Title of Invention

A RECOMBINANT CANCER-ASSOCIATED EPITOPE, VACCINES COMPRISING IT, ANTIBODIES BINDING TO SAID EPITOPE, KITS AND COMPOSITIONS COMPRISING SAID ANTIBODIES

Abstract This invention discloses an isolated cancer-associated epitope comprising recombinant cytokeratin 8 and cytokeratin 18 polypeptide fragments, wherein the cytokeratin 8 polypeptide is shorter than 482 amino acids and comprises SEQ ID NO:3 or SEQ ID NO:5, and the cytokeratin 18 polypeptide is shorter than 429 amino acids and comprises SEQ ID NO:4 or SEQ ID NO:6. The invention is also for vaccines comprising it, antibodies binding to said epitope, kits and compositions comprising said antibodies.
Full Text A RECOMBINANT CANCER-ASSOCIATED EPITOPE, VACCINES
COMPRISING IT, ANTIBODIES BINDING TO SAID EPITOPE, KITS AND
COMPOSITIONS COMPRISING SAID ANTIBODIES
FIELD OF THE INVENTION
The present invention relates to cancer-associated epitopes, antibodies and
polypeptide binding entities directed against such epitopes. The invention also relates to
diagnostic agents comprising the epitopes, antibodies or binding entities, and to the use of
the epitope, antibodies or binding entities for a variety of diagnostic or therapeutic
purposes. Pharmaceutical compositions are also contemplated by the invention, where the
composition include the epitopes, antibodies or binding entities.
BACKGROUND OF THE INVENTION
Malignant tumors sometimes express characteristic antigens or "markers" that
offers a means for detecting and possibly treating the tumors. For example, antigens that
are characteristics of the tumor may be purified and formulated and used to generate
antibodies. The antibodies raised by these antigens can be used as detection tools to
monitor the level of tumor marker in the host to track the course of the disease or the
effectiveness of treatment. Antibodies have also been coupled to toxins and administered
to treat cancer. In some instances, the antigens can be used as vaccines to stimulate an
antibody response and a celluler immune response within a cancer patient and thereby
discourage the growth and spread of the cancer.
Glandular epithelia cells contain a network of intermediate filaments that
predominantly consists of complexes of cytokeratin 8 (K8) and cytokeratin 18 (K18).
These filaments provide resilience in response to mechanical stress by forming a stable
network attached to specific cell-cell contacts of the desmosome eukaryotes are
expressed in a tissue-specific and cell type-restricted pattern (2). In epithelia cells,
intermediate filaments consist of stoichiometrically equal amounts of type I (smaller,
acidic) and type II (larger, neutral or basic) cytokeratin polypeptides that form strongly
interacting heterodimers (3-5).
Each cytokeratin polypeptide consists of a central 300-350 amino acid α-helical
rod domain that is flanked by non-helical head (N-terminal) and tail (C-

terminal) domains of various lengths and compositions. The rod domain can be
further subdivided into four sub-domains (1 A, 1B, 2A and 2B), which are
interspaced by short non-helical linkers (L1, L12, L2). Likewise, the head
domain can be subdivided into three domains: the end domain (El), the variable
domain (V1) and a region of sequence homology (H1) nearest to the rod domain
(6).
The assembly of intermediate filaments appears to involve several
association steps that depend on interactions between different domains. In
general, a type I and a type II cytokeratin polypeptide align in parallel to yield a
coiled coil heterodimer (7,8). Subsequently, a tetramer is formed by anti-
parallel, staggered, side-by-side aggregation of two dimers. The tetramers
polymerize end to end to form a protofilament, and eight protofilaments then
combine to produce the final 10 nm filament (9).
The assembled rods form a protofilament backbone structure from two
coiled coil subunits. However, the head and the tail domain are not thought to be
part of the filamentous backbone. Instead, the head and tail domain appear to
protrude laterally and to contribute to protofilament and intermediate filaments
packing. The head and tail domains may also contribute to intermediate filament
interaction with other cellular components (10-12). Thus, cytokeratins lacking
the head and tail domains are generally capable of coiled coil and higher order
lateral interactions, but are deficient in filament elongation (13).
Cytokeratin 8 (K8) type I and cytokeratin 18 (K18) type II are the major
components of intermediate filaments of simple or single layer epithelia, such as
those of the intestine, liver and breast ducts (4). These two cytokeratins form
heterodimers and filaments. Deletion studies of K8 and K18 cytokeratins have
shown that the head domains play a crucial role in forming heterodimers and
filaments. Co-transfection of head-deleted K8 and head-deleted Kl 8 resulted in
the formation of a dispersed non-fibrillar pattern, while co-transfection of a
combination of one headless plus one intact cytokeratin resulted in the formation
of cytoplasmic granules or fibrils (12). More detailed analysis showed that only
short and irregular intermediate filaments were generated when K8 and K18
were N-terminally truncated by deleting the first 66 amino acid of each of the

cytokeratins (13). The whole, or nearly the whole, H1 region of the head domain
was required for generation of these short filaments. Only tetramers were
generated when a major part of the H1 domain was additionally removed to form
a complex between a truncated K8 (amino acids 75-483) and a truncated K18
(amino acids 67-385) (13). The importance of H1 region apparently relates to its
involvement in the alignment of the two heterodimers and to the stabilization of
the formed heterotetramer complexes.
The precise function of K8 and K18 remains largely unclear, although
recent data indicates that both cytokeratins are important in natural development.
A mutant of K18 (arg89 cys), expressed as a dominant trait in transgenic mice,
resulted in marked disruption of the liver and pancreas intermediate filament
network, leading to hepatocyte instability and associated liver inflammation and
necrosis (14,15). The phenotype of K8 or K19 knockout mice included complete
or partial midgestational embryonic lethality depending on the genetic
background, female sterility and adult colorectal hyperplasia in the surviving
animals (16-18). Other data have suggested that K8/K18 filaments play a role in
multiple drug resistance (19-21).
During cell transformation and tumor development, the cell type
specificities of K8 and K18 are conserved, making them useful as clinical
histopathological markers for tissue type identification (22-24). Given that the
cell type specificities of K8 and K18 are conserved during cell transformation
and tumor development, one would not expect that the KS and K18 cytokeratins
would exhibit a new antigenic epitope in cancerous cells.
SUMMARY OF THE INVENTION
The invention provides an isolated cancer-associated epitope comprising
two separate polypeptides. The first polypeptide can have SEQ ID NO:3 of
cytokeratin 8 and the second polypeptide can have SEQ ID NO:4 of cytokeratin
18. Alternatively, the first polypeptide can have SEQ ID NO:5 of cytokeratin 8
and the second polypeptide can have SEQ ID NO:6 of cytokeratin 18. Moreover,
the first polypeptide can have SEQ ID NO:3 of cytokeratin 8 and the second
polypeptide can have SEQ ID NO:6 of cytokeratin 18. The first polypeptide can

also have SEQ ID NO:5 of cytokeratin 8 and the second polypeptide can have
SEQ ID NO:4 of cytokeratin 18.
Such isolated epitopes can be detected in filamentous cytoplasmic
structures of adenocarcinoma cells but are not substantially detected in normal
cells. Examples of adenocarcinomas where these epitopes can be detected
include colon adenocarcinoma, ovarian adenocarcinoma, renal adenocarcinoma,
mammary adenocarcinoma, lung adenocarcinoma, pancreatic adenocarcinoma
and non-seminomal testis carcinoma cells. These epitopes are useful for making
cancer-specific antibodies, and for.diagnosing cancer by detecting either the
antigenic epitopes or antibodies directed against these epitopes in the blood,
serum, feces or urine of a cancer patient. Accordingly, in one embodiment, the
epitopes are provided in a kit.
In another embodiment, the invention provides an antibody or other
binding entity that can bind any of the cancer-associated epitopes of the
invention. In one embodiment, the antibody or binding entity can include a
polypeptide comprising any one of SEQ ID NO:7-35. Preferred antibodies or
binding entities include polypeptides comprising any one of SEQ ED NO:21-35,
or a combination thereof. In another embodiment, the invention is directed to a
polypeptide comprising any combination of SEQ ID NO:7-33, wherein the
polypeptide that can bind an epitope of the invention. The antibody or binding
entity can be encoded by a nucleic acid comprising any one of SEQ ID NO.36-
39. Such a binding entity or antibody can detect the cancer-associated epitope in
filamentous cytoplasmic structures of colon adenocarcinoma, ovarian
adenocarcinoma, renal adenocarcinoma, mammary adenocarcinoma, lung
adenocarcinoma, pancreatic adenocarcinoma and non-seminomal testis
carcinoma cells. The antibody or binding entity can have a label or diagnostic
imaging agent attached to it. The invention also provides such compositions and
kits containing the binding entities or antibodies. Preferably, when antibodies
are employed, the antibody is not a COU-1 monoclonal antibody.
The invention further provides a method of detecting adenocarcinoma by
contacting an antibody or binding entity of the invention with a test sample and
detecting whether the antibody or binding entity binds to a cancer-associated

epitope. The antibodies and binding entities can have a label or diagnostic
imaging agent attached thereto.
The invention also provides a method of treating cancer in a mammal by
administering a therapeutically effective amount of an antibody or binding entity
of the invention that can bind to the cancer-associated epitope. Such an antibody
or binding entity can have a therapeutically useful agent attached thereto.
The invention further provides a method of treating cancer in a mammal
comprising administering a therapeutically effective amount of a cancer-
associated epitope of the invention.
The invention also provides a method of treating cancer in a mammal
comprising administering a therapeutically effective amount of a protease
inhibitor that can inhibit formation of the cancer-associated epitopes of the
invention by inhibiting the protease(s) that cleave cytokeratin 8 and cytokeratin
18. In one embodiment, the protease is a trypsin-like protease and the inhibitor
is a serine protease inhibitor or a trypsin inhibitor.
The invention also provides a method of identifying a mutant antibody
comprising fusing a nucleic acid encoding a polypeptide having any one of SEQ
ID NO:7-35 to a nucleic acid encoding a phage coat protein to generate a
recombinant nucleic acid encoding a fusion protein, mutating the recombinant
nucleic acid encoding the fusion protein to generate a mutant nucleic acid
encoding a mutant fusion protein, expressing the mutant fusion protein on the
surface of a phage and selecting phage that bind to a cancer-associated epitope of
the invention.
DESCRIPTION OF THE FIGURES
Figure 1 illustrates the purification of cytokeratin from colon cancer
tissue.
Figure 1A provides an elution profile (OD280, solid line) of cytokeratin-
enriched material applied to a QFF anion-exchange column (100-ml bed volume)
in SDS-containing buffer and eluted with a linear gradient to 1 M NaCl (dashed
line). Fractions containing reactivity with COU-1 were observed in the first and
second peaks of the salt gradient.

Figure 1B provides an expanded view of a region the elution profile
shown in Figure 1A. This expanded profile provides a comparison of OD280
absorption (solid line), salt gradient (dashed line) and COU-1 reactivity (dotted
line) for fractions (30-53). Proteins from these fractions were coated onto
ELISA wells and reacted with the COU-1 antibody.
Figure 1C provides a Coomassie-stained blot of electrophoretically
separated proteins from fractions 41-50, raw homogenate (H), and material
applied to the QFF anion-exchange column (S). An extract of the colon cancer
cell line Colon 137 (C137) was included as control.
Figure 1D provides a Western blot (stained with COU-1) of
electrophoretically separated proteins from fractions 41-50, raw homogenate (H),
and material applied to the QFF anion-exchange column (S). An extract of the
colon cancer cell line Colon 137 (C137) was included as control. This blot
illustrates the purity of the cytokeratins obtained from the QFF column and the
reactivity of the COU-1 antibodies with 3 bands at molecular weights of 42-46
kDa.
Figure 2 provides a Western blot analysis of cytokeratin preparations
purified from colon cancer tissue or from normal colon epithelia under identical
conditions. Homogenate (homog) and anion-exchange chromatography-purified
material (QFF) was applied to the SDS-PAGE gel at 10-fold dilutions (1:1,1:10,
1:100). The proteins were transferred to PVDF membranes and stained with
COU-1 or murine anti-K18 Mab. While the anti-K18 antibody intensely stained
cytokeratin preparations from normal and malignant colon epithelia, COU-1
intensely stained only cytokeratin proteins from cancer tissue (three bands of
about 42-46 kDa), and not cytokeratin proteins from normal epithelia.
Figure 3 illustrates the SDS-PAGE separation and N-terminal sequencing
of cytokeratins purified from colon cancer tissue and further illustrates the
presence of N-terminally-truncated K8, K18 and K19 cytokeratins in these colon
cancer tissues.
Figure 3 A is a PVDF membrane blot of SDS-PAGE-separated purified
cytokeratin. Region (a) of this membrane was stained with Coomassie. Strips of
the membrane were also stained with the COU-1 antibody (region b), a mouse

anti-K8 antibody (region c) and a mouse anti-K18 antibody (region d). These
data illustrate that ten different protein bands (1-10) can be detected. These ten
bands were each N-terminally sequenced.
Figure 3B provides the amino acid sequences of cytokeratin proteins
isolated from colon cancer tissues as determined by N-tenninal sequencing (SEQ
ID NOs:54-6l). In addition, the reactivity of the different isolated cytokeratin
proteins with a panel of K8/K18 antibodies is shown.
Figure 4 provides a map of the structural domains of the K8 and K18
cytokeratins (center). The secondary structural domains of the cytokeratin
polypeptides were identified from the amino acid sequences. A central rod
domain is shown that is flanked by a non-helical N-tenninal head domain and a
non-helical C-terminal tail domain. Domains 1A, 1B, 2A and 2B are α-helical
subdomains of the rod interspaced by linkers L1, L12 and L2. This figure also
provides a schematic representation of the K8 and K18 N-terminal and C-
terminal deletion proteins. Deletion protein names provide the starting and
ending amino acid residue numbers of the deletion protein. All deletion proteins
were expressed as GST fusion proteins. The positive (+) and negative (-)
reactivity of the deletion protein fragments with COU-1 after incubation with the
complementary keratin is shown in parentheses.
Figure 5 provides a Western blot analysis of a panel of C-terminal deleted
K18 fragments. SDS-PAGE gels containing a panel of C-terminal deleted K18
fragments expressed as GST fusion proteins were run in parallel, transferred to
PVDF membranes and stained with either a goat anti-GST antibody (A), COU-1
(B) or a mouse anti-KI8 antibody (CY-90) (D).
Figure 5 A is a Western blot of an electrophoretically separated panel of
C-terminal deleted K18 protein fragments expressed as GST fusion proteins that
was stained with a goat anti-GST antibody. The identity of the different K18
protein fragments is provided at the top, where the numbers indicate which
amino acids are present in the different K18 protein fragments. A GST protein
preparation was used a positive control for GST antibody staining. An MCF7
cancer cell lysate was used as positive control for the cytokeratin epitope (no
staining is visible because the GST protein is not present in the lysate). The GST

antibody staining demonstrated that all K18 fragments were expressed well and
at approximately the same levels.
Figure 5B is a replica of the Western blot of an electrophoretically
separated panel of C-terminally deleted K18 fragments provided in Figure 5A
that was stained with the COU-1 antibody. On this blot, COU-1 only reacted
with MCF7 cancer cell lysate used as positive control. No substantial binding of
COU-1 to individual K18 fragments was observed.
Figure 5C is a replica of the Western blot of an electrophoretically
separated panel of C-terminally deleted K18 fragments provided in Figure 5A.
However, this blot was incubated with purified intact K8 prior to staining COU-
1. COU-1 bound strongly to some of the K18 fragments when complexed with
K8.
Figure 5D is a replica of the Western blot of an electrophoretically
separated panel of C-terminally deleted K18 fragments provided in Figure 5A
that was stained with a mouse anti-K18 antibody (CY-90). The CY-90 antibody
reacted with an epitope corresponding to residues 340-390 in the C-terminal part
of non-complexed K18.
Figure 6 provides an SDS-PAGE and Western blot analysis of a panel of
C-terminal deleted K8 fragments. SDS-PAGE gels containing a panel of C-
terminal deleted K8 fragments expressed as GST fusion proteins were run in
parallel and stained with Coomassie Blue (A), or transferred to PVDF
membranes and stained with COU-1 (B) or incubated with purified intact K18
prior to staining HMab COU-1 (C).
Figure 6A is an SDS-PAGE gel of an electrophoretically separated panel
of C-terminal deleted K8 fragments stained with Coomassie Blue. The identity
of the different K8 protein fragments is provided at the top, where the numbers
indicate the range of amino acids present in the different K18 protein fragments.
Coomassie Blue staining demonstrated that all fragments were expressed
approximately equally well.
Figure 6B is a replica of the Western blot of an electrophoretically-
separated panel of C-terminally deleted K8 fragments provided in Figure 6A that
was stained with the COU-1 antibody. No binding of COU-1 to individual K8

fragments was observed. On this blot, COU-1 only reacted with the positive
control, a MCF7 cancer cell iysate.
Figure 6C is a replica of the Western blot of an electrophoretically
separated panel of C-terminally deleted K8 fragments provided in Figure 6A that
was incubated with purified intact K18 prior to staining HMab COU-1. COU-1
bound strongly to some of the K8 fragments because when they had formed
complexes with K18.
Figure 7 provides a Western blot analysis of a panel of C-terminally
deleted K8 or K18 fragments that were electrophoretically separated, transferred
to a PVDF membrane and then incubated with different purified C-terminally
deleted K8 or K18 fragments to form K8/K18 complexes prior to staining with
COU-1
Figure 7A provides a Western blot analysis of a panel of C-terminally
deleted Kl8(l-72), K18(l-124), Kl8(1-187) and intact K18 protein fragments
that were electrophoretically separated and transferred to a PVDF membrane.
The membrane was then incubated with purified C-terminally deleted K8(l-129)
fragment and stained with COU-1. The COU-1 antibodies bound strongly to
K18(1-124)/K8(1-129) complexes.
Figure 7B is a replica of the Western blot of an electrophoretically-
separated panel of C-terminally deleted K18 fragments provided in Figure 7A
that was incubated with purified C-terminally deleted K8(l-233) fragment and
stained with COU-1. COU-1 staining was absent or only minimally observed for
the K18(l-187)/K8(l-233) complex.
Figure 7C is a replica of the Western blot of an electrophoretically-
separated panel of C-terminally deleted K18 fragments provided in Figure 7A
that was incubated with purified intact K8 and stained with COU-1. COU-1
staining was absent or only minimally observed for the K18(l-187)/intact K8
complex.
Figure 7D provides a Western blot analysis of a panel of C-terminally
deleted K8(l-85), K8(l-129), K8(l-233) and intact K8 polypeptides that were
electrophoretically separated and transferred to a PVDF membrane. The
membrane was then incubated with purified K18(1-124) and stained with COU-

1. The COU-1 antibody recognized K8(l-129) complexed with K18(l-124), and
K8(l-233) complexed with K18(l-124). No COU-1 binding was observed with
complexes containing K8(l-85).
Figure 7E is a replica of the Western blot of an electrophoretically-
separated panel of C-terminally deleted K8 fragments provided in Figure 7D that
was incubated with purified K18(l-187) and stained with COU-1. The COU-1
antibody recognized K8(l-129) complexed with K18(l-187) and K8(l-233)
complexed with K18(l-187). No COU-1 binding was observed with any
complexes containing K8(l-85).
Figure 7F is a replica of the Western blot of an electrophoretically-
separated panel of C-terminally deleted K8 fragments provided in Figure 7D that
was incubated with purified K18(1-213) and stained with COU-1. The COU-1
antibody recognized K18(l-213) complexed with K8(l-129) and K18(l-213)
complexed with K8(l-233) K18(l-187). No COU-1 binding was observed with
any complexes containing K8(l-85).
Figure 8 provides a schematic representation of the N-terminal head
domain and the adjacent rod domain of K8/K18 heterotypic complex.
Figure 8A identifies the sites where K8 (SEQ ID NO:62) and K18 (SEQ
ID NO:63) are proteolytically cleaved (arrows). For cytokeratin K8, cleavage
was after Arg-22, after Arg-39, after Val-65 and after Leu-75. For cytokeratin
K18, cleavage was after Arg-49 and after IIe-67. The positions of residues that
are post-translationally phosphorylated (PO4, P) or glycosylated (GlcNac, G) are
also identified.
Figure 8B is a schematic diagram illustrating how cleavage of the N-
terminal head domain of K8 and K18 cytokeratins can cause a conformational
change allowing the COU-1 antibody to access the epitope. This diagram is
consistent with observations made in vivo in cancer cells and made on the
formation of recombinant K8/K18 complexes. This diagram further illustrates
how in vitro deletion of a major portion of the C-terminal domain of one of the
two cytokeratin proteins may also artificially expose the COU-1 epitope. This
diagram is also consistent with complex formation studies on a panel of
recombinant K8/K18 deletion proteins.

Figure 9 illustrates that COU-1 binds preferentially to heterotypic
complexes containing N-terminally deleted K8 fragments. This figure provides a
PVDF membrane blot of SDS-PAGE-separated K8 and K18 proteins. Region A
has intact K8 or K8 (66-483) proteins that were electrophoretically-separated and
then reacted with purified K18 (50-430) prior to staining with COU-1. Region B
has intact K8 or K8 (66-483) proteins incubated with purified intact K18 prior to
staining with COU-1. Region C has K18 (50-430) and intact K18 that were
electrophoretically-separated and then incubated with purified K8 (66-483) prior
to staining with COU-1. Strong staining of a band of about 75 kDa (K8/K18
proteins + the GST fusion protein) was observed in lanes containing K8(66-
483)/K18(50-430) and K8(66-4S3)/intact K18 with COU-1. Region D has
Kl8(50-430) and intact K18 proteins that were electrophoretically-separated and
then incubated with purified intact K8 prior to staining with COU-1. Only weak
staining was observed to intact K8/K18 (50-430) and intact K8/intact K18.
Figure 10 illustrates that COU-1 binds preferentially to truncated forms
of recombinant heterotypic K8/K18 complexes as measured by ELISA.
Heterotypic complexes were generated by combining purified recombinant K8(l-
129) or intact K8 with purified recombinant K18(1-124) or intact K18 in equal
molar ratio. A 5 µg/ml solution of these complexes was used to coat ELISA
plates. The ELISA plates were incubated with COU-1 in serial dilutions. Bound
COU-1 was detected and visualized with an AP-labeled secondary anti-human
kappa antibody and para-nitrophenylphosphate. As shown, the K8(1-129)/intact
K18 complex (diamond symbols) and the K8/K18(l-124) (circles) bound more
COU-1 antibodies than the intact K8/intact K18 complex.
Figure 11 illustrates the distribution of N-terminally-truncated K8/K18
complexes (A and E) and K18 (B and F) in breast cancer cells. Ethanol-fixed
MCF7 breast cancer cells were separately incubated with COU-1 antibodies (A
and E) and CY90 monoclonal antibodies (B and F), which binds to intact K18.
Bound COU-1 was detected with FITC-goat anti-human γ-chain antibody (green)
and bound CY90 detected with a Texas Red-goat-anti-mouse IgG antibody (red).
DIC images (D and H) were included to visualize the composition of the cells.
Partial co-localization, as visualized by yellow in the merged images (C and G),

was observed between the two antibodies. However, N-terminally truncated
K8/K18 complexes were predominantly found in newly-formed, proliferating
cancer cells (arrows), whereas K18 structures were equally present in all cells
(arrowheads).
Figure 12 shows the cellular distribution of the N-terminally-truncated
K8/K18 complex recognized by COU-1 (A and E) and K18 recognized by Mab
CY-90 (B and F) in breast cancer cells. MCF7 breast cancer cells were
processed and stained as described in Figure 11. DIC images (D and H) were
included to visualize the composition of the cells. While whole intermediate
filaments were stained with Mab CY-90 (arrowheads), COU-1 (arrows) only
stained short fibrils and globular structures. Some co-localization of the two
antibodies, as visualized by yellow in the merged images (C and G), was
observed.
Figure 13 provides the amino acid sequences for the variable heavy and
light chain of the human monoclonal antibody COU-1 in comparison to closest
known germ-line sequences (SEQ ID NOs:7-20).
DETAILED DESCRIPTION OF THE INVENTION
According to the invention, the separate K8 cytokeratin polypeptide joins
with the K18 cytokeratin polypeptide to form an antigenic epitope that is only
visible in cancerous, and not in normal, cells. Such neoepitopes are generated by
specific proteolytic cleavage of K8/K18 complexes in carcinoma cells. The new
epitopes visible in cancer cells are used to generate antibodies or binding entities
that are diagnostic of cancer and that are useful for treatment of cancer patients.
Definitions
The term "antibody" is used in the broadest sense, and specifically covers
monoclonal antibodies (including full length monoclonal antibodies), polyclonal
antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody
fragments (e.g., Fab, F(ab')2 and Fv) so long as they exhibit the desired biological
activity.

A "binding entity," as used herein, is a polypeptide that can bind to the
epitope identified by the invention. For example, a binding entity of the
invention is a polypeptide that can bind to an epitope comprising two separate
polypeptides, a cytokeratin S polypeptide and a cytokeratin 18 polypeptide,
wherein the cytokeratin 8 polypeptide comprises SEQ ID NO:3 or SEQ ID NO:5,
and the cytokeratin 18 polypeptide comprises SEQ ID NO:4 or SEQ ID NO:6.
The terms "cancer" and "cancerous" refer to or describe the physiological
condition in mammals that is typically characterized by unregulated cell growth.
Examples of cancer include but are not limited to, carcinoma, lymphoma,
blastoma, sarcoma, and leukemia. More particular examples of such cancers
include in colon adenocarcinoma, ovarian adenocarcinoma, renal
adenocarcinoma, mammary adenocarcinoma, lung adenocarcinoma, pancreatic
adenocarcinoma and non-seminomal testis carcinoma tissues.
The COU-1 antibody is a monoclonal antibody produced by the human
hybridoma cell line B9165 (ECACC 87040201). It can bind to a carcinoma-
associated antigen that has an apparent molecular weight of about 43,000 and an
isoelectric point in the range of about 5.4-6.2.
The expression "control sequences" refers to DNA sequences necessary
for the expression of an operably linked coding sequence in a particular host
organism. The control sequences that are suitable for prokaryotes, for example,
include a promoter, optionally an operator sequence, and a ribosome binding site.
Eukaryotic cells are known to utilize promoters, polyadenylation signals, and
enhancers.
The "derivative" of a reference antigenic epitope, antibody, nucleic acid,
protein, polypeptide or peptide, has related but different sequence or chemical
structure than the respective reference antigenic epitope, antibody, nucleic acid,
protein, polypeptide or peptide. Such a derivative antigenic epitope, antibody,
nucleic acid, protein, polypeptide or peptide is generally made purposefully to
enhance or incorporate some chemical, physical or functional property that is
absent or only weakly present in the reference antigenic epitope, antibody,
nucleic acid, protein, polypeptide or peptide. A derivative nucleic acid differs in
nucleotide sequence from a reference nucleic acid whereas a derivative antigenic

epitope, antibody, protein, polypeptide or peptide differs in amino acid sequence
from the reference antigenic epitope, antibody, protein, polypeptide or peptide,
respectively. Such sequence differences include one or more substitutions,
insertions, additions, deletions, fusions and truncations, which can be present in
any combination. Differences can be minor (e.g., a difference of one nucleotide
or amino acid), or more substantial, involving several or many nucleotides or
amino acids. However, the sequence of the derivative is not so different from
the reference that one of skill in the art would not recognize that the derivative
and reference are related in structure and/or function. Generally, differences are
limited so that the reference and the derivative are closely similar overall and, in
many regions, identical. A "variant" differs from a "derivative" nucleic acid,
protein, polypeptide or peptide in that the variant can have silent structural
differences that do not significantly change the chemical, physical or functional
properties of the reference nucleic acid, protein, polypeptide or peptide. In
contrast, the differences between the reference and derivative nucleic acid,
protein, polypeptide or peptide are intentional changes made to improve one or
more chemical, physical or functional properties of the reference nucleic acid,
protein, polypeptide or peptide.
The term "identity" or "homology" shall be construed to mean the
percentage of amino acid residues in the candidate sequence that are identical
with the residue of a corresponding sequence to which it is compared, after
aligning the sequences and introducing gaps, if necessary to achieve the
maximum percent identity for the entire sequence, and not considering any
conservative substitutions as part of the sequence identity. Neither N- or C-
terminal extensions nor insertions shall be construed as reducing identity or
homology. Methods and computer programs for the alignment are well known in
the art. Sequence identity may be measured using sequence analysis software
(e.g., Sequence Analysis Software Package, Genetics Computer Group,
University of Wisconsin Biotechnology Center, 1710 University Ave., Madison,
Wis. 53705). This software matches similar sequences by assigning degrees of
homology to various substitutions, deletions, and other modifications.

A "liposome" is a small vesicle composed of various types of lipids,
phospholipids and/or surfactant that is useful for delivery of a drug (such as the
antigenic epitopes and antibody mutants disclosed herein and, optionally,
chemotherapeutic agent) to a mammal. The components of the liposome are
commonly arranged in a bilayer formation, similar to the lipid arrangement of
biological membranes.
"Mammal" refers to any animal classified as a mammal, including
human, domestic and farm animals, nonhuman primates, and zoo, sports, or pet
animals, such as dogs, horses, cats, cows, etc.
Nucleic acid is "operably linked" when it is placed into a functional
relationship with another nucleic acid sequence. This can be a gene and a
regulatory sequence(s) that are connected in such a way as to permit gene
expression when the appropriate molecules (e.g., transcriptional activator
proteins) are bound to the regulatory sequences(s). For example, DNA for a
presequence or secretory leader is operably linked to DNA for a polypeptide if it
is expressed as a preprotein that participates in the secretion of the polypeptide; a
promoter or enhancer is operably linked to a coding sequence if it affects the
transcription of the sequence; or a ribosome binding site is operably linked to a
coding sequence if it affects the transcription of the sequence; or a ribosome
binding site is operably linked to a coding sequence if it is positioned so as to
facilitate translation. Generally, "operably linked" means that the DNA
sequences being linked are contiguous, and, in the case of a secretory leader,
contiguous and in reading phase. However, enhancers do not have to be
contiguous. Linking is accomplished by ligation at convenient restriction sites. If
such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used
in accordance with conventional practice.
The terms "protein," "polypeptide" and "peptide" are used
interchangeably. They refer to a chain of two (2) or more amino acids that are
linked together with peptide or amide bonds, regardless of post-translational
modification (e.g., glycosylation or phosphorylation). Antigens, epitopes and
antibodies are specifically intended to be within the scope of this definition. The

polypeptides of this invention may comprise more than one subunit, where each
subunit is encoded by a separate DNA sequence.
The phrase "substantially identical" with respect to an antigen, antibody
or binding entity polypeptide sequence shall be construed as a polypeptide
exhibiting at least 70%, preferably 75%, more preferably 80%, more preferably
85%, even more preferably 90%, even more preferably 95% and especially
preferably 97% or 98% sequence identity to the reference polypeptide sequence.
For polypeptides, the length of the comparison sequences will generally be at
least 25 amino acids. For nucleic acids, the length will generally be at least 75
nucleotides.
The "variant" of a reference antigenic epitope, antibody segment, binding
entity, nucleic acid, protein, polypeptide or peptide, is an antigenic epitope,
antibody segment, binding entity, nucleic acid, protein, polypeptide or peptide,
respectively, with a related but different sequence than the respective reference
antigenic epitope, antibody segment, binding entity, nucleic acid, protein,
polypeptide or peptide. The differences between variant and reference antigenic
epitopes, antibody segments, binding entities, nucleic acids, proteins,
polypeptides or peptides are silent or conservative differences. A variant
nucleic acid differs in nucleotide sequence from a reference nucleic acid whereas
a variant antigenic epitope, antibody segment, binding entity, protein,
polypeptide or peptide differs in amino acid sequence from the reference
antigenic epitope, antibody segment, binding entity, protein, polypeptide or
peptide, respectively. A variant and reference antigenic epitope, antibody
segment, binding entity, nucleic acid, protein, polypeptide or peptide may differ
in sequence by one or more substitutions, insertions, additions, deletions, fusions
and truncations, which may be present in any combination. Differences can be
minor (e.g., a difference of one nucleotide or amino acid), or more substantial.
However, the structure and function of the variant is not so different from the
reference that one of skill in the art would not recognize that the variant and
reference are related in structure and/or function. Generally, differences are
limited so that the reference and the variant are closely similar overall and, in
many regions, identical.

Epitope
According to the invention, one or more novel neoepitopes that are
immunologically recognizable are generated in a variety of adenocarcinoma- cells
through specific proteolytic cleavage of cytokeratin K8 and cytokeratin K18
proteins. Normal, non-cancerous cells do not display such neoepitopes. The
cytokeratin KS and cytokeratin K18 proteins are separate proteins. However,
they do form a cytokeratin K8/cytokeratin K18 complex. The immunologically
recognizable neoepitope contains amino acids from both the cytokeratin K8 and
cytokeratin K18 proteins.
The epitope of the invention is not substantially present in normal tissues.
However, the epitope becomes exposed in colon adenocarcinoma, ovarian
adenocarcinoma, renal adenocarcinoma, mammary adenocarcinoma, lung
adenocarcinoma, pancreatic adenocarcinoma and non-seminomal testis
carcinoma tissues. The epitope of the invention is predominantly present in
filamentous cytoplasmic structures of these types of cells during proliferation.
Testing indicates that the epitope is not detected in certain sarcomas, malignant





The epitopes of the invention consist of two polypeptides, a cytokeratin
K8 polypeptide and a cytokeratin K18 polypeptide. However, the cytokeratin K8
polypeptide is shorter than the full-length cytokeratin K.8 polypeptide that has
482 amino acids. Moreover, the cytokeratin K18 polypeptide is shorter than the
full-length cytokeratin K18 polypeptide that has 429 amino acids. In some

embodiments, the cytokeratin K8 polypeptide is shorter than about 475 amino
acids, or shorter than about 450 amino acids, or shorter than about 425 amino
acids, or shorter than about 400 amino acids. In some embodiments, the
cytokeratin K18 polypeptide is shorter than about 425 amino acids, or shorter
than about 415 amino acids, or shorter than about 400 amino acids, or shorter
than about 375 amino acids.
One example of an epitope of the invention constitutes two peptidyl
regions of two separate proteins, cytokeratin K.8 (SEQ ID NO:3) and cytokeratin
K18 (SEQ ID NO:3). The epitope involves amino acids 85-129 of cytokeratin 8
sequence, designated SEQ ID NO:3 and provided below.
1 AVRTQEKEQI KTLNNKFASF IDKVRFLEQQ NKMLETKWSL
41 LQQQ
The epitope further involves amino acids 72-124 of cytokeratin 18, designated
SEQ ID NO.4 and provided below.
1 AGMGGIQNEK ETMQSLNDRL ASYLDRVRSL ETENRRLESK
41 IREHLEKKGP QVR
In some instances an appropriate three dimensional structure permitting
interaction between cytokeratin K8 and cytokeratin K18 polypeptides may be
needed to obtain optimal immunoreactivity. Hence, longer cytokeratin
polypeptides can be used as antigens. For example, a cytokeratin K8 polypeptide
having SEQ ID NO:5 can be used with an appropriate cytokeratin K18
polypeptide to generate antibodies. SEQ ID NO:5 is as follows.



Antigenic epitope "fragments" are also contemplated by the invention.
Such fragments do not encompass a full-length cytokeratin but do encode an
antigen that has similar or improved immunological properties relative to an
antigenic epitope having SEQ ID NO:3-6. Thus, fragments of antigenic epitopes
such as SEQ ID NO:3-6 may be as small as about 6 amino acids, about 9 amino
acids, about 12 amino acids, about 15 amino acids, about 17 amino acids, about
18 amino acids, about 20 amino acids, about 25 amino acids, about 30 amino
acids or more. In general, a fragment antigenic epitope of the invention can have
any upper size limit so long as it is has similar or immunological properties
relative to an epitope form by a combination of any one of SEQ ED NO:3-6.
The invention also contemplates a fusion protein comprising a
combination of the SEQ ID NO:3 and the SEQ ID NO:4 peptide. Such a fusion
protein links the two peptides together so that the peptides can more easily form
the cancer associated epitope of the invention.
Fusion polypeptides may generally be prepared using standard
techniques, including chemical conjugation. A fusion polypeptide can also
expressed as a recombinant polypeptide, allowing the production of increased
levels, relative to a non-fused polypeptide, in an expression system. Briefly,
DNA sequences encoding the polypeptide components may be assembled
separately, and ligated into an appropriate expression vector. The 3' end of the
DNA sequence encoding one polypeptide component is ligated, with or without a

peptide linker, to the 5' end of a DNA sequence encoding the second polypeptide
component so that the reading frames of the sequences are in phase. This permits
translation into a single fusion polypeptide that retains the biological activity of
both component polypeptides.
A linker sequence may be employed to separate the first and second
polypeptide components by a distance sufficient to ensure that each polypeptide
folds into its secondary and tertiary structures. Such a linker can be a peptide,
polypeptide, alkyl chain or other convenient spacer molecule.
A polypeptide or peptide linker sequence is incorporated into the fusion
polypeptide using standard techniques well known in the art. Suitable peptide
linker sequences may be chosen based on the following factors: (1) their ability
to adopt a flexible extended conformation; (2) their inability to adopt a secondary
structure that could interact with functional epitopes on the first and second
polypeptides; and (3) the lack of hydrophobic or charged residues that might
react with the polypeptide functional epitopes. In some embodiments, peptide
linker sequences contain Gly, Asn and Ser residues. Other near neutral amino
acids, such as Thr and Ala may also be used in the linker sequence. Amino acid
sequences that may be usefully employed as linkers include those disclosed in
Maratea et al., Gene 40:39-46,1985; Murphy et al., Proc. Natl. Acad. Sci. USA
83:8258-8262, 1986; U.S. Pat. No. 4,935,233 and U.S. Pat. No. 4,751,180. The
linker sequence may generally be from 1 to about 50 amino acids in length.
Linker sequences are generally not required when the first and second
polypeptides have non-essential N-terminal amino acid regions that can be used
to separate the functional domains and prevent steric interference.
The fusion polypeptide can comprise the polypeptide epitope (e.g. SEQ
ID NO:3 and SEQ ID NO:4 peptides) as described herein together with an
unrelated immunogenic protein, such as an immunogenic protein capable of
eliciting a recall response. Examples of such proteins include tetanus,
tuberculosis and hepatitis proteins (see, for example, Stoute et al. New Engl. J
Med., 336:86-91,1997).
In one embodiment, a peptide or polypeptide that can facilitate
development of an immune response against the SEQ ID NO:3 and SEQ ID

N0:4 peptide epitope is used as the linker. Such an immunological fusion
partner can be derived from a Mycobacterium sp. For example, the
immunological fusion partner can be a Mycobacterium tuberculosis-derived
Ral2 fragment. Ral2 compositions and methods for their use in enhancing the
expression and/or immunogenicity of heterologous polynucleotide/polypeptide
sequences is described in U.S. patent application Ser. No. 60/158,585, the
disclosure of which is incorporated herein by reference in its entirety. Briefly,
Ral2 refers to a polynucleotide region that is a subsequence of a Mycobacterium
tuberculosis MTB32A nucleic acid. MTB32A is a serine protease of 32 KD
molecular weight encoded by a gene in virulent and avirulent strains of M.
tuberculosis. The nucleotide sequence and amino acid sequence of MTB32A
have been described (for example, U.S. patent application Ser. No. 60/158,585;
see also, Skeiky et al., Infection and Immun. (1999) 67:3998-4007, incorporated
herein by reference). C-terminal fragments of the MTB32A coding sequence
express at high levels and remain as a soluble polypeptides throughout the
purification process. Moreover, Ral2 may enhance the immunogenicity of
heterologous immunogenic polypeptides with which it is fused. One useful Ral2
fusion polypeptide comprises a 14 KD C-terminal fragment corresponding to
amino acid residues 192 to 323 of MTB32A. Other useful Ral2 polynucleotides
generally comprise at least about 15 consecutive nucleotides, at least about 30
nucleotides, at least about 60 nucleotides, at least about 100 nucleotides, at least
about 200 nucleotides, or at least about 300 nucleotides that encode a portion of
a Ral2 polypeptide.
Ral2 polynucleotides may comprise a native sequence (i.e., an
endogenous sequence that encodes a Ral2 polypeptide or a portion thereof) or
may comprise a variant of such a sequence. Ral2 polynucleotide variants may
contain one or more substitutions, additions, deletions and/or insertions such that
the biological activity of the encoded fusion polypeptide is not substantially
diminished, relative to a fusion polypeptide comprising a native Ral2
polypeptide. Variants preferably exhibit at least about 70% identity, more
preferably at least about 80% identity and most preferably at least about 90%

identity to a polynucleotide sequence that encodes a native Ral2 polypeptide or a
portion thereof.
In another embodiment, an immunological fusion partner is derived from
protein D, a surface protein of the gram-negative bacterium Haemophilus
influenza B (WO 91/18926). Useful portions of protein D comprise
approximately the first third of the protein (e.g., the first N-terminal 100-110
amino acids). Moreover, such a protein D fusion partner may be lipidated.
Within certain preferred embodiments, the first 109 residues of a Lipoprotein D
fusion partner is included on the N-terminus to provide the polypeptide with
additional exogenous T-cell epitopes and to increase the expression level in E.
coli (thus functioning as an expression enhancer). The lipid tail ensures optimal
presentation of the antigen to antigen presenting cells. Other fusion partners
include the non-structural protein from influenzae virus, NS1 (hemaglutinin).
Typically, the N-terminal 81 amino acids are used, although different fragments
that include T-helper epitopes may be used.
In another embodiment, the immunological fusion partner is the protein
known as LYTA, or a portion thereof (preferably a C-terminal portion). LYTA is
derived from Streptococcus pneumoniae, which synthesizes an N-acetyl-L-
alanine amidase known as amidase LYTA (encoded by the LytA gene; Gene
43:265-292,1986). LYTA is an autolysin that specifically degrades certain
bonds in the peptidoglycan backbone. The C-terminal domain of the LYTA
protein is responsible for the affinity to the choline or to some choline analogues
such as DEAE. This property has been exploited for the development of E. coli
C-LYTA expressing plasmids useful for expression of fusion proteins.
Purification of hybrid proteins containing the C-LYTA fragment at the amino
terminus has been described (see Biotechnology 10:795-798,1992). Within a
preferred embodiment, a repeat portion of LYTA may be incorporated into a
fusion polypeptide. A repeat portion is found in the C-terminal region starting at
residue 178. A particularly preferred repeat portion incorporates residues 188-
305.
Another illustrative embodiment involves fusion polypeptides, and the
polynucleotides encoding them, wherein the fusion partner comprises a targeting

signal capable of directing a polypeptide to the endosomal/lysosomal
compartment, as described in U.S. Pat. No. 5,633,234. An immunogenic
polypeptide of the invention, when fused with this targeting signal, will associate
more efficiently with MHC class II molecules and thereby provide enhanced in
vivo stimulation of CD4.sup.+ T-cells specific for the polypeptide.
Polypeptides and fusion proteins of the invention are prepared using any
of a variety of well-known synthetic and/or recombinant techniques.
Polypeptides and fusion proteins that are less than about 150 amino acids can be
generated by synthetic means, using techniques well known to those of ordinary
skill in the art. In one illustrative example, such polypeptides are synthesized
using any of the commercially available solid-phase techniques, such as the
Merrifield solid-phase synthesis method, where amino acids are sequentially
added to a growing amino acid chain. See Merrifield, J. Am. Chem. Soc.
85:2149-2146,1963. Equipment for automated synthesis of polypeptides is
commercially available from suppliers such as Perkin Elmer/Applied
BioSystems Division (Foster City, Calif), and may be operated according to the
manufacturer's instructions.
Small and large fusion proteins and polypeptide epitopes of the invention
can be produced by any other method available to one of skill in the art. For
example, the fusion proteins and polypeptide epitopes can be made
recombinantly by inserting a nucleic acid encoding a selected fusion protein or
polypeptide epitope into an expression vector using any of a variety of
procedures. In general, a nucleic acid encoding the desired protein or polypeptide
is inserted into an appropriate restriction endonuclease site(s) using techniques
known in the art. See generally, Sambrook et al., 1989, Molecular Cloning, A
Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, Cold Spring
Harbor, N.Y.; Sambrook and Russell, Molecular Cloning: A Laboratory Manual,
3rd edition (January 15, 2001) Cold Spring Harbor Laboratory Press, ISBN:
0879695765; Ausubel et al., Current Protocols in Molecular Biology, Green
Publishing Associates and Wiley Interscience, NY (1989)). Construction of
suitable expression vectors containing a fusion protein or a polypeptide epitope
employs standard ligation techniques that are known to the skilled artisan.

The ligated nucleic acid sequences are operably linked to suitable
transcriptional or translational regulatory elements that facilitate expression of
the fusion proteins and polypeptide epitopes of the invention. The regulatory
elements responsible for expression of proteins are located only 51 to the coding
region for the polypeptide. Similarly, stop codons required to end translation and
transcription termination signals are only present 3' to the nucleic acid sequence
encoding the fusion protein or polypeptide epitope. After construction of a
nucleic acid encoding the polypeptide of interest with the operably linked
regulatory elements, this expression cassette can be introduced into a host cell
and the encoded polypeptide can be expressed.
In general, polypeptide compositions (including fusion polypeptides) of
the invention are isolated. An "isolated" polypeptide is one that is removed from
its original environment. For example, a naturally-occurring protein or
polypeptide is isolated if it is separated from some or all of the coexisting
materials in the natural system. Such polypeptides can also be purified. For
example, the polypeptide epitopes and fusion proteins can be at least about 90%
pure, or at least about 95% pure or at least about 99% pure.
Antibodies and Binding Entities
The cytokeratin epitopes of the invention are displayed in a uniform
punctate pattern on the surface of viable carcinoma and adenocarcinoma cells.
Immunohistological studies have demonstrated that the cancer associated epitope
of the invention, in contrast to normal cytokeratin 8 and 18, can be used to
differentiate between malignant and normal colon epithelia, and between colon
cancer metastasis in the liver and surrounding normal hepatocytes. In addition,
the cancer associated epitope of the invention is associated with the membranes
of proliferating cells within the malignant area of biopsies, while resting cells
had a filamentous pattern when stained for the epitope.
The invention provides antibody preparations and binding entities
directed against the epitopes of the invention, for example, antibodies or binding
entities capable of binding an antigenic mixture of at least one peptide from
cytokeratin K8 and at least one peptide from cytokeratin Kl8. Examples of

peptides from cytokeratin K8 include SEQ ID NO:3 and SEQ ID NO:5.
Examples of peptides from cytokeratin K18 include SEQ ED NO:4, and SEQ ID
NO:6.
In one embodiment, the antibody or binding entity can include a
polypeptide comprising any one of SEQ ID NO:7-35,47-49. In some
embodiments, antibodies and binding entities include a polypeptide consisting
essentially of any one of SEQ ID NO:21-35, 47-49. In other embodiments,
antibodies and binding entities include a polypeptide consisting essentially of
any one of SEQ ID NO:8, 10, 12,15, 17, 19, 22, 24, 27, 29 or 32. In another
embodiment, the invention is directed to a binding entity polypeptide comprising
any combination of SEQ ID NO:7-33,47-49, wherein the polypeptide that can
bind an epitope of the invention.
The invention also provides nucleic acids encoding antibody-like
polypeptides of the invention. In one embodiment, the nucleic acid encodes a
polypeptide comprising any one of SEQ ID NO:7-35, 47-49 wherein such a
nucleic acid encodes a polypeptide that can bind an epitope of the invention. In
another embodiment, the nucleic acid encodes a combination of two or more of
SEQ ID NO:7-33, 47-49 wherein such a nucleic acid encodes a binding entity
polypeptide that can bind an epitope of the invention. Preferred nucleic acids
encode a polypeptide consisting essentially of any one of SEQ ID NO:21-33 or
any one of SEQ ID NO:8,10, 12,15, 17, 19, 22, 24,27, 29 or 32. Other nucleic
acids of the invention include nucleotide sequences SEQ ID NO:36-39.
The invention also provides antibodies made by available procedures that
can bind an epitope of the invention.
Antibody molecules belong to a family of plasma proteins called
immunoglobulins, whose basic building block, the immunoglobulin fold or
domain, is used in various forms in many molecules of the immune system and
other biological recognition systems. A standard antibody is a tetrameric
structure consisting of two identical immunoglobulin heavy chains and two
identical light chains and has a molecular weight of about 150,000 daltons.
The heavy and light chains of an antibody consist of different domains.
Each light chain has one variable domain (VL) and one constant domain (CL),

while each heavy chain has one variable domain (VH) and three or four constant
domains (CH). See, e.g., Alzari, P. N., Lascombe, M.-B. & Poljak, R. J. (1988)
Three-dimensional structure of antibodies. Annu. Rev. Immunol. 6, 555-580.
Each domain, consisting of about 110 amino acid residues, is folded into a
characteristic β-sandwich structure formed from two β-sheets packed against
each other, the immunoglobulin fold. The VH and VL domains each have three
complementarity determining regions (CDR1-3) that are loops, or turns,
connecting β-strands at one end of the domains. The variable regions of both the
light and heavy chains generally contribute to antigen specificity, although the
contribution of the individual chains to specificity is not always equal. Antibody
molecules have evolved to bind to a large number of molecules by using six
randomized loops (CDRs).
Depending on the amino acid sequences of the constant domain of their
heavy chains, immunoglobulins can be assigned to different classes. There are at
least five (5) major classes of immunoglobulins: IgA, IgD, IgE, IgG and IgM,
and several of these may be further divided into subclasses (isotypes), e.g. IgG-1,
IgG-2, IgG-3 and IgG-4; IgA-1 and IgA-2. The heavy chains constant domains
that correspond to the different classes of immunoglobulins are called alpha (a),
delta (δ), epsilon (ε), gamma (γ) and mu (µ), respectively. The light chains of
antibodies can be assigned to one of two clearly distinct types, called kappa (K)
and lambda (λ), based on the amino sequences of their constant domain. The
subunit structures and three-dimensional configurations of different classes of
immunoglobulins are well known.
The term "variable" in the context of variable domain of antibodies,
refers to the fact that certain portions of the variable domains differ extensively
in sequence among antibodies. The variable domains are for binding and
determine the specificity of each particular antibody for its particular antigen.
However, the variability is not evenly distributed through the variable domains
of antibodies. It is concentrated in three segments called complementarity
determining regions (CDRs) also known as hypervariable regions both in the
light chain and the heavy chain variable domains.

The more highly conserved portions of variable domains are called the
framework (FR). The variable domains of native heavy and light chains each
comprise four FR regions, largely a adopting a β-sheet configuration, connected
by three CDRs, which form loops connecting, and in some cases forming part of,
the P-sheet structure. The CDRs in each chain are held together in close
proximity by the FR regions and, with the CDRs from the other chain, contribute
to the formation of the antigen-binding site of antibodies. The constant domains
are not involved directly in binding an antibody to an antigen, but exhibit various
effector functions, such as participation of the antibody in antibody-dependent
cellular toxicity.
An antibody that is contemplated for use in the present invention thus can
be in any of a variety of forms, including a whole immunoglobulin, an antibody
fragment such as Fv, Fab, and similar fragments, a single chain antibody which
includes the variable domain complementarity determining regions (CDR), and
the like forms, all of which fall under the broad term "antibody", as used herein.
The present invention contemplates the use of any specificity of an antibody,
polyclonal or monoclonal, and is not limited to antibodies that recognize and
immunoreact with a specific antigen. In preferred embodiments, in the context
of both the therapeutic and screening methods described below, an antibody or
fragment thereof is used that is immunospecific for an antigen or epitope of the
invention. In some embodiments, the antibody is not the COU-1 antibody.
The term "antibody fragment" refers to a portion of a full-length
antibody, generally the antigen binding or variable region. Examples of antibody
fragments include Fab, Fab', F(ab') 2 and Fv fragments. Papain digestion of
antibodies produces two identical antigen binding fragments, called the Fab
fragment, each with a single antigen binding site, and a residual "Fc" fragment,
so-called for its ability to crystallize readily. Pepsin treatment yields an F(ab')2
fragment that has two antigen binding fragments that are capable of cross-linking
antigen, and a residual other fragment (which is termed pFc'). Additional
fragments can include diabodies, linear antibodies, single-chain antibody
molecules, and multispecific antibodies formed from antibody fragments. As

used herein, "functional fragment" with respect to antibodies, refers to Fv, F(ab)
and F(ab')2 fragments.
Antibody fragments contemplated by the invention are therefore not full-
length antibodies but do have similar or improved immunological properties
relative to an antibody such as the COU-1 antibody. Thus, fragments of the
COU-1 antibody and/or fragments of polypeptides having any one of SEQ ID
NO:7-35 antibody are contemplated by the invention. Such antibody fragments
may be as small as about 4 amino acids, 5 amino acids, 6 amino acids, 7 amino
acids, 9 amino acids, about 12 amino acids, about 15 amino acids, about 17
amino acids, about 18 amino acids, about 20 amino acids, about 25 amino acids,
about 30 amino acids or more.
In general, an antibody fragment of the invention can have any upper size
limit so long as it is has similar or immunological properties relative to antibody
that binds with specificity to an epitope formed by a combination of any one of
SEQ ID NO:3-6. Such a reference antibody can be the COU-1 antibody. For
example, binding entities and light chain antibody fragments can have less than
about 200 amino acids, less than about 175 amino acids, less than about 150
amino acids, or less than about 120 amino acids if the antibody fragment is
related to a light chain antibody subunit. Moreover, binding entities and heavy
chain antibody fragments can have less than about 425 amino acids, less than
about 400 amino acids, less than about 375 amino acids, less than about 350
amino acids, less than about 325 amino acids or less than about 300 amino acids
if the antibody fragment is related to a heavy chain antibody subunit.
Antibody fragments retain some ability to selectively bind with its
antigen, epitope or receptor. Some types of antibody fragments are defined as
follows:
(1) Fab is the fragment that contains a monovalent antigen-binding
fragment of an antibody molecule. A Fab fragment can be produced by digestion
of whole antibody with the enzyme papain to yield an intact light chain and a
portion of one heavy chain.
(2) Fab' is the fragment of an antibody molecule can be obtained by
treating whole antibody with pepsin, followed by reduction, to yield an intact

light chain and a portion of the heavy chain. Two Fab' fragments are obtained
per antibody molecule. Fab' fragments differ from Fab fragments by the addition
of a few residues at the carboxyl terminus of the heavy chain CHI domain
including one or more cysteines from the antibody hinge region.
(3) (Fab')2 is the fragment of an antibody that can be obtained by
treating whole antibody with the enzyme pepsin without subsequent reduction.
F(ab')2 is a dimer of two Fab' fragments held together by two disulfide bonds.
(4) Fv is the minimum antibody fragment that contains a complete
antigen recognition and binding site. This region consists of a dimer of one
heavy and one light chain variable domain in a right, non-covalent association
(VH -VL dimer). It is in this configuration that the three CDRs of each variable
domain interact to define an antigen binding site on the surface of the VH -VL
dimer. Collectively, the six CDRs confer antigen binding specificity to the
antibody. However, even a single variable domain (or half of an Fv comprising
only three CDRs specific for an antigen) has the ability to recognize and bind
antigen, although at a lower affinity than the entire binding site.
(5) Single chain antibody ("SCA"), defined as a genetically
engineered molecule containing the variable region of the light chain, the
variable region of the heavy chain, linked by a suitable polypeptide linker as a
genetically fused single chain molecule. Such single chain antibodies are also
referred to as "single-chain Fv" or "sFv" antibody fragments. Generally, the Fv
polypeptide further comprises a polypeptide linker between the VH and VL
domains that enables the sFv to form the desired structure for antigen binding.
For a review of sFv see Pluckthun in The Pharmacology of Monoclonal
Antibodies, vol. 113. Rosenburg and Moore eds. Springer-Verlag, N.Y., pp. 269-
315 (1994).
The term "diabodies" refers to a small antibody fragments with two
antigen-binding sites, which fragments comprise a heavy chain variable domain
(VH) connected to a light chain variable domain (VL) in the same polypeptide
chain (VH-VL). By using a linker that is too short to allow pairing between the
two domains on the same chain, the domains are forced to pair with the
complementary domains of another chain and create two antigen-binding sites.

Diabodies are described more fully in, for example, EP 404,097; WO 93/11161,
and Hollinger et al., Proc. Natl. Acad Sci. USA 90: 6444-6448 (1993).
Methods for the preparation of polyclonal antibodies are availableto
those skilled in the art. See, for example, Green, et al., Production of Polyclonal
Antisera, in: Immunochermcal Protocols (Manson, ed.), pages 1-5 (Humana
Press); Coligan, et al., Production of Polyclonal Antisera in Rabbits, Rats Mice
and Hamsters, in: Current Protocols in Immunology, section 2.4.1 (1992), which
are hereby incorporated by reference.
The preparation of monoclonal antibodies likewise is conventional. See,
for example, Kohler & Milstein, Nature, 256:495 (1975); Coligan, et al., sections
2.5.1-2.6.7; and Harlow, et al., in: Antibodies: A Laboratory Manual, page 726
(Cold Spring Harbor Pub. (1988)), which are hereby incorporated by reference.
Monoclonal antibodies can be isolated and purified from hybridoma cultures by
a variety of well-established techniques. Such isolation techniques include
affinity chromatography with Protein-A Sepharose, size-exclusion
chromatography, and ion-exchange chromatography. See, e.g., Coligan, et al.,
sections 2.7.1-2.7.12 and sections 2.9.1-2.9.3; Barnes, et al., Purification of
Immunoglobulin G (IgG), in: Methods in Molecular Biology. Vol. 10, pages 79-
104 (Humana Press (1992).
Methods of in vitro and in vivo manipulation of monoclonal antibodies
are well known to those skilled in the art. For example, the monoclonal
antibodies to be used in accordance with the present invention may be made by
the hybridoma method first described by Kohler and Milstein, Nature 256, 495
(1975), or may be made by recombinant methods, e.g., as described in U.S. Pat.
No. 4,816,567. The monoclonal antibodies for use with the present invention
may also be isolated from phage antibody libraries using the techniques
described in Clackson et al. Nature 352:624-628 (1991), as well as in Marks et
al., J. Mol Biol. 222: 581-597 (1991). Another method involves humanizing a
monoclonal antibody by recombinant means to generate antibodies containing
human specific and recognizable sequences. See, for review, Holmes, et al., J.
Immunol., 158:2192-2201 (1997) and Vaswani, et al., Annals Allergy, Asthma &
Immunol., 81:105-115 (1998).

The term "monoclonal antibody" as used herein refers to an antibody
obtained from a population of substantially homogeneous antibodies, i.e., the
individual antibodies comprising the population are identical except for passible
naturally occurring mutations that may be present in minor amounts. Monoclonal
antibodies are highly specific, being directed against a single antigenic site.
Furthermore, in contrast to conventional polyclonal antibody preparations that
typically include different antibodies directed against different determinants
(epitopes), each monoclonal antibody is directed against a single determinant on
the antigen. In additional to their specificity, the monoclonal antibodies are
advantageous in that they are synthesized by the hybridoma culture,
uncontaminated by other immunoglobulins. The modifier "monoclonal" indicates
the character of the antibody indicates the character of the antibody as being
obtained from a substantially homogeneous population of antibodies, and is not
to be construed as requiring production of the antibody by any particular method.
The monoclonal antibodies herein specifically include "chimeric"
antibodies (immunoglobulins) in which a portion of the heavy and/or light chain
is identical with or homologous to corresponding sequences in antibodies
derived from a particular species or belonging to a particular antibody class or
subclass, while the remainder of the chain(s) is identical with or homologous to
corresponding sequences in antibodies derived from another species or belonging
to another antibody class or subclass, as well as fragments of such antibodies, so
long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567);
Morrison et al. Proc. Natl. Acad Sci. 81, 6851-6855 (1984).
Methods of making antibody fragments are also known in the art (see for
example, Harlow and Lane, Antibodies: A Laboratory Manual. Cold Spring
Harbor Laboratory, New York, (1988), incorporated herein by reference).
Antibody fragments of the present invention can be prepared by proteolytic
hydrolysis of the antibody or by expression in E. coli of DNA encoding the
fragment. Antibody fragments can be obtained by pepsin or papain digestion of
whole antibodies conventional methods. For example, antibody fragments can
be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S
fragment denoted F(ab')2. This fragment can be further cleaved using a thiol

reducing agent, and optionally a blocking group for the sulfhydryl groups
resulting from cleavage of disulfide linkages, to produce 3.5S Fab' monovalent
fragments. Alternatively, an enzymatic cleavage using pepsin produces two
monovalent Fab' fragments and an Fc fragment directly. These methods are
described, for example, in US Patents No. 4,036,945 and No. 4,331,647, and
references contained therein. These patents are hereby incorporated in their
entireties by reference.
Other methods of cleaving antibodies, such as separation of heavy chains
to form monovalent light-heavy chain fragments, further cleavage of fragments,
or other enzymatic, chemical, or genetic techniques may also be used, so long as
the fragments bind to the antigen that is recognized by the intact antibody. For
example, Fv fragments comprise an association of VH and VL chains. This
association may be noncovalent or the variable chains can be linked by an
intermolecular disulfide bond or cross-linked by chemicals such as
glutaraldehyde. Preferably, the Fv fragments comprise VH and VL chains
connected by a peptide linker. These single-chain antigen binding proteins (sFv)
are prepared by constructing a structural gene comprising DNA sequences
encoding the VH and VL domains connected by an oligonucleotide. The
structural gene is inserted into an expression vector, which is subsequently
introduced into a host cell such as E. coli. The recombinant host cells synthesize
a single polypeptide chain with a linker peptide bridging the two V domains.
Methods for producing sFvs are described, for example, by Whitlow, et al.,
Methods: a Companion to Methods in Enzvmologv. Vol. 2, page 97 (1991);
Bird, et al., Science 242:423-426 (1988); Ladner, et al, US Patent No. 4,946,778;
and Pack, et al., Bio/Technology 11:1271-77 (1993).
Another form of an antibody fragment is a peptide coding for a single
complementarity-deteimining region (CDR). CDR peptides ("minimal
recognition units") are often involved in antigen recognition and binding. CDR
peptides can be obtained by cloning or constructing genes encoding the CDR of
an antibody of interest. Such genes are prepared, for example, by using the
polymerase chain reaction to synthesize the variable region from RNA of

antibody-producing cells. See, for example, Larrick, et al., Methods: a
Companion to Methods in Enzvmology, Vol. 2, page 106 (1991).
The invention contemplates human and humanized forms of non-human
(e.g. murine) antibodies. Such humanized antibodies are chimeric
immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab,
Fab', F(ab')2 or other antigen-binding subsequences of antibodies) that contain
minimal sequence derived from non-human immunoglobulin. For the most part,
humanized antibodies are human immunoglobulins (recipient antibody) in which
residues from a complementary determining region (CDR) of the recipient are
replaced by residues from a CDR of a nonhuman species (donor antibody) such
as mouse, rat or rabbit having the desired specificity, affinity and capacity.
In some instances, Fv framework residues of the human immunoglobulin
are replaced by corresponding non-human residues. Furthermore, humanized
antibodies may comprise residues that are found neither in the recipient antibody
nor in the imported CDR or framework sequences. These modifications are made
to further refine and optimize antibody performance. In general, humanized
antibodies will comprise substantially all of at least one, and typically two,
variable domains, in which all or substantially all of the CDR regions correspond
to those of a non-human immunoglobulin and all or substantially all of the FR
regions are those of a human immunoglobulin consensus sequence. The
humanized antibody optimally also will comprise at least a portion of an
immunoglobulin constant region (Fc), typically that of a human
immunoglobulin. For further details, see: Jones et al., Nature 321, 522-525
(1986); Reichmann et al., Nature 332,323-329 (1988); Presta, Curr. Op. Struct.
Biol. 2, 593-596 (1992); Holmes, et al., J. Immunol., 158:2192-2201 (1997) and
Vaswani, et al., Annals Allergy, Asthma & Immunol, 81:105-115 (1998).
While standardized procedures are available to generate antibodies, the
size of antibodies, the multi-stranded structure of antibodies and the complexity
of six binding loops present in antibodies constitute a hurdle to the improvement
and the manufacture of large quantities of antibodies. Hence, the invention
further contemplates using binding entities, which comprise polypeptides that
can recognize and bind to the epitope of the invention.

The invention is therefore further directed to antibodies and other binding
entities that can bind the cancer-associated epitope of the invention. In some
embodiments, the antibodies and binding entities have SEQ ID NO.7-33: "The
sequences for SEQ ID NO.7-33 are provided below.



Nucleic acids encoding peptides SEQ ID NO:7-33 were isolated from cells that
secrete the COU-1 antibody. While not all of the polypeptides encoded by the nucleic
acids isolated in this screen could bind the cancer-associated epitope, peptides SEQ ID
NO:7-33 were shown to play a role in binding by phage display and other experiments.
Moreover, several differences were found in similar regions of different antibody
fragment clones. For example, variable light chain CDR1 fragments that were isolated
had RASQSVSSSYLA (SEQ ID NO:15) as well as KSSQSLLYSSNNKNYLA (SEQ
ID NO:27). Similarly, variable light chain CDR2 fragments isolated had DASNRAT
(SEQ ID NO: 17), GASSRAT (SEQ ID NO:22) or WASTRES (SEQ ID NO:29).
Moreover, variable light chain CDR3 fragments isolated had QQYGNSPPYT (SEQ ID
NO:24) or QQYYSTPPM (SEQ ID NO:32). Hence, not all clones were identical.
A number of proteins can serve as protein scaffolds to which binding domains
(e.g. any of the SEQ ID NO:7-33,47-49 peptides or variants thereof) can be attached.
The binding domains bind or interact with the cancer-associated epitope of the
invention while the protein scaffold merely holds and stabilizes the binding domains so
that they can bind. A number of protein scaffolds can be used. For example, phage
capsid proteins can be used. Review in Clackson & Wells, Trends Biotechnol. 12:173-
184 (1994). Indeed, such phage capsid proteins were used as described herein to screen
for the SEQ ID NO:7-33 peptides (see Examples). Phage capsid proteins have also
been used as scaffolds for displaying random peptide sequences, including bovine
pancreatic trypsin inhibitor (Roberts et al., PNAS 89:2429-2433 (1992)), human growth
hormone (Lowman et al., Biochemistry 30:10832-10838 (1991)), Venturini et al.,
Protein Peptide Letters 1:70-75 (1994)), and the IgG binding domain of Streptococcus
(O'Neil et al., Techniques in Protein Chemistry V (Crabb, L,. ed.) pp. 517-524,
Academic Press, San Diego (1994)). These scaffolds have displayed a single
randomized loop or region that can be modified to include the binding domains
provided herein (e.g. SEQ ID NO:7-33, 47-49).

Researchers have also used the small 74 amino acid α-amylase inhibitor
Tendamistat as a presentation scaffold on the filamentous phage M13. McConnell, S.
J., & Hoess, R. H., J.Mol. Biol. 250:460-470 (1995). Tendamistat is a fcaheet protein
from Streptomyces tendae. It has a number of features that make it an attractive scaffold
for binding peptides, including its small size, stability, and the availability of high
resolution NMR and X-ray structural data. The overall topology of Tendamistat is
similar to that of an immunoglobulin domain, with two β-sheets connected by a series
of loops. In contrast to immunoglobulin domains, the β-sheets of Tendamistat are held
together with two rather than one disulfide bond, accounting for the considerable
stability of the protein. By analogy with the CDR loops found in immunoglobulins, the
loops of Tendamistat may serve a similar function and can be easily randomized by in
vitro mutagenesis. Tendamistat, however, is derived from Streptomyces tendae and
may be antigenic in humans. Its small size, however, may reduce or inhibit its
antigenicity.
Fibronectin type III domain has also been used as a protein scaffold to which
binding entities can be attached. Sequences, vectors and cloning procedures for using
such a fibronectin type III domain as a protein scaffold for binding entities (e.g. CDR
peptides) are provided, for example, in U.S. Patent Application Publication
20020019517. Fibronectin is a large protein that plays an essential role in the
formation of extracellular matrix and cell-cell interactions. Fibronectin consists of
many repeats of three types (I, II and III) of small domains. Baron, M., Norman, D. G.
& Campbell, I. D. (1991) Protein modules Trends Biochem. Sci. 16,13-17.
Fibronectin type H1 is part of a large subfamily (Fn3 family or s-type Ig family) of the
immunoglobulin superfamily. The Fn3 family includes cell adhesion molecules, cell
surface hormone and cytokine receptors, chaperonins, and carbohydrate-binding
domains. For reviews, see Bork, P. & Doolittle, R. F. (1992) Proposed acquisition of
an animal protein domain by bacteria. Proc. Natl. Acad. Sci. USA 89, 8990-8994;
Jones, E. Y. (1993) The immunoglobulin superfamily Curr. Opinion Struct. Biol. 3,
846-852; Bork, P., Horn, L. & Sander, C. (1994) The immunoglobulin fold. Structural
classification, sequence patterns and common core. J. Mol. Biol. 242, 309-320;
Campbell, LD.& Spitzfaden, C. (1994) Building proteins with fibronectin type H1
modules Structure 2,233-337; Harpez, Y. & Chothia, C. (1994) Many of the

immunoglobulin superfamily domains in cell adhesion molecules and surface receptors
belong to a new structural set which is close to that containing variable domains J. Mol.
Biol. 238, 528-539.
In the immune system, specific antibodies are selected and amplified from a
large library (affinity maturation). The combinatorial techniques employed in immune
cells can be mimicked by mutagenesis and generation of combinatorial libraries of
binding entities. Binding entities, antibody fragments and antibodies therefore can be
generated through display-type technologies, including, without limitation, phage
display, retroviral display, ribosomal display, and other techniques, using techniques
well known in the art and the resulting molecules can be subjected to additional
maturation, such as affinity maturation, as such techniques are well known in the art.
Wright and Harris, supra., Hanes and Plucthau PNAS USA 94:4937-4942 (1997)
(ribosomal display), Parmley and Smith Gene 73:305-318 (1988) (phage display), Scott
T1BS 17:241-245 (1992), Cwirla et al. PNAS USA 87:6378-6382 (1990), Russel et al.
Nucl. Acids Research 21:1081-1085 (1993), Hoganboom et al. Immunol. Reviews
130:43-68 (1992), Chiswell and McCafferty T1BTECH 10:80-84 (1992), and U.S. Pat.
No. 5,733,743.
The invention therefore also provides methods of mutating antibodies to
optimize their affinity, selectivity, binding strength and/or other desirable properties. A
mutant antibody refers to an amino acid sequence variant of an antibody. In general,
one or more of the amino acid residues in the mutant antibody is different from what is
present in the reference antibody. Such mutant antibodies necessarily have less than
100% sequence identity or similarity with the reference amino acid sequence. In
general, mutant antibodies have at least 75% amino acid sequence identity or similarity
with the amino acid sequence of either the heavy or light chain variable domain of the
reference antibody. Preferably, mutant antibodies have at least 80%, more preferably at
least 85%, even more preferably at least 90%, and most preferably at least 95% amino
acid sequence identity or similarity with the amino acid sequence of either the heavy or
light chain variable domain of the reference antibody. One method of mutating
antibodies involves affinity maturation using phage display.
For example, affinity maturation using phage display can be utilized as one
method for generating mutant antibodies. Affinity maturation using phage display

refers to a process described in Lowman et al., Biochemistry 30(45): 10832-10838
(1991), see also Hawkins et al., J. Mol Biol. 254: 889-896 (1992). While not strictly
limited to the following description, this process can be described briefly as involving
mutation of several antibody hypervariable regions in a number of different sites with
the goal of generating all possible amino acid substitutions at each site. The antibody
mutants thus generated are displayed in a monovalent fashion from filamentous phage
particles as fusion proteins. Fusions are generally made to the gene IE product of M13.
The phage expressing the various mutants can be cycled through several rounds of
selection for the trait of interest, e.g. binding affinity or selectivity. The mutants of
interest are isolated and sequenced. Such methods are described in more detail in U.S.
Patent 5,750,373, U.S. Patent 6,290,957 and Cunningham, B. C. et al., EMBO J.
13(11), 2508-2515 (1994).
In one embodiment, the invention provides methods of manipulating
antibody polypeptides or antibody-encoding nucleic acids to generate antibodies
and antibody fragments with improved binding properties that recognize the
same epitope as COU-1 antibodies.
Such methods of mutating portions of a COU-1 antibody involve fusing a
nucleic acid encoding a polypeptide having any one of SEQ ED NO:7-35 or any one of
SEQ ID NO:8,10, 12, 15, 17,19, 22, 24, 27, 29, 32, 47, 48 or 49 to a nucleic acid
encoding a phage coat protein to generate a recombinant nucleic acid encoding a fusion
protein, mutating the recombinant nucleic acid encoding the fusion protein to generate a
mutant nucleic acid encoding a mutant fusion protein, expressing the mutant fusion
protein on the surface of a phage and selecting phage that bind to an epitope of the
invention.
In one embodiment, the method involves fusing a nucleic acid encoding a
polypeptide having any combination of SEQ ID NO:7-35 or any combination of SEQ
ID NO:8,10,12,15,17,19,22,24,27, 29,32, 47,48 or 49 to a nucleic acid encoding
a phage coat protein to generate a recombinant nucleic acid encoding a fusion protein,
mutating the recombinant nucleic acid encoding the fusion protein to generate a mutant
nucleic acid encoding a mutant fusion protein, expressing the mutant fusion protein on
the surface of a phage and selecting phage that bind to an epitope of the invention.

In another embodiment, the method involves fusing a nucleic acid encoding a
polypeptide having each one of SEQ ID NO:26, 15, 21, 22, 23, 24 and 25 to a nucleic
acid encoding a phage coat protein to generate a recombinant nucleic acid encoding a
fusion protein, mutating the recombinant nucleic acid encoding the fusion protein to
generate a mutant nucleic acid encoding a mutant fusion protein, expressing the mutant
fusion protein on the surface of a phage and selecting phage that bind to an epitope of
the invention.
In another embodiment, the method involves fusing a nucleic acid encoding
a polypeptide having each one of SEQ ID NO:26, 27, 28, 29, 30, 31, 32 and 33 to a
nucleic acid encoding a phage coat protein to generate a recombinant nucleic acid
encoding a fusion protein, mutating the recombinant nucleic acid encoding the fusion
protein to generate a mutant nucleic acid encoding a mutant fusion protein, expressing
the mutant fusion protein on the surface of a phage and selecting phage that bind to an
epitope of the invention.
In another embodiment, the method involves fusing a nucleic acid
encoding a polypeptide having SEQ ID NO.34 or SEQ ID NO:35 to a nucleic acid
encoding a phage coat protein to generate a recombinant nucleic acid encoding a
fusion protein, mutating the recombinant nucleic acid encoding the fusion protein
to generate a mutant nucleic acid encoding a mutant fusion protein, expressing the
mutant fusion protein on the surface of a phage and selecting phage that bind to
an epitope of the invention. SEQ ID NO:34 and 35 encode useful variable light
chains that may bind to epitopes of the invention. SEQ ID NO:34 is provided
below.


The method can also involve fusing a nucleic acid comprising a variable heavy
or light chain relating to COU-1 (e.g. any one of SEQ ID NO:36-39) to a nucleic acid
encoding a phage coat protein to generate a recombinant nucleic acid encoding a fusion
protein, mutating the recombinant nucleic acid encoding the fusion protein to generate a
mutant nucleic acid encoding a mutant fusion protein, expressing the mutant fusion
protein on the surface of a phage and selecting phage that bind to an epitope of the
invention.
Hence, the invention is directed to a nucleic acid encoding a variable heavy
chain relating to COU-1, for example, SEQ ID NO:36 provided below.


In another embodiment the invention is directed to a nucleic acid
encoding a variable light chain relating to COU-1, for example, SEQ ID NO:38
(also called L8) provided below.

In another embodiment the invention is directed to a nucleic acid encoding
a variable light chain related to COU-1, for example, SEQ ID NO:39 (also called
T5).

Such methods can further include constructing a replicable expression
vector containing a nucleic acid encoding a polypeptide of the invention, for
example, a polypeptide comprising any one of SEQ ID NO:7-35, or a nucleic
acid comprising any one of SEQ ID NO:36-39. The nucleic acid can also encode
a fusion protein comprising a polypeptide of the invention (e.g. any of SEQ ID
NO:7-35) and at least a portion of a natural or wild-type phage coat protein. The
expression vector can also have a transcription regulatory element operably
linked to the nucleic acids encoding the fusion protein. The vector is mutated at

one or more selected positions within the nucleic acid encoding the antibody
polypeptide to form a family or "library" of plasmids containing related nucleic
acids, each encoding a slightly different antibody polypeptide. Suitable hest
cells are transformed with the family of plasmids. The transformed host cells are
infected with a helper phage having a gene encoding the phage coat protein and
the transformed, infected host cells are cultured under conditions suitable for
forming recombinant phagemide particles. Each recombinant phagemid displays
approximately one copy of the fusion protein on the surface of the phagemid
particle. To screen the phagemids, phagemid particles are contacted with an
epitope or antigen of the invention. Phagemid particles that bind are separated
from those that do not bind the epitope or antigen. Preferably, further rounds of
selection are performed by separately cloning phagemids with acceptable binding
properties and re-testing their binding affinity one or more times. The plasmids
from phagemid particles that appropriately bind the epitope or antigen can also
be isolated, cloned and even mutated again to further select for the antibody
properties desired, e.g. with good binding affinity.
The method is applicable to polypeptide complexes that are composed of
more than one subunit polypeptides. In this case, a nucleic acid encoding each
subunit of interest is separately fused to a phage coat protein and separately
analyzed for its binding properties.
Any cloning procedure used by one of skill in the art can be employed to
make the expression vectors used in such affinity maturation/phage display
procedures. For example, one of skill in the art can readily employ known
cloning procedures to fuse a nucleic acid encoding, an antibody hypervariable
region to a nucleic acid encoding a phage coat protein. See, e.g., Sambrook et
al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory,
N.Y., 1989; Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold
Spring Harbor Laboratory, N.Y., 2001.
The invention is therefore directed to a method for selecting antibodies
and/or antibody fragments or polypeptides with desirable properties. Such
desirable properties can include increased binding affinity or selectivity for the
epitopes of the invention

The antibodies and antibody fragments of the invention are isolated
antibodies and antibody fragments. An isolated antibody is one that has been
identified and separated and/or recovered from a component of the environment
in which it was produced. Contaminant components of its production
environment are materials that would interfere with diagnostic or therapeutic
uses for the antibody, and may include enzymes, hormones, and other
proteinaceous or nonproteinaceous solutes. The term "isolated antibody" also
includes antibodies within recombinant cells because at least one component of
the antibody's natural environment will not be present. Ordinarily, however,
isolated antibody will be prepared by at least one purification step
If desired, the antibodies of the invention can be purified by any available
procedure. For example, the antibodies can be affinity purified by binding an
antibody preparation to a solid support to which the antigen used to raise the
antibodies is bound. After washing off contaminants, the antibody can be eluted
by known procedures. Those of skill in the art will know of various techniques
common in the immunology arts for purification and/or concentration of
polyclonal antibodies, as well as monoclonal antibodies (see for example,
Coligan, et al., Unit 9, Current Protocols in Immunology. Wiley Interscience,
1991, incorporated by reference).
In preferred embodiments, the antibody will be purified as measurable by
at least three different methods: 1) to greater than 95% by weight of antibody as
determined by the Lowry method, and most preferably more than 99% by
weight; 2) to a degree sufficient to obtain at least 15 residues of N-terminal or
internal amino acid sequence by use of a spinning cup sequentator; or 3) to
homogeneity by SDS-PAGE under reducing or non-reducing conditions using
Coomasie blue or, preferably, silver stain.
Antigen, Binding Entity and Antibody Variants and Derivatives
The invention also provides variants and derivative of the antigenic
epitopes, binding entities and antibody segments identified herein. For example,
any derivative or variant of a SEQ ID NO:3,4, 5 or 6 antigenic epitope is
contemplated as being within the scope of the invention, particularly when the

variant or derivative retains, or has improved, specificity as a vaccine for
preventing or treating adenocarcinomas or is an improved marker for detecting
adenocarcinomas. Similarly, any derivative or variant of a SEQ ID NO:735
antibody polypeptide is contemplated by the invention, particularly when the
variant or derivative antibody polypeptide has improved specificity or binding
affinity for an antigenic epitope of the invention, for example, an antigenic
epitope having SEQ ID NO:3, 4, 5 or 6.
Derivative and variant antigenic epitopes and antibody segments of the
invention are derived from the reference antigenic epitopes and antibody
segments by deletion or addition of one or more amino acids to the N-terminal
and/or C-terminal end of the reference antigenic epitopes and antibody segments;
deletion or addition of one or more amino acids at one or more sites within the
reference antigenic epitopes and antibody segments; or substitution of one or
more amino acids at one or more sites within the reference antigenic epitopes
and antibody segments. Thus, the antigenic epitopes and antibody segments of
the invention may be altered in various ways including amino acid substitutions,
deletions, truncations, and insertions.
Such variant and derivative antigenic epitopes and antibody segments
may result, for example, from human manipulation. For example, the affinity
maturation techniques using phage display described above may be used to
generate variants and derivatives of both the antigenic epitopes and antibody
segments of the invention. Other methods for mutating or altering the sequence
of polypeptide are generally available in the art. For example, amino acid
sequence variants of the antigenic epitopes and antibody segments can be
prepared by mutations in the DNA encoding these antigenic epitopes and
antibody segments. Methods for mutagenesis and nucleotide sequence
alterations are also available in the art. See, for example, Kunkel, Proc. Natl.
Acad. Sci. USA, 82,488 (1985); Kunkel et al., Methods in Enzymol., 154,367
(1987); U. S. Patent No. 4,873,192; Walker and Gaastra, eds., Techniques in
Molecular Biology, MacMillan Publishing Company, New York (1983) and the
references cited therein. Guidance as to appropriate amino acid substitutions that
do not adversely affect the structural integrity and/or biological activity of the

peptide of interest may be found in the model of Dayhoff et al., Atlas of Protein
Sequence and Structure, Natl. Biomed. Res. Found., Washington, CD. (1978),
herein incorporated by reference.
The derivatives and variants of the antigenic epitopes and antibody
segments of the invention have identity with at least about 90%, 91%, 92%, 93%
or 94% of the amino acid positions of any one of SEQ ID NO.3-35 and generally
have similar or improved immunological properties relative to those of the
antigenic epitopes and antibody segments having any one of SEQ ID NO:3-35.
In a desirable embodiment, the antigenic epitopes and antibody segment
derivatives and variants have identity with at least about 95% or 96% of the
amino acid positions of any one of SEQ ID NO:3-35 and generally have
immunological properties that are similar or better than the antigenic epitopes
and antibody segments having SEQ ID NO: 3-3 5. In a more desirable
embodiment, the antigenic epitopes and antibody segments derivatives and
variants have identity with at least about 97% or 98% of the amino acid positions
of any one of SEQ ID NO:3-35 and generally have similar or improved
immunological properties relative to those of the antigenic epitopes and antibody
segments having SEQ ID NO.3-35.
By "similar or improved immunological properties" is meant that a
derivative or variant of a SEQ ID NO:3,4, 5 or 6 antigenic epitope retains, or has
improved, activity as a vaccine for preventing or treating adenocarcinomas or is
an improved marker for detecting adenocarcinomas. Similarly, derivatives or
variants of a SEQ ID NO:7-35 antibody polypeptide have "similar or improved
immunological properties" when they have improved specificity or binding
affinity for an antigenic epitope of the invention, for example, an antigenic
epitope having SEQ ID NO:3,4, 5 or 6.
Amino acid.residues of the antigenic epitopes, binding entities and
antibody segments and of the derivatives and variants thereof can be genetically
encoded L-amino acids, naturally occurring non-genetically encoded L-amino
acids, synthetic L-amino acids or D-enantiomers of any of the above. The amino
acid notations used herein for the twenty genetically encoded L-amino acids and
common non-encoded amino acids are conventional and are as shown in Table 1.




Variants of the present antigenic epitopes and antibody segments that are
encompassed within the scope of the invention can have one or more amino
acids substituted with an amino acid of similar chemical and/or physical
properties, so long as the backbone portions of these variant peptides have
similar or improved immunological properties relative to those of antigenic

epitopes and antibody segments having any one of SEQ ID NO.3-35. Derivative
antigenic epitopes and antibody segments can have additional peptide or
chemical moieties as well as one or more amino acids substituted with amino
acids having different chemical and/or physical properties, so long as these
derivative antigenic epitopes and antibody segments have similar or improved
immunological properties relative to those of antigenic epitopes and antibody
segments having any one of SEQ ID NO:3-35.
Amino acids that are substitutable for each other to form a variant
antigenic epitopes and antibody segments of the invention generally reside
within similar classes or subclasses. As known to one of skill in the art, amino
acids can be placed into three main classes: hydrophilic amino acids,
hydrophobic amino acids and cysteine-like amino acids, depending primarily on
the characteristics of the amino acid side chain. These main classes may be
further divided into subclasses. Hydrophilic amino acids include amino acids
having acidic, basic or polar side chains and hydrophobic amino acids include
amino acids having aromatic or apolar side chains. Apolar amino acids may be
further subdivided to include, among others, aliphatic amino acids. The
definitions of the classes of amino acids as used herein are as follows:
"Hydrophobic Amino Acid" refers to an amino acid having a side chain
that is uncharged at physiological pH and that is repelled by aqueous solution.
Examples of genetically encoded hydrophobic amino acids include IIe, Leu and
Val. Examples of non-genetically encoded hydrophobic amino acids include t-
BuA.
"Aromatic Amino Acid" refers to a hydrophobic amino acid having a
side chain containing at least one ring having a conjugated π-electron system
(aromatic group). The aromatic group may be further substituted with substituent
groups such as alkyl, alkenyl, alkynyl, hydroxyl, sulfonyl, nitro and amino
groups, as well as others. Examples of genetically encoded aromatic amino acids
include phenylalanine, tyrosine and tryptophan. Commonly encountered non-
genetically encoded aromatic amino acids include phenylglycine, 2-
naphthylalanine, P-2-tbienyialanine, l,2,3,4-tetrahydroisoquinohne-3-carboxylic

acid, 4-chlorophenylalanine, 2-fluorophenylalanine, 3-fluorophenylalanine and
4-fluorophenylalanine.
"Apolar Amino Acid" refers to a hydrophobic amino acid having a side
chain that is generally uncharged at physiological pH and that is not polar.
Examples of genetically encoded apolar amino acids include glycine, proline and
methionine. Examples of non-encoded apolar amino acids include Cha.
"Aliphatic Amino Acid" refers to an apolar amino acid having a saturated
or unsaturated straight chain, branched or cyclic hydrocarbon side chain.
Examples of genetically encoded aliphatic amino acids include Ala, Leu, Val and
IIe. Examples of non-encoded aliphatic amino acids include Nle.
"Hydrophilic Amino Acid" refers to an amino acid having a side chain
that is attracted by aqueous solution. Examples of genetically encoded
hydrophilic amino acids include Ser and Lys. Examples of non-encoded
hydrophilic amino acids include Cit and hCys.
"Acidic Amino Acid" refers to a hydrophilic amino acid having a side
chain pK value of less than 7. Acidic amino acids typically have negatively
charged side chains at physiological pH due to loss of a hydrogen ion. Examples
of genetically encoded acidic amino acids include aspartic acid (aspartate) and
glutamic acid (glutamate).
"Basic Amino Acid" refers to a hydrophilic amino acid having a side
chain pK value of greater than 7. Basic amino acids typically have positively
charged side chains at physiological pH due to association with hydronium ion.
Examples of genetically encoded basic amino acids include arginine, lysine and
histidine. Examples of non-genetically encoded basic amino acids include the
non-cyclic amino acids ornithine, 2,3-diaminopropionic acid, 2,4-diaminobutyric
acid and homoarginine.
"Polar Amino Acid" refers to a hydrophilic amino acid having a side
chain that is uncharged at physiological pH, but which has a bond in which the
pair of electrons shared in common by two atoms is held more closely by one of
the atoms. Examples of genetically encoded polar amino acids include
asparagine and glutamine. Examples of non-genetically encoded polar amino
acids include citrulline, N-acetyl lysine and methionine sulfoxide.

"Cysteine-Like Amino Acid" refers to an amino acid having a side chain
capable of forming a covalent linkage with a side chain of another amino acid
residue, such as a disulfide linkage. Typically, cysteine-like amino acids
generally have a side chain containing at least one thiol (SH) group. Examples
of genetically encoded cysteine-like amino acids include cysteine. Examples of
non-genetically encoded cysteine-like amino acids include homocysteine and
penicillamine.
As will be appreciated by those having skill in the art, the above
classifications are not absolute. Several amino acids exhibit more than one
characteristic property, and can therefore be included in more than one category.
For example, tyrosine has both an aromatic ring and a polar hydroxyl group.
Thus, tyrosine has dual properties and can be included in both the aromatic and
polar categories. Similarly, in addition to being able to form disulfide linkages,
cysteine also has apolar character. Thus, while not strictly classified as a
hydrophobic or apolar amino acid, in many instances cysteine can be used to
confer hydrophobicity to a polypeptide.
Certain commonly encountered amino acids that are not genetically
encoded and that can be present, or substituted for an amino acid, in the variant
polypeptides of the invention include, but are not limited to, β-alanine (b-Ala)
and other omega-amino acids such as 3-aminopropionic acid (Dap), 2,3-
diaminopropionic acid (Dpr), 4-aminobutyric acid and so forth; α-
aminoisobutyric acid (Aib); e-aminohexanoic acid (Aha); δ-aminovaleric acid
(Ava); N-methylglycine (MeGly); ornithine (Orn); citrulline (Cit); t-butylalanine
(t-BuA); t-butylglycine (t-BuG); N-methylisoleucine (MeIle); phenylglycine
(Phg); cyclohexylalanine (Cha); norleucine (Nle); 2-naphthylalanine (2-Nal); 4-
chlorophenylalanine (Phe(4-Cl)); 2-fluorophenylalanine (Phe(2-F)); 3-
fluorophenylalanine (Phe(3-F)); 4-fluorophenylalanine (Phe(4-F)); penicillamine
(Pen); l,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (Tic); P-2-thienylalanine
(Thi); methionine sulfoxide (MSO); homoarginine (hArg); N-acetyl lysine
(AcLys); 2,3-diaminobutyric acid (Dab); 2,3-diaminobutyric acid (Dbu); p-
aminophenylalanine (Phe(pNH2)); N-methyl valine (MeVal); homocysteine

(hCys) and homoserine (hSer). These amino acids also fall into the categories
defined above.
The classifications of the above-described genetically encoded and fton-
encoded amino acids are summarized in Table 2, below. It is to be understood
that Table 2 is for illustrative purposes only and does not purport to be an
exhaustive list of amino acid residues that may comprise the variant and
derivative antigenic epitopes and antibody segments described herein. Other
amino acid residues that are useful for making the variant and derivative
polypeptides described herein can be found, e.g., in Fasman, 1989, CRC
Practical Handbook of Biochemistry and Molecular Biology, CRC Press, Inc.,
and the references cited therein. Amino acids not specifically mentioned herein
can be conveniently classified into the above-described categories on the basis of
known behavior and/or their characteristic chemical and/or physical properties as
compared with amino acids specifically identified.

Antigenic epitopes and antibody segments of the invention can have any amino
acid substituted by any similarly classified amino acid to create a variant
antigenic epitope or a variant antibody segment, so long as the variant has similar

or improved immunological properties relative to those of an antigenic epitope or
antibody segment having any one of SEQ ID NO:3-35.
The invention is therefore also directed to binding entities and antibodies with
binding domains related to the variable light or heavy chain CDR fragments isolated
according to the invention. For example, the variable light chain CDR1 fragments can
be aligned as follows:

Related variable light chain CDR1 fragments and binding entities are of the following
formula (SEQ ID NO:47).




Detecting the Cancer-Associated Epitope
The invention also provides methods of detecting the cancer-associated
epitopes of the invention in biological test samples. Any immunoassay or in
vivo imaging procedure known to one of skill in the art can be used to detect the
cancer-associated epitopes of the invention in a biological test sample. For
example, the cancer-associated epitopes of the invention can be detected by
immunochemical, immunohistological, ELISA, radioimmunoassay, nuclear
magnetic resonance, magnetic resonance imaging, surface plasmon resonance
and related procedures.
Such methods can include the steps of contacting a test sample with an
antibody or binding entity capable of binding to a cancer-associated epitope of

the invention, and determining whether the antibody or binding entity binds to a
component of the sample. These methods can also include the steps of obtaining
a biological sample (e.g., cells, blood, plasma, tissue, etc.) from a patient
suspected of having cancer, contacting the sample with a labeled antibody or a
labeled binding entity that is specific for the cancer-associated epitope of the
invention, and detecting the epitope using standard immunoassay and/or
diagnostic imaging techniques. Binding of the antibody or binding entity to the
biological sample indicates that the sample contains the epitope.
In another embodiment, the cancer-associated epitope can be used to
detect antibodies in the blood, serum or tissues of a mammal with cancer. Such
antibodies can arise naturally within the mammal when the cancer-associated
epitope becomes exposed during malignant transformation.
Accordingly, the invention provides a method of detecting cancer in a
mammal by contacting a test sample with a cancer-associated epitope of the
invention and detecting whether an antibody from the test sample has bound to
the cancer-associated epitope.
Antibodies or binding entities that are reactive with cancer-associated
epitope of the invention and/or polypeptides comprising a cancer-associated
epitope of the invention can be labeled or coupled to a diagnostic imaging agent
for convenient detection of cancer.
The words "label" and diagnostic imaging agent refer to a detectable
compound or composition that is conjugated directly or indirectly to an antibody
or antigen or epitope. The label may itself be detectable (e.g., radioisotope labels
or fluorescent labels) or, in the case of an enzymatic label, may catalyze
chemical alteration of a substrate compound or composition that is detectable.
Such labels or diagnostic imaging agents are useful for imaging of cells
and tissues that express the cancer-associated epitope. Such labels can also be
used with a cancer-associated epitope of the invention in standard
immunoassays. Labels and diagnostic imaging agents include, but are not
limited to barium sulfate, iocetamic acid, iopanoic acid, ipodate calcium,
diatrizoate sodium, diatrizoate meglumine, metrizamide, tyropanoate sodium and
radiodiagnostics including positron emitters such as fluorine-18 and carbon-11,

gamma emitters such as iodine-123, technitium-99m, iodine-131 and indium-
111, nuclides for nuclear magnetic resonance such as fluorine and gadolinium.
Paramagnetic isotopes for purposes of in vivo diagnosis can be used
according to the methods of this invention. There are numerous examples of
elements that are useful in magnetic resonance imaging. For discussions on in
vivo nuclear magnetic resonance imaging, see, for example, Schaefer et al.,
(1989) JACC 14, 472-480; Shreve et al., (1986) Magn. Reson. Med. 3, 336-340;
Wolf, G. L., (1984) Physiol. Chem. Phys. Med. NMR 16,93-95; Wesbey et al.,
(1984) Physiol. Chem. Phys. Med. NMR 16,145-155; Runge et al., (1984)
Invest. Radiol. 19,408-415. Examples of suitable fluorescent labels include a
fluorescein label, an isothiocyalate label, a rhodamine label, a phycoerythrin
label, a phycocyanin label, an allophycocyanin label, an ophthaldehyde label, a
fluorescamine label, etc. Examples of chemiluminescent labels include a luminal
label, an isoluminal label, an aromatic acridinium ester label, an imidazole label,
an acridinium salt label, an oxalate ester label, a luciferin label, a luciferase label,
an aequorin label, etc. Those of ordinary skill in the art will know of other
suitable labels that may be employed in accordance with the present invention.
The attachment of these labels to antibodies or fragments thereof can be
accomplished using standard techniques commonly known to those of ordinary
skill in the art. Typical techniques are described by Kennedy et al., (1976) Clin.
Chim. Acta 70,1-31; and Schurs et al., (1977) Clin. Chim. Acta 81, 1-40.
Coupling techniques mentioned in the latter are the glutaraldehyde method, the
periodate method, the dimaleimide method, the m-maleimidobenzyl-N-hydroxy-
succinimide ester method. All of these methods are incorporated by reference
herein.
A solid phase or a solid support can be used in conjunction with the
antibodies, binding entities, antigens or epitopes of the invention. Such a solid
phase or solid support refers to a non-aqueous matrix to which the antibody,
binding entity, antigen or epitope can adhere. Examples of solid phases and
supports encompassed herein include those formed partially or entirely of glass
(e.g. controlled pore glass), polysaccharides (e.g., agarose), polyacrylamides,
polystyrene, polyvinyl alcohol and silicones. In certain embodiments, depending

on the context, the solid phase or support can comprise the well of an assay
plate; in others it is a purification column (e.g. an affinity chromatography
column). This term also includes a discontinuous solid phase of discrete
particles, such as those described in U.S. Pat. No. 4,275,149.
Therapy
According to the invention, the antigenic epitopes of the invention,
antibodies or binding entities directed against such epitopes and protease
inhibitors that inhibit formation of the epitopes of the invention can be used for
cancer prevention and/or therapy. The antigenic epitopes of the invention can be
used as vaccines to stimulate an immunological response in a mammal that is
directed against cells having the cancer-associated epitope. Antibodies or
binding entities directed against the antigenic epitopes of the invention can
combat or prevent adenocarcinomas. Moreover, the invention contemplates
administering protease inhibitors that inhibit cleavage of cytokeratin 8 and/or
cytokeratin 18 to prevent or treat adenocarcinomas.
In one embodiment, the invention provides a method of preventing or
treating adenocarcinoma in a mammal by administering an antigenic epitope
comprising any one of SEQ ID NO:3-6 to the mammal in an amount sufficient
to stimulate an immunological response against the antigenic epitope. Two or
more polypeptides comprising SEQ ID NO:3-6 can be combined in a therapeutic
composition and administered in several doses over a period of time that
optimizes the immunological response of the mammal. Such an immunological
response can be detected and monitored by observing whether antibodies
directed against the epitopes of the invention are present in the bloodstream of
the mammal.
Antibodies and binding entities generated as provided herein that react
selectively with the cancer-associated epitope of the invention also be used for
cancer therapy. Accordingly, the invention provides methods of preventing or
treating adenocarcinoma in a mammal by administering to the mammal a
therapeutically effective amount of an antibody or binding entity that can bind an
antigenic epitope comprising any one of SEQ ID NO:3-6.

Such antibodies or binding entities can be used alone or coupled to, or
combined with, therapeutically useful agents. Antibodies and/or binding entities
can be administered to mammals suffering from any cancer that displays the
cancer-associated epitope of the invention. Such administration can provide
both therapeutic treatment, and prophylactic or preventative measures. For
example, the therapeutic methods of the invention can be used to deter the spread
of a cancer and lead to its remission.
As used herein, "therapeutically useful agents" include any therapeutic
molecule that can beneficially be targeted to a cell expressing the cancer epitope
disclosed herein, including antineoplastic agents, radioiodinated compounds,
toxins, chemotherapeutic agents, cytostatic or cytolytic drugs.
Such therapeutically useful agents include, for example, adrimycin,
aminoglutethimide, aminopterin, azathioprine, bleomycin sulfate, bulsulfan,
carboplatin, carminomycin, carmustine, chlorambucil, cisplatin,
cyclophosphamide, cyclosporine, cytarabidine, cytosine arabinoside, cytoxin
dacarbazine, dactinomycin, daunomycin, daunorubicin, doxorubicin,
esperamicins (see U.S. Pat. No. 4,675,187), etoposide, fluorouracil, ifosfamide,
interferon-a, lomustine, melphalan, mercaptopurine, methotrexate, mitomycin C,
mitotane, mitoxantrone, procarbazine HC1, taxol, taxotere (docetaxel),
teniposide, thioguanine, thiotepa, vinblastine sulfate, vincristine sulfate and
vinorelbine. Additional agents include those disclosed in Chapter 52,
Antineoplastic Agents (Paul Calabresi and Bruce A. Chabner), and the
introduction thereto, pp.1202-1263, of Goodman and Gilman's "The
Pharmacological Basis of Therapeutics", Eighth Edition, 1990, McGraw-Hill,
Inc. (Health Professions Division). Toxins can be proteins such as, for example,
pokeweed anti-viral protein, cholera toxin, pertussis toxin, ricin, gelonin, abrin,
diphtheria exotoxin, or Pseudomonas exotoxin. Toxin moieties can also be high
energy-emitting radionuclides such as cobalt-60,1-131,1-125, Y-90 and Re-186,
and enzymatically active toxins of bacterial, fungal, plant or animal origin, or
fragments thereof.
According to the invention, such chemotherapeutic agents can be used to
reduce the growth or spread of cancer cells and tumors that express the tumor

associated epitope of the invention. Animals that can be treated by the
chemotherapeutic agents of the invention include humans, non-human primates,
cows, horses, pigs, sheep, goats, dogs, cats, rodents and the like. In all
embodiments human tumor antigens and human subjects are preferred.
The invention also contemplates using species-dependent antibodies for
use in the present therapeutic methods. Such a species-dependent antibody has
constant regions that are substantially non-immunologically reactive with the
chosen species. Such species-dependent antibody is particularly useful for
therapy because it gives rise to substantially no immunological reactions. The
species-dependent antibody can be of any of the various types of antibodies as
defined above, but preferably is mammalian, and more preferably is a humanized
or human antibody.
Therapeutically useful agents can be formulated into a composition with
the antibodies of the invention and need not be directly attached to the antibodies
of the invention. However, in some embodiments, therapeutically useful agents
are attached to the antibodies of the invention using methods available to one of
skill in the art, for example, standard coupling procedures.
The invention further provides methods of preventing or treating
adenocarcinoma in a mammal by administering to the mammal a therapeutically
effective amount of a protease inhibitor that prevents formation of an antigenic
epitope comprising any one of SEQ ID NO:3-6. According to the invention, the
sites of protease cleavage at amino acids 22 and 40 on cytokeratin K8, and at
amino acid 50 on cytokeratin K18, all contained consensus sequence
Xaa1SR↓Xaa4 (SEQ ID NO.40), where Xaa1 is serine, phenylalanine or valine
and Xaa4 is serine or valine. The structure of these cleavage sites indicates that
the enzyme responsible for these cleavages is a trypsin-like protease. Trypsin
inhibitors are available to one of skill in the art. See, e.g., U.S. Patent 6,239,106;
U.S. Patent 6,159,938; U.S. Patent 5,962,266. Such trypsin inhibitors include
inhibitors available for serine proteases such as kallikrein, chymotrypsins A and
B, trypsin, elastase, subtilisin, coagulants and procoagulants, particularly those in
active form, including coagulation factors such as factors VIIa, IXa, Xa, XIa, and

XIIa, plasmin, thrombin; proteinase-3, enterokinase, acrosin, cathepsin,
urokinase, and tissue plasminogen activator.
According to the invention, any inhibitor capable of inhibiting a protease
that can cleave Xaa1SR↓Xaa4 (SEQ ID NO:40) may be used to prevent or treat
adenocarcinomas. For example, peptides with homology to Xaa1SR↓Xaa4 (SEQ
ID NO:40) but that cannot be cleaved may be used as inhibitors in the present
therapeutic methods. Other examples of inhibitors that may be used include, for
example, soybean trypsin inhibitor (or STI, from Sigma Chemical Co.), alpha-2-
macroglobulin, alpha-1-antitrypsin, aprotinin, pancreatic secretory trypsin
inhibitor (PSTI) corn and pumpkin trypsin inhibitors (Wen, et al., Protein Exp. &
Purif. 4:215 (1993); Pedersen, et al., J. Mol. Biol. 236:385 (1994)), and so forth.
One candidate for a useful inhibitor of human origin is found in circulating
isoforms of the human amyloid P-protein precursor (APPI), also known as
protease nexin-2. APPI contains a Kunitz serine protease inhibitor domain
known as KPI (Kunitz Protease Inhibitor). See Ponte et al., Nature, 331:525
(1988); Tanzi et al., Nature 331:528 (1988); Johnstone et al., Biochem. Biophys.
Res. Commun. 163:1248 (2989); Oltersdorf et al., Nature 341.144 (1989).
Human KPI shares about 45% amino acid sequence identity with aprotinin. The
isolated KPI domain has been prepared by recombinant expression in a variety of
systems, and has been shown to be an active serine protease inhibitor. See, for
example, Sinha, et al., J. Biol. Chem. 265:8983 (1990).
Progression of adenocarcinoma cancer and/or the therapeutic efficacy of
chemotherapy may be measured using procedures available in the art. For
example, the efficacy of a particular chemotherapeutic agent can be determined
by measuring the amount of cancer-associated epitope released from
adenocarcinoma cells undergoing cell death. The concentration of antigenic
epitope (e.g. a polypeptide having any one of SEQ ID NO:3-6, or a combination
of such polypeptides) released from cells can be compared to standards from
healthy, untreated patients to assess whether heightened levels of the present
epitopes are present in a patient. Fluid samples can be collected at discrete
intervals during treatment and compared to a standard. It is contemplated that
changes in the level of a cancer-associated antigenic epitope of the invention,

will be indicative of the efficacy of treatment (that is, the rate of cancer cell
death). It is contemplated that the release of cancer-associated antigenic epitopes
can be measured in many test samples, including blood, plasma, serum, feces,
urine, sputum, vaginal secretions, seminal fluids, semen and any tissue sample.
Where the assay is used to monitor tissue viability or progression of
adenocarcinoma, the step of detecting the presence and abundance of the
antigenic epitope in samples of interest is repeated at intervals and these values
then are compared, the changes in the detected concentrations reflecting changes
in the status of the tissue. For example, an increase in the level of
adenocarcinoma-associated epitope may correlate with progression of the
adenocarcinoma. Where the assay is used to evaluate the efficacy of a therapy,
the monitoring steps occur following administration of the therapeutic agent or
procedure (e.g., following administration of a chemotherapeutic agent or
following radiation treatment). Similarly, a decrease in the level of
adenocarcinoma cancer-associated epitopes of the invention may correlate a
regression of the adenocarcinoma.
Thus, adenocarcinomas may be identified by the presence of cancer-
associated antigenic epitopes as provided herein. Once identified, the
adenocarcinoma may be treated using antibodies and protease inhibitors that
reduce cleavage of cytokeratins 8 and 18. Moreover, the methods provided
herein can be used to monitor the progression of the disease and/or treatment of
the disease.
Compositions
The invention is further directed to compositions containing the present
antibodies, binding entities, antigenic epitopes or trypsin-like protease inhibitors.
Such compositions are useful for detecting the antigenic epitopes of the
invention and for therapeutic methods involving prevention and treatment of
cancers associated with the presence of the antigenic epitopes of the invention.
The antibodies, binding entities, antigenic epitopes and protease
inhibitors of the invention can be formulated as pharmaceutical compositions
and administered to a mammalian host, such as a human patient in a variety of

forms adapted to the chosen route of administration. Routes for administration
include, for example, intravenous, intra-arterial, subcutaneous, intramuscular,
intraperitoneal and other routes selected by one of skill in the art.
Solutions of the antibodies, binding entities, antigenic epitopes and
protease inhibitors of the invention can be prepared in water or saline, and
optionally mixed with a nontoxic surfactant. Formulations for intravenous or
intra-arterial administration may include sterile aqueous solutions that may also
contain buffers, liposomes, diluents and other suitable additives.
The pharmaceutical dosage forms suitable for injection or infusion can
include sterile aqueous solutions or dispersions comprising the active ingredient
that are adapted for administration by encapsulation in liposomes. In all cases,
the ultimate dosage form must be sterile, fluid and stable under the conditions of
manufacture and storage.
Sterile injectable solutions are prepared by incorporating the antibodies,
binding entities, antigenic epitopes and protease inhibitors in the required
amount in the appropriate solvent with various of the other ingredients
enumerated above, as required, followed by filter sterilization.
Useful dosages of the antibodies, binding entities, antigenic epitopes and
protease inhibitors can be determined by observing their in vitro activity, and in
vivo activity in animal models. Methods for the extrapolation of effective
dosages in mice, and other animals, to humans are known to the art; for example,
see U.S. Patent No. 4,938,949.
In general, a suitable dose of the antibodies, binding entities, antigenic
epitopes and protease inhibitors will be in the range of from about 1 to about
2000 µg/kg, for example, from about 2.0 to about 1500 µg/kg of body weight per
treatment. Preferred doses are in the range of about 3 to about 500 µg per
kilogram body weight of the recipient per treatment, more preferably in the range
of about 10 to about 300 μg/kg/treatment, most preferably in the range of about
20 to about 200 μg/kg/treatment.
The antibodies, binding entities, antigenic epitopes and protease
inhibitors are conveniently administered in unit dosage form; for example,

containing 5 to 1000 μg, conveniently 10 to 750 μg, most conveniently, 50 to
500 µg of active ingredient per unit dosage form.
Ideally, the antibodies, binding entities, antigenic epitopes and protease
inhibitors should be administered to achieve peak plasma concentrations of from
about 0.1 to about 10 nM, preferably, about 0.2 to 10 nM, most preferably, about
0.5 to about 5 nM. This may be achieved, for example, by the intravenous
injection of a 0.05 to 25% solution of the antibodies, optionally in saline.
Desirable blood levels may be maintained by continuous infusion to provide
about 0.01-10.0 μg/kg/hr or by intermittent infusions containing about 0.4-50
μg/kg of the antibodies.
The desired dose may conveniently be presented in a single dose or as
divided doses administered at appropriate intervals, for example, as two, three,
four or more sub-doses per day. The sub-dose itself may be further divided, for
example, into a number of discrete loosely spaced administrations; such as
multiple intravenous doses. For example, it is desirable to administer the present
compositions intravenously over an extended period, either by continuous
infusion or in separate doses.
Kits
The invention further provides kits for detection of the antigenic epitope
of the invention and for treatment of adenocarcinomas.
A kit for detection of the antigenic epitope of the invention may contain a
container containing an antibody or binding entity capable of binding to an
antigenic epitope of the invention. Such an antibody or binding entity may be
labeled for easy detection. Individual kits may be adapted for performing one or
more of the methods of the invention.
Optionally, the subject kit may further comprise at least one other reagent
required for performing the method that the kit is adapted to perform. Examples
of such additional reagents include: a label, a standard, a control, a buffer, a
solution for diluting the test sample, or a reagent that facilitates detection of the
label. The reagents included in the kits of the invention may be supplied in
premeasured units so as to provide for greater precision and accuracy. Typically,

kits reagents and other components are placed and contained in separate vessels.
A reaction vessel, test tube, microwell tray, microtiter dish or other container can
also be included in the kit. Different labels can be used on different reagents so
that each reagent can be distinguished from another.
A further aspect of the invention relates to a kit for treatment of
adenocarcinomas comprising a pharmaceutical composition of the invention and
an instructional material. Such a kit may contain a container having an antigenic
epitope, an antibody, a binding entity or an inhibitor of the invention. The
antigenic epitope may act as a vaccine for preventing formation of metastatic
adenocarcinoma. The antibody or binding entity is directed against an antigenic
epitope of the invention and can be administered to treat or prevent the spread of
adenocarcinomas. An inhibitor of cytokeratin 8 or 18 cleavage can also inhibit
the formation and spread of adenocarcinomas. Any one of these antigenic
epitopes, antibodies, binding entities or inhibitors may be contained within an
appropriate container in the kit. Alternatively, a combination of antigenic
epitopes, antibodies, binding entities or inhibitors may be contained within an
appropriate container in the kit.
As used herein, an "instructional material" includes a publication, a
recording, a diagram, or any other medium of expression that is used to
communicate the usefulness of the pharmaceutical composition of the invention
for inhibiting cleavage of cytokeratin 8 or 18 or for stimulating the immune
system to recognize the epitopes of the invention in a mammal or patient. The
instructional material may also, for example, describe an appropriate dose of the
pharmaceutical composition of the invention. The instructional material of the
kit of the invention may, for example, be affixed to a container that contains a
pharmaceutical composition of the invention or be shipped together with a
container that contains the pharmaceutical composition. Alternatively, the
instructional material may be shipped separately from the container with the
intention that the instructional material and the pharmaceutical composition be
used cooperatively by the recipient.
The invention also includes a kit comprising a pharmaceutical
composition of the invention and a delivery device for delivering the

composition to a mammal, for example, a human patient who may have an
adenocarcinoma. By way of example, the delivery device may be a squeezable
spray bottle, a metered-dose spray bottle, an aerosol spray device, an atomizer, a
dry powder delivery device, a self-propelling solvent/powder-dispensing device,
a syringe, a needle, a tampon, or a dosage measuring container.
The invention will be further described by reference to the following
detailed examples, which are given for illustration of the invention, and are not
intended to be limiting thereof.
EXAMPLE 1: Cancer-Associated Epitope Characterization
Isolation of COU-1 Monoclonal Antibodies
The IgM HMab, COU-1, is secreted by the hybridoma cell line, B9165,
derived by fusing the human lymphoblastoid cell line WI-L2-729-HF2 with
lymphocytes obtained from mesenteric lymph nodes from a colon cancer patient
(35). Mesenteric lymph nodes draining the tumor region in patients with
colorectal cancer were minced under sterile conditions. Debris was removed by
filtration through cotton wool and the lymphocytes were purified by
centrifugation on Ficoll-Isopaque (Boehringer-Mannheim, Mannheim, Federal
Republic of Germany).
The lymphocytes were fused with the human fusion cell line W1-L2-729-
HF2 (referred to as HF2) (from Tecniclone Int., Santa Ana, Calif., USA)
according to Kohler, Immunological Methods Vol. II, Academic Press, 1981, pp.
285-298. The ratio between the HF2 and lymphocytes (107) was 1:2.
After washing the HF2 and the lymphocytes together in RPMI-1640
medium and collecting the cells by centrifugation, the cell pellet was
resuspended in 0.5 ml of 50% polyethylene glycol (PEG) 6000 over a period of 1
minute with constant shaking. Before dilution of the PEG with RPMI-1640, the
cells were incubated for another 2 minutes. The resulting fusion product was
washed and resuspended in solution medium [RPMI-1640,10% FCS (fetal calf
serum) supplemented with HAT (2 x 10-4 M hypoxanthine, 4 x 10-7 M
aminopterin, 3.2 x 10-6 M thymidine)]. The cells were plated in 96-well

microtiter plates using 200 µl containing 2 x 105 cells per well. The cells were
maintained in selective medium for two weeks. Further culturing was carried out
in RPMI-1640 with 10% FCS supplemented with hypoxanthine and thymidine.
Growing hybrids appeared 10 days to 4 weeks after fusion. Cloning was
performed by limiting dilution without feeder cells.
Supernatants from wells with growing clones were analyzed for
immunoglobulin production by ELISA using microtiter plates coated with rabbit
anti-human Ig (H and L chain) (Dakopatts, Copenhagen, Denmark) diluted
1:10,000 in 0.1M bicarbonate, pH 9.6. Coated wells were washed with PBS-
Tween (phosphate buffered saline—0.05% Tween 20) and incubated for 2 hours
at room temperature with supernatants diluted 1:10 in PBS-Tween. Development
was carried out with alkaline phosphatase (AP)-coupled antibody specific for
IgM, IgA or IgG (Dakopatts, Copenhagen) diluted 1:3000 in PBS-Tween. After
incubation for 1 hour at room temperature, the substrate p-nitrophenylphosphate
(PNPP), 1 mg/ml 10% diethanolamine, 1 mM MgC12, pH 9.6, was added.
Optical density was measured at 405 run after 1 hour of incubation at 37°C.
Standard curves for quantification were constructed with dilution of IgM
(Cappel) or IgG (Kabi AB, Stockholm, Sweden). Hybrids producing
immunoglobulin (Ig) assayed by ELISA were propagated by transfer to 24-well
macroplates (Nunc A/S, Denmark). The hybridoma cell line B9165 (ECACC
87040201) selected by the methods secreted the COU-1 antibodies described
below and was shown by ELISA to produce between 1 and 5 µg of IgM per ml
when allowed to grow for two weeks without change of media.
The hybridoma cell line B9165 was deposited with European Collection
of Cell Cultures (ECACC), CAMR, Salisbury, Wiltshire, SP4 OJG, UK, Deposit
no. ECACC 87040201.
COU-1 hybridoma supernatants were further analyzed by
immunocytochemical analysis for reaction with tumor cells or by
immunohistochemical analysis for reactions with tumor tissues as described
below.

Immunocytochemical Analysis of COU-1 Antibodies
Immunocytochemical analysis was performed on cell smears prepared
from different human tumor cell lines and from peripheral human blood
leukocytes. Cells were fixed on slides by treatment with formol-acetone (9.5%
formaldehyde, 43% acetone in 86 mM phosphate buffer, pH 7.2).
Approximately 50 µl of COU-1 supernatant (from the hybridoma B9165;
ECACC 87040201) was placed on the smear of fixed cells and incubated
overnight at 4°C in a humidified chamber before rinsing and incubation for 1
hour at room temperature with horseradish peroxidase (HRP)-labeled rabbit anti-
human IgM (Dakopatts) diluted to 1:80 in PBS-Tween. Finally, peroxidase
substrate (0.01% H2O2 and diaminobenzidine at 0.6 µg/ml in PBS) was added.
The smears were lightly counterstained with hematoxylin and mounted. Table 3
shows the results obtained by analysis of COU-1 on smears of various cells.

A selective reactivity with colon and mammary adenocarcinomas was apparent.
Live COLO 201 cells (colonic adenocarcinoma cells) were incubated
with the COU-1 antibody at 4°C, followed by the enzyme-labeled anti-Ig
antibody. The cells were then smeared on slides, fixed with glutaraldehyde

(0.17% in PBS) and incubated with substrate. COLO 201 cells stained with
COU-1 while control cells did not (data not shown).
Immunohistochemical analysis.
A preliminary immunohistochemical analysis was performed on frozen
tissue sections fixed in acetone. Endogenous IgM was blocked by incubation
with Fab' fragments of anti-µ-chain antibody (purchased from Dakopatts,
Copenhagen, Denmark) before incubation with the COU-1 antibody (0.5 μg/ml).
The anti-mu-chain antibody Fab' fragment was prepared according to B. Nielsen
et al., Hybridoma 6 (1), 1987, pp. 103-109). While clear-cut specificity for
cancerous tissues was observed using the COU-1 antibody, some non-specific
binding was observed in certain tissue types (for example, mammary tubules).
An improved fixation procedure was used that substantially eliminated
non-specific cross-reactivity with certain tissue types, including mammary
ductuli and tubules. Tissue specimens were obtained from colorectal cancer
patients undergoing surgical resection. Normal colon tissue was taken from the
resectate approximately 15 cm away from the site of the tumor. Tissues were
fixed in 96% alcohol for 6 h at 4°C. Afterwards, tissues were paraffin embedded
and cut into 5 µm sections. Sections were deparaffinized in xylol, rehydrated
through graded alcohol and washed in PBS-Tween. Sections were incubated for
2 h at room temperature in a humidified chamber with 100 µl of murine
monoclonal antibody, human monoclonal IgM antibody or normal polyclonal
human IgM, all at 0.5-10 μg/ml. The slides were washed and incubated with AP-
Iabeled rabbit anti-human IgM (Dako, Glostrup, Denmark), horse-radish
peroxidase (HRP) labeled rabbit anti-human IgM (Dako) or HRP-labeled rabbit
anti-mouse IgG (Dako) diluted in PBS with 10% (w/v) bovine serum albumin for
1 h at room temperature. After washing, the HRP was visualized by development
with chromogenic substrate (0.6 mg diaminobenzidine per ml PBS with 0.01%
H2O2) and AP with 0.2 mg naphthol-AS-Mx phosphate (Sigma), 1 mg Fast Red
TR Salt (Sigma), 20ug dimethylformamide per ml 0.1M Tris/HCl, 1M
levamisole, pH 8.2. The sections were counterstained with Mayer's

haematoxylin, dehydrated in xylene and mounted in Aquamount (Gurr, Poole,
England).
Bound antibody was visualized as described above for the
immunocytochemical analysis. Only the tumor cells in sections of colon
adenocarcinomas were stained COU-1. No staining was observed in tonsillar
tissue. Tables 4A and 4B summarize the reactivity of the COU-1 antibody with a
variety of tissues, where the reactivity of malignant tissues is provided in Table
4A and the lack of reactivity of non-malignant tissues is provided in Table 4B.



Normal colon epithelium showed binding of all analyzed human IgM,
monoclonal antibodies, myeloma IgM as well as normal polyclonal human IgM.
This general binding of IgM to normal colon epithelium was thus judged to be
non-specific.
EXAMPLE 2: Cancer-Associated Epitope Mapping
Materials and Methods
Antibodies
The IgM HMab, COU-1, is secreted by the hybridoma cell line, B9165,
derived by fusing the human lymphoblastoid cell line WI-L2-729-HF2 with
lymphocytes obtained from mesenteric lymph nodes from a colon cancer patient,
as described above. The hybridoma cell line B9165 was deposited with European

Collection of Cell Cultures 'ECACC), CAMR, Salisbury, Wiltshire, SP4 OJG,
UK, Deposit no. ECACC. 040201. More information about ECACC can be
obtained on the website at ecacc.org.
The human-human hybridoma cell line was grown in protein-free
medium: RPMI1640 medium (Gibco, Grand Island, NY) supplemented with
SSR3 serum replacement (Medicult, Copenhagen, Denmark). HMab COU-1 was
purified from cell culture supernatant by affinity chromatography on Sepharose-
coupled murine anti-human µ-chain monoclonal antibody (Mab)(HB57, ATCC,
Rockville, MD). The antibody was eluted with 0.1 M diethylamine, pH 10.5,
followed by fractionation by FPLC. IgM purified from normal human serum
(Cappel, Cochranville, PA) was used as a control. Murine Mabs, M20 directed
against normal K8 and CY-90 directed against normal K18, were obtained from
Sigma Chemical Co. (St. Louis, MO).
ELISA
ELISA wells (Costar, Cambridge, MA) were coated overnight at 4°C
with fractions from cytokeratin purification procedures or with different
recombinant K8/K18 complexes (5 μg/ml) in PBS, pH 7.4. The wells were
washed twice with PBS, blocked with 3% BSA in PBS for 1 h at 37°C, and
incubated with HMab COU-1 antibody for 2 h at 37°C. Plates were washed 10X
with PBS-0.05% Tween 20 and bound antibody was detected with alkaline
phosphatase (AP)-labeled goat anti-human kappa-chain (Sigma) diluted 1000
fold in PBS. Bound antibody was visualized with para-nitrophenylphosphate
(Sigma)(l mg/ml 1 mM MgCl2,10% (w/v) diethanolamine, pH 9.6) and read at
405 nm.
Cell culture
The human breast adenocarcinoma cell line MCF7 (ATCC) was
maintained in Eagle's MEM (Gibco), supplemented with 10% FCS, non-
essential amino acids, lmM sodium pyruvate, lmM HEPES buffer, 100 U
penicillin/ml, 100 mg streptomycin/ml and 2mM L-glutamine. The human colon
adenocarcinoma cell line Colon 137 (kindly provided by Dr. Ebbesen, Aarhus

University, Denmark) was maintained in RPMI1640 (Gibco), supplemented
with FCS, penicillin, streptomycin and L-glutamine as above.
Purification of cytokeratin from normal and malignant tissue
Cytokeratin were prepared from fresh, surgically-removed, colon cancer
tissue or normal colon epithelia. Tissue samples (1-5 g) were minced with a
shears and homogenized in 10-30 ml of Tris-buffered saline (TBS)(10 mM Tris,
0.14 M NaCl, 15 mM NaN3,pH 7.6) containing 1% (v/v) Emulphogene (Sigma)
using a blade rotor (Euro Turrax T20b basic, DCA Labortechnik, Staufen,
Germany) for 3 x 5 sec at 27.000 rpm on ice. Enzyme inhibitors: 5 mM
iodoacetamide, 10 mM PMSF, 5 mM EDTA (all Sigma), 5 mM Cyclocapron
(KABI, Stockholm, Sweden), and 10 U Aprotinin (Bayer, Leverkusen, Germany)
per ml were included in the buffers during the homogenization, sonication and
ion exchange chromatography. The suspension was pelleted by centrifugation at
10.000 g for 10 min at 4°C, washed twice in TBS containing 1% Emulphogene
and resuspended in buffer A (10 mM Tris pH 8.6 containing 0.1% SDS (w/v)
and 0.05% Emulphogene). The suspension was sonicated for 3 x 15 sec on ice
and centrifuged at 12.000 g for 10 min at 4°C. The supernatant was applied to an
anion exchange column (20 ml Q-Sepharose Fast Flow column, QFF,
(Pharmacia Upjohn, Uppsala, Sweden)) pre-equilibrated with buffer A. After
washing the column with 10 column volumes of buffer A, bound proteins were
eluted with a linear gradient to 1M NaCl in buffer A. Fractions of 1ml were
collected and further analyzed by SDS-PAGE/Western blotting and ELISA. For
ELISA, 10µl of each fraction was added to wells containing 10 µl of SM2 beads
(BioRad) in 100 µl TBS, followed by incubation with COU-1 as described
above. The beads bind the detergent and thus allow for the direct coating of the
proteins in the fractions.
SDS-PAGE and Western Blot analysis
Electrophoresis was performed in a discontinuous buffer system on 8 cm
4-20% or 10% (w/v) polyacrylamide gels for analysis and on 15 cm 14%
polyacrylamide gels for N-terminal sequencing (36). Samples were mixed with 2

x sample buffer (4% SDS, 0.2% bromophenol blue, 20% glycerol in 100 mM
Tris buffered saline), boiled for 5 rain and resolved under denaturing and
reducing (100 mM DTT) conditions. Protein bands were visualized with
Coomassie Brilliant Blue. Separated proteins were also electroblotted onto
polyvinylidene difluoride membranes (PVDF, Immobilon P, Millipore, Bedford,
MA), at 100 Volts for 1 h in ice, using transfer buffer (10% (v/v) ethanol, 25 mM
Tris, 200 mM glycine). Prior to transfer, the membrane was pre-soaked in
ethanol for 2 min and the membranes and the gel were incubated in transfer
buffer for 10 min. Following transfer, the membrane was blocked for 2 h in
Western blot buffer (50 mM Tris, 350 mM NaCl, 15 mM NaN3, 0.1% Tween-
20) washed 3x with Western blot buffer and incubated with COU-1 antibody (5
μg/ml), mouse anti-K8 antibody (diluted 1/2000), mouse anti K-18 antibody
(diluted 1/2000) or goat-anti-GST antibody (diluted 1/1000, Pharmacia Upjohn)
overnight at room temperature. The membrane was washed in Western blot
buffer and incubated with AP-conjugated rabbit-anti-goat IgG antibody (diluted
1/1000, Sigma), or AP-conjugated rabbit-anti-human IgM antibody (diluted
1/500, DAKO, Glostrup, Denmark) for 2 h at room temperature. Following 3
washes in PBS, the membrane was fixed with 0.2% glutaraldehyde in PBS for 15
min at room temperature and finally washed in PBS. Bound AP conjugate was
visualized by NBT/BCIP (Bio-Rad, Hercules, CA). MCF7 or Colon 137 cells,
resuspended in SDS sample buffer and sonicated, were used as antigen control.
A low range protein marker (Bio-Rad) was used to indicate the molecular weight
of the fragments.
Amino Acid Sequencing and Amino Acid Analysis
Previously described procedures (37) were employed for amino acid
sequencing and amino acid analysis. For N-terminal sequencing, purified
cytokeratin was run on SDS-PAGE and electroblotted onto PVDF membranes
prior to detection with Coomassie. The different bands were excised from the
blot and sequenced in an Applied Biosystems 470 A protein sequencer (ABI,
Forster City, CA). Sequences similar to cytokeratins were searched for in
GenBank/EBI/DDBI/PDB databases using the BLAST program.

Expression and purification of recombinant K8 and K18 proteins
E. coli DH5a harboring plasmids encoding a panel of K8 and K18
proteins were analyzed. The panel consisted of the full length and several N-
terminal and C-terminal deleted fragments of K8 and K18, cloned as GST fusion
proteins into a modified pGEX-2T vector (38). The E. coli cultures were grown
in Super Broth medium, supplemented with 20 mM MgCl2 and 50 mg
carbenicillin/ml at 37°C until OD600 reached 0.6. Protein expression was then
induced with 1 mM IPTG (Sigma) and 4 µM cAMP and the culture allowed to
grow for an additional 3 h at 30°C. The bacteria were pelleted at 4.000 g for 15
min at 4°C. For SDS-PAGE, the pellet was resuspended in sample buffer and
sonicated 5 x 10 sec before electrophoresis. For purification of the recombinant
K8 or K18 proteins, the pellet of a 400 ml culture grown and processed as
described above was resuspended in 50 ml lysis buffer (50 mM Tris-HCl, 100
mM NaCl, 1 mM EDTA, 5 mM P-mercaptoethanol, pH 8.0) containing I mg/ml
lysozyme and incubated for 30 min at 4°C. The suspension was sonicated 3 x 20
sec and pelleted at 20.000 g at 4°C. The pellet was washed twice in a high salt
buffer (50 mM Tris-HCl, 2 M NaCl, 10 mM EDTA, 5 mM p-mercaptoethanol,
1% NP40, pH 8.0) and once in lysis buffer. The pellet was subsequently washed
twice in lysis buffer containing 2 M urea and stored at 4°C in lysis buffer
containing 8 M urea.
Heterotypic association assay
Panels of different C- or N-terminal-deleted or intact K8 and K18
proteins were separated by SDS-PAGE and transferred to a PVDF membrane, as
described above. After blocking, the membrane was incubated for 16 h at 4°C
with 100 µg/ml of purified K8 or K18 protein in PBS, 2% BSA and 4 M urea; if
K8 proteins were transferred to the membrane, the membrane was subsequently
incubated with a purified K18 protein, and vice versa (38). The membrane was
then washed with PBS, incubated with COU-1 (5 μg/ml) in Western blot buffer
containing 10% FCS for 2 h at room temperature and binding detected as
described above.

Surface plasmon resonance
The kinetics of HMab COU-1 binding to heterotypic complexes of
recombinant intact K8 or K18 (and fragments thereof) was determined by surface
plasmon resonance measurements using the BIAcore instrument (Pharmacia).
The sensor chip was activated for immobilization with N-hydroxysuccinimide
and N-ethyl-NT-(3-diethyl aminopropyl) carbodiimide. The heterotypic
cytokeratin complexes were coupled to the surface by injection of 50 µl of a 50
μg/ml sample. Excess activated esters were quenched with 30 µl 1 M
ethanolamine, pH 8.5. Typically, 3000 resonance units were immobilized.
Binding of COU-1 to immobilized heterotypic cytokeratin complexes was
studied by injecting COU-1 in a range of concentrations (0.5-80 μg/ml) at a flow
rate of 5 ul/min. The association was monitored as the increase in resonance
units per unit time. Dissociation measurements were obtained following the end
of the association phase with a flow rate of 20 ul/min. The binding surface was
regenerated with 10 mM HC1,1M NaCl, pH 2.0, and remained active for 10
measurements. The association and dissociation rate constants, kon and koff,
were determined from a series of measurements, as described previously (39).
Association and dissociation constants were deduced from kinetic data using the
Bioevaluation program version 3.1 (Pharmacia).
Confocal laser scanning microscopy
Cells were seeded into Lab Tek chamber slides (Nalge Nunc, Naperville,
DL) and allowed to grow and adhere to the glass slides for 48 h at 37° C, 5% CO2.
Cells were fixed with ice-cold 96% ethanol for 5 min, washed 3x with PBS and
blocked with 10% normal goat serum in PBS for 1 h at room temperature. COU-
1 (5 μg/ml) together with either mouse anti-K8 antibody (1/1000) or mouse anti
K-18 antibody (1/1000) were incubated overnight at 4 °C. After washing with
PBS, the cells were incubated with FITC-labeled goat-anti-human γ-chain and
Texas Red-labeled goat anti-mouse IgG antibody (diluted 1/200, both from
Jackson ImmunoResearch, West Grove, PA) for 1 h at room temperature in the
dark. The cells were washed with PBS for 3 x 5 min and the slides mounted with

anti-fading reagent Slow Fade™ in PBS/glycerol (Molecular Probes, Eugene,
OR). Results were analyzed using a MRC-1024 confocal laser scanning
microscope (Bio-Rad), attached to a Zeiss Anyvert 100TV. As a control, all.
experiments were also performed omitting the primary antibody or including
species and isotype matched control antibody instead of the primary antibody. In
addition, differential interference contrast (DIC) images of analyzed cells were
obtained.
Results
Purification of cytokeratins from colon cancer and normal colon
epithelia
Fresh, surgically-removed colon cancer tissue and normal colon epithelia
was used to separately extract cytokeratin K8 an K18 by taking advantage of the
fact that cytokeratins and other cytoskeletal proteins are present as insoluble
filamentous structures in buffer at physiological salt concentrations. Non-ionic
detergent was added to the buffer to improve homogenization, partially by
disrupting cell membranes. The insoluble intermediate filaments proteins were
precipitated by centrifugation, subsequently solubilized in an SDS -containing
buffer, and separated by QFF anion-exchange chromatography using a linear salt
gradient.
Figure 1A shows the elution profile from the QFF anion-exchange
column. The fractions containing COU-1 reactivity were found in the first and
second peak of the gradient (fractions 41-48). COU-1 reactivity was detected by
coating the proteins in these fractions onto ELISA wells followed by incubation
with COU-1 (Figure 1B). Western blot analysis demonstrated reactivity of COU-
1 with three main bands in the same fractions (Figure 1D). The proteins in these
three bands represented only a portion of the proteins with molecular weight in
the 41-46 kDa range found in these fractions, as revealed by Coomassie staining
of the SDS-separated gels (Figure 1C).
Western blot analysis and Coomassie staining of cytokeratin purified
from colon cancers of four different patients revealed a similar pattern of protein
bands, reactive and non-reactive with HMab COU-1. Cytokeratin was also

isolated from normal colon epithelia obtained from three individuals using the
same purification procedure to compare the nature of the KS/K18 in colon cancer
versus normal colon tissues. Tissue homogenate and purified cytokeratin
preparations (QFF eluate) from the two sources were examined by Western
blotting using a panel of anti-K8 and anti-K18 antibodies. When approximately
equal amounts of cytokeratin from cancer and the normal epithelia were
analyzed, protein bands (in the 42-46 kDa range) of equal intensity were
observed following staining with the anti-K18 antibody, CYK-90, which
recognizes a linear epitope in the C-terminal part of K18 (Figure 2). In contrast,
when the same preparations were stained with COU-1, three bands at molecular
weights of 42-46 kDa were stained in the cytokeratin preparations from the colon
cancer tissue but not from the normal colon epithelia (Figure 2).
To determine the nature of the different K8/K18-like proteins found in
the colon cancer tissues, an improved separation of the individual protein bands
of the purified cytokeratin preparations was used. Purified cytokeratin
preparations from different colon cancer tissues were, therefore, individually
separated on large 14% SDS-PAGE gels, the proteins were blotted onto filters
and Coomassie stained. Strips of the blot were incubated with either the anti-K8
antibody M20, the anti-K18 antibody CY-90, or the COU-1. Figure 3 A exhibits a
typical blot of a colon cancer tissue sample, displaying approximately 10
different bands visualized by Coomassie staining. At this increased separation, 5
bands showed clear COU-1 reactivity. Additional bands, not stained with COU-
1, were stained either with the anti-K8 antibody, the anti-K18 antibody or both
(Figure 3A).
All ten bands were N-termmal sequenced. As shown in Figure 3B, the
bands corresponded to different forms of K8, K18 and K19, except for one band
that was identified as migration inhibitory factor-related protein 8 (MRPS, also
known as calretinin), a calcium binding protein that may bind to cytokeratins
(40). Most of the bands were N-tenninally truncated K8 or K18, as
demonstrated by the identified amino acid sequence starting at residue 23 to 76,
instead of at the expected residue 1.

The amino terminal truncations of K8 corresponded to residues 23, 40,
66, and 76, while the truncations of K18 corresponded to residues 50 and 68.
Significantly, the K8 and K18 truncations were found at the same residue in
three different colon cancers, indicating that the truncations were caused by
specific proteases.
Analysis of the sequence surrounding the cleavage site suggested that at
least two different proteases were responsible for the cleavage, including one
trypsin-like protease. The bands recognized by COU-1 were N-terminally
truncated K8 and K18. Interestingly, however, not all the N-tenninal truncated
K8 and K18 proteins were recognized by COU-1. For example, no COU-1
binding was observed to an N-terminally truncated K8 protein where the first 22
amino acids were missing, nor did the antibody react with intact K8 or K18. The
latter two observations were made by staining with anti-K8 and anti-K18
antibodies, respectively (bands 1 and 3 in Figure 3A), but not by N-terminal
sequencing, because the proteins were N-terminally blocked (K18 contains an
acetylated serine at its N-terminus).
Mapping the COU-1 epitope using recombinant K8 and K18
fragments
To detail the nature of the epitope recognized by COU-1, this epitope was
mapped using a panel of recombinant N- or C-terminally-deleted K8 and K18
fragments or intact K8 and K18 expressed as GST-fusion proteins. The nature of
these fragments is depicted in Figure 4.
Initially, the panel of K8 and K18 fragments were separated by SDS-
PAGE and blotted onto PVDF membranes. Subsequent analyses of these
Western blots, surprisingly, showed that the COU-1 antibody did not bind to any
of the individual K8 or K18 fragments- Nor did the COU-1 antibody bind to the
intact K8 or K18 molecules (Figures 5B and 6B).
In each experiment, MCF7 cell lysate was included as a positive control,
providing positively reacting bands at molecular weights of 42-46 kDa. To
assure that the K8 and K18 fragment were evenly expressed, gels containing the
fragment panel were run in parallel and the gels for the Western blots were

stained with Coomassie blue (Figure 6A). Moreover, blots of SDS-PAGE-
separated K8/GST or K18/GST fusion proteins were stained with an anti-GST
antibody (Figure 5A). The results demonstrated an approximately even
expression of the different fusion proteins, and that the lack of signal with COU1
was not due to a low expression level of the cytokeratin fragments or to
incomplete transfer of proteins.
In addition, Mabs to K8 and K18, respectively, were tested for binding to
the panel of K8 or K18 fragments. As shown in Figure 5D, the anti-K18 Mab
reacted strongly with K18(l-356), Kl8(l-385) and intact K18, but not with
K18(l-312), indicating that its epitope was located in the region 312-356, Next,
cytokeratins K8 and K18 complexes were tested to see whether those complexes
were recognized by COU-1. Western blots of the panel of K18 fragments were
incubated with the intact purified K8 and the unbound K8 was washed away
before staining with COU-1 (Figure 5C). COU-1 bound strongly to complexes
formed between intact K8 and the K18 fragments Kl8(1-213) through K18(l-
385). In contrast, COU-1 bound only weakly to intact K8/K18(1-187) and intact
K8/intact K18, and no binding was observed to intact K8/K18(1-65) and intact
K8/K18(l-124) (Figure 5C).
Likewise, blots containing the panel of K8 fragments were incubated
with intact K18 before staining with COU-1. COU-1 bound strongly to
complexes formed between the intact K18 and the K8 fragments K8(l-213)
through K8(l-385). In contrast, COU-1 bound weakly to intact K8/intact K18
and no binding was observed for K8(l-65)/intact K18 complexes (Figure 6C).
N-terminal sequencing demonstrated that both K8 and K18 proteins from
colon cancer patients were truncated. Experiments were performed to identify
the K8/K18 heterotypic epitope bound by COU-1. In parallel, Western blots
containing the C-terminal-deleted fragments surrounding the COU-1 epitope,
K18(l-72), K18(l-124), K18(l-187) and intact K18 were generated. These blots
were then incubated with one of the K8 fragments surrounding the COU-1
epitope, K8(l-85), K8(l-129) or K8(l-233), or the intact K8 protein. After
permitting K8-K18 complex formation, the blots were incubated with COU-1
antibodies.

As shown in Figure 7 (A-C), the epitope recognized by COU-1 is not
exposed, or is only minimally exposed, on K18(l-124)/intact K8 or K18(l-
124)/K8(l-233) complexes. In contrast, strong binding of COU-1 was observed
for K18(l-124)/K8(l-129) complexes. No COU-1 binding was observed for any
of the heterotypic complexes containing K8(l-85) or K18(l-72).
Taken together, these results confirm that the epitope recognized by
COU-1 involves the K8 region 85-129 and the K18 region 72-124. As shown in
Figures 4 and 8, this region involves the C-terminal part of the N-terminal head
domain and the N-terminal part of the first helical domain, 1A, of the alpha-
helical rod domain of both K8 and K18.
The results further demonstrate that this epitope is poorly exposed on
heterotypic complexes of intact K8 and K18, even when intact K8 is cornplexed
with Kl8(1-124). The COU-1 epitope is revealed when the first domain, Al, of
the alpha helical rod is not in its normal coil-coil structure. This can be caused by
truncation that removed essential contact points for the existing association
leaving the COU-1 binding region of the K8/K18 complex in an unfolded state.
The combination described above was reversed such that Western blots
of the C-terminal deleted fragments of K8(l-85), K8(l-129), K8(l-233) and
intact K8 were incubated with the fragments of K18 surrounding the COU-1
epitope, K18(l-72), K18(l-124), K18(l-187) and intact K18. These blots were
then incubated with the COU-1 antibody. As shown in Figure 7 (D-F), the
epitope recognized by COU-1 was equally exposed when K8(l-129) was
cornplexed with K18(l-72), K18(l-124), and K18(l-187) or intact K18. Again,
no COU-1 binding was observed with any heterotypic complexes containing
K8(l-85).
COU-1 binding was tested using a panel of heterotypic complexes
consisting of N-terminal deleted K8 and K18 combined with intact K8 and K18
using the heterotypic Western blot assay. These fragments were missing the first
129 atnino acids or more as detailed in Figure 4. However, no COU-1 binding
was observed to any of these N-terminal-deleted heterotypic K8/K18 complexes,
indicating that the COU-1 epitope was located within the N-terminal 129 amino

acids (data not shown). The control showed that the N-terminal-deleted
fragments were well recognized by the murine anti-K8 and anti-K18 Mabs.
The N-terminal sequencing data and the recombinant mapping datas
indicated that the COU-1 epitope were well exposed when the first 6.5 amino
acids of K8 and the first 49 amino acids of K18 were missing.
Two additional N-terminal deleted fragments, K8(66-483) and K18(50-
430) were generated as GST fusion proteins. Figure 9 shows blots of intact K8
and K8(66-483) incubated with K18(50-430) (A) or intact K18 (B). Figure 9
also shows the blots of K18(50-430) and intact K18 incubated with K8(66-483)
(C) or intact K8. Significantly stronger COU-1 binding was observed for K8(66-
483)/K18(50-430) and K8(66-483)/intact K18 complexes than for intact
K8/K18(50-430) or intact K8/intact K18 complexes.
Further investigations were made to determine whether the N-terminal
cleavage of K8 and K18 observed in cancer cells might be caused by adenovirus
infection. The adenovirus L3 23-kDa proteinase promotes specific cleaving of
the N-terminal domain of K18, while leaving K8 intact in adenovirus infection
of HeLa cells (41, 42). This cleavage resulted in removal of region 1-73 of the
head-domain of K18 and the disassembly of the cytokeratin network into
spheroid globules. Tests were performed to examine whether the fragmentation
caused by adenovirus infection would result in the conformation change that
allowed COU-1 binding.
Previous data indicated that COU-1 antibodies do not bind to K8/K18
from HeLa cells. Cytokeratin from HeLa cells infected with adenovirus were
purified and separated by SDS-PAGE, demonstrating a band at a molecular
weight of 41 kDa, in accordance with previous reports. Incubation of COU-1
with Western blot of the adenovirus-infected HeLa cells resulted in no staining
(data not shown), suggesting that the cytokeratin fragments found in the
adenocarcinomas were not a result of adenovirus infection.
It seemed clear that the COU-1 epitope was only present when
heterotypic K8/K18 complexes were formed. The epitope is not present on
individual K8 and K18 molecules. However, the question remained as to why
COU-1 binding to Western blots of SDS-separated cancer cell lysate was

observed where the K8/K18 complexes may have dissociated. A possible
explanation was that during the incubation steps, part of the different
cytokeratins dissociate from the membrane and subsequently attach to and form
high affinity heterotypic complexes with its complementary cytokeratin still
bound to the membrane.
To examine this hypothesis, Western blots of lysate of the colon cancer
cell line, colon 137, were separated into halves. One half was fixed with ethanol
before incubation with the antibodies, while the other half was processed as
usual without fixation. Staining was observed with anti-K18 antibody (CY-90)
on both the fixed and the unfixed blots, while staining with COU-1 was only
observed on the unfixed blot.
Earlier immunohistochemical studies had showed that ethanol fixation of
tissue sections had no effect on the COU-1 antigen. Dot blots of the cancer lysate
were tested for detection of the cancer-associated epitope with or without
fixation. Staining with COU-1 was observed both with and without fixation,
confirming that the COU-1 epitope was not affected by ethanol treatment. In
conclusion, it seems the initial hypothesis was correct, i.e. that heterodimer
formation of cytokeratins takes place during the development of Western blots
and that such heterodimer formation by partially truncated cytokeratin is required
for the formation of the COU-1 epitope.
COU-1 binding to the different recombinant heterotypic K8/K18
complexes was also measured by ELISA. Purified recombinant fragments of K8
or intact K.8 were combined with purified recombinant fragments of K18 or
intact K18 in a molar ratio of 1:1 to generate heterotypic complexes in urea. The
samples were then dialyzed against PBS to allow the formation of the heterotypic
complex, and coated at 5 μg/ml on ELISA plates. Intact K8 was combined with
K18(l-124), Kl8(1-187), K18(l-213), and intact K18. In addition, intact K18
was combined with K8(l-65), K8(l-85), K8(l-129), and K8(l-233).
COU-1 bound with various intensity to all the complexes in this ELISA
assay, except to K8(l-65)/intact K18 and intact K8(l-85)/ intact K18 complexes.
These data are in accord with the results from Western blot analysis. Figure 10
shows the titration of COU-1 on three of the heterotypic complexes,

demonstrating significantly stronger binding to the fragmented K8/K18 than to
the intact K8/K18 complexes.
The kinetic parameters for the binding of COU-1 to different
recombinant heterotypic K8/K18 complexes were measured by real-time
biospecific interaction analysis (BIAcore). COU-1 exhibited high affinity
binding to the heterotypic complexes of K8(l-124)/intact K18 and K8(l-

Cellular distribution of truncated heterotypic K8/K18 complexes
To evaluate the cellular distribution of normal K8 and K18 compared to
truncated K8/K18 heterotypic complexes, breast and colon cancer cell lines
MCF-7 and BrCaOl were co-stained with COU-1 and either Mab M20 (anti-K8)
or Mab CY-90 (anti-K18) and an analyzed by high resolution confocal
microscopy (Figure 11 and 12). Mabs M20 and CY-90 both stained long fibers
of intermediate filaments forming complex interconnecting networks. The fibers
emanate from a perinuclear ring, from which the filaments appear to connect to
the nuclear surface and extend throughout the cytoplasm, terminating at the
plasma membrane. In contrast, COU-1 exhibited a speckled pattern, with
staining of short filament fragments and rod-like particles, indicative of
fragmented intermediate filaments.
Examining the staining pattern of MCF7 cells within cell clusters, only
the peripheral, newly-formed, proliferating cells were strongly positive for COU-
1, while all cells were stained with anti-K18 and anti-K8 Mabs (Figure 11).
Within the proliferating cells of a cluster, COU-1 staining was must prominent at
the outward cell surface, facing away from the cluster. In contrast, Mabs M20

and CY-90 stained the intermediate filamentous network throughout the cells.
The speckled COU-1 staining was seen in close association to the intact
intermediate filament network, as determined by overlay of images stained with
COU-1 and Mab M20 or Mab CY-90 (Figure 12).
Accordingly, N-terminally truncated forms of K8/K18 complexes were
identified only in cancerous epithelia, whereas intact K8/K18 complexes were
observed in both normal and cancerous simple glandular epithelia. The cleavage
of both cytokeratin K8 and cytokeratin K18 at identical sites in different cancer
patients indicates that specific proteases are involved. The cleavage sites at
amino acids 22 and 40 on K8, and at amino acid 50 on K18, all contained the
(S/F/V)XSR↓X(S/V) (SEQ ID NO:50) consensus sequence, suggesting that the
enzyme responsible for these cleavages is a trypsin-like protease (Figure 8).
Analysis of the amino acid sequences in the vicinity of the cleavage sites
revealed one other site on K8 that had the same general sequence (amino acid 32,
GSR↓I (SEQ ID NO:64), but was not cleaved. This suggests that the amino acids
at P3 or P1' positions of the substrate are also influencing the recognition by this
protease.
A consensus sequence was not apparent at the three remaining cleavage
sites on K8 and K18 (TAV↓T (SEQ ID NO:51), SPL↓V (SEQ ID NO:52),
TGI↓A (SEQ ID NO:53)). A protease that requires less stringent recognition
conditions or several different proteases may be responsible for these cleavages.
One such protease may be elastase-type protease that accepts valine, leucine and
isoleucine in the P1 position.
It is unlikely that cleavage of cytokeratins K8 and K18 fragments
occurred during the purification of cytokeratin from the tissue samples for
several reasons. First, a cocktail of five enzyme-inhibitors was present at all
times. Second, cytokeratin fragments were not observed following purification of
cytokeratin from normal colon epithelia using identical purification conditions.
Third, the HMab COU-1, which only recognizes the truncated form of K8/K18,
can detect its epitope in cancerous, but not in normal, epithelia when tissue
samples were minimally handled and immediately fixed.

In contrast to the earlier views, the maintenance of the cytokeratin
network in epithelial cells is a dynamic process involving constant restructuring
by assembly and disassembly of intermediate bundles (45). Microinjection of
biotin-labeled cytokeratin or transfection with fluorescence-labeled cytokeratin
has demonstrated an inward-directed flow of diffuse material at the cell
periphery moving in the form of dots and thin filaments towards the deeper
cytoplasm, where it coalesces with other filaments and filament bundles (46).
While this process occurs in both normal and malignant epithelia cells, the
results provided by the invention indicate the presence of a second degradation
pathway specifically within cancer cells.
Also according to the invention, the human antibody, COU-1, cloned
from a tumor-draining lymph node of a colon cancer patient, specifically
recognizes the N-terminal truncated form of K8 and K18 when the two
cytokeratins formed a heterotypic complex. Previous analysis of COU-1
indicated selective reaction of COU-1 with K18 (35,48), or a modifiedKlS (31,
32,49). Proteolytic cleavage of K18 in association with apoptosis has been
reported (56). However, the cleavage sites for the apoptotic proteases, caspase-
3, -6 and -7, are located in the conserved L1-2 linker and in C-terminal tail
domain, and quite distant to the N-tenninal cleavage sites, as we have studied in
vital tumor tissue (56). Recently, an antibody (M30) was reported to recognize a
neoepitope only exposed in apoptotic cancer cells and not vital or necrotic cells
(57). The neoepitope become exposed when the C-terminus tail domain was
liberated after cleavage by caspase-3, -6 or -7 into 26,22 and 19 kDa fragments.
The cleavage sites observed in colon cancer cells were also different from the
one reported for adenovirus infected HeLa cells, where the N-terminal 73 amino
acids of K18 were removed (41,42). Surprisingly, no COU-1 binding to cleaved
K8/K18 heterotypic complexes from infected HeLa cells was observed, while
COU-1 bound K8/K18 complexes where the 67 most N-terminal amino acids of
K18 were removed. This suggests, although the cleavage sites seem close,
additional removal of 6 amino acids may cause conformational changes that
prevent COU-1 from binding.

Some evidence indicates that K8/K18 is intimately associated with cell
migration and invasiveness. N-terminal cleavage of K8/K18 may influence these
processes. Moreover, the missing N-terminal head domain of K8/K18 contains
several important phosphorylation sites, including ser52 on K.18, which has been
associated with filament reorganization and compartment localization and a
second phosphorylation site important for binding to the 14-3-3 protein (58, 59).
In K8 the phosphorylation site ser23 has been associated with mitogen activation
(60).
The abbreviations used herein are: K8, cytokeratin 8; K18, cytokeratin
18; IF, intermediate filaments; HMab, human monoclonal antibody; FCS, fetal
calf serum; AP, alkaline phosphatase; QFF, Q-Sepharose fast flow; ELISA,
enzyme-linked immunosorbent assay; PBS, phosphate-buffered saline; TBS,
Tris-buffered saline; PVDF, polyvinylidene difluoride membranes; FITC,
fluorescein isothiocyanate.
EXAMPLE 3: Cloning Antibody Fragments that Bind
the Cancer-Associated Epitope
To further develop antibodies useful for detection of cancer, nucleic acids
encoding portions of antibodies were cloned and screened by phage display
selection for binding to the cancer-associated epitope of the invention. These
nucleic acids encode human Fab and other fragments.
Materials and Methods
Antibodies. The human monoclonal IgM antibody, COU-1, is secreted
by the hybridoma cell line, B9165, derived by fusing the human lymphoblastoid
cell line WI-L2-729-HF2 and lymphocytes obtained from mesenteric lymph
nodes from a patient with colon cancer as described above. See also Borup-
Christensen, P., Erb, K., Jensenius, J. C, Nielsen, B. & Svehag, S. -E. (1986)
Int. J. Cancer 37, 683-688. The human-human hybridoma cell line was grown in
protein free medium: RPMI1640 medium (G1BCO, Grand Island, N. Y.)
supplemented with SSR3 serum replacement (Medicult, Copenhagen, Denmark).
The COU-1 antibody was purified from cell culture supernatant by affinity
chromatography on Sepharose-coupled murine monoclonal anti-human u chain

antibody (HB57, American Type Culture Collections, Rockville, MD). The
antibody was eluted with 0.1 M diethylamine, pH 10.5, followed by fractionation
by FPLC. IgM purified from nonnal human serum (Cappel, Cochranville, PA.)
was used as a control.
The human monoclonal IgM antibody, 16.88 was obtained from Dr. R.
McCabe. See Haspel, et al., (1985) Cancer Res. 45, 3951-3961. This antibody
has been used successfully for tumor imaging in humans. See Steis et al. (1990)
J. Clin. Oncol. 8,476-490; Boven et al. (1991) Eur. J. Cancer 27,1430-1436;
Rosenblum et al. (1994) Cancer Immunol. Immunother. 39,397-400). Two
murine monoclonal antibodies, M20 directed against normal cytokeratin 8 and
CY-90 directed against normal cytokeratin 18, were obtained from Sigma
Chemical Co. (St. Louis, MO).
PCR amplification and cloning of the variable heavy and light chain
genes. Total RNA was prepared from the B9165 hybridoma cell line by the
guanidinium method. After reverse transcription, the µ (Fd region) and K chains
were amplified by the polymerase chain reaction (PCR) using a set of family-
specific primers using methods described in Persson et al., (1991) Proc. Natl.
Acad. Sci. USA 88,2432-2436. The amplified light chain DNA was cut with the
restriction enzymes Sac I and Xba I and ligated with Sac I/Xba 1-lmea.rized
pComb3 vector for 3 h as described in Burton et al., (1991) Proc. Nail. Acad. Sci.
USA 88,10134-10137, and Barbas et al. (1991) Proc. Natl. Acad. Sci. USA 88,
7978-7982. The ligated material was purified and transformed by electroporation
into 200 µl Escherichia coli XL1-Blue cells. After transformation, the cells were
grown overnight and phagemid DNA was prepared.
Subsequently, the PCR amplified heavy chain and isolated phagemid
DNA containing the light chain were digested with the restriction enzymes Spe I
and Xho I. The heavy chain phagemid fragments were ligated and used to
transform XLl-Blue. The Fab library was grown in SOC medium for 1 h at 37°C
following addition of SB medium containing carbenicillin (50 μg/ml) and
tetracycline (10 μg/ml). After 3 h, helper phage VCS-M13 (1012 plaque-forming
units) was added and the culture was shaken for an additional 2 h. Kanamycin (70

µg/ml) was added and the culture was incubated at 30°C overnight. The
supernatant was cleared by centrifugation (4000 x g for 20 min) at 4°C. Phage
were precipitated by a second round of centrifugation after the addition of 5%
polyethylene glycol and 0.15 M NaCl and incubation on ice for 30 min. Phage
pellets were resuspended in phosphate-buffered saline, pH 7.4 (PBS) containing
1% (w/v) bovine serum albumin (BSA) and centrifuged for 3 min at 10,000 x g to
pellet debris.
Enrichment of antigen-binding phage through panning. Panning of
the B9165 antibody library was carried out using methods described in Burton et
al. (1991) Proc. Natl. Acad. Sci. USA 88,10134-10137. In brief, microtiter wells
were coated overnight with ultrasonicated lysate of a colon cancer cell line
(colol37) in 0.1 M bicarbonate buffer, pH 8.6 at 4°C. See Ditzel et al. (1992)
Eur. J. Nucl. Med. 19,409-417. Following blocking with PBS containing 3%
BSA for 1 h at 37°C, 50 ul phage suspension in PBS was added to each well and
incubated for 2 h. Unbound phage were removed by vigorous washing 10 times
with PBS containing 0.05% (w/v) Tween 20 (PBS-Tween)(Merck, Darmstadt,
FRG). Bound phage, enriched for those bearing antigen-binding Fabs, were eluted
with 0.2 M glycine/HCl, pH 2.2. The eluted phage were amplified by infection of
E. coli and recovered by superinfection with VCS-M13 helper phage. The
panning procedure was carried out twice. Phagemid DNA was isolated from the
last round of panning, cut with NheI and SpeI and religated. This step excised the
cpIII gene, resulting in a vector producing soluble Fab fragments.
ELISA analysis of B9165 Fab and intact antibodies. Fabs were
prepared as bacterial supematants through a freeze-thawing procedure, using
methods reported by Burton et al. (1991) Proc. Natl. Acad. Sci. USA 88,10134-
10137, and Barbas et al. (1991) Proc. Natl. Acad. Sci. USA 88, 7978-7982.
To assess specificity, supematants and purified Fabs were screened in an
ELISA system for binding to ultrasonicates of colon cancer cells (Colon 137),
BSA (Sigma), ovalbumin (Sigma), recombinant HIV-1 gpl20 (IIIB) (Intracel,
Issaquah, WA) and human placental DNA (Sigma). ELISA wells (Costar) were

coated overnight at 4 °C with 50 µl of antigen (1-10 μg/ml) in 0.1 M bicarbonate
buffer, pH 8.6. DNA in PBS was dried on the ELISA wells at 37°C. The wells
were washed twice with PBS, blocked by filling the wells with 3% BSA in-PBS
for 1 h at 37°C, and incubated with human Fab samples or intact human IgM
antibody for 2 h at 37°C. Plates were washed 10 times with PBS-Tween and
bound Fab was detected with alkaline phosphatase (AP) labeled goat anti-human
IgG F(ab')2 (Pierce Chemical Co, Rockford, EL) diluted 500 fold in PBS or
alkaline phosphatase-labeled rabbit anti-human K-chain (Sigma) diluted 1000 fold
in PBS. Bound antibody was visualized with para-nitrophenylphosphate
(Sigma)(l mg/ml, 1 mM MgCl2,10% (w/v) diethanolamine, pH 9.6) and read at
405 nm.
Purification of Fab. Recombinant B9165 Fab was purified losing methods
described in Ditzel et al. (1995) J. Immunol. 154, 895-908 with some
modifications. In brief, E. coli containing the appropriate clone was inoculated
into one liter cultures of superbroth containing carbenicillin (50 μg/ml),
tetracycline (10µg/ml) and MgCl2 (20 mM), and grown at 37°C, with shaking, for
6 h. Protein expression was then induced with 2 mM isopropyl p-D-
thiogalactopyranoside and growth continued at 30°C overnight. Soluble Fab was
purified from bacterial supernatants by affinity chromatography using a goat
antibody against human IgG F(ab')2 (Pierce) cross-linked to protein G
gammabind matrix (Pharmacia). The column was washed with PBS and bound
Fab eluted with 0.2 M glycine/HCl, pH 2.2, and immediately neutralized with 1
M Tris/HCl, pH 9.0.
Nucleotide sequencing. Sequencing was carried out on a 373A automated
DNA sequencer (ABI, Foster City, Ca) using a Taq fluorescent dideoxy
terminator cycle sequencing kit (ABI). Primers for the elucidation of light chain
sequence were the SEQKb primer (5'-ATAGAAGTTGTTCAGCAGGCA-3',
SEQ ID NO.41), hybridizing to the (+) strand and the KEF primer (5'-
GAATTCTAAACTAGCTAGTTCG-3', SEQ ID NO:42) hybridizing to the (-)
strand. For the heavy chain, the CMHD primer (5'-


Analysis by confocal laser scanning microscopy. Human colon cancer
cell-lines (H3619 and colol37), and breast cancer cell-lines (MCF-7 and H3396)
were grown in Iscove's modified Dulbecco's medium containing 10% FBS and
allowed to adhere to chambered coverslips (Nunc, Kamstrup, Denmark) for 48 h
at 37°C, 5% CO2, in order to form monolayers. Experiments were performed
using the primary COU-1 antibodies, B9165 Fab, murine anti-cytokeratin S,
murine anti-cytokeratin 18, and HuMab 16.88 as indicated below. All antibodies
were tested at 10 µg/ml except B9165 Fab (30 μg/ml).
1) Intracellular staining. H3619 and colo 137 cells were permeabilized
with methanol at -20°C for 5 min, blocked with normal goat serum followed by
incubation with primary antibodies at room temperature for 1 h. The cells were
then washed 3 times with culture medium and incubated with FITC-labeled goat
anti-human K-chain antibody (Southern biotech) or FITC-labeled goat anti-mouse
IgG (BioSource) diluted 1:100 and 1:50 respectively in PBS for 1 h at room
temperature.
2) Surface staining. Live H3619 cells were incubated with COU-1
antibodies at 4°C for 2 h, washed 3 times with cold culture medium and incubated
with secondary FITC-labeled antibody at 4°C for 1 h.
3) Internalization. Live H3619 and colol37 cells were incubated with
COU-1 antibodies or B9165 Fab at 37°C for 6 h, followed by washing 3 times
and permeabilization with methanol at -20°C for 5 min. Cells were blocked with
normal goat serum and incubated with secondary FITC-labeled antibody at RT for
1 h. For all experiments, following primary and secondary antibody incubations,
the cells were washed, fixed with 2% paraformaldehyde in PBS for 15 min at
room temperature, washed twice and mounted in anti-fading reagent (30mM
dithioerythritol:PBS:glycerol, 2:9:1). Staining of cells was evaluated by confocal
laser scanning microscopy. As control all experiments were carried out omitting
the primary antibody.

Immunohistochemical analysis. Tissue specimens were obtained from
colorectal cancer patients undergoing surgical resection. Normal colon tissue was
taken from the resectate approximately 15 cm away from the site of the tumor.
Tissues were fixed in 96% alcohol for 6 h at 4°C. Afterwards, tissues were
paraffin embedded and cut into 5 um sections. Sections were deparaffinized in
xylol, rehydrated through graded alcohol and washed in PBS-Tween. Sections
were incubated for 2 h at room temperature in a humidified chamber with 100 µl
of murine monoclonal antibody, human monoclonal IgM antibody or normal
polyclonal human IgM, all at 0.5-10 μg/ml. The slides were washed and
incubated with AP-labeled rabbit anti-human IgM (Dako, Glostrup, Denmark),
horse-radish peroxidase (HRP) labeled rabbit anti-human IgM (Dako) or HRP-
labeled rabbit anti-mouse IgG (Dako) diluted in PBS with 10% (w/v) bovine
serum albumin for 1 h at room temperature. After washing, the HRP was
visualized by development with chromogenic substrate (0.6 mg
diaminobenzidine per ml PBS with 0.01% H2O2) and AP with 0.2 mg naphthol-
AS-Mx phosphate (Sigma), 1 mg Fast Red TR Salt (Sigma), 20µg
dimethylformamide per ml 0.1M Tris/HCl, 1M levamisole, pH 8.2. The sections
were counterstained with Mayer's haematoxylin, dehydrated in xylene and
mounted in Aquamount (Gurr, Poole, England). The staining intensity was
graded as follows: (-) no staining, (+) weak staining, (++) moderate staining,
(+++) strong staining.
Results
Phage display expression and sequencing of HuMab that can bind the
Cancer-Associated Epitope. RNA was extracted from the B9165 cell line and
the heavy (µ, Fd region) and light (k)-chain genes from the corresponding cDNA
were amplified by PCR using 3' family specific primers and a 5' constant primer.
The light and heavy chain products were then sequentially cloned into the M13
phage surface expression vector pComb3 to generate a library of 2x106 members.
The phage library was selected twice on an ultrasonicate of the COU-1 antigen
positive colon cancer cell line (colon 137). Eluted phage from the last round of

selection were used to infect E. coli XLI-blue cells. DNA was prepared from
these cells and gene H1 fragment removed by NheI/SpeI digestion and ligation.
The reconstructed phagemids were used to transform XLI-Blue to produce clones
secreting soluble Fab fragments. Supernatants of three of the 80 single Fab
expression clones tested, exhibited binding to colon 137 lysate and no binding to
ovalbumin in ELISA.
The sequences of these three clones were identical. Sequence analysis
showed that the B9165 hybridoma cell light chain belongs to the VKIII family
and that it exhibits 97% (269/276) nucleotide homology to L6 as closest germ-
line (Figure 13). The B9165 light chain contained an extra serine inserted
corresponding to codon 30. The light chain J segment showed 95% (36/38)
nucleotide homology to the germ-line JK5 segment. Further, sequence analysis
showed that the heavy chain belongs to the VHI family, exhibiting 98%
nucleotide homology to the heavy chain germ-line DP-7. The heavy chain J
segment showed 96% (53/55) nucleotide homology to the germ-line JH6b
segment. The D segment of COU-1 showed closest homology to the D2 germ-line
D segment with a 16 nucleotide stretch of complete homology.
Purified recombinant B9165 Fab was tested in parallel with the intact
COU-1 antibodies and normal polyclonal IgM for binding to lysate of colon
cancer cells (colol37) and irrelevant antigens in ELISA. The B9165 Fab and
COU-1 exhibited strong binding to colon 137 lysate, but not to a panel of other
antigens including BSA, ovalbumin, human DNA and HIV-1 gpl20 (data not
shown). In contrast, normal human IgM did not bind to any of the antigens. The
concentration needed for saturation was significantly higher for the B9165 Fab
(20 μg/ml) than for the intact antibody (1 μg/ml) and was similar to that
previously measured for chemical derived half-monomeric fragments, exhibiting
a Ka of 2x106 M-1 (Ditzel, H., Erb, K., Leslie, G. & Jensenius, J. C. (1993) Hum.
Antibod. Hybridomas 4, 86-93).
COU-1 binds preferentially to malignant carcinoma cells. The
subcellular localization of the antigen recognized by COU-1 in tissue biopsies of
colon and rectal adenocarcinomas was studied using an indirect

The antibodies were compared for staining of colon metastases in liver
versus surrounding normal liver tissue. COU-1 gave intense staining of the
immunoperoxidase and alkaline-phosphatase techniques. At high magnification,
distinct fibrillar staining of intermediated filaments by COU-1 was observed. In
small cell clusters or individual cells, intense staining was seen at the periphery,
possibly associated with the cell surface. In addition, enhanced staining associated
with the junctional zone between adjacent cells was seen. No staining was
observed in adjacent normal colon crypt epithelial cells in five of eight colon or
rectal cancer. In the other three cancers, weak staining of a few individual cell
surrounded by negative cells was observed in adjacent morphologically normal
colon tissues in addition to strong staining of the cancer tissue. Although these
colon epithelia looked morphologically normal, this may not be the case. Murine
anti-cytokeratin 8 antibodies and anti-cytokeratin 18 antibodies (not shown) gave
intense staining of the adjacent normal colon epithelia as well as of the colon
cancer tissue. COU-1, however, reacted only with the malignant cells and not
with the normal epithelia. A comparison of the staining levels for COU-1, murine
anti-cytokeratin 8 and 18, and 16.88 is given in Table 3. The 16.88 antibody
showed strong staining of the colon cancer cells, weak staining only in some areas
of the normal colon epithelia, but in addition stained the smooth muscle fibers
and myoepithelia derived connective tissue was observed (Table 5).


metastasis whereas no staining of the majority of hepatocytes was observed. A
few hepatocytes in the periportal zones were weakly positive. Similarly, the 16.88
antibody did not stain the majority of the hepatocytes. However, the myoepithelia
connective tissue was stained by 16.88, but not with COU-1. Both human
antibodies stained the biliary ducts. The murine anti-cytokeratin 8 and 18 (not
shown) antibodies stained the metastases as well as the normal hepatocytes
strongly and with equal intensity. The staining decreased towards the
centrilobular area. Particular strong staining was seen associated with the cell
membrane of the hepatocytes with the murine Mabs.
Phage display and bacterial expression was therefore used to clone and
further characterize Fab and other antibody fragments from a hybridoma cell line
expressing the human monoclonal antibody COU-1. The binding characteristics
of the cloned B9165 Fab were very similar to previous reports for the half-
monomeric fragments generated by chemical reduction and alkylation (Ditzel, H.,
Erb, K., Leslie, G. & Jensenius, J. C. (1993) Hum. Antibod. Hybridomas 4, 86-
93). Sequence analysis showed that the variable region of the heavy and light
chain had minimal somatic mutations with 98% and 97% nucleotide homology to
the closest germ-line V genes, respectively. This is in accordance with COU-1
being an IgM antibody, and indicates that substantial affinity maturation through
site directed mutagenesis is possible.
The foregoing specification, including the specific embodiments and
examples, is intended to be illustrative of the present invention and is not to be
taken as limiting. Numerous other variations and modifications can be effected
without departing from the true spirit and scope of the present invention.
References
The following references, to the extent that they provide exemplary
procedural or other details supplementary to those set forth herein, are
specifically incorporated herein by reference.
1. Schmidt, A., Heid, H. W., Schafer, S., Nuber, U. A., and Zimbelmann, R.
Desmosomes and cytoskeletal architecture in epithelial differentiation:
cell type-specific plaque components and intermediate filament
anchorage. European Journal of Cell Biology, 65:229-245,1994.

2. Fuchs, E. and Cleveland, D. W. A structural scaffolding of intermediate
filaments in health and disease. Science, 279: 514-519,1998.
3. Hatzfeld, M. and Franke, W. W. Pair formation and promiscuity of
cytokeratins: formation in vitro of heterotypic complexes and
intermediate-sized filaments by homologous and heterologous
recombinations of purified polypeptides. J. Cell Biol., 101: 1826-1841,
1985.
4. Fuchs, E. and Weber, K. Intermediate filaments: structure, dynamics,
function, and disease. Annu. Rev. Biochem., 63: 345-382,1994.
5. Moll, R. Cytokeratins as markers of differentiation in the diagnosis of
epithelial tumors. Subcell. Biochem., 31:205-262,1998.
6. Steinert, P. M. and Roop, D. R. Molecular and cellular biology of
intermediate filaments. Ann. Rev. Biochem., 57: 593-625,1988.
7. Hatzfeld, M. and Weber, K. The coiled coil of in vitro assembled keratin
filaments is a heterodimer of type I & II keratins; use of site-specific
mutagenesis and recombinant protein expression. J. Cell Biol., 110:
1199-1210,1990.

8. Steinert, P. M. The two-chain coiled-coil molecule of native epidermal
keratin intermediate filaments is a type I-type II heterodimer. J. Biol.
Chem., 265: 8766-8774,1990.
9. Traub, P. Intermediate filaments: a review, vol. ed. Heidelberg,
Germany: Springer-Verlag, 1985.

10. Hatzfeld, M. and Weber, K. Tailless keratins assemble into regular
intermediate filaments in vitro. J. Cell Sci., 97: 317-324,1990.
11. Shoeman, R. L., Mothes, E., Kesselmeier, C, and Traub, P. Intermediate
filament assembly and stability in vitro: effect and implications of the
removal of head and tail domains of vimentin by the human

immunodeficiency virus type I protease. Cell Biol. Int. Rep., 14: 583-594,
1990.
12. Bader, B. L., Magin, T. M, Freudenmann, M., Stumpp, S., and Franke,
W. W. Intermediate filaments formed de novo from tail-less cytokeratins
in the cytoplasm and in the nucleus. J. Cell Biol., 115:1293-1307, 1991.
13. Hatzfeld, M. and Burba, M. Function of type I and type II keratin head
domains: their role in dimer, tetramer and filament formation. J. Cell
Science, 107: 1959-1972,1994.
14. Ku, N. O., Michie, S., Oshima, R. G., and Omary, M. B. Chronic
hepatitis, hepatocyte fragility, and increased soluble
phosphoglycokeratins in trans genie mice expressing a keratin 18
conserved arginine mutant. J. Cell Biology, 131: 1303-1314, 1995.
15. Ku, N. O., Gish, R., Wright, T. L., and Omary, M. B. Keratin 8 mutations
in patients with cryptogenic liver disease. N. Engl. J. Med., 344: 1580-
1587,2001.
16. Baribault, H., Price, J., and Oshima, R. G. Mid-gestational lethality in
mice lacking keratin 8. Genes and Development, 7: 1191-1202,1993.
17. Baribault, H., Penner, J., Iozzo, R. V., and Wilson-Heiner, M. Colorectal
hyperplasia and inflammation in keratin 8-deficient FVB/N mice. Genes
and Development, 8: 2964-2973,1994.
18. Hesse, M, Franz, T., Tamai, Y., Taketo, M. M., and Magin, T. M.
Targeted deletion of keratins 18 and 19 leads to trophoblast fragility and
early embryonic lethality. EMBO J., 19: 5060-5070, 2000.
19. Bauman, P. A., Dalton, W. S., Anderson, J. M., and Cress, A. E.
Expression of cytokeratin confers multiple drug resistance. PNAS, 91:
5311-5314,1994.

20. Parekh, H. K. and Simpkins, H. The differential expression of cytokeratin
18 in cisplatin-sensitive and -resistant human ovarian adenocarcinoma
cells and its association with drug sensitivity. Cancer Research, 55: 5203-
5206,1995.
21. Anderson, J. M., Heindl, L. M., Bauman, P. A., Ludi, C. W., Dalton, W.
S., and Cress, A. E. Cytokeratin expression results in a drug-resistant
phenotype to six different chemotherapeutic agents. Clinical Cancer
Research, 2: 97-105, 1996.
22. Schaafsma, H. E., Ramaekers, F. C. S., van Muijen, G. N. P., Lane, E. B.,
Leigh, I. M, Robben, H., Huijsmans, A., Ooms, E. C. M, and Ruiter, D.
J. Distribution of cytokeratin polypeptides in human transitional cell
carcinomas, with special emphasis on changing expression patterns
during tumor progression. Am. J. Pathology, 136: 329-343,1990.
23. Moll, R. Cytokeratins in the histological diagnosis of malignant tumors.
Int. J. Biol. Markers, 9: 63-69, 1994.
24. Moll, R., Franke, W. W., Schiller, D. L., Geiger, B., and Krepler, R. The
catalog of human cytokeratins: Patterns of expression in normal epithelia,
tumors and cultured cells. Cell, 31: 11-24,1982.
25. Pankov, R., Umezawa, A., Maki, R., Der, C. J., Hauser, C. A., and
Oshima, R. G. Keratin 18 activation by Ha-ras is mediated through ETS
and Jun binding sites. PNAS, 91: 873-877,1994.

26. Oshima, R. G., Abrams, L., and Kulesh, D. Activation of an intron
enhancer within the keratin 18 gene by expression of c-fos and c-jun in
undifferentiated F9 embryonal carcinoma cells. Genes. Dev., 4: 835-848,
1990.
27. Oshima, R. G., Baribault, H., and Caulin, C. Oncogenic regulation and
function of keratins 8 and 18. Cancer and Metastasis Reviews, 15: 445-
471,1996.

28. Chu, Y. W., Runyan, R. B., Oshima, R. G., and Hendrix, M. J. C.
Expression of complete keratin filaments in mouse L cells augments cell
migration and invasion. PNAS, 90: 4261-4265,1993.
29. Pankov, R., Simcha, I., Zoller, M., Oshima, R. G., and Ben-Ze'ev, A.
Contrasting effects of k8 and k18 on stabilizing k19 expression, cell
motility and tumorigenicity in the BSp73 adenocarcinoma. J. Cell
Science, 110: 965-974,1997.
30. Hendrix, M. J. C, Seftor, E. A., Chu, Y. W., Seftor, R. E. B., Nagle, R.
B., McDaniel, K. M., Leong, S. P. L., and Yohem, K. H. Co-expression
of vimentin and keratins by human melanoma tumor cells: Correlation
with invasive and metastatic potential. J. Natl. Cancer Inst., 84:165-174,
1992.
31. Ditzel, H. J., Garrigues, U., Andersen, C. B., Larsen, M. K., Garrigues,
H. J., Svejgaard, A., Hellstrom, L, Hellstrom, K. E., and Jensenius, J. C.
Modified cytokeratins expressed on the surface of carcinoma cells
undergo endocytosis upon binding of human monoclonal antibody and its
recombinant Fab fragment. PNAS USA, 94: 8110-8115,1997a.
32. Ditzel, H. J., Rasmussen, J. W., Erb, K., Borup-Christensen, P., Titlestad,
I., Lassen, E., Fenger, C, Kronborg, O., and Jensenius, J. C. Tumor
detection with 1311-labeled human monoclonal antibody COU-1 in
patients with suspected colorectal carcinoma. Cancer Research, 53: 5920-
5928,1993.
33. Chan, R., Rossitto, P. V., Edwards, B. F., and Cardiff, R. I). Presence of
proteolytically processed keratins in the culture medium of MCF-7 cells.
Cancer Research, 46:6353-6359, 1986.
34. Hembrough, T. A., Vasudevan, J., Allietta, M. M., Glass, W. F.H, and
Gonias, S. L. A cytokeratin 8-like protein with plasminogen-binding
activity is present on the external surfaces of hepatocytes, HepG2 cells
and breast carcinoma cell lines. J. Cell Science, 108: 1071-1082,1995.

35. Borup-Christensen, P., Erb, K., Ditzel, H., Nielsen, B., Larsen, J. K.,
Svehag, S. E., and Jensenius, J. C. Human hybridoma producing
monoclonal antibodies against colorectal cancer-associated antigens.
APMIS, 98: 674-684,1990.
36. Towbin, H., Staehelin, T., and Gordon, J. Electrophoretic transfer of
proteins from polyacrylamide gels to nitrocellulose sheets: procedure and
some applications. PNAS, 76: 4350-4354,1979.
37. Crawford, C, Brown, N. R., and Willis, A. C. Investigation of the
structural basis of the interaction of calpain II with phospholipid and with
carbohydrate. Biochem. J., 265: 575-579,1990.
38. Waseem, A., Lane, E. B., Harrison, D., and Waseem, N. A keratin
antibody recognizing a heterotypic complex: Epitope mapping to
complementary locations on both components of the complex.
Experimental Cell Research, 223: 203-214,1996.
39. Karlsson, R. A., Michaelsson, A., and Mattsson, L. Kinetic analysis of
monoclonal antibody-antigen interactions with a new biosensor based
analytical system. J. Immunol. Methods, 145: 229-240,1991.
40. Marilley, D. and Schwaller, B. Association between the calcium-binding
protein calretinin and cytoskeletal components in the human colon
adenocarcinoma cell line WiDr. Experimental Cell Research, 259: 12-22,
2000.
41. Chen, P. H., Ornelles, D. A., and Shenk, T. The adenovirus L3 23-
kilodalton proteinase cleaves the amino-terminal head domain from
cytokeratin 18 and disrupts the cytokeratin network of HeLa cells. J.
Virology, 67: 3507-3514, 1993.
42. Zhang, Y. and Schneider, R. J. Adenovirus inhibition of cell translation
facilitates release of virus particles and enhances degradation of the
cytokeratin network. J. Virology, 68: 2544-2555,1994.

43. Broers, J. L. V., Ramaekers, F. C. S., Rot, M. K., Oostendorp, T.,
Huysmans, A., van Muijen, G. N. P., Wagenaar, S. S., and Vooijs, G. P.
Cytokeratins in different types of human lung cancer as monitored by
chain-specific monoclonal antibodies. Cancer Research, 48: 3221-3229,
1988.
44. Shitara, K., Fujiwara, K., Kusano, A., Yamaguchi, K., Yoshida, H., Sato,
S., and Hanai, N. Application of anti lung adenocarcinoma monoclonal
antibody recognizing cytokeratin-like cytoplasmic antigen for tumor
diagnosis. Anticancer Res, 12: 1121-1129, 1992.
45. Lane, E. B., Goodman, S. L., and Trejdosiewicz, L. K. Disruption of the
keratin filament network during epithelial cell division . EMBO Journal,
1: 1365-1372,1982.
46. Miller, R. K., Khuon, S., and Goldman, R. D. Dynamics of keratin
assembly: Exogenous type I keratin rapidly associates with type II keratin
in vivo. J. Cell Biology, 122: 123-135,1993.
47. Nishibori, H., Matsuno, Y., Iwaya, M., Osada, T., Kubomura, N.,
Iwamatsu, A., Kohno, H., Sato, S., Kitajima, M., and Hirohashi, S.
Human colorectal carcinomas specifically accumulate Mr 42,000
ubiquitin-conjugated cytokeratin 8 fragments. Cancer Res., 56: 2752-
2757,1996.

48. Erb, K., Borup-Christensen, P., Ditzel, H., Chemnitz, J., Haas, H., and
Jensenius, J. C. Characterization of a human-human hybridoma antibody,
C-OU1, directed against a colon tumor-associated antigen. Hybridoma,
11: 121-134, 1992.
49. Ditzel, H. J. Human monoclonal antibodies: a took for cancer detection
in vivo. APMIS Suppl., 94: 5-42,1999.
50. Hembrough, T. A., Kralovich, K. R., Li, L., and Gonias, S. L.
Cytokeratin 8 released by breast carcinoma cells in vitro binds

plasminogen and tissue-type plasminogen activator and promotes
plasminogen activation. Biochem. J., 317: 763-769,1996.
51. Kralovich, K. R., Li, L., Hembrough, T. A., Webb, D. J., Karns, L. R.,
and Gonias, S. L. Characterization of the binding sites for plasminogen
and tissue-type plasminogen activator in cytokeratin 8 and cytokeratin 18.
J. Protein Chem., 17: 845-854,1998.
52. Wells, M. J., Hatton, M. W. C, Hewlett, B., Podor, T. J., Sheffield, W.
P., and Blajchman, M. A. Cytokeratin 18 is expressed on the hepatocyte
plasma membrane surface and interacts with thrombin-antithrombin
complexes. J. Biological Chemistry, 272: 28574-28581,1997.
53. Rydlander, L., Ziegler, E., Bergman, T., Schoberl, E., Steiner, G.,
Bergman, A. C, Zetterberg, A., Marberger, M., Bjorklund, P., Skern, T.,
Einarsson, R., and Jornvall, H. Molecular characterization of a tissue-
polypeptide-specific-antigen epitope and its relationship to human
cytokeratin 18. Eur. J. Biochem., 241: 309-314,1996.
54. Silen, A., Wiklund, B., Norlen, B. J., and Nilsson, S. Evaluation of a new
tumor marker for cytokeratin 8 and 18 fragments in healthy individuals
and prostate cancer patients. Prostate, 24: 326-332,1994.
55. Godfroid, E., Geuskens, M., Dupressoir, T., Parent, L, and Szpirer, C.
Cytokeratins are exposed on the outer surface of established human
mammary carcinoma cells. J. Cell Science, 99: 595-607,1991.
56. Caulin, C, Salvesen, G. S., and Oshima, G. Caspase cleavage of keratin
18 and reorganization of intermediate filaments during epithelial cell
apoptosis. J. Cell Biol., 138:1379-1394,1997.
57. Leers, M. P., Kolgen, W., Bjorklund, V., Bergman, T., Tribbick, G.,
Persson, B., Bjorklund, P., Ramaekers, F. C, Bjorklund, B., Nap, M.,
Jomvall, H., and Schutte, B. Immunocytochemical detection and

mapping of a cytokeratin 18 neo-epitope exposed during early apoptosis.
J. Pathol., 187: 567-572,1999.
58. Ku, N. O. and Omary, M. B. Identification of the major physiologic
phosphorylation site of human keratin 18: potential kinases and a role in
filament reorganization. J. CellBiol., 727: 161-171, 1994.
59. Liao, J., Lowthert, L. A., Ku, N. O., Fernandez, R., and Omary, M. B.
Dynamics of human keratin 18 phosphorylation: Polarized distribution of
phosphorylated keratins in simple epithelial tissues. J. Cell Biology, 131:
1291-1301,1995.
60. Ku, N. O., Liao, J., Chou, C. F., and Omary, M. B. Implications of
intermediate filament protein phosphorylation. Cancer and Metastasis
Reviews, 15: 429-444,1996.

We claim:
1. An isolated cancer-associated epitope comprising recombinant cytokeratin 8 and
cytokeratin 18 polypeptide fragments, wherein the cytokeratin 8 polypeptide is shorter than 482
amino acids and comprises SEQ ID NO:3 or SEQ ID NO:5, and the cytokeratin 18 polypeptide
is shorter than 429 amino acids and comprises SEQ ID NO:4 or SEQ ID NO:6.
2. The isolated epitope as claimed in claim 1, wherein the cytokeratin 8 polypeptide is
shorter than about 475 amino acids and the cytokeratin 18 polypeptide is shorter than about 425
amino acids.
3. The isolated epitope as claimed in claim 1, wherein the cancer-associated epitope is
detected in filamentous cytoplasmic structures of adenocarcinoma cells but is substantially
undetected in normal cells.
4. The isolated epitope as claimed in claim 1, wherein the cancer-associated epitope is
detected in filamentous cytoplasmic structures of colon adenocarcinoma, ovarian
adenocarcinoma, renal adenocarcinoma, mammary adenocarcinoma, lung adenocarcinoma,
pancreatic adenocarcinoma and non-seminomal testis carcinoma cells.
5. A vaccine composition for preventing or treating adenocarcinoma comprising
recombinant cytokeratin 8 and cytokeratin 18 polypeptide fragments, wherein the cytokeratin 8
polypeptide is shorter than about 482 amino acids and comprises SEQ ID NO:3 or SEQ ID
NO:5, and the cytokeratin 18 polypeptide is shorter than about 429 amino acids and comprises
SEQ ID NO:4 or SEQ ID NO:6.
6. The vaccine composition as claimed in claim 5, wherein the cytokeratin 8 polypeptide is
shorter than about 475 amino acids and the cytokeratin 18 polypeptide is shorter than about 425
amino acids.
7. The vaccine composition as claimed in claim 5, wherein the adenocarcinoma is colon
adenocarcinoma, ovarian adenocarcinoma, renal adenocarcinoma, mammary adenocarcinoma,
lung adenocarcinoma, pancreatic adenocarcinoma or non-seminomal testis carcinoma.

8. An isolated antibody that can bind to a cancer-associated epitope consisting of a complex
between two separate polypeptides, a cytokeratin 8 polypeptide missing the first 65 amino acids
and a cytokeratin 18 polypeptide missing the first 49 amino acids, wherein the antibody
specifically binds cytokeratin 8 SEQ ID NO:3 or SEQ ID NO:5, and cytokeratin 18 SEQ ID
NO:4 or SEQ ID NO:6, wherein the antibody is not a COU-I IgM antibody secreted by a B9165
cell line and the antibody specifically binds to adenocarcinoma tissue but has substantially
weaker binding to normal epithelial cells and smooth muscle fibers.
9. The isolated antibody as claimed in claim 8, wherein the is shorter than about 425 amino
acids.
10. The isolated antibody as claimed in claim 8, wherein the antibody is shorter than about
200 amino acids.
11. The isolated as claimed in claim 8, wherein the antibody is an IgG, IgA, IgD, IgE or IgM
antibody.
12. The isolated antibody as claimed in claim 8, wherein the antibody can detect the cancer-
associated epitope in filamentous cytoplasmic structures of adenocarcinoma cells but in
substantially no filamentous structures of normal cells.
13. The binding entity as claimed in claim 8, wherein the antibody can detect the cancer-
associated in filamentous cytoplasmic structures of colon adenocarcinoma, ovarian
adenocarcinoma, renal adenocarcinoma, mammary adenocarcinoma, lung adenocarcinoma,
pancreatic adenocarcinoma and non-seminomal testis carcinoma cells.
14. The antibody as claimed in claim 8, wherein the antibody comprises a polypeptide having
an amino acid sequence with at least 98% homology to any one of SEQ ID NO:7-35.
15. The antibody as claimed in claim 8, wherein the antibody comprises a polypeptide having
any one of SEQ ID NO:7-35 or SEQ ID NO:47-49.
16. The antibody as claimed in claim 8, wherein the antibody is encoded by a nucleic acid
comprising any one of SEQ ID NO:36-39.

17. A kit for detecting cancer comprising a container containing an antibody that can bind to
a cancer-associated epitope, wherein the cancer-associated epitope consists of a complex
between two separate polypeptides, a cytokeratin 8 polypeptide missing the first 65 amino acids
and a cytokeratin 18 polypeptide missing the first 49 amino acids, wherein the antibody
specifically binds cytokeratin 8 polypeptide SEQ ID NO:3 or SEQ ID NO:5, and cytokeratin 18
polypeptide SEQ ID NO.4 or SEQ ID NO.6, wherein the antibody is not a COU-1 IgM antibody
secreted by a B9165 cell line and the antibody specifically binds to adenocarcinoma tissue but
has substantially weaker binding to normal epithelial cells and smooth muscle fibers
18. The kit as claimed in claim 17, wherein the antibody is shorter than about 425 amino
acids.
19. The kit as claimed in claim 17, wherein the antibody is shorter than about 200 amino
acids.
20. The kit as claimed in claim 18, wherein the antibody is an IgG antibody.
21. The kit as claimed in claim 17, wherein the cancer is an adenocarcinoma.
22. The kit as claimed in claim 17, wherein the cancer is colon adenocarcinoma, ovarian
adenocarcinoma, renal adenocarcinoma, mammary adenocarcinoma, lung adenocarcinoma or
non-seminomal testis carcinoma.
23. The kit as claimed in claim 17, wherein the antibody further comprises a label or
diagnostic imaging agent.
24. The kit as claimed in claim 17, wherein the antibody further comprises barium sulfate,
iocetamic acid, iopanoic acid, ipodate calcium, diatrizoate sodium, diatrizoate meglumine,
metrizamide, tyropanoate sodium, fluorine-18, carbon-11, iodine-123, technitium-99m, iodine-
131, indium-111, fluorine, gadolinium, fluorescein, isothiocyalate, rhodamine, phycoerythrin,
phycocyanin, allophycocyanin, ophthaldehyde, fluorescamine, luminal, isoluminal, luciferin,
luciferase or aequorin.
25. The kit as claimed in claim 17, wherein the antibody comprises a polypeptide having an
amino acid sequence with at least 98% homology to any one of SEQ ID NO:7-35.

26. The kit as claimed in claim 18, wherein the antibody comprises a polypeptide having any
one of SEQ ID NO:7-35 or SEQ ID NO:47-49.
27. The kit as claimed in claim 17, wherein the antibody is encoded by a nucleic acid
comprising any one of SEQ ID NO:36-39.
28. A therapeutic composition comprising an antibody and a pharmaceutically acceptable
carrier, wherein the antibody can bind to a cancer-associated epitope consisting of a complex
between two separate polypeptides, a cytokeratin 8 polypeptide missing the first 65 amino acids
and a cytokeratin 18 polypeptide missing the first 49 amino acids, wherein the cytokeratin 8
polypeptide comprises SEQ ID NO:3 or SEQ ID NO:5, and the cytokeratin 18 polypeptide
comprises SEQ ID NO:4 or SEQ ID NO:6, wherein the antibody is not a COU-I IgM antibody
secreted by a B9165 cell line, and the antibody specifically binds to adenocarcinoma tissue but
has substantially weaker binding to normal epithelial cells and smooth muscle fibers
29. The therapeutic composition as claimed in claims 28, wherein the antibody is shorter than
about 425 amino acids.
30. The therapeutic composition as claimed in claims 28, wherein the antibody is shorter than
about 200 amino acids.
31. The therapeutic composition as claimed in claims 28, wherein the antibody is an IgG
antibody.
32. The therapeutic composition as claimed in claims 28, wherein the antibody can bind to
the cancer-associated epitope in filamentous cytoplasmic structures of adenocarcinoma cells but
in substantially no filamentous structures of normal cells.
33. The therapeutic composition as claimed in claims 28, wherein the antibody can bind to
the cancer-associated epitope in filamentous cytoplasmic structures of colon adenocarcinoma,
ovarian adenocarcinoma, renal adenocarcinoma, mammary adenocarcinoma, lung
adenocarcinoma, pancreatic adenocarcinoma and non-seminomal testis carcinoma cells.

34. The therapeutic composition as claimed in claim 28, wherein the antibody comprises a
polypeptide having an amino acid sequence with at least 98% homology to any one of SEQ ID
NO:7-35.
35. The therapeutic composition as claimed in claim 28, wherein the antibody comprises a
polypeptide having any one of SEQ ID NO.7-35 or SEQ ID NO.47-49.
36. The therapeutic composition as claimed in claim 28, wherein the antibody is encoded by
a nucleic acid comprising any one of SEQ ID NO.36-39.
37. A therapeutic composition for treating adenocarcinoma comprising an inhibitor of a
protease that cleaves Xaa1SR↓Xaa4 (SEQ ID NO:40) and a pharmaceutically acceptable carrier,
wherein Xaa1 is serine, phenylalanine or valine and Xaa4 is serine or valine.

38. The therapeutic composition as claimed in claim 37, wherein the protease is a trypsin-like
protease.
39. The therapeutic composition as claimed in claim 37, wherein the inhibitor is soybean
trypsin inhibitor, alpha-2-macroglobulin, alpha-1-antitrypsin, aprotinin, pancreatic secretory
trypsin inhibitor, corn trypsin inhibitor, pumpkin trypsin inhibitor or human amyloid β-protein
precursor inhibitor.
40. The therapeutic composition as claimed in claim 37, wherein the adenocarcinoma is
colon adenocarcinoma, ovarian adenocarcinoma, renal adenocarcinoma, mammary
adenocarcinoma, lung adenocarcinoma, pancreatic adenocarcinoma or non-seminomal testis
carcinoma.
41. An in vitro method of identifying a mutant binding entity comprising:
fusing a nucleic acid encoding a polypeptide having any one of SEQ ID NO:7-35 to a
nucleic acid encoding a display protein to generate a recombinant nucleic acid encoding a fusion
protein;
mutating the recombinant nucleic acid encoding the fusion protein to generate a mutant
nucleic acid encoding a mutant fusion protein;
expressing the mutant fusion protein; and

selecting a mutant fusion protein that can bind to a cancer-associated epitope comprising
two separate polypeptides, the first polypeptide comprising SEQ ID NO:3 of cytokeratin 8 and
the second polypeptide comprising SEQ ID NO:4 of cytokeratin 18.
42. The method as claimed in claim 41, wherein the binding entity is a CDR or Fab fragment.
43. The method as claimed in claim 41, wherein the display protein is a phage display
protein, retroviral display protein, or a ribosomal display protein.
44. A composition comprising an antibody and a pharmaceutically acceptable carrier,
wherein the antibody can bind with specificity to a cancer-associated epitope consisting of a
fusion protein consisting of two separate polypeptides, a cytokeratin 8 polypeptide missing the
first 65 amino acids and a cytokeratin 18 polypeptide missing the first 49 amino acids, wherein
the antibody specifically binds cytokeratin 8 SEQ ID NO:3 and cytokeratin 18 SEQ ID NO:4 and
wherein the antibody is not a COU-I IgM antibody secreted by a B9165 cell line.
45. The composition of claim 30, wherein the antibody is a Fab, Fab', F(ab')2, Fv, single
chain antibody (scFv) or diabody.
46. The antibody of claim 8, wherein the antibody is a Fab, Fab', F(ab')2, Fv, single
chain antibody (scFv) or diabody.

This invention discloses an isolated cancer-associated epitope comprising recombinant
cytokeratin 8 and cytokeratin 18 polypeptide fragments, wherein the cytokeratin 8
polypeptide is shorter than 482 amino acids and comprises SEQ ID NO:3 or SEQ ID
NO:5, and the cytokeratin 18 polypeptide is shorter than 429 amino acids and comprises
SEQ ID NO:4 or SEQ ID NO:6.

Documents:

924-kolnp-2004-granted-abstract.pdf

924-kolnp-2004-granted-assignment.pdf

924-kolnp-2004-granted-claims.pdf

924-kolnp-2004-granted-correspondence.pdf

924-kolnp-2004-granted-description (complete).pdf

924-kolnp-2004-granted-drawings.pdf

924-kolnp-2004-granted-examination report.pdf

924-kolnp-2004-granted-form 1.pdf

924-kolnp-2004-granted-form 13.pdf

924-kolnp-2004-granted-form 18.pdf

924-kolnp-2004-granted-form 3.pdf

924-kolnp-2004-granted-form 5.pdf

924-kolnp-2004-granted-gpa.pdf

924-kolnp-2004-granted-reply to examination report.pdf

924-kolnp-2004-granted-sequence listing.pdf

924-kolnp-2004-granted-specification.pdf


Patent Number 226798
Indian Patent Application Number 924/KOLNP/2004
PG Journal Number 52/2008
Publication Date 26-Dec-2008
Grant Date 24-Dec-2008
Date of Filing 02-Jul-2004
Name of Patentee THE SCRIPPS RESEARCH INSTITUTE
Applicant Address 10555 NORTH TORREY PINES ROAD, LA JOLLA, CA
Inventors:
# Inventor's Name Inventor's Address
1 DITZEL HENRIK 4411 CAMINITO SANA, #4, SAN DIEGO, CA 92122
2 JENSENIUS JENS C FINSENS ALLE 28, 5320 ODENSE M
PCT International Classification Number C07K 5/00
PCT International Application Number PCT/US03/00297
PCT International Filing date 2003-01-03
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/345,208 2002-01-03 U.S.A.