancer Therapy", in Monoclonal Antibodies For Cancer Detection And Therapy,
Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al.,
"The
Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates", Immunol.
Rev. 62:119-58 (1982).
Alternatively, an antibody can be conjugated to a second antibody to form an
antibody heteroconjugate as described by Segal in U.S. Patent No. 4,676,980,
which
is incorporated herein by reference in its entirety.
An antibody, with or without a therapeutic moiety conjugated to it,
administered alone or in combination with cytotoxic factors) and/or
cytokine(s) can
be used as a therapeutic.
Irnnauyaophenotypihg
The antibodies of the invention may be utilized for immunophenotyping of
cell lines and biological samples. The translation product of the gene of the
present
invention may be useful as a cell specific marker, or more specifically as a
cellular
marker that is differentially expressed at various stages of differentiation
and/or
maturation of particular cell types. Monoclonal antibodies directed against a
specific
epitope, or combination of epitopes, will allow for the screening of cellular
populations expressing the marker. Various techniques can be utilized using
monoclonal antibodies to screen for cellular populations expressing the
marker(s), and
include magnetic separation using antibody-coated magnetic beads, "panning"
with
antibody attached to a solid matrix (i.e., plate), and flow cytometry (See,
e.g., U.S.
Patent 5,985,660; and Morrison et czl., 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 leulcemic patients) and "non-self' cells in
transplantations to

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
130
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
tecluziques
such as western blots, radioimmunoassays, ELISA (enzyme linked irmnunosorbent
assay), "sandwich" immunoassays, immunoprecipitation assays, precipitin
reactions,
gel diffusion precipitin reactions, immunodiffusion assays, agglutination
assays,
complement-fixation assays, immunoradiometric assays, fluorescent
immunoassays,
protein A immunoassays, to name but a few. Such assays are routine and well
known in the art (see, e.g., Ausubel et al, eds, 1994, Current Protocols in
Molecular
Biology, Vol. l, John Wiley & Sons, Inc., New York, which is incorporated by
reference herein in its entirety). Exemplary immunoassays are described
briefly
below (but are not intended by way of limitation).
T_mmunoprecipitation protocols generally comprise lysing a population of cells
in a lysis buffer such as RIPA buffer (1 % NP-40 or Triton ~- 100, 1 % sodium
20. deoxycholate, 0.1% SDS, 4.15 M NaCI, 0.01 M sodium phosphate at pH 7.2, 1%
Trasylol) supplemented with protein phosphatase and/or protease inhibitors
(e.g.,
EDTA, PMSF, aprotinin, sodium vanadate), adding the antibody of interest to
the cell
lysate, incubating for a period of time (e.g., 1-4 hours) at 4° C,
adding protein A
and/or protein G sepharose beads to the cell lysate, incubating for about an
hour or
more at 4° C, washing the beads in lysis buffer and resuspending the
beads in
SDS/sample buffer. The ability of the antibody of interest to
immunoprecipitate a
particular antigen can be assessed by, e.g., western blot analysis. One of
skill in the
art would be knowledgeable as to the parameters that can be modified to
increase the
binding of the antibody to an antigen and decrease the background (e.g., pre-
clearing
the cell lysate with sepharose beads). For further discussion regarding
immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current
Protocols in
Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.16.1.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
131
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, P~DF
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
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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
132
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 IZSI} in the presence
of
increasing amounts of an unlabeled second antibody.
The~~apeutic 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
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
133
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.
The antibodies of this invention may be advantageously utilized in
combination with other monoclonal or chimeric antibodies, or with lympholcines
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,
including fragments thereof. Preferred binding affinities include those with a
dissociation constant or Kd less than 5 X 10'2 M, 10'Z M, 5 X 10'3 M, 10'3 M,
5 X 10'
4 M, 10'4 M, 5 X 10-5 M, 10'5 M, 5 X 10'6 M, 10'6 M, 5 X 10'' M, 10'' M, 5 X
10-8 M,
10-8 M, 5 X 10'~ M, 10'~ M, 5 X 10'1° M, 10'1° M, 5 X 10-11 M,
10''1 M, 5 X 10-12 M,
10-12 M, 5 X 10'13 M, 10' 13 M, 5 X 10'14 M, 10-14 M, 5 X 10-15 M, and 10'15
M.
Gene Tlzef-apy
In a specific embodiment, nucleic acids comprising sequences encoding
antibodies or functional derivatives thereof, are administered to treat,
inhibit or

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
134
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-S05 (1993); Wu and ~u, Biotherapy 3:87-95 (1991};
Tolstoshev, Ann. Rev. Pharmacol. Toxicol. 32:5?3-596 (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 ~iley & Sons, NY (1993);
and
Kriegler, Gene Transfer and Expression, A Laboratory Manual, Stockton Press,
NY
( 1990).
In a preferred aspect, the compound comprises nucleic acid sequences
encoding an antibody, said nucleic acid sequences being part of expression
vectors
that express the antibody or fragments or chimeric proteins or heavy or light
chains
thereof in a suitable host. In particular, such nucleic acid sequences have
promoters
operably linked to the antibody coding region, said promoter being inducible
or
constitutive, and, optionally, tissue- specific. In another particular
embodiment,
nucleic acid molecules are used in which the antibody coding sequences and any
other
desired sequences are flanked by regions that promote homologous recombination
at a
desired site in the genome, thus providing for intrachromosomal expression of
the
antibody encoding nucleic acids (Koller and Smithies, Proc. Natl. Acad. Sci.
USA
86:8932-8935 (1989); Zijlstra et al., Nature 342:435-438 (1989). In specific
embodiments, the expressed antibody molecule is a single chain antibody;
alternatively, the nucleic acid sequences include sequences encoding both the
heavy
and light chains, or fragments thereof, of the antibody.
Delivery of the nucleic acids into a patient may be either direct, in which
case
the patient is directly exposed to the nucleic acid or nucleic acid- carrying
vectors, or

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
135
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
inj ection 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
(Roller 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
136
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 Gmzberg, 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. I~ozarsky 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.
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
137
carned out by any method known in the art, including but not limited to
transfection,
electroporation, microinjection, infection with a viral or bacteriophage
vector
containing the nucleic acid sequences, cell fusion, chromosome-mediated gene
transfer, microcell-mediated gene transfer, spheroplast fusion, etc. Numerous
techniques are known in the art for the introduction of foreign genes into
cells (see,
e.g., Loeffler and Behr, Meth. Enzymol. 217:599-618 (1993); Cohen et al.,
Meth.
Enzymol. 217:618-644 (1993); Cline, Pharmac. Ther. 29:69-92m (1985) and may be
used in accordance with the present invention, provided that the necessary
developmental and physiological functions of the recipient cells are not
disrupted.
The technique should provide for the stable transfer of the nucleic acid to
the cell, so
that the nucleic acid is expressible by the cell and preferably heritable and
expressible by its cell progeny.
The resulting recombinant cells can be delivered to a patient by various
methods known in the art. Recombinant blood cells (e.g., hematopoietic stem or
progenitor cells) are preferably administered intravenously. The amount of
cells
envisioned for use depends on the desired effect, patient state, etc., and can
be
determined by one skilled in the art.
Cells into which a nucleic acid can be introduced for purposes of gene therapy
encompass any desired, available cell type, and include but axe not limited to
epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells,
hepatocytes;
blood cells such as Tlymphocytes, Blymphocytes, monocytes, macrophages,
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
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
138
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 Activi y
The compounds or pharmaceutical compositions of the invention are
preferably tested in vitro, and then in vivo for the desired therapeutic ar
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
I 5 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.
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 subj
ect 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;

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
139
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 intradennal, intramuscular,
intraperitoneal,
intravenous, subcutaneous, intranasal, epidural, and oral routes. The
compounds or
compositions may be administered by any convenient route, for example by
infusion
or bolus injection, by absorption through epithelial or mucocutaneous linings
(e.g.,
oral mucosa~ rectal and intestinal mucosa, etc.) and may be administered
together
with other biologically active agents. Administration can be systemic or
local. In
addition, it may be desirable to introduce the pharmaceutical compounds or
compositions of the invention into the central nervous system by any suitable
route,
including intraventricular and intrathecal injection; intraventricular
injection may be
facilitated by an intraventricular catheter, for example, attached to a
reservoir, such
as an Ommaya reservoir. Pulmonary administration can also be employed, e.g.,
by
use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
In a specific embodiment, it may be desirable to administer the pharmaceutical
compounds or compositions of the invention locally to the area in need of
treatment;
this may be achieved by, for example, and not by way of limitation, local
infusion
during surgery, topical application, e.g., in conjunction with a wound
dressing after
surgery, by inj ection, 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
140
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
Longer,
S supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al.,
Surgery
88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)). In another
embodiment, polymeric materials can be used (see Medical Applications of
Controlled Release, Longer 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 (I985);
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)).
Othemcontrolled release systems are discussed in the review by Longer
(Science 249:1527-1533 (1990)).
In a specific embodiment where the compound of the invention is a nucleic
acid encoding a protein, the nucleic acid can be administered in vivo to
promote
expression of its encoded protein, by constructing it as part of an
appropriate nucleic
acid expression vector and administering it so that it becomes intracellular,
e.g., by
use of a retroviral vector (see U.S. Patent No. 4,980,286), or by direct
injection, or by
use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or
coating
with lipids or cell-surface receptors or transfecting agents, or by
administering it in
linkage to a homeobox- like peptide which is known to enter the nucleus (see
e.g.,
Joliot et al., Proc. Natl. Acad. Sci. USA 88:1864-1868 (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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
141
"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 fox use in animals, and more particularly in humans. The term
"carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the
therapeutic
is administered. Such pharmaceutical carriers can be sterile liquids, such as
water and
oils, including those of petroleum, animal, vegetable or synthetic origin,
such as
peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a
preferred
carrier when the pharmaceutical composition is administered intravenously.
Saline
solutions and aqueous dextrose and glycerol solutions can also be employed as
liquid
carriers, particularly for injectable solutions. Suitable pharmaceutical
excipients
include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk,
silica gel,
sodium stearate, glycerol monostearate, talc, sodium chloride, dried slum
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 earners such as
pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium
saccharine, cellulose, magnesium carbonate, etc. Examples of suitable
pharmaceutical earners are described in "Remington's Pharmaceutical Sciences"
by
E.W. Martin. Such compositions will contain a therapeutically effective amount
of
the compound, preferably in purified form, together with a suitable amount of
carrier
so as to provide the form for proper administration to the patient. The
formulation
should suit the mode of administration.
In a preferred embodiment, the composition is formulated in accordance with
routine procedures as a pharmaceutical composition adapted for intravenous
administration to human beings. Typically, compositions for intravenous
administration are solutions in sterile isotonic aqueous buffer. Where
necessary, the
composition may also include a solubilizing 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
142
lyophilized powder or water free concentrate in a hermetically sealed
container such
as an ampoule or sachette indicating the quantity of active agent. Where the
composition is to be administered by infusion, it can be dispensed with an
infusion
bottle containing sterile pharmaceutical grade water or saline. Where the
composition
is administered by injection, an ampoule of sterile water for injection or
saline can be
provided so that the ingredients may be mixed prior to administration.
The compounds of the invention can be formulated as neutral or salt forms.
Pharmaceutically acceptable salts include those formed with anions such as
those
derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc.,
and those
formed with canons such as those derived from sodium, potassium, ammonium,
calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino
ethanol,
histidine, procaine, etc.
The amount of the compound of the invention which will be effective in the
treatment, inhibition and prevention of a disease or disorder associated with
aberrant
expression and/or activity of a polypeptide of the invention can be determined
by
standard clinical techniques. In addition, in vitro assays may optionally be
employed
to help identify optimal dosage ranges. The precise dose to be employed in the
formulation will also depend on the route of administration, and the
seriousness of the
disease or disorder, and should be decided according to the judgment of the
practitioner and each patient's circumstances. Effective doses may be
extrapolated
from dose-response curves derived from in vitro or animal model test systems.
For antibodies, the dosage administered to a patient is typically 0. I 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 mglkg to 10 mglkg of the patient's body weight. Generally, human
antibodies have a longer half life within the human body than antibodies from
other
species due to the immune response to the foreign polypeptides. Thus, lower
dosages
of human antibodies and less frequent administration is often possible.
Further, the
dosage and frequency of administration of antibodies of the invention may be
reduced
by enhancing uptake and tissue penetration (e.g., into the brain) of the
antibodies by
modifications such as, for example, lipidation.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
143
The invention also provides a pharmaceutical pack or lcit 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 ayad 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
Ievel,
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
144
(e.g., see Jallcanen, 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
imrnunosorbent 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, I21I), carbon (14C), sulfur (35S),
tritium (3H),
indium (112In), and technetium (99Tc); luminescent labels, such as Iuminol;
and
fluorescent labels, such as fluorescein and rhodamine, and biotin.
One aspect of the invention is the detection and diagnosis of a disease or
disorder associated with aberrant expression of a polypeptide of interest in
an animal,
preferably a mammal and most preferably a human. In one embodiment, diagnosis
comprises: a) administering (for example, parenterally, subcutaneously, or
intraperitoneally) to a subject an effective amount of a labeled molecule
which
specifically binds to the polypeptide of interest; b) waiting for a time
interval
following the administering for permitting the labeled molecule to
preferentially
concentrate at sites in the subject where the polypeptide is expressed (and
for
unbound labeled molecule to be cleared to background level); c) determining
background level; and d) detecting the labeled molecule in the subject, such
that
detection of labeled molecule above the background level indicates that the
subject
has a particular disease or disorder associated with aberrant expression of
the
polypeptide of interest. Background level can be determined by various methods
including, comparing the amount of labeled molecule detected to a standard
value
previously determined for a particular system.
It will be understood in the art that the size of the subject and the imaging
system used will determine the quantity of imaging moiety needed to produce
diagnostic images. W the case of a radioisotope moiety, for a human subject,
the
quantity of radioactivity inj ected 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:

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
145
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
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 lcit comprises an antibody of the invention, preferably a
purified
antibody, in one or more containers. In a specific embodiment, the kits of the
present

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
146
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
and/or cancerous polynucleotides and polypeptides. Such a kit may include a
control
antibody that does not react with the polypeptide of interest. Such a kit may
include a
substantially isolated polypeptide antigen comprising an epitope which is
specifically
immunoreactive with at least one anti-polypeptide antigen antibody. Further,
such a
kit includes means for detecting the binding of said antibody to the antigen
(e.g., the
antibody may be conjugated to a fluorescent compound such as fluorescein or
rhodamine which can be detected by flow cytometry). In specific embodiments,
the
kit may include a recombinantly produced or chemically synthesized polypeptide
antigen. The polypeptide antigen of the kit may also be attached to a solid
support.
In a more specific embodiment the detecting means of the above-described kit
includes a solid support to which said polypeptide antigen is attached. Such a
kit may
also include a non-attached reporter-labeled anti-human antibody. W 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
147
monoclonal antibody. Alternatively, or in addition, the~detecting means rnay
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
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
148
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 (CHl, 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).)
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
149
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); I~. 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
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
150
vector is a virus, it may be paclcaged in vitro using an appropriate packaging
cell line
and then transduced into host cells.
The polynucleotide insert should be operatively linked to an appropriate
promoter, such as the phage lambda PL promoter, the E. coli lac, trp, phoA and
tac
promoters, the SV40 early and late promoters and promoters of retroviral LTRs,
to
name a few. Other suitable promoters will be known to the skilled artisan. The
expression constructs will further contain sites for transcription initiation,
termination,
and, in the transcribed region, a ribosome binding site for translation. The
coding
portion of the transcripts expressed by the constructs will preferably include
a
translation initiating codon at the beginning and a termination codon (UAA,
UGA or
UAG) appropriately positioned at the end of the polypeptide to be translated.
As indicated, the expression vectors will preferably include at least one
selectable marker. Such markers include dihydrofolate reductase, 6418 or
neomycin
resistance for eukaryotic cell culture and tetracycline, kanamycin or
ampicillin
resistance genes for culturing in E. coli and other bacteria. Representative
examples
of appropriate hosts include, but are not limited to, bacterial cells, such as
E. coli,
Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast
cells
(e.g., Saccharomyces cerevisiae or Pichia pastoris (ATCC Accession No.
201178));
insect cells such as Drosophila S2 and Spodoptera Sf~ 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,
pNH8A,
pNHl6a, pNHl8A, pNH46A, available from Stratagene Cloning Systems, Inc.; and
ptrc99a, pKK223-3, pKK233-3, pDR540, pRITS available from Pharmacia Biotech,
Inc. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl
and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available
from Pharmacia. Preferred expression vectors for use in yeast systems include,
but are
not limited to pYES2, pYDl, pTEFl/Zeo, pYES2/GS, pPICZ,pGAPZ, pGAPZaIph,
pPIC9, pPIC3.5, pHIL-D2, PHIL-S1, pPIC3.5K, pPIC9K, and PA0815 (all available
from Invitrogen, Carlbad, CA). Other suitable vectors will be readily apparent
to the
skilled artisan.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
151
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 cation exchange
chromatography,
phosphocellulose chromatography, hydrophobic interaction chromatography,
affinity
chromatography, hydroxylapatite chromatography and lectin chromatography. Most
preferably, high performance liquid chromatography ("HPLC") is employed for
purification.
Polypeptides of the present invention, 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-
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 prolcaryotes,
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 pastoy-is is used to express the
polypeptide of the present invention in a eukaryotic system. Pichia pasto~is
is a
methylotrophic yeast which can metabolize methanol as its sole carbon source.
A

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
152
main step in the methanol metabolization pathway is the oxidation of methanol
to
formaldehyde using OZ. 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 Iow affinity of
alcohol
oxidase far 02. 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
pastor is. See, Ellis, S.B., et al., Mol. Cell. Biol. S:1 111-21 (1985);
Koutz, P.J, et al.,
J'cast 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.
h1 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
AOXI
promoter linked to the Piclaia pastoris alkaline phosphatase (PHO) secretory
signal
peptide (i.e., leader) located upstream of a multiple cloning site.
Many other yeast vectors could be used in place of pPIC9K, such as, pYES2,
pYDl, 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
153
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 pohynucleotides. For
example,
techniques known in the art may be used to operabhy associate heterohogous
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; Internationah Publication No. WO 9612941 l,
published September 26, 1996; International Publication No. WO 94/12650,
. published August 4, 1994; Kolher et al., Proc. Natl. Acad. Sci. USA 86:8932-
8935
(1989); and Zijlstra et a1., Nature 342:435-438 (1989), the disclosures of
each of
which are incorporated by reference in their entireties).
In addition, polypeptides of the invention can be chemically synthesized using
techniques known in the art (e.g., see Creighton, 1983, Proteins: Structures
and
Molecular Principles, W.H. Freeman & Co., N.Y., and Hunkapiller et al.,
Nature,
310:105-111 (1984)). For example, a polypeptide corresponding to a fragment of
a
polypeptide sequence of the invention can be synthesized by use of a peptide
synthesizer. Furthermore, if desired, nonclassicah 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, citrulhine, homocitrulline, cysteic
acid, t-
butylglycine, t-butylalanine, phenylglycine, cyclohexylahanine, b-alanine,
fluoro-
amino acids, designer amino acids such as b-methyl amino acids, Ca-methyl
amino

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
154
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 specif c 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
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 I kDa and about 100 kDa (the term "about" indicating that in
preparations of
polyethylene glycol, some molecules will weigh more, some Iess, than the
stated
molecular weight) for ease in handling and manufacturing. Other sizes may be
used,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
1ss
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. Bioclaem. Biotechhol. 56:59-72 (1996);
Vorobjev et
al., Nucleosides Nucleotides 18:2745-2750 (1999); and Caliceti et al.,
Biocohjug.
Chetra. 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
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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
156
As suggested above, polyethylene glycol may be attached to proteins via
Iiu~age 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 rnonopegylated 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.
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.,
Cy°it. Rev.
Theta. Drug ~'a~~ie~ Sys. 9:249-304 (2992); Francis et al., hcte~h. J.
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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
157
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 l,l'-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,
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. They°a. Drug
Ca~~rief- 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
158
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 axe 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
cI?NA
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
1 S polypeptides having different amino acid sequences. In specific
embodiments, the
multimer of the invention is a homodimer (e.g., containing polypeptides having
identical or different amino acid sequences) or a homotrimer (e.g., containing
polypeptides having identical and/or different amino acid sequences). In
additional
embodiments, the homomeric multimer of the invention is at least a homodimer,
at
least a homotrimer, or at least a homotetramer.
As used herein, the term heteromer refers to a multimer containing one or
more heterologous polypeptides (i. e., polypeptides of different proteins) in
addition to
the polypeptides of the invention. In a specific embodiment, the multimer of
the
invention is a heterodimer, a heterotrimer, or a heterotetramer. In additional
embodiments, the heteromeric 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
159
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 entixety). 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.
5,473,627
(hereby incorporated by reference). Proteins comprising multiple polypeptides
of the
invention separated by peptide linkers may be produced using conventional
recombinant DNA technology.
Another method for preparing multimer polypeptides of the invention involves
use of polypeptides of the invention fused to a leucine zipper or isoleucine
zipper
polypeptide sequence. Leucine zipper and isoleucine zipper domains are
polypeptides
that promote multimerization of the proteins in which they are found. Leucine
zippers were originally identified in several DNA-binding proteins (Landschulz
et al.,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
160
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 ar trimerizes in solution are expressed in suitable
host cells,
and the resulting soluble multimenic 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 I~oppe et al. (FEBS Letters
344:I9I,
(1994)) and in U.S. patent application Ser. No. 08/446,922, hereby
incorporated by
I S 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, polypeptades desired to be contained in the
multimers
of the invention may be chemically cross-linked using Linker molecules and
linker
molecule length optimization techniques known in the art (see, e.g., US Patent
Number 5,478,925, which is herein incorporated by reference in its entirety).
Additionally, multimers of the invention may be generated using techniques
known in
the art to form one or more inter-molecule cross-links between the cysteine
residues
located within the sequence of the polypeptides desired to be contained in the
multimer (see, e.g., US Patent Number 5,478,925, which is herein incorporated
by
reference in its entirety). Further, polypeptides of the invention may be
routinely

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
16I
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,
S 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, multirners 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
1 S 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
liposomes (see, e.g., US Patent Number 5,478,925, which is herein incorporated
by
2S reference in its entirety).
Uses of the Polynucleotides
Each of the polynucleotides identified herein can be used in numerous ways as
reagents. The following description should be considered exemplary and
utilizes
known techniques.
The polynucleotides of the present invention are useful for chromosome
identification. There exists an ongoing need to identify new chromosome
markers,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
162
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 (Sae, e.g., Shuler,
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
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
163
(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.
McI~usick, Mendelian Inheritance in Man (available on line through Johns
Hopkins
University Welch Medical Library) .) Assuming 1 megabase mapping resolution
and
one gene per 20 kb, a cDNA precisely localized to a chromosomal region
associated
with the disease could be one of 50-500 potential causative genes.
Thus, once coinheritance is established, differences in 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.
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
164
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 lcit for analyzing
samples
for the presence of proliferative and/or cancerous polynucleotides derived
from a test
subject. W 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
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
165
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,
thymine and cytosine are available commercially (Perceptive Biosystems).
Certain
components of DNA, such as phosphorus, phosphorus oxides, or deoxyribose
derivatives, axe 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,
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
166
baclcbone is more flexible. Because of this, PNA/DNA duplexes bind under a
wider
range of stringency conditions than DNA/DNA duplexes, malting it easier to
perform
multiplex hybridization. Smaller probes can be used than with DNA due to the
strong
binding. In addition, it is more likely that single base mismatches can be
determined
with PNA/DNA hybridization because a single mismatch in a PNA/DNA 15-mer
lowers the melting point (T<sub>m</sub>) by 8°-20° C, vs. 4°-
16° C for the DNA/DNA 15-
mer duplex. Also, the absence of charge groups in PNA means that hybridization
can
be done at low ionic strengths and reduce possible interference by salt during
the
analysis.
The present invention 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-I82
(1985)).
Neoplasias are now believed to result from the qualitative alteration of a
normal
cellular gene product, or from the quantitative modification of gene
expression by
insertion into the chromosome of a viral sequence, by chromosomal
translocation of a
gene to a more actively transcribed region, or by some other mechanism.
(Gelmann
et al., supra) It is likely that mutated or altered expression of specific
genes is
involved in the pathogenesis of some leukemias, among other tissues and cell
types.
(Gelmann et al., supra) Indeed, the human counterparts of the oncogenes
involved in
some animal neoplasias have been amplified or translocated in some cases of
human
leukemia and carcinoma. (Gelmann et al., supra)

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
167
For example, c-myc expression is highly amplified in the non-Iymphocytic
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
(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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
168
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.
W 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 DJ database is established for an
individual, positive identification of that individual, living or dead, can be
made from
extremely small tissue samples.
Forensic biology also benefits from using DNA-based identification
techniques as disclosed herein. DNA sequences talcen 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
169
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
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 fox 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 (I25I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium
(112In), and

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
170
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,
I l2ln, 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 In.c. (1982).)
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
171
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
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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
172
Gene Therauy 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 inj ectable materials to the cells of an
animal,
such as, injection into the interstitial space of tissues (heart, muscle,
skin, lung, liver,
and the like). The polynucleotide constructs may be delivered in a
pharmaceutically
acceptable liquid or aqueous carrier.
In one embodiment, the polynucleotide of the 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,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
173
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 fox replication. Appropriate vectors include
pWLNEO, pSV2CAT, pOG44, pXTl 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
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,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
174
fluid, rnucopolysaccharide 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
inj ection 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. 1h. 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 inj ection. The appropriate and effective dosage of nucleic
acid sequence
can readily be determined by those of ordinary skill in the art and may depend
on the
condition being treated and the route of administration.
The preferred route of administration is by the parenteral route of injection
into the interstitial space of tissues. However, other parenteral routes may
also be
used, such as, inhalation of an aerosol formulation particularly for delivery
to lungs or
bronchial tissues, throat or mucous membranes of the nose. In addition, naked
DNA
constructs can be delivered to arteries during angioplasty by the catheter
used in the
procedure.
The naked polynucleotides are delivered by any method known in the art,
including, but not limited to, direct needle injection at the delivery site,
intravenous
injection, topical administration, catheter infusion, and so-called "gene
guns". These
delivery methods are known in the art.
The constructs may also be delivered with delivery vehicles such as viral
sequences, viral particles, liposome formulations, lipofectin, precipitating
agents, etc.
Such methods of delivery are known in the art.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
175
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.
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
176
starting materials in appropriate ratios. Methods for making liposomes using
these
materials are well known in the art.
For example, commercially dioleoylphosphatidyl choline (DOPC),
dioleoylphosphatidyl glycerol (DOPG), and dioleoylphosphatidyl ethanolamine
(DOPE) can be used in various combinations to make conventional liposomes,
with or
without the addition of cholesterol. Thus, for example, DOPG/DOPC vesicles can
be
prepared by drying 50 mg each of DOPG and DOPC under a stream of nitrogen gas
into a sonication vial. The sample is placed under a vacuum pump overnight and
is
hydrated the following day with 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.
SLTVs are prepared by extended sonication of MLVs to produce a homogeneous
population of unilamellar liposomes. The material to be entrapped is added to
a
suspension of preformed MLVs and then sonicated. When using liposomes
containing
cationic lipids, the dried lipid film is resuspended in an appropriate
solution such as
sterile water or an isotonic buffer solution such as 10 mM Tris/NaCl,
sonicated, and
then the preformed liposomes are mixed directly with the DNA. The liposome and
DNA form a very stable complex due to binding of the positively charged
liposomes
to the cationic DNA. SUVs find use with small nucleic acid fragments. LUVs are
prepared by a number of methods, well known in the art. Commonly used methods
include Ca2+-EDTA chelation (Papahadjopoulos et al., Biochim. Biophys. Acta,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
177
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 ih 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
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 PE501, PA317, R-2, R-AM, PAl2, 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
178
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
aclininistered 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 vitYO
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);
Rosenfeld et al., Cell , 68:143-155 (1992); Engelhardt et al., Human Genet.
Ther.,
4:759-769 (1993); Yang et al., Nature Genet., 7:362-369 (1994); Wilson et al.,
Nature , 365:691-692 (1993); and U.S. Patent NO: 5,652,224, which are herein
incorporated by reference. For example, the adenovirus vector Ad2 is useful
and can
be grown in human 293 cells. These cells contain the E1 region of adenovirus
and
constitutively express Ela and Elb, which complement the defective
adenoviruses by
providing the products of the genes deleted from the vector. In addition to
Ad2, other

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
179
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 L1 through L5.
W 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. Irnrrmnol., 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
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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
180
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
96/29411, published September 26, 1996; International Publication NO: WO
94/12650, published August 4, 1994; Koller et al., Proc. Natl. Acad. Sci. USA,
86:8932-8935 (1989); and Zijlstra et al., Nature, 342:435-438 (1989). This
method
involves the activation of a gene which is present in the target cells, but
which is not
normally expressed in the cells, or is expressed at a lower level than
desired.
Polynucleotide constructs are made, using standard tecluliques 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.
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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
181
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
(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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
182
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.
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
183
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 Actiyities
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.
Polynucleotides, translation products and antibodies corresponding to this
gene may be useful for the diagnosis, prognosis, prevention, and/or treatment
of
diseases and/or disorders associated with the following systems.
Immune Activity
Polynucleotides, polypeptides, antibodies, andJor agonists or antagonists of
the
present invention may be useful in treating, preventing, diagnosing and/or
prognosing
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 the present invention can be used as a marker or detector of a
particular
immune system disease or disorder.
In another embodiment, a polypeptide of the invention, or polynucleotides,
antibodies, agonists, or antagonists corresponding to that polypeptide, may be
used to
treat diseases and disorders of the immune system and/or to inhibit or enhance
an

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
184
immune response generated by cells associated with the tissues) in which the
polypeptide of the invention is expressed, including one, two, three, four,
five, or
more tissues disclosed in Table 1, column 8 (Tissue Distribution Library
Code).
Polynucleotides, polypeptides, antibodies, andlor agonists or antagonists of
the
present invention may be useful in treating, preventing, diagnosing, and/or
prognosing
immunodeficiencies, including both congenital and acquired immunodeficiencies.
Examples of B cell immunodeficiencies in which immunoglobulin levels B cell ,
function and/or B cell numbers are decreased include: X-linked
agamrnaglobulinemia
(Bruton's disease), X-linked infantile agammaglobulinemia, X-linked
immunodeficiency with hyper IgM, non X-linked immunodeficiency with hyper IgM,
X-linked lymphoproliferative syndrome (XLP), agammaglobulinemia including
congenital and acquired agammaglobulinemia, adult onset agammaglobulinemia,
late-
onset agammaglobulinemia, dysgammaglobulinemia, hypogammaglobulinemia,
unspecified hypogammaglobulinemia, recessive agammaglobulinemia (Swiss type),
Selective IgM deficiency, selective IgA deficiency, selective IgG subclass
deficiencies, IgG subclass deficiency (with or without IgA deficiency), Ig
deficiency
with increased IgM, IgG and IgA deficiency with increased IgM, antibody
deficiency
with normal or elevated Igs, Ig heavy chain deletions, kappa chain deficiency,
B cell
lymphoproliferative disorder (BLPD), common variable immunodeficiency (CVID),
common variable immunodeficiency (CVI) (acquired), and transient
hypogammaglobulinemia of infancy.
In specific embodiments, ataxia-telangiectasia or conditions associated with
ataxia-telangiectasia are treated, prevented, diagnosed, and/or prognosing
using the
polypeptides or polynucleotides of the invention, and/or agonists or
antagonists
thereof.
Examples of congenital immunodeficiencies in which T cell and/or B cell
function and/or number is decreased include, but are not limited to: DiGeorge
anomaly, severe combined immunodeficiencies (SCID) (including, but not limited
to,
X-linked SCID, autosomal recessive SLID, adenosine deaminase deficiency,
purine
nucleoside phosphorylase (PNP) deficiency, Class II MHC deficiency (Bare
lymphocyte syndrome), Wiskott-Aldrich syndrome, and ataxia telangiectasia),
thyrnic
hypoplasia, third and fourth pharyngeal pouch syndrome, 22q11.2 deletion,
chronic

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
185
mucocutaneous candidiasis, natural killer cell deficiency (NK), idiopathic
CD4+ T-
lymphocytopenia, immunodeficiency with predominant T cell defect
(unspecified),
and unspecified immunodeficiency of cell mediated immunity.
ha specific embodiments, DiGeorge anomaly or conditions associated with
DiGeorge anomaly are treated, prevented, diagnosed, and/or prognosed using
polypeptides or polynucleotides of the invention, or antagonists or agonists
thereof.
Other immunodeficiencies that may be treated, prevented, diagnosed, and/or
prognosed using polypeptides or polynucleotides of the invention, and/or
agonists or
antagonists thereof, include, but are not limited to, chronic granulomatous
disease,
Chediak-Higashi syndrome, myeloperoxidase deficiency, leukocyte glucose-6-
phosphate dehydrogenase deficiency, X-linked lyrnphoproliferative syndrome
(XLP),
leukocyte adhesion deficiency, complement component deficiencies (including
C1,
C2, C3, C4, C5, C6, C7, C8 and/or C9 deficiencies), reticular dysgenesis,
thymic
alymphoplasia-aplasia, immunodeficiency with thymoma, severe congenital
leukopenia, dysplasia with immunodeficiency, neonatal neutropenia, short
limbed
dwarfism, and Nezelof syndrome-combined immunodeficiency with Igs.
In a preferred embodiment, the immunodeficiencies and/or conditions
associated with the immunodeficiencies recited above are treated, prevented,
diagnosed and/or prognosed using polynucleotides, polypeptides, antibodies,
and/or
agonists or antagonists 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 immunodeficient individuals. In specific
embodiments, 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.
The polynucleotides, polypeptides, antibodies, and/or agonists or antagonists
of the present invention may be useful in treating, preventing, diagnosing
and/or
prognosing autoimmune disorders. Many autoimmune disorders result from
inappropriate recognition of self as foreign material by immune cells. This
inappropriate recognition results in an immune response leading to the
destruction of
the host tissue. Therefore, the administration of polynucleotides and
polypeptides of

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
186
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, diagnosed
and/or prognosed 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: systemic lupus erythematosus, rheumatoid arthritis, ankylosing
spondylitis, multiple sclerosis, autoimmune thyroiditis, Hashimoto's
thyroiditis,
autoimmune hemolytic anemia, hemolytic anemia, thrombocytopenia, autoimmune
thrombocytopenia purpura, autoimmune neonatal thrombocytopenia, idiopathic
thrombocytopenia purpura, purpura (e.g., Henloch-Scoenlein purpura),
autoimmunocytopenia, Goodpasture's syndrome, Pemphigus vulgaris, myasthenia
gravis, Grave's disease (hyperthyroidism), and insulin-resistant diabetes
mellitus.
Additional disorders that are likely to have an autoimmune component that
may be treated, prevented, and/or diagnosed with the compositions of the
invention
include, but are not limited to, type II collagen-induced arthritis,
antiphospholipid
syndrome, dermatitis, allergic encephalomyelitis, myocarditis, relapsing
polychondritis, rheumatic heart disease, neuritis, uveitis ophthalmia,
polyendocrinopathies, Reiter's Disease, Stiff Man Syndrome, autoimmune
pulmonary
inflammation, autism, Guillain-Barre Syndrome, insulin dependent diabetes
mellitus,
and autoimmune inflammatory eye disorders.
Additional disorders that are likely to have an autoimmune component that
may be treated, prevented, diagnosed and/or prognosed With the compositions of
the
invention include, but are not limited to, 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
187
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 mellitus
(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 disorders that may have an autoimmune component that may be
treated, prevented, diagnosed and/or prognosed 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 mitochondria 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, granulomatous, degenerative, and atrophic disorders.
In a preferred embodiment, the autoimmune diseases and disorders and/or
conditions associated with the diseases and disorders recited above are
treated,
prevented, diagnosed and/or prognosed using for example, antagonists or
agonists,
polypeptides or polynucleotides, or antibodies of the present invention. 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 erythematosus 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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
188
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.
Iii a preferred embodiment, the autoimmune diseases and disorders and/or
conditions associated with the diseases and disorders recited above are
treated,
prevented, diagnosed and/or prognosed using polynucleotides, polypeptides,
antibodies, and/or agonists or antagonists of the present invention
In preferred embodiments, polypeptides, antibodies, polynucleotides and/or
agonists or antagonists of the present invention are used as a
irmnunosuppressive
agent(s).
Polynucleotides, polypeptides, antibodies, and/or agonists or antagonists.of
the
present invention may be useful in treating, preventing, prognosing, 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, including but not limited to, leukopenia, neutropenia, anemia, and
thrombocytopenia. Alternatively, 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 an increase in certain (or many) types of hematopoietic cells,
including but not limited to, histiocytosis.
Allergic reactions and conditions, such as asthma (particularly allergic
asthma) or other respiratory problems, may also be treated, prevented,
diagnosed
and/or prognosed using polypeptides, antibodies, or polynucleotides of the
invention,
and/or agonists or antagonists thereof. Moreover, these molecules can be used
to
treat, prevent, prognose, and/or diagnose anaphylaxis, hypersensitivity to an
antigenic
molecule, or blood group incompatibility.
Additionally, polypeptides or polynucleotides of the invention, and/or
agonists
or antagonists thereof, may be used to treat, prevent, diagnose and/or
prognose

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
189
IgE-mediated allergic reactions. Such allergic reactions include, but are not
limited
to, asthma, rhinitis, and eczema. In specific embodiments, polynucleotides,
polypeptides, antibodies, and/or agonists or antagonists of the present
invention may
be used to modulate IgE concentrations in vitro or in vivo.
Moreover, polynucleotides, polypeptides, antibodies, and/or agonists or
antagonists of the present invention have uses in the diagnosis, prognosis,
prevention,
and/or treatment of inflammatory conditions. 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 prevent
and/or
treat chronic and acute inflammatory conditions. Such inflammatory conditions
include, but are not limited to, for example, inflammation associated with
infection
(e.g., septic shock, sepsis, or systemic inflammatory response syndrome),
ischemia-
reperfusion injury, endotoxin lethality, complement-mediated hyperacute
rejection,
nephritis, cytokine or chemokine induced lung injury, inflammatory bowel
disease,
Crohn's disease, over production of cytokines (e.g., TNF or IL-l.),
respiratory
disorders (e.g., asthma and allergy); gastrointestinal disorders (e.g.,
inflammatory
bowel disease); cancers (e.g., gastric, ovarian, lung, bladder, liver, and
breast); CNS
disorders (e.g., multiple sclerosis; ischemic brain injury and/or stroke,
traumatic brain
injury, neurodegenerative disorders (e.g., Parkinson's disease and Alzheimer's
disease); AIDS-related dementia; and prion disease); cardiovascular disorders
(e.g.,
atherosclerosis, myocarditis, cardiovascular disease, and cardiopulmonary
bypass
complications); as well as many additional diseases, conditions, and disorders
that are
characterized by inflammation (e.g., hepatitis, rheumatoid arthritis, gout,
trauma,
pancreatitis, sarcoidosis, dermatitis, renal ischemia-reperfusion injury,
Grave's
disease, systemic lupus erythematosus, diabetes mellitus, and allogenic
transplant
rejection).
Because inflammation is a fundamental defense mechanism, inflammatory
disorders can effect virtually any tissue of the body. Accordingly,
polynucleotides,
polypeptides, and antibodies of the invention, as well as agonists or
antagonists
thereof, have uses in the treatment of tissue-specific inflammatory disorders,
including, but not limited to, adrenalitis, alveolitis, angiocholecystitis,
appendicitis,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
190
balanitis, blepharitis, bronchitis, bursitis, carditis, cellulitis,
cervicitis, cholecystitis,
chorditis, cochlitis, colitis, conjunctivitis, cystitis, dermatitis,
diverticulitis,
encephalitis, endocarditis, esophagitis, eustachitis, fibrositis,
folliculitis, gastritis,
gastroenteritis, gingivitis, glossitis, hepatosplenitis, keratitis,
labyrinthitis, laryngitis,
lymphangitis, mastitis, media otitis, meningitis, metritis, mucitis,
myocarditis,
myosititis, myringitis, nephritis, neuritis, orchitis, osteochondritis,
otitis, pericarditis,
peritendonitis, peritonitis, pharyngitis, phlebitis, poliomyelitis,
prostatitis, pulpitis,
retinitis, rhinitis, salpingitis, scleritis, sclerochoroiditis, scrotitis,
sinusitis, spondylitis,
steatitis, stomatitis, synovitis, syringitis, tendonitis, tonsillitis,
urethritis, and vaginitis.
In specific embodiments, polypeptides, antibodies, or polynucleotides of the
invention, andlor agonists or antagonists thereof, are useful to diagnose,
prognose,
prevent, and/or treat organ transplant rejections and graft-versus-host
disease. 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. In specific embodiments, 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
experimental allergic and hyperacute xenograft rejection.
In other embodiments, polypeptides, antibodies, or polynucleotides of the
invention, and/or agonists or antagonists thereof, are useful to diagnose,
prognose,
prevent, and/or treat immune complex diseases, including, but not limited to,
serum
sickness, post streptococcal glomerulonephritis, polyarteritis nodosa, and
immune
complex-induced vasculitis.
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,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
191
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.
In another embodiment, polypeptides, antibodies, polynucleotides and/or
agonists or antagonists of the present invention are used as a vaccine
adjuvant that
enhances immune responsiveness to an antigen. In a specific embodiment,
polypeptides, antibodies, polynucleotides and/or agonists or antagonists of
the present
invention are used as an adjuvant to enhance tumor-specific immune responses.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as 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 of: AIDS, meningitis, Dengue, EBV, and hepatitis (e.g., hepatitis
B). In
another specific embodiment, the compositions of the invention are used as an
adjuvant to enhance an immune response to a virus, disease, or symptom
selected
from the group consisting of HIV/AIDS, respiratory syncytial virus, Dengue,
rotavirus, Japanese B encephalitis, influenza A and B, parainfluenza, measles,
cytomegalovirus, rabies, Junin, Chikungunya, Rift Valley Fever, herpes
simplex, and
yellow fever.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
192
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 Vibf°io cholerae,
Mycobacterium
leprae, Salmonella typhi, Salmonella paratyphi, MeisseYia meningitidis,
StYeptococcus pneumoniae, Group B streptococcus, Shigella spp.,
Enterotoxigenic
Eschef-ichia coli, Enterohemorrhagic E. coli, and BoY~elia burgdorferi.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as an
adjuvant to
enhance anti-parasitic immune responses. Anti-parasitic immune responses that
may
be enhanced using the compositions of the invention as an adjuvant, include
parasite
and parasite associated diseases or symptoms described herein or otherwise
known in
the art. In specific embodiments, the compositions of the invention are used
as an
adjuvant to enhance an immune response to a parasite. In another specific
embodiment, the compositions of the invention are used as an adjuvant to
enhance an
immune response to Plasmodium (malaria) or Leishmania.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention may also be employed
to treat
infectious diseases including silicosis, sarcoidosis, and idiopathic pulmonary
fibrosis;
for example, by preventing the recruitment and activation of mononuclear
phagocytes.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as an antigen
for the
generation of antibodies to inhibit or enhance immune mediated responses
against
polypeptides of the invention.
In one embodiment, polypeptides, antibodies, polynucleotides and/or agonists
or antagonists of the present invention are administered 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 and

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
193
immunoglobulin class switching (e.g., IgG, IgA, IgM, and IgE), and/or to
increase an
immune response.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a
stimulator of B
cell responsiveness to pathogens.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as an
activator of T
cells.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as an agent
that
elevates the immune status of an individual prior to their receipt of
immunosuppressive therapies.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as an agent
to induce
higher affinity antibodies.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as an agent
to increase
serum immunoglobulin concentrations.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as an agent
to
accelerate recovery of immunocompromised individuals.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as an agent
to boost
immunoresponsiveness among aged populations and/or neonates.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used 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,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
194
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.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as an agent
to boost
irnmunoresponsiveness among individuals having an acquired loss of B cell
function.
Conditions resulting in an acquired loss of B cell function that may be
ameliorated or
treated by administering the polypeptides, antibodies, polynucleotides and/or
agonists
or antagonists thereof, include, but are not limited to, HIV Infection, AIDS,
bone
marrow transplant, and B cell chronic lymphocytic leukemia (CLL).
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used 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, and recovery from surgery.
In another specific embodiment, polypeptides, antibodies, polynucleotides
ancUor agonists or antagonists of the present invention are used as a
regulator of
antigen presentation by monocytes, dendritic cells, andlor 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 antagonism of antigen presentation may be useful as an anti-tumor treatment
or to
modulate the immune system.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as an agent
to direct
an individual's immune system towards development of a humoral response (i.e.
TH2) as opposed to a TH1 cellular response.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
195
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used 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.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a
stimulator of B
cell production in pathologies such as AIDS, chronic lymphocyte disorder
and/or
Common Variable Immunodificiency.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a therapy
for
generation and/or regeneration of lymphoid tissues following surgery, trauma
or
genetic defect. In another specific embodiment, polypeptides, antibodies,
polynucleotides and/or agonists or antagonists of the present invention are
used in the
pretreatment of bone marrow samples prior to transplant.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a gene-
based
therapy for genetically inherited disorders resulting in immuno-
incompetence/irrnnunodeficiency such as observed among SLID patients.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a means of
activating monocytes/macrophages to defend against parasitic diseases that
effect
monocytes such as Leishmania.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a means of
regulating secreted cytokines that are elicited by polypeptides of the
invention.
In another embodiment, polypeptides, antibodies, polynucleotides and/or
agonists or antagonists of the present invention are used in one or more of
the
applications decribed herein, as they may apply to veterinary medicine.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a means of

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
196
blocking various aspects of immune responses to foreign agents or self.
Examples of
diseases or conditions in which blocking of certain aspects of immune
responses may
be desired include autoirmnune disorders such as lupus, and arthritis, as well
as
immunoresponsiveness to skin allergies, inflammation, bowel disease, injury
and
diseases/disorders associated with pathogens.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a therapy
for
preventing the B cell proliferation and Ig secretion associated with
autoimmune
diseases such as idiopathic thrombocytopenic purpura, systemic lupus
erythematosus
and multiple sclerosis.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a
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.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a therapy
for
chronic hypergammaglobulinemia evident in such diseases as monoclonal
gammopathy of undetermined significance (MGUS), Waldenstrom's disease, related
idiopathic monoclonal gammopathies, and plasmacytomas.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or a~atagonists of the present invention 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 autoimrnune and
chronic
inflammatory and infective diseases. Examples of autoimmune diseases are
described
herein and include multiple sclerosis, and insulin-dependent diabetes.
The polypeptides, antibodies, polynucleotides and/or agonists or antagonists
of the present invention may also be employed to treat idiopathic hyper-
eosinophilic
syndrome by, for example, preventing eosinophil production and migration.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
197
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used to enhance or
inhibit
complement mediated cell lysis.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used to enhance or
inhibit
antibody dependent cellular cytotoxicity.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention may also be employed
for
treating atherosclerosis, for example, by preventing monocyte infiltration in
the artery
wall.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention may be employed to
treat adult
respiratory distress syndrome CARDS).
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention may be useful for
stimulating
wound and tissue repair, stimulating angiogenesis, and/or stimulating the
repair of
vascular or lymphatic diseases or disorders. Additionally, agonists and
antagonists of
the invention may be used to stimulate the regeneration of mucosal surfaces.
In a specific embodiment, polynucleotides or polypeptides, and/or agonists
thereof are used to diagnose, prognose, treat, and/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, CVID, 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: Other diseases and disorders that may be
prevented, diagnosed, prognosed, and/or treated with polynucleotides or

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
198
polypeptides, and/or agonists of the present invention include, but are not
limited to,
HIV infection, HTLV-BLV infection, lymphopenia, phagocyte bactericidal
dysfunction anemia, thrombocytopenia, and hemoglobinuria.
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 diagnose,
prognose,
prevent, and/or treat cancers or neoplasms including immune cell or immune
tissue
related cancers or neoplasms. Examples of cancers or neoplasms that may be
prevented, diagnosed, or treated by polynucleotides, polypeptides, antibodies,
and/or
agonists or antagonists of the present invention include, but are not limited
to, acute
myelogenous leukemia, chronic myelogenous leukemia, Hodgkin's disease, non-
Hodgkin's lymphoma, acute lymphocytic anemia (ALL) Chronic lymphocyte
leukemia, plasmacytomas, multiple myeloma, Burkitt's lymphoma, EBV-transformed
diseases, and/or diseases and disorders described in the section entitled
"Hyperproliferative Disorders" elsewhere herein.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a therapy
for
decreasing cellular proliferation of Large B-cell Lymphomas.
In another specific embodiment, polypeptides, antibodies, polynucleotides
and/or agonists or antagonists of the present invention are used as a means of
decreasing the involvement of B cells and Ig associated with Chronic
Myelogenous
Leukemia.
In specific embodiments, the compositions of the invention are used as an
agent to boost immunoresponsiveness among B cell immunodeficient individuals,
such as, for example, an individual who has undergone a partial or complete
splenectomy.
Antagonists of the invention include, for example, binding and/or inhibitory
antibodies, antisense nucleic acids, ribozymes or soluble forms of the
polypeptides of

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
199
the present invention (e.g., Fc fusion protein; see, e.g., Example 9).
Agonists of the
invention include, for example, binding or stimulatory antibodies, and soluble
forms
of the polypeptides (e.g., Fc fusion proteins; see, e.g., Example 9).
polypeptides,
antibodies, polynucleotides and/or agonists or antagonists of the present
invention
may be employed in a composition with a pharmaceutically acceptable carrier,
e.g., as
described herein.
In another embodiment, polypeptides, antibodies, polynucleotides and/or
agonists or antagonists of the present invention are administered 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 irmnune system from another animal (see, e.g., published PCT
Application Nos. W098/24893, WO/9634096, WO/9633735, and WO/9110741).
Administration of polypeptides, antibodies, polynucleotides and/or agonists or
antagonists of the present invention to such animals is useful for the
generation of
monoclonal antibodies against the polypeptides, antibodies, polynucleotides
and/or
agonists or antagonists of the present invention in an organ system listed
above.
Slood-Related Disorders
The polynucleotides, polypeptides, antibodies, and/or agonists or antagonists
of the present invention may be used to modulate hemostatic (the stopping of
bleeding) or thrombolytic (clot dissolving) activity. 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, hemophilia), 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
200
important in the treatment or prevention of heart attacks (infarction),
strolces, or
scarring.
In specific embodiments, the polynucleotides, polypeptides, antibodies, and/or
agonists or antagonists of the present invention may be used to prevent,
diagnose,
pragnose, and/or treat thrombosis, arterial thrombosis, venous thrombosis,
thromboembolism, pulmonary embolism, atherosclerosis, myocardial infarction,
transient ischemic attack, unstable angina. In specific embodiments, the
polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of
the
present invention may be used for the prevention of occulsion of saphenous
grafts, for
reducing the risk of periprocedural thrombosis as might accompany angioplasty
procedures, for reducing the risk of stroke in patients with atrial
fibrillation including
nonrheumatic atrial fibrillation, for reducing the risk of embolism associated
with
mechanical heart valves and or mural valves disease. Other uses for the
polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of
the
present invention, include, but are not limited to, the prevention of
occlusions in
extrcorporeal devices (e.g., intravascular canulas, vascular access shunts in
hemodialysis patients, hemodialysis machines, and cardiopulmonary bypass
machines).
W another embodiment, a polypeptide of the invention, or polynucleotides,
antibodies, agonists, or antagonists corresponding to that polypeptide, may be
used to
prevent, diagnose, prognose, and/or treat diseases and disorders of the blood
and/or
blood forming organs associated with the tissues) in which the polypeptide of
the
invention is expressed, including one, two, three, four, five, or more tissues
disclosed
in Table 1, column 8 (Tissue Distribution Library Code).
The polynucleotides, polypeptides, antibodies, andlor agonists or antagonists
of the present invention may be used to modulate hematopoietic activity (the
formation of blood cells). For example, the polynucleotides, polypeptides,
antibodies,
andlor agonists or antagonists of the present invention may be used to
increase the
quantity of all or subsets of blood cells, such as, for example, erythrocytes,
lymphocytes (B or T cells), myeloid cells (e.g., basophils, eosinophils,
neutrophils,
mast cells, macrophages) and platelets. The ability to decrease the quantity
of blood
cells or subsets of blood cells may be useful in the prevention, detection,
diagnosis

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
201
and/or treatment of anemias and leulcopenias described below. Alternatively,
the
polynucleotides, polypeptides, antibodies, and/or agonsts or antagonists of
the
present invention may be used to decrease the quantity of all or subsets of
blood cells,
such as, for example, erythrocytes, lymphocytes (B or T cells), myeloid cells
(e.g.,
basophils, eosinophils, neutrophils, mast cells, macrophages) and platelets..
The
ability to decrease the quantity of blood cells or subsets of blood cells may
be useful
in the prevention, detection, diagnosis and/or treatment of leukocytoses, such
as, fox
example eosinophilia.
The polynucleotides, polypeptides, antibodies, and/or agonists or antagonists
of the present invention may be used to prevent, treat, or diagnose blood
dyscrasia.
Anemias are conditions in which the number of red blood cells or amount of
hemoglobin (the protein that carries oxygen) in them is below normal. Anemia
may
be caused by excessive bleeding, decreased red blood cell production, or
increased
red blood cell destruction (hemolysis). The polynucleotides, polypeptides,
antibodies,
and/or agonists or antagonists of the present invention may be useful in
treating,
preventing, and/or diagnosing anemias. Anemias that may be treated prevented
or
diagnosed by the polynucleotides, polypeptides, antibodies, and/or agonists or
antagonists of the present invention include iron deficiency anemia,
hypochromic
anemia, microcytic anemia, chlorosis, hereditary siderob;astic anemia,
idiopathic
acquired sideroblastic anemia, red cell aplasia, megaloblastic anemia (e.g.,
pernicious
anemia, (vitamin B12 deficiency) and folic acid deficiency anemia), aplastic
anemia,
hemolytic amemias (e.g., autoimmune helolytic anemia, microangiopathic
hemolytic
anemia, and paroxysmal nocturnal hemoglobinuria). The polynucleotides,
polypeptides, antibodies, and/ox agonists or antagonists of the present
invention may
be useful in treating, preventing, and/or diagnosing anemias associated with
diseases
including but not limited to, anemias associated with systemic lupus
erythematosus,
cancers, lymphomas, chronic renal disease, and enlarged spleens. The
polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of
the
present invention may be useful in treating, preventing, and/or diagnosing
anemias
arising from drug treatments such as anemias associated with methyldopa,
dapsone,
and/or sulfadrugs. Additionally, the polynucleotides, polypeptides,
antibodies, and/or
agonists or antagonists of the present invention may be useful in treating,
preventing,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
202
and/or diagnosing anemias associated with abnormal red blood cell architecture
including but not limited to, hereditary spherocytosis, hereditary
elliptocytosis,
glucose-6-phosphate dehydrogenase deficiency, and sickle cell anemia.
The polynucleotides, polypeptides, antibodies, and/or agonists or antagonists
of the present invention may be useful in treating, preventing, and/or
diagnosing
hemoglobin abnormalities, (e.g., those associated with sickle cell anemia,
hemoglobin
C disease, hemoglobin S-C disease, and hemoglobin E disease). Additionally,
the
polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of
the
present invention may be useful in diagnosing, prognosing, preventing, and/or
treating
thalassemias, including, but not limited to major and minor forms of alpha-
thalassemia and beta-thalassemia.
In another embodiment, the polynucleotides, polypeptides, antibodies, and/or
agonists or antagonists of the present invention may be useful in diagnosing,
prognosing, preventing, and/or treating bleeding disorders including, but not
limited
to, thrombocytopenia (e.g., idiopathic thrombocytopenic purpura, and
thrombotic
thrombocytopenic purpura), Von Willebrand's disease, hereditary platelet
disorders
(e.g., storage pool disease such as Chediak-Higashi and Hermansky-Pudlak
syndromes, thromboxane A2 dysfunction, thromboasthenia, and Bernard-Soulier
syndrome), hemolytic-uremic syndrome, hemophelias such as hemophelia A or
Factor
VII deficiency and Christmas disease or Factor IX deficiency, Hereditary
Hemorhhagic Telangiectsia, also known as Rendu-Osler-Weber syndrome, allergic
purpura (Henoch Schonlein purpura) and disseminated intravascular coagulation.
The effect of the polynucleotides, polypeptides, antibodies, and/or agonists
or
antagonists of the present invention on the clotting time of blood may be
monitored
using any of the clotting tests known in the art including, but not limited
to, whole
blood partial thromboplastin time (PTT), the activated partial thromboplastin
time
(aPTT), the activated clotting time (ACT), the recalcified activated clotting
time, or
the Lee-White Clotting time.
Several diseases and a variety of drugs can cause platelet dysfunction. Thus,
in
a specific embodiment, the polynucleotides, polypeptides, antibodies, and/or
agonists
or antagonists of the present invention may be useful in diagnosing,
prognosing,
preventing, and/or treating acquired platelet dysfunction such as platelet
dysfunction

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
203
accompanying kidney failure, leukemia, multiple myeloma, cirrhosis of the
liver, and
systemic lupus erythematosus as well as platelet dysfunction associated with
drug
treatments, including treatment with aspirin, ticlopidine, nonsteroidal anti-
inflammatory drugs (used for arthritis, pain, and sprains), and penicillin in
high doses.
In another embodiment, the polynucleotides, polypeptides, antibodies, and/or
agonists or antagonists of the present invention may be useful in diagnosing,
prognosing, preventing, and/or treating diseases and disorders characterized
by or
associated with increased or decreased numbers of white blood cells.
Leukopenia
occurs when the number of white blood cells decreases below normal.
Leukopenias
include, but are not limited to, neutropenia and lymphocytopenia. An increase
in the
number of white blood cells compared to normal is known as leukocytosis. The
body
generates increased numbers of white blood cells during infection. Thus,
leukocytosis
may simply be a normal physiological parameter that reflects infection.
Alternatively,
leukocytosis may be an indicator of injury or other disease such as cancer.
Leokocytoses, include but are not limited to, eosinophilia, and accumulations
of
macrophages. In specific embodiments, the polynucleotides, polypeptides,
antibodies, and/or agonists or antagonists of the present invention may be
useful in
diagnosing, prognosing, preventing, and/or treating leukopenia. In other
specific
embodiments, the polynucleotides, polypeptides, antibodies, and/or agonists or
antagonists of the present invention may be useful in diagnosing, prognosing,
preventing, and/or treating leukocytosis.
Leukopenia may be a generalized decreased in all types of white blood cells,
or may be a specific depletion of particular types of white blood cells. Thus,
in
specific embodiments, the polynucleotides, polypeptides, antibodies, and/or
agonists
or antagonists of the present invention may be useful in diagnosing,
prognosing,
preventing, and/or treating decreases in neutrophil numbers, known as
neutropenia.
Neutropenias that may be diagnosed, prognosed, prevented, and/or treated by
the
polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of
the
present invention include, but are not limited to, infantile genetic
agranulocytosis,
familial neutropenia, cyclic neutropenia, neutropenias resulting from or
associated
with dietary deficiencies (e.g., vitamin B 12 deficiency or folic acid
deficiency),
neutropenias resulting from or associated with drug treatments (e.g.,
antibiotic

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
204
regimens such as penicillin treatment, sulfonamide treatment, anticoagulant
treatment,
anticonvulsant drugs, anti-thyroid drugs, and cancer chemotherapy), and
neutropenias
resulting from increased neutrophil destruction that may occur in association
with
some bacterial or viral infections, allergic disorders, autoimmune diseases,
conditions
in which an individual has an enlarged spleen (e.g., Felty syndrome, malaria
and
sarcoidosis), and some drug treatment regimens.
The polynucleotides, polypeptides, antibodies, and/or agonists or antagonists
of the present invention may be useful in diagnosing, prognosing, preventing,
and/or
treating lymphocytopenias (decreased numbers of B and/or T lymphocytes),
including, but not limited lymphocytopenias resulting from or associated with
stress,
drug treatments (e.g., drug treatment with corticosteroids, cancer
chemotherapies,
and/or radiation therapies), AIDS infection andlor other diseases such as, for
example,
cancer, rheumatoid arthritis, systemic lupus erythematosus, chronic
infections, some
viral infections and/or hereditary disorders (e.g., DiGeorge syndrome, Wiskott-
Aldrich Syndome, severe combined immunodeficiency, ataxia telangiectsia).
The polynucleotides, polypeptides, antibodies, and/or agonists or antagonists
of the present invention may be useful in diagnosing, prognosing, preventing,
and/or
treating diseases and disorders associated with macrophage numbers and/or
macrophage function including, but not limited to, Gaucher's disease, Niemann-
Pick
disease, Letterer-Siwe disease and Hand-Schuller-Christian disease.
hi another embodiment, the polynucleotides, polypeptides, antibodies, andlor
agonists or antagonists of the present invention may be useful in diagnosing,
prognosing, preventing, and/or treating diseases and disorders associated with
eosinophil numbers andlor eosinophil function including, but not limited to,
idiopathic hypereosinophilic syndrome, eosinophilia-myalgia syndrome, and Hand-
Schuller-Christian disease.
In yet another embodiment, the polynucleotides, polypeptides, antibodies,
and/or agonists or antagonists of the present invention may be useful in
diagnosing,
prognosing, preventing, and/or treating leukemias and lymphomas including, but
not
limited to, acute lymphocytic (lymphpblastic) leukemia (ALL), acute myeloid
(myelocytic, myelogenous, myeloblastic, or myelomonocytic) leukemia, chronic
lymphocytic leukemia (e.g., B cell leukemias, T cell leukemias, Sezary
syndrome, and

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
205
Hairy cell leukenia), chronic myelocytic (myeloid, myelogenous, or
granulocytic)
leukemia, Hodglcin's lymphoma, non-hodgkin's lymphoma, Burkitt's lymphoma, and
mycosis fungoides.
In other embodiments, the polynucleotides, polypeptides, antibodies, and/or
agonists or antagonists of the present invention may be useful in diagnosing,
prognosing, preventing, and/or treating diseases and disorders ofplasma cells
including, but not limited to, plasma cell dyscrasias, monoclonal
gammaopathies,
monoclonal gammopathies of undetermined significance, multiple myeloma,
macroglobulinemia, Waldenstrom's macroglobulinemia, cryoglobulinemia, and
Raynaud's phenomenon.
In other embodiments, the polynucleotides, polypeptides, antibodies, and/or
agonists or antagonists of the present invention may be useful in treating,
preventing,
and/or diagnosing myeloproliferative disorders, including but not limited to,
polycythemia vera, relative polycythemia, secondary polycythemia,
myelofibrosis,
acute myelofibrosis, agnogenic myelod metaplasia, thrombocythemia, (including
both
primary and seconday thrombocythemia) and chronic myelocytic leukemia.
In other embodiments, the polynucleotides, polypeptides, antibodies, and/or
agonists or antagonists of the present invention may be useful as a treatment
prior to
surgery, to increase blood cell production.
In other embodiments, the polynucleotides, polypeptides, antibodies, and/or
agonists or antagonists of the present invention may be useful as an agent to
enhance
the migration, phagocytosis, superoxide production, antibody dependent
cellular
cytotoxicity of neutrophils, eosionophils and macrophages.
In other embodiments, the polynucleotides, polypeptides, antibodies, and/or
agonists or antagonists of the present invention may be useful as an agent to
increase
the number of stem cells in circulation prior to stem cells pheresis. In
another specific
embodiment, the polynucleotides, polypeptides, antibodies, and/or agonists or
antagonists of the present invention may be useful as an agent to increase the
number
of stem cells in circulation prior to platelet pheresis.
In other embodiments, the polynucleotides, polypeptides, antibodies, and/or
agonists or antagonists of the present invention may be useful as an agent to
increase
cytokine production.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
206
In other embodiments, the polynucleotides, polypeptides, antibodies, and/or
agonists or antagonists of the present invention may be useful in preventing,
diagnosing, and/or treating primary hematopoietic disorders.
Hyperproliferative Disorders
In certain embodiments, polynucleotides or polypeptides, or agonists or
antagonists of the present invention can be used to treat or detect
hyperproliferative
disorders, including neoplasms. Polynucleotides or polypeptides, or agonists
or
antagonists of the present invention may inhibit the proliferation of the
disorder
through direct or indirect interactions. Alternatively, Polynucleotides or
polypeptides,
or agonists or antagonists of the present invention may proliferate other
cells which
can inhibit the hyperproliferative disorder.
For example, by increasing an immune response, particularly increasing
antigenic qualities of the hyperproliferative disorder or by proliferating,
differentiating, or mobilizing T-cells, hyperproliferative disorders can be
treated.
This immune response may be increased by either enhancing an existing immune
response, or by initiating a new immune response. Alternatively, decreasing an
immune response may also be a method of treating hyperproliferative disorders,
such
as a chemotherapeutic agent.
Examples of hyperproliferative disorders that can be treated or detected by
polynucleotides or polypeptides, or agonists or antagonists of the present
invention
include, but are not limited to neoplasms located in the: colon, abdomen,
bone, breast,
digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal,
parathyroid,
pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous
(central and
peripheral), lymphatic system, pelvis, skin, soft tissue, spleen, thorax, and
urogenital
tract.
Similarly, other hyperproliferative disorders can also be treated or detected
by
polynucleotides or polypeptides, or agonists or antagonists of the present
invention.
Examples of such hyperproliferative disorders include, but are not limited to:
Acute
Childhood Lymphoblastic Leukemia, Acute Lymphoblastic Leukemia, Acute
Lymphocytic Leukemia, Acute Myeloid Leukemia, Adrenocortical Carcinoma, Adult
(Primary) Hepatocellular Cancer, Adult (Primary) Liver Cancer, Adult Acute

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
207
Lymphocytic Leukemia, Adult Acute Myeloid Leukemia, Adult Hodgkin's Disease,
Adult Hodgkin's Lymphoma, Adult Lymphocytic Leukemia, Adult Non-Hodgkin's
Lymphoma, Adult Primary Liver Cancer, Adult Soft Tissue Sarcoma, AIDS-Related
Lymphoma, AIDS-Related Malignancies, Anal Cancer, Astrocytoma, Bile Duct
Cancer, Bladder Cancer, Bone Cancer, Brain Stem Glioma, Brain Tumors, Breast
Cancer, Cancer of the Renal Pelvis and Ureter, Central Nervous System
(Primary)
Lymphoma, Central Nervous System Lymphoma, Cerebellar Astrocytoma, Cerebral
Astrocytoma, Cervical Cancer, Childhood (Primary) Hepatocellular Cancer,
Childhood (Primary) Liver Cancer, Childhood Acute Lymphoblastic Leukemia,
Childhood Acute Myeloid Leukemia, Childhood Brain Stem Glioma, Childhood
Cerebellar Astrocytoma, Childhood Cerebral Astrocytoma, Childhood Extracranial
Germ Cell Tumors, Childhood Hodgkin's Disease, Childhood Hodgkin's Lymphoma,
Childhood Hypothalamic and Visual Pathway Glioma, Childhood Lymphoblastic
Leukemia, Childhood Medulloblastoma, Childhood Non-Hodgkin's Lymphoma,
Childhood Pineal and Supratentorial Primitive Neuroectodermal Tumors,
Childhood
Primary Liver Cancer, Childhood Rhabdomyosarcoma, Childhood Soft Tissue
Sarcoma, Childhood Visual Pathway and Hypothalamic Glioma, Chronic
Lymphocytic Leukemia, Chronic Myelogenous Leukemia, Colon Cancer, Cutaneous
T-Cell Lymphoma, Endocrine Pancreas Islet Cell Carcinoma, Endometrial Cancer,
Ependymoma, Epithelial Cancer, Esophageal Cancer, Ewing's Sarcoma and Related
Tumors, Exocrine Pancreatic Cancer, Extracranial Germ Cell Tumor, Extragonadal
Germ Cell Tumor, Extrahepatic Bile Duct Cancer, Eye Cancer, Female Breast
Cancer, Gaucher's Disease, Gallbladder Cancer, Gastric Cancer,
Gastrointestinal
Carcinoid Tumor, Gastrointestinal Tumors, Germ Cell Tumors, Gestational
Trophoblastic Tumor, Hairy Cell Leukemia, Head and Neck Cancer, Hepatocellular
Cancer, Hodgkin's Disease, Hodgkin's Lymphoma, Hypergasnmaglobulinemia,
Hypopharyngeal Cancer, Intestinal Cancers, Intraocular Melanoma, Islet Cell
Carcinoma, Islet Cell Pancreatic Cancer, Kaposi's Sarcoma, Kidney Cancer,
Laryngeal Cancer, Lip and Oral Cavity Cancer, Liver Cancer, Lung Cancer,
Lymphoproliferative Disorders, Macroglobulinemia, Male Breast Cancer,
Malignant
Mesothelioma, Malignant Thymoma, Medulloblastoma, Melanoma, Mesothelioma,
Metastatic Occult Primary Squamous Neck Cancer, Metastatic Primary Squamous

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
208
Neclc Cancer, Metastatic Squamous Neck Cancer, Multiple Myeloma, Multiple
MyelomalPlasma Cell Neoplasm, Myelodysplastic Syndrome, Myelogenous
Leukemia, Myeloid Leukemia, Myeloproliferative Disorders, Nasal Cavity and
Paranasal Sinus Cancer, Nasopharyngeal Cancer, Neuroblastoma, Non-Hodgkin's
Lymphoma During Pregnancy, Nonmelanoma Skin Cancer, Non-Small Cell Lung
Cancer, Occult Primary Metastatic Squamous Neck Cancer, Oropharyngeal Cancer,
Osteo-(Malignant Fibrous Sarcoma, Osteosarcoma/Malignant Fibrous Histiocytoma,
OsteosarcomalMalignant Fibrous Histiocytoma of Bone, Ovarian Epithelial
Cancer,
Ovarian Germ Cell Tumor, Ovarian Low Malignant Potential Tumor, Pancreatic
Cancer, Paraproteinemias, Purpura, Parathyroid Cancer, Penile Cancer,
Pheochromocytoma, Pituitary Tumor, Plasma Cell Neoplasm/Multiple Myeloma,
Primary Central Nervous System Lymphoma, Primary Liver Cancer, Prostate
Cancer,
Rectal Cancer, Renal Cell Cancer, Renal Pelvis and Ureter Cancer,
Retinoblastoma,
Rhabdomyosarcoma, Salivary Gland Cancer, Sarcoidosis Sarcomas, Sezary
Syndrome, Skin Cancer, Small Cell Lung Cancer, Small Intestine Cancer, Soft
Tissue
Sarcoma, Squamous Neclc Cancer, Stomach Cancer, Supratentorial Primitive
Neuroectodermal and Pineal Tumors, T-Cell Lymphoma, Testicular Cancer,
Thymoma, Thyroid Cancer, Transitional Cell Cancer of the Renal Pelvis and
Ureter,
Transitional Renal Pelvis and Ureter Cancer, Trophoblastic Tumors, Ureter and
Renal
Pelvis Cell Cancer, Urethral Cancer, Uterine Cancer, Uterine Sarcoma, Vaginal
Cancer, Visual Pathway and Hypothalamic Glioma, Vulvar Cancer, Waldenstrom's
Macroglobulinemia, Wilms' Tumor, and any other hyperproliferative disease,
besides
neoplasia, located in an organ system listed above.
In another preferred embodiment, polynucleotides or polypeptides, or agonists
or antagonists of the present invention are used to diagnose, pragnose,
prevent, and/or
treat premalignant conditions and to prevent progression to a neoplastic or
malignant
state, including but not limited to those disorders described above. Such uses
are
indicated in conditions known or suspected of preceding progression to
neoplasia or
cancer, in particular, where non-neoplastic cell growth consisting of
hyperplasia,
metaplasia, or most particularly, dysplasia has occurred (for review of such
abnormal
growth conditions, see Robbins and Angell, 1976, Basic Pathology, 2d Ed., W.
B.
Saunders Co., Philadelphia, pp. 68-79.)

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
209
Hyperplasia is a form of controlled cell proliferation, involving an increase
in
cell number in a tissue or organ, without significant alteration in structure
or function.
Hyperplastic disorders which can be diagnosed, prognosed, prevented, and/or
treated
with compositions of the invention (including polynucleotides, polypeptides,
agonists
or antagonists) include, but are not limited to, angiofollicular mediastinal
lymph node
hyperplasia, angiolymphoid hyperplasia with eosinophilia, atypical melanocytic
hyperplasia, basal cell hyperplasia, benign giant lymph node hyperplasia,
cementum
hyperplasia, congenital adrenal hyperplasia, congenital sebaceous hyperplasia,
cystic
hyperplasia, cystic hyperplasia of the breast, denture hyperplasia, ductal
hyperplasia,
endometrial hyperplasia, fibromuscular hyperplasia, focal epithelial
hyperplasia,
gingival hyperplasia, inflammatory fibrous hyperplasia, inflarmnatory
papillary
hyperplasia, intravascular papillary endothelial hyperplasia, nodular
hyperplasia of
prostate, nodular regenerative hyperplasia, pseudoepitheliomatous hyperplasia,
senile sebaceous hyperplasia, and verrucous hyperplasia.
Metaplasia is a form of controlled cell growth in which one type of adult or
fully differentiated cell substitutes for another type of adult cell.
Metaplastic disorders
which can be diagnosed, prognosed, prevented, and/or treated with compositions
of
the invention (including polynucleotides, polypeptides, agonists or
antagonists)
include, but are not limited to, agnogenic myeloid metaplasia, apocrine
metaplasia,
atypical metaplasia, autoparenchymatous metaplasia, connective tissue
metaplasia,
epithelial metaplasia, intestinal metaplasia, metaplastic anemia, metaplastic
ossification, metaplastic polyps, myeloid metaplasia, primary myeloid
metaplasia,
secondary myeloid metaplasia, squamous metaplasia, squamous metaplasia of
amnion, and symptomatic myeloid metaplasia.
Dysplasia is frequently a forerunner of cancer, and is found mainly in the
epithelia; it is the most disorderly form of non-neoplastic cell growth,
involving a loss
in individual cell uniformity and in the architectural orientation of cells.
Dysplastic
cells often have abnormally large, deeply stained nuclei, and exhibit
pleomorphism.
Dysplasia characteristically occurs where there exists chronic irritation or
inflammation. Dysplastic disorders which can be diagnosed, prognosed,
prevented,
and/or treated with compositions of the invention (including polynucleotides,
polypeptides, agonists or antagonists) include, but are not limited to,
anhidrotic

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
210
ectodermal dysplasia, anterofacial dysplasia, asphyxiating thoracic dysplasia,
atriodigital dysplasia, bronchopulmonary dysplasia, cerebral dysplasia,
cervical
dysplasia, chondroectodermal dysplasia, cleidocranial dysplasia, congenital
ectodermal dysplasia, craniodiaphysial dysplasia, craniocarpotarsal dysplasia,
craniometaphysial dysplasia, dentin dysplasia, diaphysial dysplasia,
ectodermal
dysplasia, enamel dysplasia, encephalo-ophthalmic dysplasia, dysplasia
epiphysialis
hemimelia, dysplasia epiphysialis multiplex, dysplasia epiphysialis punctata,
epithelial dysplasia, faciodigitogenital dysplasia, familial fibrous dysplasia
of jaws,
familial white folded dysplasia, fibromuscular dysplasia, fibrous dysplasia of
bone,
florid osseous dysplasia, hereditary renal-retinal dysplasia, hidrotic
ectodermal
dysplasia, hypohidrotic ectodennal dysplasia, lyrnphopenic thyrnic dysplasia,
mammary dysplasia, mandibulofacial dysplasia, metaphysial dysplasia, Mondini
dysplasia, monostotic fibrous dysplasia, mucoepithelial dysplasia, multiple
epiphysial
dysplasia, oculoauriculovertebral dysplasia, oculodentodigital dysplasia,
oculovertebral dysplasia, odontogenic dysplasia, ophthalmomandibulomelic
dysplasia, periapical cemental dysplasia, polyostotic fibrous dysplasia,
pseudoachondroplastic spondyloepiphysial dysplasia, retinal dysplasia, septo-
optic
dysplasia, spondyloepiphysial dysplasia, and ventriculoradial dysplasia.
Additional pre-neoplastic disorders which can be diagnosed, prognosed,
prevented, and/or treated with compositions of the invention (including
polynucleotides, polypeptides, agonists or antagonists) include, but are not
limited to,
benign dysproliferative disorders (e.g., benign tumors, fibrocystic
conditions, tissue
hypertrophy, intestinal polyps, colon polyps, and esophageal dysplasia),
leukoplakia,
keratoses, Bowen's disease, Farmer's Skin, solar cheilitis, and solar
keratosis.
In another embodiment, a polypeptide of the invention, or polynucleotides,
antibodies, agonists, or antagonists corresponding to that polypeptide, may be
used to
diagnose and/or prognose disorders associated with the tissues) in which the
polypeptide of the invention is expressed, including one, two, three, four,
five, or
more tissues disclosed in Table l, column 8 (Tissue Distribution Library
Code).
In another 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 cancers and neoplasms,
including,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
211
but not limited to those described herein. 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 acute myelogenous leukemia.
Additionally, polynucleotides, polypeptides, and/or agonists 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 diagnosed, prognosed, prevented, and/or
treated
by polynucleotides, polypeptides, andlor agonists 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, biliaiy cirrhosis, Behcet's disease,
Crohn's
disease, polymyositis, systemic lupus erythematosus and immune-related
glomerulonephritis and rheumatoid arthritis) and viral infections (such as
herpes
viruses, pox viruses and adenoviruses), inflammation, graft v. host disease,
acute graft
rejection, and chronic graft rejection.
In. preferred embodiments, polynucleotides, polypeptides, and/or agonists or
antagonists of the invention are used to inhibit growth, progression, and/or
metastasis
of cancers, in particular those listed above.
Additional diseases or conditions associated with increased cell survival that
could be diagnosed, prognosed, prevented, and/or treated by polynucleotides,
polypeptides, and/or 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
212
(granulocytic) leukemia and chronic lymphocytic leulcemia)), 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, leiomyosarcorna, 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, emangioblastoma, acoustic neuroma, oligodendroglioma, menangioma,
melanoma, neuroblastoma, and retinoblastoma.
Diseases associated with increased apoptosis that could be diagnosed,
prognosed, prevented, and/or treated by polynucleotides, polypeptides, and/or
agonists or antagonists of the invention, include AIDS; neurodegenerative
disorders
(such as Alzheimer's, disease, Parkinson's disease, amyotrophic lateral
sclerosis,
retinitis pigmentosa, cerebellar degeneration and brain tumor or prior
associated
disease); autoimmune disorders (such as, multiple sclerosis, Sjogren's
syndrome,
Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crolm'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,
ischemialreperfusion injury, cholestosis (bile duct injury) and liver cancer);
toxin
induced liver disease (such as that caused by alcohol), septic shock, cachexia
and
anorexia.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
213
Hypeiproliferative diseases and/or disorders that could be diagnosed,
prognosed, prevented, and/or treated by polynucleotides, polypeptides, and/or
agonists 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 system (central and peripheral), lymphatic system,
pelvis, skin, soft tissue, spleen, thorax, and urogenital tract.
Similarly, other hyperproliferative disorders can also be diagnosed,
prognosed,
prevented, and/or treated by polynucleotides, polypeptides, and/or agonists 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.
Another preferred embodiment utilizes polynucleotides of the present
invention to inhibit aberrant cellular division, by gene therapy using the
present
invention, and/or protein fusions or fragments thereof.
Thus, the present invention provides a method for treating cell proliferative
disorders by inserting into an abnormally proliferating cell a polynucleotide
of the
present invention, wherein said polynucleotide represses said expression.
Another embodiment of the present invention provides a method of treating
cell-proliferative disorders in individuals comprising administration of one
or more
active gene copies of the present invention to an abnormally proliferating
cell or cells.
In a preferred embodiment, polynucleotides of the present invention is a DNA
construct comprising a recombinant expression vector effective in expressing a
DNA
sequence encoding said polynucleotides. Tn another preferred embodiment of the
present invention, the DNA construct encoding the poynucleotides of the
pxesent
invention is inserted into cells to be treated utilizing a retrovirus, or more
preferably
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
214
inserted into proliferating cells either alone, or in combination with or
fused to other
polynucleotides, can then be modulated via an external stimulus (i.e.
magnetic,
specific small molecule, chemical, or drug administration, etc.), which acts
upon the
promoter upstream of said polynucleotides to induce expression of the encoded
protein product. As such the beneficial therapeutic affect of the present
invention
may be expressly modulated (i.e. to increase, decrease, or inhibit expression
of the
present invention) based upon said external stimulus.
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 l~nown 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
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 (Pates 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
aiid 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
215
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.
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 fox treating one or more of the described
disorders. Methods for producing anti-polypeptides and anti-polynucleotide
antibodies polyclonal and monoclonal antibodies are described in detail
elsewhere
herein. Such antibodies may be provided in pharmaceutically acceptable
compositions as known in the art or as described herein.
A summary of the ways in which the antibodies of the present invention may
be used therapeutically includes binding polynucleotides or polypeptides of
the
present invention locally or systemically in the body or by direct
cytotoxicity of the
antibody, e.g. as mediated by complement {CDC) or by effector cells {ADCC).
Some

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
216
of these approaches are described in more detail below. Armed with the
teachings
provided herein, one of ordinary skill in the art will know how to use the
antibodies of
the present invention for diagnostic, monitoring or therapeutic purposes
without
undue experimentation.
In particular, the antibodies, fragments and derivatives of the present
invention
are useful for treating a subject having or developing cell proliferative
and/or
differentiation disorders as described herein. Such treatment comprises
administering
a single or multiple doses of the antibody, or a fragment, derivative, or a
conjugate
thereof.
The antibodies of this invention may be advantageously utilized in
combination with other monoclonal or chimeric antibodies, or with lymphokines
or
hematopoietic growth factors, for example., which serve to increase the number
or
activity of effector cells which interact with the antibodies.
It is preferred to use high affinity and/or potent in vivo inhibiting and/or
neutralizing antibodies against polypeptides or polynucleotides of the present
invention, fragments or regions thereof, for both immunoassays directed to and
therapy of disorders related to polynucleotides or polypeptides, including
fragements
thereof, of the present invention. Such antibodies, fragments, or regions,
will
preferably have an affinity for polynucleotides or polypeptides, including
fragements
thereof. Preferred binding affinities include those with a dissociation
constant or Kd
less than SX10-6M, 10-6M, SX10-~M, 10-~M, SX10-8M, 10-8M, SX10-9M, 10-9M,
SX10-1°M, 10-1°M, SX10-11M, 10-11M, SX10-12M, 10~12M, SX10-13M,
10-13M, SX10-
14M, 10-14M, SX10-15M, and 10-15M.
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.,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
217
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 (TR.AMP) 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, anti-inflammatory
proteins
(See for example, Mutat Res 400(1-2):447-55 (1998), Med Hypotheses.50(5):423-
33
(1998), Chem Biol Interact. Apr 24;111-112:23-34 (1998), J Mol Med.76(6):402-
12
(1998), Int J Tissue React;20(1):3-15 (1998), which are all hereby
incorporated by
reference).
Polypeptides, including protein fusions to, or fragments thereof, of the
present
invention are useful in inhibiting the metastasis of proliferative cells or
tissues.
Inhibition may occur as a direct result of administering polypeptides, or
antibodies
directed to said polypeptides as described elsewere herein, or indirectly,
such as
activating the expression of proteins known to inhibit metastasis, for example
alpha 4
integrins, (See, e.g., Curr Top Microbiol Immunol 1998;231:125-41, which is
hereby
incorporated by reference). Such thereapeutic affects of the present invention
may be
achieved either alone, or in combination with small molecule drugs or
adjuvants.
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
218
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.
Renal Disorders
Polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of
the
present invention, may be used to treat, prevent, diagnose, and/or prognose
disorders
of the renal system. Renal disorders which can be diagnosed, prognosed,
prevented,
and/or treated with compositions of the invention include, but are not limited
to,
kidney failure, nephritis, blood vessel disorders of kidney, metabolic and
congenital
kidney disorders, urinary disorders of the kidney, autoirnlnune disorders,
sclerosis and
necrosis, electrolyte imbalance, and kidney cancers.
Kidney diseases which can be diagnosed, prognosed, prevented, and/or treated
with compositions of the invention include, but are not limited to, acute
kidney
failure, chronic kidney failure, atheroembolic renal failure, end-stage renal
disease,
inflammatory diseases of the kidney (e.g., acute glomerulonephritis,
postinfectious
glomerulonephritis, rapidly progressive glomerulonephritis, nephrotic
syndrome,
membranous glomerulonephritis, familial nephrotic syndrome,
membranoproliferative
glomerulonephritis I and II, mesangial proliferative glomerulonephritis,
chronic
glomerulonephritis, acute tubulointerstitial nephritis, chronic
tubulointerstitial
nephritis, acute post-streptococcal glomerulonephritis (PSGN), pyelonephritis,
lupus
nephritis, chronic nephritis, interstitial nephritis, and post-streptococcal
glomerulonephritis), blood vessel disorders of the kidneys (e.g., kidney
infarction,
atheroembolic kidney disease, cortical necrosis, malignant nephrosclerosis,
renal vein
thrombosis, renal underperfusion, renal retinopathy, renal ischemia-
reperfusion, renal

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
219
artery embolism, and renal artery stenosis), and kidney disorders resulting
form
urinary tract disease (e.g., pyelonephritis, hydronephrosis, urolithiasis
(renal lithiasis,
nephrolithiasis), reflux nephropathy, urinary tract infections, urinary
retention, and
acute or chronic unilateral obstructive uropathy.)
In addition, compositions of the invention can be used to diagnose, prognose,
prevent, and/or treat metabolic and congenital disorders of the kidney (e.g.,
uremia,
renal amyloidosis, renal osteodystrophy, renal tubular acidosis, renal
glycosuria,
nephrogenic diabetes insipidus, cystinuria, Fanconi's syndrome, renal
fibrocystic
osteosis (renal rickets), Hartnup disease, Banter's syndrome, Liddle's
syndrome,
1-0 polycystic kidney disease, medullary cystic disease, medullary sponge
kidney,
Alport's syndrome, nail-patella syndrome, congenital nephrotic syndrome, CRUSH
syndrome, horseshoe kidney, diabetic nephropathy, nephrogenic diabetes
insipidus,
analgesic nephropathy, kidney stones, and membranous nephropathy), and
autoimmune disorders of the kidney (e.g., systemic lupus erythematosus (SLE),
15 Goodpasture syndrome, IgA nephxopathy, and IgM mesangial proliferative
glomerulonephritis).
Compositions of the invention can also be used to diagnose, prognose,
prevent, andlor treat sclerotic or necrotic disorders of the kidney (e.g.,
glomerulosclerosis, diabetic nephropathy, focal segmental glomerulosclerosis
20 (FSGS), necrotizing glomerulonephritis, and renal papillary necrosis),
cancers of the
kidney (e.g., nephroma, hypernephroma, nephroblastoma, renal cell cancer,
transitional cell cancer, renal adenocarcinoma, squamous cell cancer, and
Wilm's
tumor), and electrolyte imbalances (e.g., nephrocalcinosis, pyuria, edema,
hydronephritis, proteinuria, hyponatremia, hypernatremia, hypokalemia,
25 hyperkalemia, hypocalcemia, hypercalcemia, hypophosphatemia, and
' hyperphosphatemia).
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
30 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
220
art. Polypeptides may be administered as part of a Therapeutic, described in
more
detail below. Methods of delivering polynucleotides are described in more
detail
herein.
Cardiovascular Disorders
Polynucleotides or polypeptides, or agonists or antagonists of the present
invention, may be used to treat, prevent, diagnose, and/or prognose
cardiovascular
disorders, including, but not limited to, peripheral artery disease, such as
limb
ischemia.
Cardiovascular disorders include, but are not limited to, cardiovascular
abnormalities, such as arterio-arterial fistula, arteriovenous fistula,
cerebral
arteriovenous malformations, congenital heart defects, pulmonary atresia, and
Scimitar Syndrome. Congenital heart defects include, but are not limited to,
aortic
coarctation, cor triatriatum, coronary vessel anomalies, crisscross heart,
dextrocardia,
patent ductus arteriosus, Ebstein's anomaly, Eisenmenger complex, hypoplastic
left
heart syndrome, levocardia, tetralogy of fallot, transposition of great
vessels, double
outlet right ventricle, tricuspid atresia, persistent truncus arteriosus, and
heart septal
defects, such as aortopulmonary septal defect, endocardial cushion defects,
Lutembacher's Syndrome, trilogy of Fallot, ventricular heart septal defects.
Cardiovascular disorders also include, but are not limited to, 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, but are not limited to, sinus arrhythmia, atrial
fibrillation, atrial flutter, bradycardia, extrasystole, Adams-Stokes
Syndrome, bundle-

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
221
branch block, sinoatrial block, long QT syndrome, parasystole, Lown-Ganong-
Levine
Syndrome, Mahaim-type pre-excitation syndrome, Wolff Parlcinson-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 diseases include, but are not limited to, aortic valve
insufficiency,
aortic valve stenosis, hear murmurs, aortic valve prolapse, mural valve
prolapse,
tricuspid valve prolapse, mitral valve insufficiency, mitral valve stenosis,
pulmonary
atresia, pulmonary valve insufficiency, pulmonary valve stenosis, tricuspid
atresia,
tricuspid valve insufficiency, and tricuspid valve stenosis.
Myocardial diseases include, but are not limited to, 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, but are not limited to, coronary disease, such
as
angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary
thrombosis,
coronary vasospasm, myocardial infarction and myocardial stunning.
Cardiovascular diseases also include vascular diseases such as aneurysms,
angiodysplasia, angiomatosis, bacillary angiomatosis, Hippel-Lindau Disease,
Klippel-Trenaunay-Weber Syndrome, Sturge-Weber Syndrome, angioneurotic edema,
aortic diseases, Takayasu's Arteritis, aortitis, Leriche's Syndrome, arterial
occlusive
diseases, arteritis, enarteritis, polyarteritis nodosa, cerebrovascular
disorders, diabetic
angiopathies, diabetic retinopathy, embolisms, thrombosis, erythromelalgia,
hemorrhoids, hepatic veno-occlusive disease, hypertension, hypotension,
ischemia,
peripheral vascular diseases, phlebitis, 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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
222
Aneurysms include, but are not limited to, dissecting aneurysms, false
aneurysms, infected aneurysms, ruptured aneurysms, aortic aneurysms, cerebral
aneurysms, coronary aneurysms, heart aneurysms, and iliac aneurysms.
Arterial occlusive diseases include, but are not limited to, arteriosclerosis,
intermittent claudication, caxotid stenosis, hbromuscular dysplasias,
mesenteric
vascular occlusion, Moyamoya disease, renal artery obstruction, retinal artery
occlusion, and thromboangiitis obliterans.
Cerebrovascular disorders include, but are not limited to, carotid artery
diseases, cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia,
cerebral
arteriosclerosis, cerebral arteriovenous malformation, cerebral artery
diseases,
cerebral embolism and thrombosis, carotid artery thrombosis, sinus thrombosis,
Wallenberg's syndrome, cerebral hemorrhage, epidural hematoma, subdural
hematoma, subaraxlmoid hemorrhage, cerebral infarction, cerebral ischemia
(including transient), subclavian steal syndrome, periventricular
leukomalacia,
vascular headache, cluster headache, migraine, and vertebrobasilar
insufficiency.
Embolisms include, but are not limited to, air embolisms, amniotic fluid
embolisms, cholesterol embolisms, blue toe syndrome, fat embolisms, pulmonary
embolisms, and thromoboembolisms. Thrombosis include, but are not limited to,
coronary thrombosis, hepatic vein thrombosis, retinal vein occlusion, carotid
artery
thrombosis, sinus thrombosis, Wallenberg's syndrome, and thrombophlebitis.
Ischemic disorders include, but are not limited to, cerebral ischemia,
ischemic
colitis, compartment syndromes, anterior compartment syndrome, myocardial
ischemia, reperfusion injuries, and peripheral limb ischemia. Vasculitis
includes, but
is not limited to, 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.
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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
223
topical applications during surgery, aerosol delivery. Such methods are known
in the
art. Polypeptides may be administered as part of a Therapeutic, described in
more
detail below. Methods of delivering polynucleotides are described in more
detail
herein.
Respiratory Disorders
Polynucleotides or polypeptides, or agonists or antagonists of the present
invention may be used to treat, prevent, diagnose, and/or prognose diseases
and/or
disorders of the respiratory system.
Diseases and disorders of the respiratory system include, but are not limited
to, nasal vestibulitis, nonallergic rhinitis (e.g., acute rhinitis, chronic
rhinitis, atrophic
rhinitis, vasomotor rhinitis), nasal polyps, and sinusitis, juvenile
angiofibromas,
cancer of the nose and juvenile papillomas, vocal cord polyps, nodules
(singer's
nodules), contact ulcers, vocal cord paralysis, laryngoceles, pharyngitis
(e.g., viral and
bacterial), tonsillitis, tonsillar cellulitis, parapharyngeal abscess,
laryngitis,
laryngoceles, and throat cancers (e.g., cancer of the nasopharynx, tonsil
cancer, larynx
cancer), lung cancer (e.g., squamous cell carcinoma, small cell (oat cell)
carcinoma,
large cell carcinoma, and adenocarcinoma), allergic disorders (eosinophilic
pneumonia, hypersensitivity pneumonitis (e.g., extrinsic allergic alveolitis,
allergic
interstitial pneumonitis, organic dust pneumoconiosis, allergic
bronchopuhnonary
aspergillosis, asthma, Wegener's granulomatosis (granulomatous vasculitis),
Goodpasture's syndrome)), pneumonia (e.g., bacterial pneumonia (e.g.,
Streptococcus
przeumoniae (pneumoncoccal pneumonia), Staphylococcus aureus (staphylococcal
pneumonia), Gram-negative bacterial pneumonia (caused by, e.g., Klebsiella and
Pseudomas spp.), Mycoplasrna prZeumoniae pneumonia, Hemoplailus influenzae
pneumonia, Legionella pneunZOplaila (Legionnaires' disease), and Clalarnydia
psittaci
(Psittacosis)), and viral pneumonia (e.g., influenza, chickenpox (varicella).
Additional diseases and disorders of the respiratory system include, but are
not
limited to bronchiolitis, polio (poliomyelitis), croup, respiratory syncytial
viral
infection, mumps, erythema infectiosum (fifth disease), roseola infantum,
progressive
rubella panencephalitis, german measles, and subacute sclerosing
panencephalitis),
fungal pneumonia (e.g., Histoplasmosis, Coccidioidomycosis, Blastomycosis,
fungal

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
224
infections in people with severely suppressed immune systems (e.g.,
cryptococcosis,
caused by Cryptococcus neofor°mans; aspergillosis, caused by
Aspergillus spp.;
candidiasis, caused by Candida; and mucormycosis)), Pneumocystis carifaii
(pneumocystis pneumonia), atypical pneumonias (e.g., Mycoplasnaa and Ghlamydia
spp.), opportunistic infection pneumonia, nosocomial pneumonia, chemical
pneumonitis, and aspiration pneumonia, pleural disorders (e.g., pleurisy,
pleural
effusion, and pneumothorax (e.g., simple spontaneous pneumothorax, complicated
spontaneous pneumothorax, tension pneumothorax)), obstructive airway diseases
(e.g., asthma, chronic obstructive pulmonary disease (COPD), emphysema,
chronic or
acute bronchitis), occupational lung diseases (e.g., silicosis, black lung
(coal workers'
pneumoconiosis), asbestosis, berylliosis, occupational asthsma, byssinosis,
and
benign pneumoconioses), Infiltrative Lung Disease (e.g., pulmonary fibrosis
(e.g.,
fibrosing alveolitis, usual interstitial pneumonia), idiopathic pulmonary
fibrosis,
desquamative interstitial pneumonia, lymphoid interstitial pneumonia,
histiocytosis X
(e.g., Letterer-Siwe disease, Hand-Schiiller-Christian disease, eosinophilic
granuloma), idiopathic pulmonary hemosiderosis, sarcoidosis and pulmonary
alveolar
proteinosis), Acute respiratory distress syndrome (also called, e.g., adult
respiratory
distress syndrome), edema, pulmonary embolism, bronchitis (e.g., viral,
bacterial),
bronchiectasis, atelectasis, lung abscess (caused by, e.g., ,Staphylococcus
auy~eus or
Legioraella praeumoplaila), and cystic fibrosis.
Anti-An~io~enesis Actiyity
The naturally occurnng balance between endogenous stimulators and
inhibitors of angiogenesis is one in which inhibitory influences predominate.
Rastinejad et al., Cell 56:345-355 (1989). In those rare instances in which
neovascularization occurs under normal physiological conditions, such as wound
healing, organ regeneration, embryonic development, and female reproductive
processes, angiogenesis is stringently regulated and spatially and temporally
delimited. Under conditions of pathological angiogenesis such as that
characterizing
solid tumor growth, these regulatory controls fail. Unregulated angiogenesis
becomes
pathologic and sustains progression of many neoplastic and non-neoplastic
diseases.
A number of serious diseases are dominated by abnormal neovascularization

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
225
including solid tumor growth and metastases, arthritis, some types of eye
disorders,
and psoriasis. See, e.g., reviews by Moses et al., Biotech. 9:630-634 (1991);
Follcman
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, Afn. J.
Opthahnol. 94:715-743 (1982); and Folkman et al., Science 221:719-725 (1983).
In a
number of pathological conditions, the process of angiogenesis contributes to
the
disease state. For example, significant data have accumulated which suggest
that the
growth of solid tumors is dependent on angiogenesis. Folkman and Klagsbrun,
Science 235:442-447 (1987).
The present invention provides for treatment of diseases or disorders
associated with neovascularization by administration of the polynucleotides
and/or
polypeptides of the invention, as well as agonists or antagonists of the
present
invention. Malignant and metastatic conditions which can be treated with the
polynucleotides and polypeptides, or agonists or antagonists of the invention
include,
but are not limited to, malignancies, solid tumors, and cancers described
herein and
otherwise known in the art (for a review of such disorders, see Fishman et
al.,
Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia (1985)).Thus, the present
invention provides a method of treating an angiogenesis-related disease and/or
disorder, comprising administering to an individual in need thereof a
therapeutically
effective amount of a polynucleotide, polypeptide, antagonist and/or agonist
of the
invention. For example, polynucleotides, polypeptides, antagonists and/or
agonists
may be utilized in a variety of additional methods in order to therapeutically
treat a
cancer or tumor. Cancers which may be treated with polynucleotides,
polypeptides,
antagonists and/or agonists include, but are not limited to solid tumors,
including
prostate, lung, breast, ovarian, stomach, pancreas, larynx, esophagus, testes,
liver,
parotid, biliary tract, colon, rectum, cervix, uterus, endometrium, kidney,
bladder,
thyroid cancer; primary tumors and metastases; melanomas; glioblastoma;
Kaposi's
sarcoma; leiomyosarcoma; non- small cell lung cancer; colorectal cancer;
advanced
malignancies; and blood born tumors such as leukemias. For example,
polynucleotides, polypeptides, antagonists and/or agonists may be delivered
topically,
in order to treat cancers such as skin cancer, head and neck tumors, breast
tumors, and

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
226
Kaposi's sarcoma.
Within yet other aspects, polynucleotides, polypeptides, antagonists and/or
agonists may be utilized to treat superficial forms of bladder cancer by, for
example,
intravesical administration. Polynucleotides, polypeptides, antagonists and/or
agonists
may be delivered directly into the tumor, or near the tumor site, via
injection or a
catheter. Of course, as the artisan of ordinary skill will appreciate, the
appropriate
mode of administration will vary according to the cancer to be treated. Other
modes
of delivery are discussed herein.
Polynucleotides, polypeptides, antagonists and/or agonists may be useful in
treating other disorders, besides cancers, which involve angiogenesis. These
disorders include, but are not limited to: benign tumors, for example
hemangiomas,
acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas;
artheroscleric plaques; ocular angiogenic diseases, for example, diabetic
retinopathy,
retinopathy of prematurity, macular degeneration, corneal graft rejection,
neovascular
glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, uvietis and
Pterygia
(abnormal blood vessel growth) of the eye; rheumatoid arthritis; psoriasis;
delayed
wound healing; endometriosis; vasculogenesis; granulations; hypertrophic scars
(keloids); nonunion fractures; scleroderma; trachoma; vascular adhesions;
myocardial
angiogenesis; coronary collaterals; cerebral collaterals; arteriovenous
malformations;
ischemic limb angiogenesis; Osler-Webber Syndrome; plaque neovascularization;
telangiectasia; hemophiliac joints; angiofibroma; fibromuscular dysplasia;
wound
granulation; Crohn's disease; and atherosclerosis.
For example, within one aspect of the present invention methods are provided
for treating hypertrophic scars and keloids, comprising the step of
administering a
polynucleotide, polypeptide, antagonist and/or agonist of the invention to a
hypertrophic scar or keloid.
Within one embodiment of the present invention polynucleotides,
polypeptides, antagonists and/or agonists of the invention 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
227
(approximately 14 days after the initial injury), but before hypertrophic scar
or keloid
development. As noted above, the present invention also provides methods for
treating neovascular diseases of the eye, including for example, corneal
neovascularization, neovascular glaucoma, proliferative diabetic retinopathy,
retrolental fibroplasia and macular degeneration.
Moreover, Ocular disorders associated with neovascularization which can be
treated with the polynucleotides and polypeptides of the present invention
(including
agonists andlor 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., Sure. Ophtlaal. 22:291-312 (1978).
Thus, within one aspect of the present invention methods are provided for
treating neovascular diseases of the eye such as corneal neovascularization
(including
corneal graft neovascularization), comprising the step of administering to a
patient a
therapeutically effective amount of a compound (as described above) to the
cornea,
such that the formation of blood vessels is inhibited. Briefly, the cornea is
a tissue
which normally lacks blood vessels. In certain pathological conditions
however,
capillaries may extend into the cornea from the pericomeal 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 disorders can result in
corneal
neovascularization, including for example, corneal infections (e.g., trachoma,
herpes
simplex keratitis, leishmaniasis and onchocerciasis), immunological processes
(e.g.,
graft rej ection and Stevens-Johnson's syndrome), alkali burns, trauma,
inflammation
(of any cause), toxic and nutritional deficiency states, and as a complication
of
wearing contact lenses.
Within particularly preferred embodiments of the invention, may be prepared
for topical administration in saline (combined with any of the preservatives
and
antimicrobial agents commonly used in ocular preparations), and administered
in
eyedrop form. The solution or suspension may be prepared in its pure form and

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
228
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-
s 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 inj ection may vary with the morphology of the
individual lesion,
but the goal of the administration would be to place the composition at the
advancing
front of the vasculature (i.e., interspersed between the blood vessels and the
normal
cornea). In most cases this would involve perilimbic corneal injection to
"protect" the
cornea from the advancing blood vessels. This method may also be utilized
shortly
after a corneal insult in order to prophylactically prevent corneal
neovascularization.
In this situation the material could be injected in the perilimbic cornea
interspersed
between the corneal lesion and its undesired potential limbic blood supply.
Such
methods may also be utilized in a similar fashion to prevent capillary
invasion of
transplanted corneas. In a sustained-release form injections might only be
required 2-
3 times per year. A steroid could also be added to the injection solution to
reduce
inflammation resulting from the injection itself.
Within another aspect of the present invention, methods are provided for
treating neovascular glaucoma, comprising the step of administering to a
patient a
therapeutically effective amount of a polynucleotide, polypeptide, antagonist
and/or
agonist to the eye, such that the formation of blood vessels is inhibited. In
one
embodiment, the compound may be administered topically to the eye in order to
treat
early forms of neovascular glaucoma. Within other embodiments, the compound
may
be implanted by injection into the region of the anterior chamber angle.
Within other
embodiments, the compound may also be placed in any location such that the

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
229
compound is continuously released into the aqueous humor. Within another
aspect of
the present invention, methods are provided for treating proliferative
diabetic
retinopathy, comprising the step of administering to a patient a
therapeutically
effective amount of a polynucleotide, polypeptide, antagonist and/or agonist
to the
eyes, such that the formation of blood vessels is inhibited.
Within particularly preferred embodiments of the invention, proliferative
diabetic retinopathy may be treated by injection into the aqueous humor or the
vitreous, in order to increase the local concentration of the polynucleotide,
polypeptide, antagonist and/or agonist in the retina. Preferably, this
treatment should
be initiated prior to the acquisition of severe disease requiring
photocoagulation.
Within another aspect of the present invention, methods are provided for
treating retrolental fibroplasia, comprising the step of administering to a
patient a
therapeutically effective amount of a polynucleotide, polypeptide, antagonist
and/or
agonist to the eye, such that the formation of blood vessels is inhibited. The
compound may be administered topically, via intravitreous injection and/or via
intraocular implants.
Additionally, disorders which can be treated with the polynucleotides,
polypeptides, agonists and/or agonists include, but are not limited to,
hemangioma,
arthritis, psoriasis, angiofibroma; atherosclerotic plaques, delayed wound
healing,
granulations, hemophilic joints, hypertrophic scars, nonunion fractures, Osler-
Weber
syndrome, pyogenic granuloma, scleroderma, trachoma, and vascular adhesions.
Moreover, disorders and/or states, which can be treated, prevented, diagnosed,
and/or prognosed with the the polynucleotides, polypeptides, agonists and/or
agonists
of the invention include, but are not limited to, solid tumors, blood born
tumors such
as leukemias, tumor metastasis, I~aposi'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,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
230
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, agousts
and/or
agonists may also be used in controlling menstruation or administered as
either a
peritoneal lavage fluid or for peritoneal implantation in the treatment of
endometriosis.
Polynucleotides, polypeptides, agonists and/or agonists of the present
invention may be incorporated into surgical sutures in order to prevent stitch
granulomas.
Polynucleotides, polypeptides, agonists and/or agonists may be utilized in a
wide variety of surgical procedures. For example, within one aspect of the
present
invention a compositions (in the form of, for example, a spray or film) may be
utilized
to coat or spray an area prior to removal of a tumor, in order to isolate
nornal
surrounding tissues from malignant tissue, and/or to prevent the spread of
disease to
surrounding tissues. Within other aspects of the present invention,
compositions (e.g.,
in the form of a spray) may be delivered via endoscopic procedures in order to
coat
tumors, or inhibit angiogenesis in a desired locale. Within yet other aspects
of the
present invention, surgical meshes which have been coated with anti-
angiogenic
compositions of the present invention may be utilized in any procedure wherein
a
surgical mesh might be utilized. For example, within one embodiment of the
invention a surgical mesh laden with an anti-angiogenic composition may be
utilized
during abdominal cancer resection surgery (e.g., subsequent to colon
resection) in
order to provide support to the structure, and to release an amount of the
anti-
angiogenic factor.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
231
Within further aspects of the present invention, methods are provided for
treating tumor excision sites, comprising administering a polynucleotide,
polypeptide,
agonist and/or agonist to the resection margins of a tumor subsequent to
excision,
such that the local recurrence of cancer and the formation of new blood
vessels at the
site is inhibited. Within one embodiment of the invention, the anti-angiogenic
compound is administered directly to the tumor excision site (e.g., applied by
swabbing, brushing or otherwise coating the resection margins of the tumor
with the
anti-angiogenic compound). Alternatively, the anti-angiogenic compounds may be
incorporated into known surgical pastes prior to administration. Within
particularly
preferred embodiments of the invention, the anti-angiogenic compounds are
applied
after hepatic resections for malignancy, and after neurosurgical operations.
Within one aspect of the present invention, polynucleotides, polypeptides,
agonists and/or agonists may be administered to the resection margin of a wide
variety of tumors, including for example, breast, colon, brain and hepatic
tumors. For
example, within one embodiment of the invention, anti-angiogenic compounds may
be administered to the site of a neurological tumor subsequent to excision,
such that
the formation of new blood vessels at the site are inhibited.
The polynucleotides, polypeptides, agonists and/or agonists of the present
invention may also be administered along with other anti-angiogenic factors.
Representative examples of other anti-angiogenic factors include: Anti-
Invasive
Factor, retinoic acid and derivatives thereof, paclitaxel, Suramin, Tissue
Inhibitor of
Metalloproteinase-1, Tissue Inhibitor of Metalloproteinase-2, Plasminogen
Activator
Inhibitor-l, 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
232
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, molybdenmn oxide, and molybdenyl
complexes. Suitable molybdate complexes include ammonium molybdate and its
hydrates, sodium molybdate and its hydrates, and potassium molybdate and its
hydrates. Suitable molybdenum oxides include molybdenum (VI) oxide, molybdenum
(VI) oxide, and molybdic acid. Suitable molybdenyl complexes include, for
example,
molybdenyl acetylacetonate. Other suitable tungsten and molybdenum complexes
include hydroxo derivatives derived from, for example, glycerol, tartaric
acid, and
sugars.
A wide variety of other anti-angiogenic factors may also be utilized within
the
context of the present invention. Representative examples include platelet
factor 4;
protamine sulphate; sulphated chitin derivatives (prepared from queen crab
shells),
(Murata et al., Cancer Res. 51:22-26, 1991); Sulphated Polysaccharide
Peptidoglycan
Complex (SP- PG) (the function of this compound may be enhanced by the
presence
of steroids such as estrogen, and tamoxifen citrate); Staurosporine;
modulators of
matrix metabolism, including for example, proline analogs, cishydroxyproline,
d,L-
3,4-dehydroproline, Thiaproline, alpha,alpha-dipyridyl, aminopropionitrile
fumarate;
4-propyl-5-(4-pyridinyl)-2(3H)-oxazolone; Methotrexate; Mitoxantrone; Heparin;
Interferons; 2 Macroglobulin-serum; ChIMP-3 (Pavloff et al., J. Bio. Chem.
267:17321-17326, 1992); Chymostatin (Tomkinson et al., Biochem J. 286:475-480,
1992); Cyclodextrin Tetradecasulfate; Eponemycin; Camptothecin; Fumagillin
(Ingber et al., Nature 348:555-557, 1990); Gold Sodium Thiomalate ("GST";
Matsubara and Ziff, J. Clin. Invest. 79:1440-1446, 1987); anticollagenase-
serum;
alpha2-antiplasmin (Holmes et al., J. Biol. Chem. 262(4):1659-1664, 1987);
Bisantrene (National Cancer Institute); Lobenzarit disodium (N-(2)-
carboxyphenyl-4-
chloroanthronilic acid disodium or "CCA"; Takeuchi et al., Agents Actions
36:312-

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
233
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, diagnosed, and/or prognosed using
polynucleotides
or polypeptides, as well as antagonists or agonists of the present invention,
include
cancers (such as follicular lymphomas, carcinomas with p53 mutations, and
hormone-
dependent tumors, including, but not limited to colon cancer, cardiac tumors,
pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer,
intestinal
cancer, testicular cancer, stomach cancer, neuroblastoma, myxoma, myoma,
lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma,
chondrosarcoma, adenoma, breast cancer, prostate cancer, I~aposi'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 aald immune-related
glomerulonephritis and rheumatoid arthritis) and viral infections (such as
herpes
viruses, pox viruses and adenoviruses), inflammation, graft v. host disease,
acute graft
rejection, and chronic graft rejection.
In preferred embodiments, polynucleotides, polypeptides, and/or antagonists
of the invention are used to inhibit growth, progression, and/or rnetasis of
cancers, in
particular those listed above.
Additional diseases or conditions associated with increased cell survival that
could be treated or detected by polynucleotides or polypeptides, or agonists
or
antagonists of the present invention include, but are not limited to,
progression, and/or
metastases of malignancies and related disorders such as leukemia (including
acute
leukemias (e.g., acute lyrnphocytic leukemia, acute myelocytic leukemia
(including
myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia))
and
chronic leukemias (e.g., chronic rriyelocytic (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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
234
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,
diagnosed, andlor prognesed using polynucleotides or polypeptides, as well as
agonists or antagonists of the present invention, include, but are not limited
to, ASS;
neurodegenerative disorders (such as Alzheimer's disease, Parkinson's disease,
Amyotrophic lateral sclerosis, Retinitis pigmentosa, Cerebellar degeneration
and
brain tumor or prior associated disease); autoimmune disorders (such as,
multiple
sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis,
Behcet's
disease, Crohn's disease, polymyositis, systemic lupus erythematosus and
immune-
related glomerulonephritis and rheumatoid arthritis) myelodysplastic syndromes
(such
as aplastic anemia), graft v. host disease, ischemic injury (such as that
caused by
myocardial infarction, stroke and reperfusion injury), liver injury (e.g.,
hepatitis
related liver injury, ischemia/reperfusion injury, cholestosis (bile duct
injury) and
liver cancer); toxin-induced liver disease (such as that caused by alcohol),
septic
shock, cachexia and anorexia.
Wound Healing and Epithelial Cell Proliferation
In accordance with yet a further aspect of the present invention, there is
provided a process for utilizing polynucleotides or polypeptides, as well as
agonists or
antagonists of the present invention, for therapeutic purposes, for example,
to

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
235
stimulate epithelial cell proliferation and basal keratinocytes for the
purpose of wound
healing, and to stimulate hair follicle production and healing of dermal
wounds.
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention, may be clinically useful in stimulating wound healing including
surgical
wounds, excisional wounds, deep wounds involving damage of the dermis and
epidermis, eye tissue wounds, dental tissue wounds, oral cavity wounds,
diabetic
ulcers, dermal ulcers, cubitus ulcers, arterial ulcers, venous stasis ulcers,
burns
resulting from heat exposure or chemicals, and other abnormal wound healing
conditions such as uremia, malnutrition, vitamin deficiencies and
complications
associated with systemic treatment with steroids, radiation therapy and
aaltineoplastic
drugs and antimetabolites. Polynucleotides or polypeptides, as well as
agonists or
antagonists of the present invention, could be used to promote dermal
reestablishment
subsequent to dermal loss
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present invention, could be used to increase the adherence of skin grafts to a
wound
bed and to stimulate re-epithelialization from the wound bed. The following
are types .
of grafts that polynucleotides or polypeptides, agonists or antagonists of the
present
invention, could be used to increase adherence to a wound bed: autografts,
artificial
skin, allografts, autodermic graft, autoepdermic grafts, avacular grafts,
Blair-Brown
grafts, bone graft, brephoplastic grafts, cutis graft, delayed graft, dennic
graft,
epidermic graft, fascia graft, full thickness graft, heterologous graft,
xenograft,
homologous graft, hyperplastic graft, lamellar graft, mesh graft, mucosal
graft, Ollier-
Thiersch graft, omenpal graft, patch graft, pedicle graft, penetrating graft,
split skin
graft, thick split graft. Polynucleotides or polypeptides, as well as agonists
or
antagonists of the present invention, can be used to promote skin strength and
to
improve the appearance of aged skin.
It is believed that polynucleotides or polypeptides, as well as agonists or
antagonists of the present invention, will also produce changes in hepatocyte
proliferation, and epithelial cell proliferation in the lung, breast,
pancreas, stomach,
small intestine, and large intestine. Polynucleotides or polypeptides, as well
as
agonists or antagonists of the present invention, could promote proliferation
of
epithelial cells such as sebocytes, hair follicles, hepatocytes, type II
pneumocytes,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
236
mucin-producing goblet cells, and other epithelial cells and their progenitors
contained within the skin, lung, liver, and gastrointestinal tract.
Polynucleotides or
polypeptides, agonists or antagonists of the present invention, may promote
proliferation of endothelial cells, keratinocytes, and basal keratinocytes.
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present invention, could also be used to reduce the side effects of gut
toxicity that
result from radiation, chemotherapy treatments or viral infections.
Polynucleotides or
polypeptides, as well as agonists or antagonists of the present invention, may
have a
cytoprotective effect on the small intestine mucosa. Polynucleotides or
polypeptides,
as well as agonists or antagonists of the present invention, may also
stimulate healing
of mucositis (mouth ulcers) that result from chemotherapy and viral
infections.
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present invention, could further be used in full regeneration of skin in full
and partial
thickness skin defects, including burns, (i.e., repopulation of hair
follicles, sweat
glands, and sebaceous glands), treatment of other skin defects such as
psoriasis.
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention, could be used to treat epidermolysis bullosa, a defect in adherence
of the
epidermis to the underlying dermis which results in frequent, open and painful
blisters
by accelerating reepithelialization of these lesions. Polynucleotides or
polypeptides,
as well as agonists or antagonists of the present invention, could also be
used to treat
gastric and doudenal ulcers and help heal by scar formation of the mucosal
lining and
regeneration of glandular mucosa and duodenal mucosal lining more rapidly.
Inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis,
are
diseases which result in destruction of the mucosal surface of the small or
large
intestine, respectively. Thus, polynucleotides or polypeptides, as well as
agonists or
antagonists of the present invention, could be used to promote the resurfacing
of the
mucosal surface to aid more rapid healing and to prevent progression of
inflammatory
bowel disease. Treatment with polynucleotides or polypeptides, agonists or
antagonists of the present invention, is expected to have a significant effect
on the
production of mucus tlmoughout the gastrointestinal tract and could be used to
protect
the intestinal mucosa from injurious substances that are ingested or following
surgery.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
237
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present
invention, could be used to treat diseases associate with the under
expression.
Moreover, polynucleotides or polypeptides, as well as agonists or antagonists
of the present invention, could be used to prevent and heal damage to the
lungs due to
various pathological states. Polynucleotides or polypeptides, as well as
agonists or
antagonists of the present invention, which could stimulate proliferation and
differentiation and promote the repair of alveoli and brochiolar epithelium to
prevent
or treat acute or chronic lung damage. For example, emphysema, which results
in the
progressive loss of aveoli, and inhalation injuries, i.e., resulting from
smoke
inhalation and burns, that cause necrosis of the bronchiolar epithelium and
alveoli
could be effectively treated using polynucleotides or polypeptides, agonists
or
antagonists of the present invention. Also, polynucleotides or polypeptides,
as well as
agonists or antagonists of the present invention, could be used to stimulate
the
proliferation of and differentiation of type II pneumocytes, which may help
treat or
prevent disease such as hyaline membrane diseases, such as infant respiratory
distress
syndrome and bronchopulmonary displasia, in premature infants.
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present invention, could stimulate the proliferation and differentiation of
hepatocytes
and, thus, could be used to alleviate or treat liver diseases and pathologies
such as
fulminant liver failure caused by cirrhosis, liver damage caused by viral
hepatitis and
toxic substances (i.e., acetaminophen, carbon tetraholoride and other
hepatotoxins
known in the art).
In addition, polynucleotides or polypeptides, as well as agonists or
antagonists
of the present invention, could be used treat or prevent the onset of diabetes
mellitus.
In patients with newly diagnosed Types I and II diabetes, where some islet
cell
function remains, polynucleotides or polypeptides, as well as agonists or
antagonists
of the present invention, could be used to maintain the islet function so as
to alleviate,
delay or prevent permanent manifestation of the disease. Also, polynucleotides
or
polypeptides, as well as agonists or antagonists of the present invention,
could be used
as an auxiliary in islet cell transplantation to improve or promote islet cell
function.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
238
Neural Activity and Neurological Diseases
The polynucleotides, polypeptides and agonists or antagonists of the invention
may be used for the diagnosis andfor treatment of diseases, disorders, damage
or
injury of the brain and/or nervous system. Nervous system disorders that can
be
treated 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 or disorders which result in either a disconnection of
axons, a
diminution or degeneration of neurons, or demyelination. Nervous system
lesions
which may be treated in a patient (including human and non-human mammalian
patients) according to the methods of 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 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, or 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
or disorders, 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
B12 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),

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
239
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 one embodiment, the polypeptides, polynucleotides, or agonists or
antagonists of the invention are used to protect neural cells from the
damaging effects
of hypoxia. In a further 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 or prevent neural cell injury
associated
with cerebral hypoxia. In one non-exclusive aspect of this embodiment, the
polypeptides, polynucleotides, or agonists or antagonists of the invention,
are used to
treat or prevent neural cell injury associated with cerebral ischemia. In
another non-
exclusive aspect of this embodiment, the polypeptides, polynucleotides, or
agonists or
antagonists of the invention are used to treat or prevent neural cell injury
associated
with cerebral infarction.
In another preferred embodiment, the polypeptides, polynucleotides, or
agonists or antagonists of the invention are used to treat or prevent neural
cell injury
associated with a stroke. In a specific embodiment, the polypeptides,
polynucleotides,
or agonists or antagonists of the invention are used to treat or prevent
cerebral neural
cell injury associated with a stroke.
In another preferred embodiment, the polypeptides, polynucleotides, or
agonists or antagonists of the invention are used to treat or prevent neural
cell injury
associated with a heart attack. In a specific embodiment, the polypeptides,
polynucleotides, or agonists or antagonists of the invention are used to treat
or prevent
cerebral neural cell injury associated with a heart attack.
The compositions of the invention which axe 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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
240
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
either in the presence or absence of hypoxia or hypoxic conditions; (2)
increased
sprouting of neurons in culture or i~2 vivo; (3) increased production of a
neuron-
s associated molecule in culture or ifa vivo, e.g., choline acetyltransferase
or
acetylcholinesterase with respect to motor neurons; or (4) decreased symptoms
of
neuron dysfunction ifs 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, in Zhang et al., Proc Natl Acad Sci LTSA 97:3637-42
(2000) or
in Arakawa et al., J. Neurosci., 10:3507-15 (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. Neu~ol., 70:65-82 (1980), or Brown et al., Ahn.
Rev.
Neu~osci., 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 disorders that may be treated
according to the invention include, but are not limited to, disorders 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 disorders 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-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

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
241
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, Alzheimer's Disease, Parkinson's Disease, Huntington's Disease,
Tourette
Syndrome, schizophrenia, mania, dementia, paranoia, obsessive compulsive
disorder,
panic disorder, learning disabilities, ALS, psychoses, autism, and altered
behaviors,
including disorders in feeding, sleep patterns, balance, and perception. In
addition,
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., multi-infarct),
leukornalacia,
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
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.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
242
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.
Additional neurologic diseases which can be treated or detected with
polynucleotides, polypeptides, agonists, and/or antagonists of the present
invention
include dementia such as A>DS 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
multi-infarct dementia, encephalitis which include encephalitis periaxialis,
viral

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
243
encephalitis such as epidemic encephalitis, Japanese Encephalitis, St. Louis
Encephalitis, tick-borne encephalitis and West Nile Fever, acute disseminated
encephalomyelitis, meningoencephalitis such as uveomeningoencephalitic
syndrome,
Postencephalitic Parkinson Disease and subacute sclerosing panencephalitis,
encephalomalacia such as periventricular leukomalacia, epilepsy such as
generalized
epilepsy which includes infantile spasms, absence epilepsy, myoclonic epilepsy
which
includes MERRF Syndrome, tonic-clonic epilepsy, partial epilepsy such as
complex
partial epilepsy, frontal lobe epilepsy and temporal lobe epilepsy, post-
traumatic
epilepsy, status epilepticus such as Epilepsia Partialis Continua, 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 AmS 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 lyrnphocytic choriomeningitis, Bacterial meningtitis which
includes
Haemophilus Meningtitis, Listeria Meningtitis, Meningococcal Meningtitis such
as
Waterhouse-Friderichsen Syndrome, Pneumococcal Meningtitis and meningeal
tuberculosis, fungal meningitis such as Cryptococcal Meningtitis, subdural
effusion,
meningoencephalitis such as uvemeningoencephalitic syndrome, myelitis such as
transverse myelitis, neurosyphilis such as tabes dorsalis, poliomyelitis which
includes
bulbar poliomyelitis and postpoliomyelitis syndrome, prion diseases (such as

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
244
Creutzfeldt-Jakob Syndrome, Bovine Spongifonn 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 Angehnan Syndrome, Cri-du-Chat Syndrome, De Lange's
Syndrome, Down Syndrome, Gangliosidoses such as gangliosidoses G(Ml), Sandhoff
Disease, Tay-Sachs Disease, Hartnup Disease, homocystinuria, Laurence-Moon-
Biedl Syndrome, Lesch-Nyhan Syndrome, Maple Syrup Urine Disease, mucolipidosis
such as fucosidosis, neuronal ceroid-lipofuscinosis, oculocerebrorenal
syndrome,
phenylketonuria such as maternal phenylketonuria, Prader-Willi Syndrome, Rett
Syndrome, Rubinstein-Taybi Syndrome, Tuberous Sclerosis, WAGR Syndrome,
nervous system abnormalities such as holoprosencephaly, neural tube defects
such as
anencephaly which includes hydrangencephaly, Arnold-Chairi Deformity,
encephalocele, meningocele, meningomyelocele, spinal dysraphism such as spina
bifida cystica and spina bifida occulta.
Additional neurologic diseases which can be treated or detected with
polynucleotides, polypeptides, agonists, and/or antagonists of the present
invention
include hereditary motor and sensory neuropathies which include Charcot-Marie

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
245
Disease, Hereditary optic atrophy, Refsum's Disease, hereditary spastic
paraplegia,
Werdnig-Hoffmann Disease, Hereditary Sensory and Autonomic Neuropathies such
as Congenital Analgesia and Familial Dysautonomia, Neurologic manifestations
(such
as agnosia that include Gerstmann's Syndrome, Amnesia such as retrograde
amnesia,
apraxia, neurogenic bladder, cataplexy, communicative disorders such as
hearing
disorders that includes deafness, partial hearing loss, loudness recruitment
and
tinnitus, language disorders such as aphasia which include agraphia, anomia,
broca
aphasia, and Wernicke Aphasia, Dyslexia such as Acquired Dyslexia, language
development disorders, speech disorders such as aphasia which includes anomia,
broca aphasia and Wernicke Aphasia, articulation disorders, communicative
disorders
such as speech disorders which include dysarthria, echolalia, mutism and
stuttering,
voice disorders such as aphonia and hoarseness, decerebrate state, delirium,
fasciculation, hallucinations, meningism, movement disorders such as angelinan
syndrome, ataxia, athetosis, chorea, dystonia, hypokinesia, muscle hypotonia,
myoclonus, tic, torticollis and tremor, muscle hypertonia such as muscle
rigidity such
as stiff man syndrome, muscle spasticity, paralysis such as facial paralysis
which
includes Herpes Zoster Oticus, Gastroparesis, Hemiplegia, ophthalmoplegia such
as
diplopia, Duane's Syndrome, Homer's Syndrome, Chronic progressive external
ophthalmoplegia such as Kearns Syndrome, Bulbax Paralysis, Tropical Spastic
Paraparesis, Paraplegia such as Brown-Sequard Syndrome, quadriplegia,
respiratory
paralysis and vocal cord paralysis, paresis, phantom limb, taste disorders
such as
ageusia and dysgeusia, vision disorders such as amblyopia, blindness, color
vision
defects, diplopia, hemianopsia, scotoma and subnormal vision, sleep disorders
such as
hypersomnia which includes Kleine-Levin Syndrome, insonnnia, and somnambulism,
spasm such as trismus, unconsciousness such as coma, persistent vegetative
state and
syncope and vertigo, neuromuscular diseases such as amyotonia congenita,
amyotrophic lateral sclerosis, Lambert-Eaton Myasthenic Syndrome, motor neuron
disease, muscular atrophy such as spinal muscular atrophy, Charcot-Marie
Disease
and Werdnig-Hoffmann Disease, Postpoliomyelitis Syndrome, Muscular Dystrophy,
Myasthenia Gravis, Myotonia Atrophica, Myotonia Confenita, Nemaline Myopathy,
Familial Periodic Paralysis, Multiplex Paramyloclonus, Tropical Spastic
Paraparesis
and Stiff Man Syndrome, peripheral nervous system diseases such as acrodynia,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
246
amyloid neuropathies, autonomic nervous system diseases such as Adie's
Syndrome,
Barre-Lieou Syndrome, Familial Dysautonomia, Homer's Syndrome, Reflex
Sympathetic Dystrophy and Shy-Drager Syndrome, Cranial Nerve Diseases such as
Acoustic Nerve Diseases such as Acoustic Neuroma which includes
Neurofibromatosis 2, Facial Nerve Diseases such as Facial Neuralgia,Melkersson-
Rosenthal Syndrome, ocular motility disorders which includes amblyopia,
nystagmus,
oculomotor nerve paralysis, ophthalmoplegia such as Duane's Syndrome, Homer's
Syndrome, Chronic Progressive External Ophthalmoplegia which includes Kearns
Syndrome, Strabismus such as Esotropia and Exotropia, Oculomotor Nerve
Paralysis,
Optic Nerve Diseases such as Optic Atrophy which includes Hereditary Optic
Atrophy, Optic Disk Drusen, Optic Neuritis such as Neuromyelitis Optica,
Papilledema, Trigeminal Neuralgia, Vocal Cord Paralysis, Demyelinating
Diseases
such as Neuromyelitis Optica and Swayback, and Diabetic neuropathies such as
diabetic foot.
Additional neurologic diseases which can be treated or detected with
polynucleotides, polypeptides, agonists, and/or antagonists of the present
invention
include nerve compression syndromes such as carpal tumzel syndrome, tarsal
tumlel
syndrome, thoracic outlet syndrome such as cervical rib syndrome, ulnar nerve
compression syndrome, neuralgia such as causalgia, cervico-brachial neuralgia,
facial
neuralgia and trigeminal neuralgia, neuritis such as experimental allergic
neuritis,
optic neuritis, polyneuritis, polyradiculoneuritis and radiculities such as
polyradiculitis, hereditary motor and sensory neuropathies such as Charcot-
Marie
Disease, Hereditary Optic Atrophy, Refsum's Disease, Hereditary Spastic
Paraplegia
and Werdnig-Hoffmann Disease, Hereditary Sensory and Autonomic Neuropathies
which include Congenital Analgesia and Familial Dysautonomia, POEMS Syndrome,
Sciatica, Gustatory Sweating and Tetany).
Endocrine Disorders
Polynucleotides or polypeptides, or agonists or antagonists of the present
invention, may be used to treat, prevent, diagnose, and/or prognose disorders
and/or
diseases related to hormone imbalance, and/or disorders or diseases of the
endocrine

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
247
system.
Hormones secreted by the glands of the endocrine system control physical
growth, sexual function, metabolism, and other functions. Disorders may be
classified
in two ways: disturbances in the production of hormones, and the inability of
tissues
to respond to hormones. The etiology of these hormone imbalance or endocrine
system diseases, disorders or conditions may be genetic, somatic, such as
cancer and
some autoimmune diseases, acquired (e.g., by chemotherapy, injury or toxins),
or
infectious. Moreover, polynucleotides, polypeptides, antibodies, and/or
agonists or
antagonists of the present invention can be used as a marker or detector of a
particular
disease or disorder related to the endocrine system andlor hormone imbalance.
Endocrine system and/or hormone imbalance and/or diseases encompass
disorders of uterine motility including, but not limited to: complications
with
pregnancy and labor (e.g., pre-term labor, post-term pregnancy, spontaneous
abortion, and slow or stopped labor); and disorders and/or diseases of the
menstrual
cycle (e.g., dysmenorrhea and endometriosis).
Endocrine system and/or hormone imbalance disorders and/or diseases include
disorders and/or diseases of the pancreas, such as, for example, diabetes
mellitus,
diabetes insipidus, congenital pancreatic agenesis, pheochromocytoma--islet
cell
tumor syndrome; disorders and/or diseases of the adrenal glands such as, for
example,
Addison's Disease, corticosteroid deficiency, virilizing disease, hirsutism,
Cushing's
Syndrome, hyperaldosteronism, pheochromocytoma; disorders and/or diseases of
the
pituitary gland, such as, for example, hyperpituitarism, hypopituitarism,
pituitary
dwarfism, pituitary adenoma, panhypopituitarism, acromegaly, gigantism;
disorders
and/or diseases of the thyroid, including but not limited to, hyperthyroidism,
hypothyroidism, Plummer's disease, Graves' disease (toxic diffuse goiter),
toxic
nodular goiter, thyroiditis (Hashimoto's thyroiditis, subacute granulomatous
thyroiditis, and silent lymphocytic thyroiditis), Pendred's syndrome,
myxedema,
cretinism, thyrotoxicosis, thyroid hormone coupling defect, thymic aplasia,
Hurthle
cell tumours of the thyroid, thyroid cancer, thyroid carcinoma, Medullary
thyroid
carcinoma; disorders and/or diseases of the parathyroid, such as, for example,
hyperparathyroidism, hypoparathyroidism; disorders and/or diseases of the
hypothalamus.

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
248
In addition, endocrine system and/or hormone imbalance disorders and/or
diseases may also include disorders and/or diseases of the testes or ovaries,
including
cancer. Other disorders and/or diseases of the testes or ovaries further
include, for
example, ovarian cancer, polycystic ovary syndrome, Klinefelter's syndrome,
vanishing testes syndrome (bilateral anorchia), congenital absence of Leydig's
cells,
cryptorchidism, Noonan's syndrome, myotonic dystrophy, capillary haemangioma
of
the testis (benign), neoplasias of the testis and neo-testis.
Moreover, endocrine system and/or hormone imbalance disorders and/or
diseases may also include disorders and/or diseases such as, for example,
polyglandular deficiency syndromes, pheochromocytoma, neuroblastoma, multiple
Endocrine neoplasia, and disorders and/or cancers of endocrine tissues.
Tn another embodiment, a polypeptide of the invention, or polynucleotides,
antibodies, agonists, or antagonists corresponding to that polypeptide, may be
used to
diagnose, prognose, prevent, and/or treat endocrine diseases and/or disorders
associated with the tissues) in which the polypeptide of the invention is
expressed,
including one, two, three, four, five, or more tissues disclosed in Table l,
column ~
(Tissue Distribution Library Code).
Reproductive System Disorders
The polynucleotides or polypeptides, or agonists or antagonists of the
invention may be used for the diagnosis, treatment, or prevention of diseases
and/or
disorders of the reproductive system. Reproductive system disorders that can
be
treated by the compositions of the invention, include, but are not limited to,
reproductive system injuries, infections, neoplastic disorders, congenital
defects, and
diseases or disorders which result in infertility, complications with
pregnancy, labor,
or parturition, and postpartum difficulties.
Reproductive system disorders and/or diseases include diseases and/or
disorders of the testes, including testicular atrophy, testicular
feminization,
cryptorchism (unilateral and bilateral), anorchia, ectopic testis,
epididymitis and
orchitis (typically resulting from infections such as, for example, gonorrhea,
mumps,
tuberculosis, and syphilis), testicular torsion, vasitis nodosa, germ cell
tumors (e.g.,
seminomas, embryonal cell carcinomas, teratocarcinomas, choriocarcinomas, yolk
sac

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
249
tumors, and teratomas), stromal tumors (e.g., Leydig cell tumors), hydrocele,
hematocele, varicocele, spermatocele, inguinal hernia, and disorders of sperm
production (e.g., immotile cilia syndrome, aspermia, asthenozoospermia,
azoospermia, oligospermia, and teratozoospermia).
Reproductive system disorders also include disorders of the prostate gland,
such as acute non-bacterial prostatitis, chronic non-bacterial prostatitis,
acute bacterial
prostatitis, chronic bacterial prostatitis, prostatodystonia, prostatosis,
granulomatous
prostatitis, malacoplakia, benign prostatic hypertrophy or hyperplasia, and
prostate
neoplastic disorders, including adenocarcinomas, transitional cell carcinomas,
ductal
carcinomas, and squamous cell carcinomas.
Additionally, the compositions of the invention may be useful in the
diagnosis,
treatment, and/or prevention of disorders or diseases of the penis and
urethra,
including inflammatory disorders, such as balanoposthitis, balanitis xerotica
obliterans, phimosis, paraphimosis, syphilis, herpes simplex virus, gonorrhea,
non-
gonococcal urethritis, chlamydia, mycoplasma, trichomonas, HIV, AIDS, Reiter's
syndrome, condyloma acuminatum, condyloma latum, and pearly penile papules;
urethral abnormalities, such as hypospadias, epispadias, and phimosis;
premalignant
lesions, including Erythroplasia of Queyrat, Bowen's disease, Bowenoid
paplosis,
giant condyloma of Buscke-Lowenstein, and varrucous carcinoma; penile cancers,
including squamous cell carcinomas, carcinoma in situ, verrucous carcinoma,
and
disseminated penile carcinoma; urethral neoplastic disorders, including penile
urethral
carcinoma, bulbomembranous urethral carcinoma, and prostatic urethral
carcinoma;
and erectile disorders, such as priapism, Peyronie's disease, erectile
dysfunction, and
impotence.
Moreover, diseases and/or disorders of the vas deferens include vasculititis
and CBAVD (congenital bilateral absence of the vas deferens); additionally,
the
polynucleotides, polypeptides, and agonists or antagonists of the present
invention
may be used in the diagnosis, treatment, and/or prevention of diseases and/or
disorders of the seminal vesicles, including hydatid disease, congenital
chloride
diarrhea, and polycystic kidney disease.
Other disorders and/or diseases of the male reproductive system include, for
example, Klinefelter's syndrome, Young's syndrome, premature ejaculation,
diabetes

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
250
mellitus, cystic fibrosis, I~artagener's syndrome, high fever, multiple
sclerosis, and
gynecomastia.
Further, the polynucleotides, polypeptides, and agonists or antagonists of the
present invention may be used in the diagnosis, treatment, and/or prevention
of
diseases and/or disorders of the vagina and vulva, including bacterial
vaginosis,
candida vaginitis, herpes simplex virus, chancroid, granuloma inguinale,
lymphogranuloma venereum, scabies, human papillomavirus, vaginal trauma,
vulvar
trauma, adenosis, chlamydia vaginitis, gonorrhea, trichomonas vaginitis,
condyloma
acuminatum, syphilis, molluscum contagiosum, atrophic vaginitis, Paget's
disease,
lichen sclerosus, lichen planus, vulvodynia, toxic shock syndrome, vaginismus,
vulvovaginitis, vulvar vestibulitis, and neoplastic disorders, such as
squamous cell
hyperplasia, clear cell carcinoma, basal cell carcinoma, melanomas, cancer of
Bartholin's gland, and vulvar intraepithelial neoplasia.
Disorders and/or diseases of the uterus include dysmenorrhea, retroverted
uterus, endometriosis, fibroids, adenomyosis, anovulatory bleeding,
amenorrhea,
Cushing's syndrome, hydatidifonn moles, Asherman's syndrome, premature
menopause, precocious puberty, uterine polyps, dysfunctional uterine bleeding
(e.g.,
due to aberrant hormonal signals), and neoplastic disorders, such as
adenocarcinomas,
keiomyosarcomas, and sarcomas. Additionally, the polypeptides,
polynucleotides, or
agonists or antagonists of the invention may be useful as a marker or detector
of, as
well as in the diagnosis, treatment, and/or prevention of congenital uterine
abnormalities, such as bicornuate uterus, septate uterus, simple unicornuate
uterus,
unicornuate uterus with a noncavitary rudimentary horn, unicornuate uterus
with a
non-communicating cavitary rudimentary horn, unicomuate uterus with a
communicating cavitary horn, arcuate uterus, uterine didelfus, and T-shaped
uterus.
Ovarian diseases andlor disorders include anovulation, polycystic ovary
syndrome (Stein-Leventhal syndrome), ovarian cysts, ovarian hypofunction,
ovarian
insensitivity to gonadotropins, ovarian overproduction of androgens, right
ovarian
vein syndrome, amenorrhea, hirutism, and ovarian cancer (including, but not
limited
to, primary and secondary cancerous growth, Sertoli-Leydig tumors, endometriod
carcinoma of the ovary, ovarian papillary serous adenocarcinoma, ovarian
mucinous
adenocarcinoma, and Ovarian Krukenberg tumors).

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
251
Cervical diseases and/or disorders include cervicitis, chronic cervicitis,
mucopurulent cervicitis, cervical dysplasia, cervical polyps, Nabothian cysts,
cervical
erosion, cervical incompetence, and cervical neoplasms (including, for
example,
cervical carcinoma, squamous metaplasia, squamous cell carcinoma,
adenosquamous
cell neoplasia, and columnar cell neoplasia).
Additionally, diseases andlor disorders of the reproductive system include
disorders and/or diseases of pregnancy, including miscarriage and stillbirth,
such as
early abortion, late abortion, spontaneous abortion, induced abortion,
therapeutic
abortion, threatened abortion, missed abortion, incomplete abortion, complete
abortion, habitual abortion, missed abortion, and septic abortion; ectopic
pregnancy,
anemia, Rh incompatibility, vaginal bleeding during pregnancy, gestational
diabetes,
intrauterine growth retardation, polyhydramnios, HELLP syndrome, abruptio
placentae, placenta previa, hyperemesis, preeclampsia, eclampsia, herpes
gestationis,
and urticaria of pregnancy. Additionally, the polynucleotides, polypeptides,
and
agonists or antagonists of the present invention may be used in the diagnosis,
treatment, and/or prevention of diseases that can complicate pregnancy,
including
heart disease, heart failure, rheumatic heart disease, congenital heart
disease, mitral
valve prolapse, high blood pressure, anemia, kidney disease, infectious
disease (e.g.,
rubella, cytomegalovirus, toxoplasmosis, infectious hepatitis, chlamydia, HIV,
AIDS,
and genital herpes), diabetes mellitus, Graves' disease, thyroiditis,
hypothyroidism,
Hashimoto's thyroiditis, chronic active hepatitis, cirrhosis of the liver,
primary biliary
cirrhosis, asthma, systemic lupus eryematosis, rheumatoid arthritis,
myasthenia
gravis, idiopathic thrombocytopenic purpura, appendicitis, ovarian cysts,
gallbladder
disorders,and obstruction of the intestine.
Complications associated with labor and parturition include premature rupture
of the membranes, pre-term labor, post-term pregnancy, postm,aturity, labor
that
progresses too slowly, fetal distress (e.g., abnormal heart rate (fetal or
maternal),
breathing problems, and abnormal fetal position), shoulder dystocia, prolapsed
umbilical cord, amniotic fluid embolism, and aberrant uterine bleeding.
Further, diseases and/or disorders of the postdelivery period, including
endometritis, myometritis, parametritis, peritonitis, pelvic thrombophlebitis,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
252
pulmonary embolism endotoxemia, pyelonephritis, saphenous thrombophlebitis,
mastitis, cystitis, postpartum hemorrhage, and inverted uterus.
Other disorders and/or diseases of the female reproductive system that may be
diagnosed, treated, and/or prevented by the polynucleotides, polypeptides, and
agonists or antagonists of the present invention include, for example,
Turner's
syndrome, pseudohermaphroditism, premenstrual syndrome, pelvic inflammatory
disease, pelvic congestion (vascular engorgement), frigidity, anorgasmia,
dyspareunia, ruptured fallopian tube, and Mittelschmerz.
Infectious Disease
Polynucleotides or polypeptides, as well as agonists or antagonists of the
present invention can be used to treat or detect infectious agents. For
example, by
increasing the immune response, particularly increasing the proliferation and
differentiation of B and/or T cells, infectious diseases may be treated. The
immune
response may be increased by either enhancing an existing immune response, or
by
initiating a new immune response. Alternatively, polynucleotides or
polypeptides, as
well as agonists or antagonists of the present invention may also directly
inhibit the
infectious agent, without necessarily eliciting an immune response.
Viruses are one example of an infectious agent that can cause disease or
symptoms that can be treated or detected by a polynucleotide or polypeptide
and/or
agonist or antagonist of the present invention. Examples of viruses, include,
but are
not limited to Examples of viruses, include, but are not limited to the
following DNA
and RNA viruses and viral families: Arbovirus, Adenoviridae, Arenaviridae,
Arterivirus, Birnaviridae, Bunyaviridae, Caliciviridae, Circoviridae,
Coronaviridae,
Dengue, EBV, HIV, Flaviviridae, Hepadnaviridae (Hepatitis), Herpesviridae
(such as,
Cytomegalovirus, Herpes Simplex, Herpes Zoster), Mononegavirus (e.g.,
Paramyxoviridae, Morbillivirus, Rhabdoviridae), Orthomyxoviridae (e.g.,
Influenza
A, Influenza B, and parainfluenza), Papiloma virus, Papovaviridae,
Parvoviridae,
Picornaviridae, Poxviridae (such as Smallpox or Vaccinia), Reoviridae (e.g.,
Rotavirus), Retroviridae (HTLV-I, HTLV-II, Lentivirus), and Togaviridae (e.g.,
Rubivirus). Viruses falling within these families can cause a variety of
diseases or
symptoms, including, but not limited to: arthritis, bronchiollitis,
respiratory syncytial

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
253
virus, encephalitis, eye infections (e.g., conjunctivitis, lceratitis),
chronic fatigue
syndrome, hepatitis (A, B, C, E, Chronic Active, Delta), Japanese B
encephalitis,
Junin, Chikungunya, Rift Valley fever, yellow fever, meningitis, opportunistic
infections (e.g., AIDS), pneumonia, Burkitt's Lymphoma, chickenpox,
hemorrhagic
fever, Measles, Mumps, Parainfluenza, Rabies, the common cold, Polio,
leukemia,
Rubella, sexually transmitted diseases, skin diseases (e.g., Kaposi's, warts),
and
viremia. polynucleotides or polypeptides, or agonists or antagonists of the
invention,
can be used to treat or detect any of these symptoms or diseases. In specific
embodiments, polynucleotides, polypeptides, or agonists or antagonists of the
invention are used to treat: meningitis, Dengue, EBV, and/or hepatitis (e.g.,
hepatitis
B). In an additional specific embodiment polynucleotides, polypeptides, or
agonists
or antagonists of the invention are used to treat patients nonresponsive to
one or more
other commercially available hepatitis vaccines. In a further specific
embodiment
polynucleotides, polypeptides, or agonists or antagonists of the invention are
used to
treat AIDS.
Similarly, bacterial and fungal agents that can cause disease or symptoms and
that can be treated or detected by a polynucleotide or polypeptide and/or
agonist or
antagonist of the present invention include, but not limited to, the following
Gram-
Negative and Gram-positive bacteria, bacterial families, and fungi:
Actinomyces
(e.g., Norcardia), Acinetobacter, Cryptococcus neoformans, Aspergillus,
Bacillaceae
(e.g., Bacillus antlarasis), Bacteroides (e.g., Bacteroides fragilis),
Blastomycosis,
Bordetella, Borrelia (e.g., Borrelia burgdorferi), Brucella, Candidia,
Campylobacter,
Chlamydia, Clostridium (e.g., Clostridiuna botulinum, Clostridiz~na dificile,
Clostridium perfringens, Clostridium tetani), Coccidioides, Corynebacterium
(e.g.,
Corynebacterium diptlzeriae), Cryptococcus, Dermatocycoses, E. coli (e.g.,
Enterotoxigenic E. coli and Enterohemorrhagic E. coli), Enterobacter (e.g.
Enterobacter aerogenes), Enterobacteriaceae (Klebsiella, Salmonella (e.g.,
Salmonella typhi, Salrnoraella entef°itidis, Salmonella typhi),
Serratia, Yersinia,
Shigella), Erysipelothrix, Haemophilus (e.g., Haemophilus influenza type B),
Helicobacter, Legionella (e.g., LegiorZella pneunZOplaila), Leptospira,
Listeria (e.g.,
Listeria motaocytogenes), Mycoplasma, Mycobacterium (e.g., Mycobacterium
leprae
aft.d Mycobacterium tuberculosis), Vibrio (e.g., Vibrio cholerae),
Neisseriaceae (e.g.,

CA 02423559 2003-03-04
WO 02/22638 PCT/USO1/01386
254
Neissef°ia gofaoYrhea, Neisseria meni~tgiticlis), Pasteurellacea,
Proteus, Pseudomonas
(e.g., Pseudomonas aef°ugifaosa), Rickettsiaceae, Spirochetes (e.g.,
Treponema spp.,
Leptospira spp., Borrelia spp.), Shigella spp., Staphylococcus (e.g.,
Staphylococcus
auf~eus), Meningiococcus, Pneumococcus and Streptococcus (e.g., Streptococcus
pneumoniae and Groups A, B, and C Streptococci), and Ureaplasmas. These
bacterial, parasitic, and fungal families can cause diseases or symptoms,
including,
but not limited to: antibiotic-resistant infections, bacteremia, endocarditis,
septicemia,
eye infections (e.g., conjunctivitis), uveitis, tuberculosis, gingivitis,
bacterial diarrhea,
opportunistic infections (e.g., AIDS related infections), paronychia,
prosthesis-related
infections, dental caries, Reiter's Disease, respiratory tract infections,
such as
Whooping Cough or Empyema, sepsis, Lyme Disease, Cat-Scratch Disease,
dysentery, paratyphoid fever, food poisoning, Legionella disease, chronic and
acute
inflammation, erythema, yeast infections, typhoid, pneumonia, gonorrhea,
meningitis
(e.g., mengitis types A and B), chlamydia, syphillis, diphtheria, leprosy,
brucellosis,
peptic ulcers, anthrax, spontaneous abortions, birth defects, pneumonia, lung
infections, ear infections, deafness, blindness, lethargy, malaise, vomiting,
chronic
diarrhea, Crohn's disease, colitis, vaginosis, sterility, pelvic inflammatory
diseases,
candidiasis, paratuberculosis, tuberculosis, lupus, botulism, gangrene,
tetanus,
impetigo, Rheumatic Fever, Scarlet Fever, sexually transmitted diseases, skin
diseases
(e.g., cellulitis, dermatocycoses), toxemia, urinary tract infections, wound
infections,
noscomial infections. Polynucleotides or polypeptides, agonists or antagonists
of the
invention, can be used to treat or detect any of these symptoms or diseases.
In
specific embodiments, polynucleotides, polypeptides, agonists or antagonists
of the
invention are used to treat: tetanus, diptheria, botulism, and/or meningitis
type B.
Moreover, parasitic agents causing disease or symptoms that can be treated,
prevented, and/or diagnosed by a polynucleotide or polypeptide and/or agonist
or
antagonist of the present invention include, but not limited to, the following
families
or class: Amebiasis, Babesiosis, Coccidiosis, Cryptosporidiosis,
Dientamoebiasis,
Dourine, Ectoparasitic, Giardias, Helminthiasis, Leishmaniasis, Schistisoma,
Theileriasis, Toxoplasmosis, Trypanosomiasis, and Trichomonas and Sporozoans
(e.g., Plasmodium virax, Plasmodium falciparium, Plasmodium Tnalar~iae and
PlasfraodiunZ ovale). These parasites can cause a variety of diseases or
symptoms,

DEMANDE OU BREVET VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.
CECI EST LE TOME 1 DE 2
CONTENANT LES PAGES 1 A 254
NOTE : Pour les tomes additionels, veuillez 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
CONTAINING PAGES 1 TO 254
NOTE: For additional volumes, please contact the Canadian Patent Office
NOM DU FICHIER / FILE NAME
NOTE POUR LE TOME / VOLUME NOTE:

Dessin représentatif

Désolé, le dessin représentatif concernant le document de brevet no 2423559 est introuvable.

États administratifs

2024-08-01 : Dans le cadre de la transition vers les Brevets de nouvelle génération (BNG), la base de données sur les brevets canadiens (BDBC) contient désormais un Historique d'événement plus détaillé, qui reproduit le Journal des événements de notre nouvelle solution interne.

Veuillez noter que les événements débutant par « Inactive : » se réfèrent à des événements qui ne sont plus utilisés dans notre nouvelle solution interne.

Pour une meilleure compréhension de l'état de la demande ou brevet qui figure sur cette page, la rubrique Mise en garde , et les descriptions de Brevet , Historique d'événement , Taxes périodiques et Historique des paiements devraient être consultées.

Historique d'événement

Description Date
Inactive : CIB expirée 2018-01-01
Demande non rétablie avant l'échéance 2008-01-17
Le délai pour l'annulation est expiré 2008-01-17
Réputée abandonnée - omission de répondre à un avis sur les taxes pour le maintien en état 2007-01-17
Inactive : CIB de MCD 2006-03-12
Lettre envoyée 2006-01-24
Toutes les exigences pour l'examen - jugée conforme 2006-01-17
Exigences pour une requête d'examen - jugée conforme 2006-01-17
Requête d'examen reçue 2006-01-17
Lettre envoyée 2005-07-27
Lettre envoyée 2005-07-27
Lettre envoyée 2005-07-27
Lettre envoyée 2005-07-27
Lettre envoyée 2005-07-27
Lettre envoyée 2005-07-27
Lettre envoyée 2005-07-27
Inactive : Supprimer l'abandon 2005-07-21
Inactive : Abandon. - Aucune rép. à lettre officielle 2005-06-08
Inactive : Transfert individuel 2005-06-07
Lettre envoyée 2004-06-28
Exigences de prorogation de délai pour l'accomplissement d'un acte - jugée conforme 2004-06-28
Inactive : Prorogation de délai lié aux transferts 2004-06-01
Modification reçue - modification volontaire 2003-10-17
Inactive : CIB attribuée 2003-05-12
Inactive : CIB attribuée 2003-05-12
Inactive : CIB en 1re position 2003-05-12
Inactive : CIB enlevée 2003-05-12
Inactive : CIB attribuée 2003-05-12
Inactive : CIB attribuée 2003-05-12
Inactive : Lettre de courtoisie - Preuve 2003-05-06
Inactive : Page couverture publiée 2003-05-05
Inactive : CIB en 1re position 2003-05-01
Inactive : Notice - Entrée phase nat. - Pas de RE 2003-05-01
Demande reçue - PCT 2003-04-24
Exigences pour l'entrée dans la phase nationale - jugée conforme 2003-03-04
Demande publiée (accessible au public) 2002-03-21

Historique d'abandonnement

Date d'abandonnement Raison Date de rétablissement
2007-01-17

Taxes périodiques

Le dernier paiement a été reçu le 2006-01-16

Avis : Si le paiement en totalité n'a pas été reçu au plus tard à la date indiquée, une taxe supplémentaire peut être imposée, soit une des taxes suivantes :

  • taxe de rétablissement ;
  • taxe pour paiement en souffrance ; ou
  • taxe additionnelle pour le renversement d'une péremption réputée.

Les taxes sur les brevets sont ajustées au 1er janvier de chaque année. Les montants ci-dessus sont les montants actuels s'ils sont reçus au plus tard le 31 décembre de l'année en cours.
Veuillez vous référer à la page web des taxes sur les brevets de l'OPIC pour voir tous les montants actuels des taxes.

Historique des taxes

Type de taxes Anniversaire Échéance Date payée
TM (demande, 2e anniv.) - générale 02 2003-01-17 2003-03-04
Taxe nationale de base - générale 2003-03-04
TM (demande, 3e anniv.) - générale 03 2004-01-19 2004-01-13
Prorogation de délai 2004-06-01
TM (demande, 4e anniv.) - générale 04 2005-01-17 2005-01-11
Enregistrement d'un document 2005-06-07
TM (demande, 5e anniv.) - générale 05 2006-01-17 2006-01-16
Requête d'examen - générale 2006-01-17
Titulaires au dossier

Les titulaires actuels et antérieures au dossier sont affichés en ordre alphabétique.

Titulaires actuels au dossier
HUMAN GENOME SCIENCES, INC.
Titulaires antérieures au dossier
CHARLES E. BIRSE
CRAIG A. ROSEN
D. ROXANNE DUAN
DANIEL R. SOPPET
GEORGE A. KOMATSOULIS
GIL H. CHOI
HENRIK S. OLSEN
JIAN NI
KEVIN P. BAKER
MICHELE FISCELLA
PAUL A. MOORE
PING WEI
REINHARD EBNER
YANGGU SHI
Les propriétaires antérieurs qui ne figurent pas dans la liste des « Propriétaires au dossier » apparaîtront dans d'autres documents au dossier.
Documents

Pour visionner les fichiers sélectionnés, entrer le code reCAPTCHA :



Pour visualiser une image, cliquer sur un lien dans la colonne description du document. Pour télécharger l'image (les images), cliquer l'une ou plusieurs cases à cocher dans la première colonne et ensuite cliquer sur le bouton "Télécharger sélection en format PDF (archive Zip)" ou le bouton "Télécharger sélection (en un fichier PDF fusionné)".

Liste des documents de brevet publiés et non publiés sur la BDBC .

Si vous avez des difficultés à accéder au contenu, veuillez communiquer avec le Centre de services à la clientèle au 1-866-997-1936, ou envoyer un courriel au Centre de service à la clientèle de l'OPIC.


Description du
Document 
Date
(aaaa-mm-jj) 
Nombre de pages   Taille de l'image (Ko) 
Description 2003-03-03 256 15 315
Abrégé 2003-03-03 1 66
Revendications 2003-03-03 5 159
Description 2003-03-03 259 13 616
Description 2003-10-16 250 14 899
Description 2003-10-16 182 10 520
Description 2003-10-16 87 3 639
Avis d'entree dans la phase nationale 2003-04-30 1 189
Demande de preuve ou de transfert manquant 2004-03-07 1 101
Courtoisie - Certificat d'enregistrement (document(s) connexe(s)) 2005-07-26 1 114
Courtoisie - Certificat d'enregistrement (document(s) connexe(s)) 2005-07-26 1 114
Courtoisie - Certificat d'enregistrement (document(s) connexe(s)) 2005-07-26 1 114
Courtoisie - Certificat d'enregistrement (document(s) connexe(s)) 2005-07-26 1 114
Courtoisie - Certificat d'enregistrement (document(s) connexe(s)) 2005-07-26 1 114
Courtoisie - Certificat d'enregistrement (document(s) connexe(s)) 2005-07-26 1 114
Courtoisie - Certificat d'enregistrement (document(s) connexe(s)) 2005-07-26 1 114
Rappel - requête d'examen 2005-09-19 1 116
Accusé de réception de la requête d'examen 2006-01-23 1 176
Courtoisie - Lettre d'abandon (taxe de maintien en état) 2007-03-13 1 175
Correspondance 2003-04-30 1 23
Correspondance 2004-06-27 1 15
PCT 2003-03-03 12 548

Listes de séquence biologique

Sélectionner une soumission LSB et cliquer sur le bouton "Télécharger la LSB" pour télécharger le fichier.

Si vous avez des difficultés à accéder au contenu, veuillez communiquer avec le Centre de services à la clientèle au 1-866-997-1936, ou envoyer un courriel au Centre de service à la clientèle de l'OPIC.

Soyez avisé que les fichiers avec les extensions .pep et .seq qui ont été créés par l'OPIC comme fichier de travail peuvent être incomplets et ne doivent pas être considérés comme étant des communications officielles.

Fichiers LSB

Pour visionner les fichiers sélectionnés, entrer le code reCAPTCHA :