CA2816437A1 - Antibodies that specifically block the biological activity of kidney associated antigen 1 - Google Patents

Antibodies that specifically block the biological activity of kidney associated antigen 1 Download PDF

Info

Publication number
CA2816437A1
CA2816437A1 CA2816437A CA2816437A CA2816437A1 CA 2816437 A1 CA2816437 A1 CA 2816437A1 CA 2816437 A CA2816437 A CA 2816437A CA 2816437 A CA2816437 A CA 2816437A CA 2816437 A1 CA2816437 A1 CA 2816437A1
Authority
CA
Canada
Prior art keywords
seq
antibody
chain variable
antigen binding
binding fragment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2816437A
Other languages
French (fr)
Inventor
Gilles Bernard Tremblay
Mario Filion
Traian Sulea
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Alethia Biotherapeutics Inc
Original Assignee
National Research Council of Canada
Alethia Biotherapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/CA2009/001586 external-priority patent/WO2010060186A1/en
Application filed by National Research Council of Canada, Alethia Biotherapeutics Inc filed Critical National Research Council of Canada
Priority to CA2816437A priority Critical patent/CA2816437A1/en
Priority claimed from PCT/CA2010/001795 external-priority patent/WO2011054112A1/en
Publication of CA2816437A1 publication Critical patent/CA2816437A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Oncology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Pregnancy & Childbirth (AREA)
  • Reproductive Health (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Novel antibodies that specifically bind to KAAG1 are described. In some embodiments, the antibodies block the biological activity of KAAG1 and are useful in composition in certain cancers, more particularly in cancers that have increased cell surface expression of KAAG1, such as ovarian, renal, lung, colorectal, breast, brain, and prostate cancer, as well as melanoma. The invention also relates to cells expressing the monoclonal antibodies and antigen binding fragments such as humanized and chimeric antibodies. Additionally, methods of detecting and treating cancer using the antibodies and fragments are also disclosed.

Description

2 ANTIBODIES THAT SPECIFICALLY BLOCK THE BIOLOGICAL ACTIVITY OF

FIELD OF THE INVENTION
The present invention relates to specific antibodies and antigen binding fragments thereof that specifically binds to KAAG1 and their use for treating certain diseases including diagnosing, preventing and treating malignant tumors related to ovarian cancer. The present invention also relates to the use of these antibodies for diagnosis, prevention and treatment of various other cancer types.
BACKGROUND OF THE INVENTION
Among gynecologic malignancies, ovarian cancer accounts for the highest tumor-related mortality in women in the United States (Jemal et al., 2005). It is the fourth leading cause of cancer-related death in women in the U.S (Menon et al., 2005). The American Cancer Society estimated a total of 22,220 new cases in 2005 and attributed 16,210 deaths to the disease (Bonome et al., 2005). For the past 30 years, the statistics have remained largely the same - the majority of women who develop ovarian cancer will die of this disease (Chambers and Vanderhyden, 2006). The disease carries a 1:70 lifetime risk and a mortality rate of >60% (Chambers and Vanderhyden, 2006). The high mortality rate is due to the difficulties with the early detection of ovarian cancer when the malignancy has already spread beyond the ovary. Indeed, >80% of patients are diagnosed with advanced staged disease (stage III or IV) (Bonome et at., 2005). These patients have a poor prognosis that is reflected in <45% 5-year survival rate, although 80% to 90% will initially respond to chemotherapy (Berek et al., 2000). This increased success compared to 20% 5-year survival rate years earlier is, at least in part, due to the ability to optimally debulk tumor tissue when it is confined to the ovaries, which is a significant prognostic factor for ovarian cancer (Bristow R. E., 2000; Brown et al., 2004). In patients who are diagnosed with early disease (stage l), the 5-yr survival ranges from >90 (Chambers and Vanderhyden, 2006).
Ovarian cancer comprises a heterogeneous group of tumors that are derived from the surface epithelium of the ovary or from surface inclusions. They are classified into serous, mucinous, endometrioid, clear cell, and Brenner (transitional) types corresponding to the different types of epithelia in the organs of the female reproductive tract (Shih and Kurman, 2005). Of these, serous tumors account for ¨60% of the ovarian cancer cases diagnosed. Each histologic subcategory is further divided into three groups: benign, intermediate (borderline tumor or low malignancy potential (LMP)), and malignant, reflecting their clinical behavior (Seidman et al., 2002). LMP represents 10% to 15% of tumors diagnosed as serous and is a conundrum as they display atypical nuclear structure and metastatic behavior, yet they are considerably less aggressive than high-grade serous tumors. The 5-year survival for patients with LMP tumors is 95% in contrast to a <45% survival for advanced high-grade disease over the same period (Berek et al., 2000).
Presently, the diagnosis of ovarian cancer is accomplished, in part, through routine analysis of the medical history of patients and by performing physical, ultrasound and x-ray examinations, and hematological screening. Two alternative strategies have been reported for early hematological detection of serum biomarkers. One approach is the analysis of serum samples by mass spectrometry to find proteins or protein fragments of unknown identity that detect the presence or absence of cancer (Mor et al., 2005; Kozak et al., 2003). However, this strategy is expensive and not broadly available. Alternatively, the presence or absence of known proteins/peptides in the serum is being detected using antibody microarrays, ELISA, or other similar approaches. Serum testing for a protein biomarker called CA-125 (cancer antigen-125) has long been widely performed as a marker for ovarian cancer. However, although ovarian cancer cells may produce an excess of these protein molecules, there are some other cancers, including cancer of the fallopian tube or endometrial cancer (cancer of the lining of the uterus), 60% of people with pancreatic cancer, and 20%-25% of people with other malignancies with elevated levels of CA-125. The CA-125 test only returns a true positive result for about 50% of Stage I ovarian cancer patients and has a 80% chance of returning true positive results from stage II, Ill, and IV ovarian cancer patients. The other 20% of ovarian cancer patients do not show any increase in CA-125 concentrations. In addition, an elevated CA-125 test may indicate other benign activity not associated with cancer, such as menstruation, pregnancy, or endometriosis. Consequently, this test has very limited clinical application for the detection of early stage disease when it is still treatable, exhibiting a positive predictive value (PPV) of <10%. Even with the addition of ultrasound screening to CA-125, the PPV only improves to around 20% (Kozak et al., 2003). Thus, this test is not an effective screening test.
Despite improved knowledge of the etiology of the disease, aggressive cytoreductive surgery, and modern combination chemotherapy, there has been only little change in mortality. Poor outcomes have been attributed to (1) lack of adequate screening tests for early disease detection in combination with only subtle presentation of symptoms at this stage - diagnosis is frequently being made only after progression to later stages, at which point the peritoneal dissemination of the cancer limits effective treatment and (2) the frequent development of resistance to standard chemotherapeutic strategies limiting improvement in the 5-year survival rate of patients. The initial chemotherapy regimen for ovarian cancer includes the combination of carboplatin (Paraplatin) and paclitaxel (taxol). Years of clinical trials have proved this combination to be most effective after effective surgery -reduces tumor volume in about 80% of the women with newly diagnosed ovarian cancer and 40% to 50% will have complete regression - but studies continue to look for ways to improve it. Recent abdominal infusion of chemotherapeutics to target hard-to-reach cells in combination with intravenous delivery has increased the effectiveness.
However, severe side effects often lead to an incomplete course of treatment.
Some other chemotherapeutic agents include doxorubicin, cisplatin, cyclophosphamide, bleomycin, etoposide, vinblastine, topotecan hydrochloride, ifosfamide, 5-fluorouracil and melphalan. More recently, clinical trials have demonstrated that intraperitoneal administration of cisplatin confers a survival advantage compared to systemic intravenous chemotherapy (Cannistra and McGuire, 2007). The excellent survival rates for women with early stage disease receiving chemotherapy provide a strong rationale for research efforts to develop strategies to improve the detection of ovarian cancer. Furthermore, the discovery of new ovarian cancer-related biomarkers will lead to the development of more effective therapeutic strategies with minimal side effects for the future treatment of ovarian cancer.
Notwithstanding these recent advances in the understanding and the treatment for ovarian cancer, the use of chemotherapy is invariably associated with severe adverse reactions, which limit their use. Consequently, the need for more specific strategies such as combining antigen tissue specificity with the selectivity of monoclonal antibodies should permit a significant reduction in off-target-associated side effects.
The use of monoclonal antibodies for the therapy of ovarian cancer is beginning to emerge with an increasing number of ongoing clinical trials (Oei et al., 2008;
Nicodennus and berek, 2005). Most of these trials have examined the use of monoclonal antibodies conjugated to radioisotopes, such as yttrium-90, or antibodies that target tumor antigens already identified in other cancer types. An example of this is the use of bevacizunnab, which targets vascular endothelial growth factor (Burger, 2007). There are very few ovarian cancer specific antigens that are currently under investigation as therapeutic targets for monoclonal antibodies. Some examples include the use of a protein termed B7-H4 (Simon et al., 2006) and more recently
3 folate receptor-alpha (Ebel et al., 2007), the latter of which has recently entered Phase II clinical trials.
Kidney associated antigen 1 (KAAG1) was originally cloned from a cDNA library derived from a histocompatibility leukocyte antigen-B7 renal carcinoma cell line as an antigenic peptide presented to cytotoxic T lymphocytes (Van den Eynde et al., 1999;
Genebank accesssion no. Q9UBP8). The locus containing KAAG1 was found to encode two genes transcribed in both directions on opposite strands. The sense strand was found to encode a transcript that encodes a protein termed DCDC2.
Expression studies by these authors found that the KAAG1 antisense transcript was tumor specific and exhibited very little expression in normal tissues whereas the DCDC2 sense transcript was ubiquitously expressed (Van den Eynde et al., 1999).
The expression of the KAAG1 transcript in cancer, and in particular ovarian cancer, renal cancer, lung cancer, colon cancer, breast cancer and melanoma was disclosed in the published patent application No. PCT/CA2007/001134. Van den Eynde et al., also observed RNA expression in renal carcinomas, colorectal carcinomas, melanomas, sarcomas, leukemias, brain tumors, thyroid tumors, mammary carcinomas, prostatic carcinomas, oesophageal carcinomas, bladder tumor, lung carcinomas and head and neck tumors. Recently, strong genetic evidence obtained through linkage disequilibrium studies found that the VMP/DCDC2/KAAG1 locus was associated with dyslexia (Schumacher et al., 2006; Cope et al., 2005). One of these reports pointed to the DCDC2 marker as the culprit in dyslexic patients since the function of this protein in cortical neuron migration was in accordance with symptoms of these patients who often display abnormal neuronal migration and maturation (Schumacher et al., 2006).
SUMMARY OF THE INVENTION
This invention relates to the expression of KAAG1 in tumor cells. The invention also relates to specific anti-KAAG1 antibodies and antigen binding fragments as well as kits useful for the treatment, detection and diagnosis of cancer. The antibodies and antigen binding fragments may more particularly be useful for the treatment, detection and diagnosis of cancer where tumor cells expresses KAAG1 or a KAAG1 variant.
Exemplary embodiments of such cancer includes, for example, ovarian cancer, skin cancer, renal cancer, colorectal cancer, sarcoma, leukemia, brain cancer, cancer of the thyroid, breast cancer, prostate cancer, cancer of the oesophagus, bladder cancer, lung cancer and head and neck cancer.
4 The present invention provides in one aspect thereof, an isolated or substantially purified antibody or antigen binding fragment which may be capable of specific binding to Kidney associated antigen 1 (KAAG1 defined in SEQ ID NO.:2) or to a KAAG1 variant.
In accordance with an embodiment of the invention, the antibody or antigen binding fragment may be capable of binding to an epitope having an amino acid sequence which is identical to at least 10 (e.g., 10 to 20 or more) consecutive amino acids of KAAG1. The 10 consecutive amino acids of KAAG1 may be domain located between amino acid 30 and amino acid 84 of KAAG1, such as, for example, between amino acids 1 to 35, 36 to 60 or 61 to 84 of KAAG1.
More specifically and in accordance with an embodiment of the invention, the antibody or antigen binding fragment may bind to a domain located between amino acid 30 and amino acid 84 of KAAG1.
In accordance with another embodiment of the invention, the antibody or antigen binding fragment may be capable of binding to an epitope comprised within amino acid 1 to 35 of KAAG1.
In accordance with a further embodiment of the invention, the antibody or antigen binding fragment may be capable of binding to an epitope comprised within amino acid 36 to 60 of KAAG1.
In accordance with yet a further embodiment of the invention, the antibody or antigen binding fragment may be capable of binding to an epitope comprised within amino acid 61 to 84 of KAAG1.
The antibody or antigen binding fragment of the present invention is especially capable of specific binding to a secreted form of KAAG1, i.e., a form of KAAG1 where the signal peptide has been cleaved.
The antibody or antigen binding fragment of the present invention is especially capable of binding to the extracellular region of KAAG1.
As such, the present invention encompasses diagnostic and/or therapeutic antibodies or antigen binding fragments having specificity for a secreted form of KAAG1 or for an extracellular region of KAAG1. Also encompassed by the present invention are antibodies or antigen binding fragments having the same epitope specificity as the antibody of the present invention. A candidate antibody may be identified by
5 determining whether it will bind to the epitope to which the antibodies described herein binds and/or by performing competition assays with antibodies or antigen binding fragments known to bind to the epitope.
Therefore another aspect the present invention provides an isolated antibody or antigen binding fragment capable of competing with the antibody or antigen binding fragment described herein.
Isolated antibodies or antigen binding fragments of the present invention include those which may be capable of inducing killing (elimination, destruction, lysis) of KAAG1-expressing tumor cells or KAAG1 variant-expressing tumor cells (e.g., in an ADCC-dependent manner).
Isolated antibodies or antigen binding fragments of the present invention also include those which are characterized by their ability to reduce spreading of tumor cells expressing KAAG1 or a variant thereof and also those which are characterized by their ability to decrease or impair formation of tumors expressing KAAG1 or a variant thereof.
The antibodies or antigen binding fragments may be particularly effective when KAAG1 or KAAG1 variant is expressed at the surface of the KAAG1-expressing tumor cells and may be particularly useful in targeting tumor cells expressing or a KAAG1 variant and characterized by anchorage-independent growth.
The invention relates to specific antibodies or antigen binding fragments having the amino acid sequences described herein. Such antibodies or antigen binding fragments may be in the form of monoclonal antibodies, polyclonal antibodies, chimeric antibodies, humanized antibodies and human antibodies (isolated) as well as antigen binding fragments having the characteristics described herein.
Antibodies or antigen binding fragments encompassing permutations of the light and/or heavy chains between a monoclonal, chimeric, humanized or human antibody are also encompassed herewith.
The antibodies or antigen binding fragments of the present invention may thus comprise amino acids of a human constant region and/or framework amino acids of a human antibody.
The term "antibody or antigen binding fragment" or similar terms such as "antibodies and antigen binding fragments" encompasses, for example "variant antibody or
6 antigen binding fragment" such as, for example, "humanized antibody or antigen binding fragment".
The term "antibody" refers to intact antibody, monoclonal or polyclonal antibodies.
The term "antibody" also encompasses multispecific antibodies such as bispecific antibodies. Human antibodies are usually made of two light chains and two heavy chains each comprising variable regions and constant regions. The light chain variable region comprises 3 CDRs, identified herein as CDRL1, CDRL2 and CDRL3 flanked by framework regions. The heavy chain variable region comprises 3 CDRs, identified herein as CDRH1, CDRH2 and CDRH3 flanked by framework regions.
The term "antigen-binding fragment", as used herein, refers to one or more fragments of an antibody that retain the ability to bind to an antigen (e.g., KAAG1, secreted form of KAAG1 or variants thereof). It has been shown that the antigen-binding function of an antibody can be performed by fragments of an intact antibody. Examples of binding fragments encompassed within the term "antigen-binding fragment" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHi domains; (ii) a F(abt)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHi domains; (iv) a Fv fragment consisting of the VL
and VH
domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR), e.g., VH CDR3. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single polypeptide chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al.
(1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci.
USA
85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antigen-binding fragment" of an antibody. Furthermore, the antigen-binding fragments include binding-domain immunoglobulin fusion proteins comprising (i) a binding domain polypeptide (such as a heavy chain variable region, a light chain variable region, or a heavy chain variable region fused to a light chain variable region via a linker peptide) that is fused to an immunoglobulin hinge region polypeptide, (ii) an immunoglobulin heavy chain CH2 constant region fused to the hinge region, and (iii) an immunoglobulin heavy chain CH3 constant region fused to the CH2 constant region. The hinge region may be modified by replacing one or more cysteine residues
7 with serine residues so as to prevent dimerization. Such binding-domain immunoglobulin fusion proteins are further disclosed in US 2003/0118592 and US

2003/0133939. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
A typical antigen binding site is comprised of the variable regions formed by the pairing of a light chain immunoglobulin and a heavy chain immunoglobulin. The structure of the antibody variable regions is very consistent and exhibits very similar structures. These variable regions are typically comprised of relatively homologous framework regions (FR) interspaced with three hypervariable regions termed Complementarity Determining Regions (CDRs). The overall binding activity of the antigen binding fragment is often dictated by the sequence of the CDRs. The FRs often play a role in the proper positioning and alignment in three dimensions of the CDRs for optimal antigen binding.
Antibodies and/or antigen binding fragments of the present invention may originate, for example, from a mouse, a rat or any other mammal or from other sources such as through recombinant DNA technologies.
Further scope, applicability and advantages of the present invention will become apparent from the non-restrictive detailed description given hereinafter. It should be understood, however, that this detailed description, while indicating exemplary embodiments of the invention, is given by way of example only, with reference to the accompanying drawings.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1A shows the expression profiling analyses using semi-quantitative RT-PCR
reactions carried out to measure the level of KAAG1 mRNA expression in RNA
samples derived from greater than 20 ovarian tumors, benign (low malignancy potential) tumors, ovarian cancer cell lines, and 30 normal tissues. The control panels show GAPDH expression, a house-keeping gene used to compare the amount of starting material in each RT-PCR reaction.
Figure 1B shows semi-quantitative RT-PCR experiments demonstrating that KAAG1 mRNA is expressed in ovarian cancer cell lines, in particular those that are derived from ascites.
8 Figure 10 shows a diagram illustrating the ability of ovarian cancer cell lines to form 3D structures called spheroids. The left panels show the cells grown in medium lacking serum whereas 5% serum stimulated the formation of the spheroid structures.
Figure 1D shows semi-quantitative RT-PCR experiments demonstrating that the KAAG1 mRNA is highly induced during the formation of spheroids in ovarian cancer cell lines.
Figure 2A shows a diagram illustrating the wound or scratch assay, a cell-based assay that is a measurement of a cell line's ability to migrate into a denuded area over a pre-determined period of time. TOV-21G cells harboring KAAG1 shRNAs display a reduced capacity to fill in the denuded area.
Figure 2B shows an illustration of the clonogenic assay, also known as a colony survival assay. It measured the survival of diluted cells over a period of several days.
TOV-21G cells harboring KAAG1 shRNAs display reduced survival.
Figure 3A shows a polyacrylamide gel that was stained with Coomassie Blue and contains a sample (10 jig) of purified Fc-KAAG1 fusion protein that was produced in transiently transfected 293E cells.
Figure 3B shows the results of an ELISA of one of the 96-well plates containing individual monoclonal antibodies selected from Omniclonal library #3 containing anti-KAAG1 Fabs. The results showed that 48 (highlighted in grey) of the Fabs interacted very efficiently with KAAG1. The wells indicated by bold numbers contained the exemplary monoclonals 3D3, 3G10, and 304.
Figure 4A shows a polyacrylamide gel that was stained with Coomassie Blue and contains a sample (10 jig) of purified Fc-KAAG1 fusion protein (lane 1), a truncated mutant of KAAG1 spanning amino acids 1-60 (lane 2), and another truncated mutant of KAAG1 spanning amino acids 1-35 (lane 3) that were produced in transiently transfected 293E cells. All proteins were Fc fusion proteins.
Figure 4B is a scheme that illustrates the truncated mutants of KAAG1 that were generated for the epitope mapping studies.
Figure 40 shows a drawing that describes the results from ELISA analyses to map the epitopes that are bound by the anti-KAAG1 antibodies contained in Omniclonal library #3. The results showed that the majority of monoclonals interact with central
9 region of KAAG1 and that certain antibodies bound to the amino- or carboxyl-termini of KAAG1.
Figure 5 presents a scheme that illustrates the steps involved to convert the mouse Fabs into IgG1 mouse-human chimeric mAbs.
Figure 6 shows drawings that compare the binding of the mouse anti-KAAG1 Fabs with the binding of the corresponding IgG1 chimeric monoclonal antibodies for exemplary antibodies 3D3, 3G10, and 3C4. The results indicate that the relative binding of the Fab variable regions was maintained when transferred to a full human IgG1 scaffold.
Figure 7 shows depictions of spheroid formation experiments using TOV-21G and OV-90 ovarian cancer cell lines in the presence of chimeric IgG1 anti-KAAG1 monoclonal antibodies. Loosely packed structures are indicative of less invasive cancer cell lines. The results show spheroids treated with the exemplary anti-antibodies 3D3, 3G10, or 304.
Figure 8A shows a scan of a tissue microarray containing approximately 70 biopsy samples obtained from ovarian tumor patients. The samples were blotted with the 3D3 anti-KAAG1 antibody and showed that the vast majority of ovarian tumors expressed very high level of KAAG1 antigen.
Figure 8B a higher magnification picture from the tissue microarray experiment. The arrows show the membrane localization of KAAG1 at the apical surface of the epithelial layer of cells in serous ovarian tumors.
Figure 80 illustrates other immunohistochemical studies that demonstrate that KAAG1 is highly expressed in all ovarian cancer types. The histotypes shown are serous, mucinous and endometroid.
Figure 9A, 9B and 9C is a summary of alignment results obtained for selected CDRL1, CDRL2 or CDRL3 sequences using the ClustalW2 program; where " * "
means that the residues in that column are identical in all sequences in the alignment, means that conserved substitutions have been observed and " . " means that semi-conserved substitutions are observed. Consensus CDRs were generated using the ClustalW program (Larkin M.A., et al., (2007) ClustalW and ClustaIX
version 2.
Bioinformatics 2007 23(21): 2947-2948).

Figure 10A, 10B and 100 is a summary of alignment results obtained for selected CDRH1, CDRH2 or CDRH3 sequences using the ClustalW2 program; where " * "
means that the residues in that column are identical in all sequences in the alignment, " : " means that conserved substitutions have been observed and " . " means that semi-conserved substitutions are observed. Consensus CDRs were generated using the ClustalW program (Larkin M.A., et al., (2007) ClustalW and ClustaIX
version 2.
Bioinformatics 2007 23(21): 2947-2948).
Figure 11 represents sequence comparison between each of the light chain variable regions generated and representative light chain variable regions identified in SEQ ID
NOs:16, 20, 24 or 105. Percent sequence identity and percent sequence similarity has been determined unsing Blast2 sequence program as indicated herein.
Figure 12 represents sequence comparison between each of the heavy chain variable regions generated and representative heavy chain variable regions identified in SEQ
ID NOs:18, 22, 26 or 132. Percent sequence identity and percent sequence similarity has been determined unsing Blast2 sequence program as indicated herein.
Figure 13 An IgGi antibody that targets KAAG1 can efficiently mediate ADCC
activity in vitro. PBMNCs (AllCells, LLC, Emoryville, CA) were incubated with 3D3 for 30 min and mixed with either OVCAR-3 or WIL2-S cells at a ratio of 1:25. The cells were incubated for 4 h at 37 C and cell lysis was determined by measuring LDH
levels in the medium. Cell cytotoxicity was calculated as follows: % cytotoxicity =
(experimental ¨ effector spontaneous ¨ target spontaneous) x 100 / (target maximum ¨ target spontaneous).
Figure 14 Anti-KAAG1 mAbs prevent the spread of TOV-112D ovarian tumors in vivo. 1 x 106 cells were implanted in the peritoneal cavity of SCID mice in a volume of 200 1.11_. Treatment with either PBS or antibodies diluted in PBS was performed 2 days later at a dose of 25 mg/kg qwk. The mice were sacrificed as soon as the tumors were detected by palpation of the abdomen. The number of tumors were scored visually (B) and the data in panel A is expressed as the average number of tumors/mouse SE.
Figure 15 shows immunohistochemistry performed with an anti-KAAG1 antibody on human skin tumor tissue microarrays (Pantomics Inc., Richmond, CA) of several sections isolated from squamous cell carcinomas and melanomas.

Figure 16 illustrates spheroid formation of melanoma cell lines (A375 and SK-MEL5) and of renal cell carcinoma cell lines (A498 and 786-0) in the presence or absence of the chimeric 3D3 antibody.
Figure 17A represents graphs illustrating the binding of increasing concentrations of the 304, 3D3 and 3G10 antibodies to cell lines (OV-90, TOV-21G and SKOV-3) fixed under condition that do not permeate the cells.
Figure 17B is a graph illustrating the results of flow cytometry performed on cell line with the 3D3 antibody.
Figure 18A is a schematic illustrating the structure of the 3D3 antibody model.
Figure 18B is a schematic illustrating the structure of the 3C4 antibody model.
Figure 19A is a graph illustrating the binding of increasing concentration of the humanized 3D3 antibody in comparison with the chimeric 3D3 antibody to recombinant KAAG1.
Figure 19B is a table summarizing the kinetics parameters of the humanized 3D3 antibody, the chimeric 3D3 antibody as well as hybrid antibodies encompassing permutations of the light and heavy chains of the chimeric or humanized antibody.
Figure 190 illustrates spheroid formation of SKOV-3 ovarian cancer cells in the presence of the humanized 3D3 antibody, chimeric 3D3 antibody or in the presence of a buffer or a control IgG.
Figure 20A represents sequence alignment of the monoclonal 3D3 light chain variable region (SEQ ID N0.:16) and the humanized 3D3 light chain variable region (SEQ ID N0.:178). The humanized 303 light chain variable region is 86%
identical (94% sequence similarity) to the monoclonal 3D3 light chain variable region and their three CDRs are 100% (indicated in bold).
Figure 20B represents sequence alignment of the monoclonal 3D3 heavy chain variable region (SEQ ID N0.:18) and the humanized 3D3 heavy chain variable region (SEQ ID N0.:179). The humanized 3D3 heavy chain variable region is 82%
identical (91% sequence similarity) to the monoclonal 3D3 heavy chain variable region and their three CDRs are 100% (indicated in bold).
Figure 21A represents sequence alignment of the monoclonal 3C4 light chain variable region (SEQ ID NO. :24) and the humanized 304 light chain variable region (SEQ ID NO.:182). The humanized 304 light chain variable region is 85%
identical (93% sequence similarity) to the monoclonal 3C4 light chain variable region and their three CDRs are 100% (indicated in bold).
Figure 21B represents sequence alignment of the monoclonal 304 heavy chain variable region (SEQ ID NO.:26) and the humanized 304 heavy chain variable region (SEQ ID NO.:183). The humanized 3C4 heavy chain variable region is 86%
identical (93% sequence similarity) to the monoclonal 3C4 heavy chain variable region and their three CDRs are 100% (indicated in bold).
Figure 22 shows the expression of the KAAG1 antigen on the surface of the ovarian cancer cell line OVCAR-3 as measured by immunofluorescence. The cells were stained with the chimeric 3D3 antibody followed by visualization with a fluorescently labelled secondary antibody. The cell surface expression was confirmed with the co-localization of an known surface protein, E-cadherin.
Figure 23 represents the detection of the KAAG1 antigen on the surface of SKOV-cells by flow cytometry. The fluorescent signal decreases with time when the cells were incubated at 37 C, which suggests that the KAAG1/antibody complex was internalized during the incubation.
Figure 24 shows that the KAAG1 antigen that was detected by the chimeric 3D3 antibody gets internalized soon after the complex is formed. Punctate pen-nuclear staining was observed at 30 minutes of incubation at 37 C which was consistent with the complex following an endosomal pathway.
DETAILED DESCRIPTION OF THE INVENTION
The expression and biological activity of KAAG1 in cancer cells The present invention relates to the use of antibodies to target tumors found in various cancer types, in particular ovarian cancer. In order to direct the antibodies to the tumors, the identification of tumor-specific antigens that are expressed at the cell surface of the cancer cells must be carried out. There are several technologies that are available to identify tumor-specific antigens and the method that was used to identify KAAG1 in ovarian tumors, an innovative discovery platform called Subtractive Transcription-based Amplification of mRNA (STAR), is described in the published patent application No. PCT/CA2007/001134.
Analysis of the ovarian cancer STAR libraries yielded many genes that encode secreted and cell surface proteins. One of these, termed AB-0447, contained an open reading frame that encoded a polypeptide of 84 amino acids, corresponding to SEQ

ID NO. :2 that was encoded by a cDNA of 885 base pairs with the nucleotide sequence shown in SEQ ID NO.:1. A search of publicly available databases revealed that the AB-0447 nucleotide sequence was identical to that of a gene called KAAG1.
Bioinformatic analysis predicted a membrane-anchored protein that presents its functional domain to the extracellular compartment. KAAG1 was originally cloned from a kidney cancer library as a cell surface antigen, a result that confirms its membrane localization. Additionally, our studies showed that the protein was processed at its amino-terminus, a result that was consistent with cleavage of a functional signal peptide at or between amino acids 30 and 34. Furthermore, transient expression of the full-length cDNA resulted in detection of cleaved KAAG1 in the culture medium. This last finding indicated that this membrane-anchored protein could be shed from the cells when expressed at high levels. In contrast, expression of an amino-truncated mutant of KAAG1 resulted in intra-cellular retention of the protein.
There are currently no published reports that shed any light on its function and the over-expression of KAAG1 in ovarian cancer, as disclosed by this invention, has never been previously documented.
We have thus investigated whether KAAG1 could be used for antibody-based diagnostics and therapeutics.
Several ovarian cancer cell-based models have been established, such as TOV-21G, TOV-112D, OV-90, and others, and are familiar to those skilled in the art.
These cells are part of a collection of human ovarian cancer cell lines derived from patients with ovarian tumors or ascites fluid. These cell lines have undergone an in-depth analysis, including global gene expression patterns on microarrays that make them excellent cell-based models for human ovarian cancer. The growth properties, gene expression patterns, and response to chemotherapeutic drugs indicated that these cell lines are very representative of ovarian tumor behavior in vivo (Benoit et al., 2007).
RT-PCR
analysis of total RNA isolated from these ovarian cancer cell lines showed that the KAAG1 transcript was weakly expressed in the cell lines derived from primary tumors.
In contrast, cell lines derived from ascitic fluid contained high levels of expression. The increased expression of KAAG1 in cells from the ascitic fluid suggested that the environment of the cells influences the regulation of the gene. Ascitic cells are associated with advanced disease and this pattern of expression implies that increased KAAG1 levels are associated with anchorage-independent growth. In concordance with this latter suggestion, KAAG1 expression was found to significantly increase in cell lines derived from primary tumors when these cells were cultured as spheroids in 3D cultures. These spheroids have been extensively characterized and were found to display many properties associated with tumors in vivo (Cody et al., 2008). Thus, expression of KAAG1 was found to be significantly increased in models that mimic tumor progression, in particular during the evolution of ovarian cancer.
With the demonstration that KAAG1 expression is regulated in ovarian cancer cells, the function of this gene in ovarian cancer cell behavior was examined in cell-based assays. To that effect, RNA interference (RNAi) was used to knock down the expression of the endogenous KAAG1 gene in the ovarian cancer cell lines and it was found that decreased expression of KAAG1 resulted in a significant reduction in the migration of the cells as determined in a standard cell motility assay, as exemplified by a wound healing (or scratch) assay. This type of assay measures the speed at which cells fill a denuded area in a confluent monolayer. Decreased expression of KAAG1 resulted in a reduction in the survival of ovarian cancer cell lines as measured by a clonogenic assay, such as a colony survival assay. Those skilled in the art may use other methods to evaluate the requirement of KAAG1 in the behavior of cancer cells, in particular ovarian cancer cells.
Based on the expression of KAAG1 in a large proportion of ovarian tumors, its limited expression in normal tissues, and a concordance between expression levels and increased malignancy, and a putative biological role for KAAG1 in the behavior of ovarian cancer cell lines, KAAG1 was chosen as a therapeutic target for the development of antibodies for the detection, prevention, and treatment of ovarian cancer. Expression of KAAG1 in cancer, other than ovarian cancer also lead the Applicant to the evaluation of therapeutic or diagnostic antibodies for other cancer indications.
Therefore, a variety of anti-KAAG1 antibodies and antigen binding fragments thereof, such as monoclonal antibodies, polyclonal antibodies, chimeric and humanized antibodies (including humanized monoclonal antibodies), antibody fragments, single chain antibodies, domain antibodies, and polypeptides with an antigen binding region, useful for targeting KAAG1 or a KAAG1 variant are provided.
KAAG1 as antigen and epitopes derived from KAAG1 The Applicant has come to the unexpected discovery that KAAG1 is expressed in several tumor types and is also found in blood and in ascitic fluid of patients. This antigen may thus be useful for targeting tumor cells expressing the antigen in vivo and in the development of detection assays for measuring the tumor associated antigen in vitro or in vivo. The KAAG1 antigen circulating in blood lacks the signal peptide.
The present invention therefore provides a KAAG1 antigen useful for generating antibodies specific for the circulating form of KAAG1 and/or specific for tumor-expressed KAAG1. The KAAG1 antigen (i.e., epitope) may comprise a fragment of at least 10 amino acids (and up to 84 amino acids) of KAAG1 and may especially correspond to the extracellular region of KAAG1.
The antigen may be made by recombinant DNA technology, by chemical synthesis, by isolation of the KAAG1 protein cells, by proteolytic digestion of a KAAG1 protein (e.g., isolated, recombinant, etc.) or by any other methods known in the art.
An exemplary antigen is the whole KAAG1 protein or a variant form having at least 80% sequence identity with SEQ ID NO.:2 or a fragment thereof.
Another exemplary antigen derived from KAAG1 is the secreted or circulating form of KAAG1 which lacks the signal peptide or the extracellular region of KAAG1.
This antigen may more particularly lack amino acids 1 to 25, 1 to 26, 1 to 27, 1 to 28, 1 to 29, 1 to 30, 1 to 31, 1 to 32, 1 to 33, 1 to 34, 1 to 35 or 1 to 36 of KAAG1.
The antigen or the epitope described herein may be fused with a carrier such as keyhole limpet (KHL), bovine serum albumin (BSA), ovalbumin (OVA) or else in order to generate antibodies and antigen binding fragments.
The present invention also provides an epitope of at least 10 amino acids comprised within amino acid 1 to 35 of SEQ ID NO.:2, within amino acid 36 to 60 of SEQ
ID
NO.:2 or within amino acid 61 to 84 of SEQ ID NO.:2 to generate antibodies and = 25 antigen binding fragments described herein. The present invention further provides a composition for generating antibodies to a secreted or circulating form of KAAG1 or to an extracellular region of KAAG1, the composition may comprise an epitope of KAAG1 comprised within amino acids 30 to 84 of SEQ ID NO.:2 and a carrier. The epitope may especially comprise at least 10 amino acids of KAAG1.
Exemplary embodiments of compositions are pharmaceutical composition for generating antibodies to a secreted or circulating form of KAAG1 or to the extracellular region of KAAG1. The pharmaceutical composition may comprise an epitope of KAAG1 comprised within amino acids 30 to 84 of SEQ ID NO.:2 and a pharmaceutically acceptable carrier.
In yet a further aspect the invention provides a method for generating antibodies to a secreted or circulating form of KAAG1. The method may comprise administering a polypeptide comprising an epitope of KAAG1 comprised within amino acids 30 to of SEQ ID NO.:2 wherein the epitope lacks a KAAG1 signal peptide.
Alternatively, the method may comprise administering an epitope which comprises the signal peptide and selecting antibodies which only binds to the secreted form or the extracellular region of the protein.
In an additional aspect, the present invention provides the use of an epitope of KAAG1 comprised within amino acids 30 to 84 of SEQ ID NO.:2 for generating antibodies to a secreted or circulating form of KAAG1.
Antibodies and antigen binding fragments that binds to KAAG1 Antibodies were initially isolated from Fab librairies for their specificity towards the antigen of interest. Comparison of the amino acid sequences of the light chain variable domains or the heavy chain variable domains of antibodies showing the greatest characteristics allowed us to derive consensus sequences within the CDRs and within the variable regions. The consensus for CDRs are provided in SEQ ID
Nos: 74 to 90.
The variable regions described herein may be fused with constant regions of a desired species thereby allowing recognition of the antibody by effector cells of the desired species. The constant region may originate, for example, from an IgG1, IgG2, IgG3, or IgG4 subtype. Cloning or synthesizing a constant region in frame with a variable region is well within the scope of a person of skill in the art and may be performed, for example, by recombinant DNA technology.
In certain embodiments of the present invention, antibodies that bind to KAAG1 may be of the IgG1, IgG2, IgG3, or IgG4 subtype. More specific embodiments of the invention relates to an antibody of the IgG1 subtype. The antibody may be a humanized antibody of the IgG1 subtype that is biologically active in mediating antibody-dependent cellular cytotoxicity (ADCC), complement-mediated cytotoxicity (CMC), or associated with immune complexes. The typical ADCC involves activation of natural killer (NK) cells and is reliant on the recognition of antibody-coated cells by Fc receptors on the surface of the NK cells. The Fc receptors recognize the Fc domain of antibodies such as is present on IgG1, which bind to the surface of a target cell, in particular a cancerous cell that expresses an antigen, such as KAAG1.
Once bound to the Fc receptor of IgG the NK cell releases cytokines and cytotoxic granules that enter the target cell and promote cell death by triggering apoptosis.
In some instances, anti-KAAG1 antibodies with substantially identical light and heavy chain variable regions to antibody 3D3, will interact with an epitope spanned by amino acids 36¨ 60, inclusively, of KAAG1. In other instances, anti-KAAG1 antibodies with substantially identical light and heavy chain variable regions to antibody 3G10, will interact with an epitope spanned by amino acids 61 ¨84, inclusively, of KAAG1.
In yet another instance, anti-KAAG1 antibodies with substantially identical light and heavy chain variable regions to antibody 3C4 will interact with an epitope spanned by amino acids 1 ¨ 35, inclusively, of KAAG1.
The present invention described a collection of antibodies that bind to KAAG1 or to a KAAG1 variant. In certain embodiments, the antibodies may be selected from the group consisting of polyclonal antibodies, monoclonal antibodies such as chimeric or humanized antibodies, antibody fragments such as antigen binding fragments, single chain antibodies, domain antibodies, and polypeptides with an antigen binding region.
In an aspect of the invention, the isolated antibody or antigen binding fragment of the present invention may be capable of inducing killing (elimination, destruction, lysis) of KAAG1-expressing tumor cells or KAAG1 variant-expressing tumor cells (e.g., in an ADCC-dependent manner).
In a further aspect of the invention, the isolated antibody or antigen binding fragment of the present invention may especially be characterized by its capacity of reducing spreading of tumor cells expressing KAAG1 or a KAAG1 variant.
In an additional aspect of the invention, the isolated antibody or antigen binding fragment of the present invention may be characterized by its capacity of decreasing or impairing formation of tumors expressing KAAG1 or a KAAG1 variant.
In accordance with an embodiment of the invention, the antibody or antigen binding fragment may be more particularly effective when KAAG1 is expressed at the surface of the tumor cells expressing KAAG1 or a KAAG1 variant.

Also in accordance with the present invention, the antibody or antigen binding fragment may be especially useful in targeting tumor cells expressing KAAG1 or a KAAG1 variant which are characterized by anchorage-independent growth.
In a further aspect, the present invention relates to an isolated antibody or antigen binding fragment for use in the treatment of cancer comprising tumor cells expressing KAAG1 or a KAAG1 variant.
In yet a further aspect, the present invention relates to an isolated antibody or antigen binding fragment for use in the detection of cancer comprising tumor cells expressing KAAG1 or a KAAG1 variant.
In an exemplary embodiment of the invention, the isolated antibody or antigen binding fragment may comprise amino acids of a constant region, which may originate, for example, from a human antibody.
In another exemplary embodiment of the invention, the isolated antibody or antigen binding fragment may comprise framework amino acids of a human antibody.
Without being limited to the exemplary embodiments presented herein, the Applicant as generated specific antibodies and antigen binding fragments which may be useful for the purposes described herein.
The present invention therefore provides in an exemplary embodiment, an isolated antibody or antigen binding fragment comprising a light chain variable domain having;
a. a CDRL1 sequence selected from the group consisting of SEQ ID
NO.:74 and SEQ ID NO.:75;
b. a CDRL2 sequence selected from the group consisting of SEQ ID
NO.:76, SEQ ID NO.: 77 and SEQ ID NO.:78, or;
c. a CDRL3 sequence selected from the group consisting of SEQ ID
NO.:79, SEQ ID NO.:80 and SEQ ID NO.:81.
The isolated antibody or antigen binding fragment may also comprise a heavy chain variable domain having;
a. a CDRH1 sequence comprising SEQ ID NO. :82;
b. a CDRH2 sequence selected from the group consisting of SEQ ID
NO.:83, SEQ ID NO.:84, SEQ ID NO.:85, SEQ ID NO.:86 and SEQ ID
NO.:87, or;

c. a CDRH3 sequence selected from the group consisting of SEQ ID
NO.:88, SEQ ID NO.:89 and SEQ ID NO.:90.
In an exemplary embodiment, the antibody or antigen binding fragment may comprise any individual CDR or a combination of CDR1, CDR2 and/or CDR3 of the light chain variable region. The CDR3 may more particularly be selected. Combination may include for example, CDRL1 and CDRL3; CDRL1 and CDRL2; CDRL2 and CDRL3 and; CDRL1, CDRL2 and CDRL3.
In another exemplary embodiment, the antibody or antigen binding fragment may comprise any individual CDR or a combination of CDR1, CDR2 and/or CDR3 of the heavy chain variable region. The CDR3 may more particularly be selected.
Combination may include for example, CDRH1 and CDRH3; CDRH1 and CDRH2;
CDRH2 and CDRH3 and; CDRH1, CDRH2 and CDRH3.
In accordance with the present invention, the antibody or antigen binding fragment may comprise at least two CDRs of a CDRL1, a CDRL2 or a CDRL3.
Also in accordance with the present invention, the antibody or antigen binding fragment may comprise one CDRL1, one CDRL2 and one CDRL3.
Further in accordance with the present invention, the antibody or antigen binding fragment may comprise:
a. At least two CDRs of a CDRL1, CDRL2 or CDRL3 and;
b. At least two CDRs of a CDRH1, one CDRH2 or one CDRH3.
The antibody or antigen binding fragment may more preferably comprise one CDRL1, one CDRL2 and one CDRL3.
The antibody or antigen binding fragment may also more preferably comprise one CDRH1, one CDRH2 and one CDRH3.
Other exemplary embodiments of the invention relates to an isolated antibody or antigen binding fragment comprising a heavy chain variable domain having;
a. a CDRH1 sequence comprising SEQ ID NO.:82;
b. a CDRH2 sequence selected from the group consisting of SEQ ID
NO.:83, SEQ ID NO.:84, SEQ ID NO.:85, SEQ ID NO.:86 and SEQ ID
NO.:87, or;
c. a CDRH3 sequence selected from the group consisting of SEQ ID
NO.:88, SEQ ID NO.:89 and SEQ ID NO.:90.

In accordance witht the present invention, the antibody or antigen binding fragment may comprise one CDRH1, one CDRH2 or one CDRH3.
In accordance witht the present invention, the antibody or antigen binding fragment may also comprise one CDRH1, one CDRH2 and one CDRH3.
When only one of the light chain variable domain or the heavy chain variable domain is available, an antibody or antigen-binding fragment may be reconstituted by screening a library of complementary variable domains using methods known in the art (Portolano et al. The Journal of Immunology (1993) 150:880-887, Clarkson et al., Nature (1991) 352:624-628).
Also encompassed by the present invention are polypeptides or antibodies comprising variable chains having at least one conservative amino acid substitution in at least one of the CDRs described herein (in comparison with the original CDR).
The present invention also encompasses polypeptides or antibodies comprising variable chains having at least one conservative amino acid substitution in at least two of the CDRs (in comparison with the original CDRs).
The present invention also encompasses polypeptides or antibodies comprising variable chains having at least one conservative amino acid substitution in the 3 CDRs (in comparison with the original CDRs).
The present invention also encompasses polypeptides or antibodies comprising variable chains having at least two conservative amino acid substitutions in at least one of the CDRs (in comparison with the original CDRs).
The present invention also encompasses polypeptides or antibodies comprising variable chains having at least two conservative amino acid substitutions in at least two of the CDRs (in comparison with the original CDRs).
The present invention also encompasses polypeptides or antibodies comprising variable chains having at least two conservative amino acid substitutions in the 3 CDRs (in comparison with the original CDRs).
In another aspect, the present invention relates to a polypeptide, antibody or antigen binding fragment comprising (on a single polypeptide chain or on separate polypeptide chains) at least one complementarity-determining region of a light chain variable domain and at least one complementarity-determining region of a heavy chain variable domain of one of the antibodies or antigen binding fragment described herein.
The present invention relates in another aspect thereof to anti-KAAG1 antibodies that may comprise (on a single polypeptide chain or on separate polypeptide chains) all six complementarity-determining regions (CDRs) of the antibody or antigen binding fragment described herein.
The antibodies or antigen binding fragment of the present invention may further comprise additional amino acids flanking the amino and/or carboxy region of the CDR(s). Those additional amino acids may be as illustrated in Table A or Table B or may include, for example, conservative amino acid substitution.
In accordance with the present invention, the antibody may comprise a CDRL1 sequence comprising or consisting of formula:
X1aSSX2,SLLX3,X4,X5aX6aX7aX8,X9aX10,LX11, (SEQ ID NO.:74) wherein Xi, may be a basic amino acid;
wherein X2, may be a basic amino acid;
wherein X3a may be H, Y or N;
wherein X4a may be S, T, N or R;
wherein X5, may be absent, S or N;
wherein Xga may be D, F or N;
wherein X7a may be G or Q;
wherein Xga may be K, L or N;
wherein Xga may be T or N;
wherein X,0a may be an aromatic amino acid, and;
wherein Xila may be A, N, E or Y.
In an exemplary embodiment of the invention Xia may be K or R.
In a further embodiment of the invention X2a may be Q or K.
In yet a further embodiment of the invention X3a may be N or H.
In an additional embodiment of the invention Xio, may be Y or F.
More specific embodiments of the invention include CDRL1 of SEQ ID NO.:74 where:
Xia is K; X2, is Q; X3a is N; X3a is H; X4a is S, X4a is T; Xga is S; Xga is absent; X6, is N;
X7, is Q; X7a is G; Xga is K; Xga is N; X9, is T; Xioa is Y; or Xlia is A.

In accordance with the present invention, the antibody may comprise a CDRL1 sequence comprising or consisting of formula:
KASQDX1I,X2bX3bX4bX5bX6b (SEQ ID NO.:75) wherein Xib may be an hydrophobic amino acid;
wherein X2b may be G or H;
wherein X3b may be T, N or R;
wherein X4b may be F, Y or A;
wherein X5b may be an hydrophobic amino acid, and;
wherein X6b may be N or A.
In an exemplary embodiment of the invention Xib may be V or I.
In another exemplary embodiment of the invention X5b may be V or L.
More specific embodiments of the invention include CDRL1 of SEQ ID NO.:75 where Xib is I, X2b is H; X3b is T; X3t, is N; X4b is Y; X4b is F; X5b is L or X6b is N.
In accordance with the present invention, the antibody may comprise a CDRL2 sequence comprising or consisting of formula:
FX1cSTX2,X3,S (SEQ ID NO. :76) Wherein X1, is A or G;
Wherein X2, is R or T, and;
Wherein X3 is E, K or A.
In an exemplary embodiment of the invention Xle may be A and X2 may be T.
In another exemplary embodiment of the invention Xlc may be A and X2b may be R.
Other specific embodiments of the invention include CDRL2 of SEQ ID NO. :76 where X1, is A; X2 is R or X3 is E.
In accordance with the present invention, the antibody may comprise a CDRL2 sequence comprising or consisting of formula:
X1dVSX2dX3dX4dS (SEQ ID NO. :77) Wherein Xid may be L or K;
Wherein X2d may be a basic amino acid;
Wherein X3d may be L or R and;
Wherein X4d may be D or F.

In an exemplary embodiment of the invention X2d may be K or N.
Other specific embodiments of the invention include CDRL2 of SEQ ID NO.:77 where Xid is L; X2d is K; X3d is L or X4d is D.
In accordance with the present invention, the antibody may comprise a CDRL2 sequence comprising or consisting of formula:
X1eANRLVX2, (SEQ ID NO.:78) Wherein Xi, may be a basic amino acid, and;
Wherein X2, may be D or A.
In an exemplary embodiment of the invention Xi, may be R or H.
Other specific embodiments of the invention include CDRL2 of SEQ ID NO. :78 where Xi, is R or X2, is D.
In accordance with the present invention, the antibody may comprise a CDRL3 sequence comprising or consisting of formula:
X1fQX2fX3fX4fX5fPLT (SEQ ID NO. :79) Wherein Xif may be Q or L;
Wherein X21 may be an aromatic amino acid;
Wherein X3f may be D, F or Y;
Wherein X4f may be E, A, N or S, and;
Wherein X5f may be I, F or T.
In an exemplary embodiment of the invention X2f may be Y or H.
In another exemplary embodiment of the invention X3f may be Y or D.
In yet another exemplary embodiment of the invention X5f may be I or T.
Other specific embodiments of the invention include CDRL3 of SEQ ID NO.:79 where Xif iS Q; X2f iS H; X3f is D; X3f IS Y; X4f iS S, X41 iS E; X4f iS A; X5f is T, or X5f iS I.
In accordance with the present invention, the antibody may comprise a CDRL3 sequence comprising or consisting of formula:
QQHX1gX2gX3gPLT (SEQ ID NO. :80) Wherein Xig may be an aromatic amino acid;
Wherein X2g may be N or S, and;
Wherein X3g may be I or T.

In an exemplary embodiment of the invention Xi9 may be F or Y
Other specific embodiments of the invention include CDRL3 of SEQ ID NO. :80 where X29 is S or X39 is T.
In accordance with the present invention, the antibody may comprise a CDRL3 sequence comprising or consisting of formula:
X1nQGX2r,HX3,-,PX4hT (SEQ ID NO.:81) Wherein X1h may be an aromatic amino acid;
Wherein X2h may be a neutral hydrophilic amino acid;
Wherein X3h may be F or V, and;
Wherein X4h may be R or L.
In an exemplary embodiment of the invention X h may be W or F.
In another exemplary embodiment of the invention X2h may be S or T.
Other specific embodiments of the invention include CDRL3 of SEQ ID NO. :81 where Xi h is W; X2h is T; X3h is F, Or X4h is R.
In accordance with the present invention, the antibody may comprise a CDRH1 sequence comprising or consisting of formula:
GYX1,FX2,X3,YX4,X5,H (SEQ ID NO. :82) Wherein X1, may be T, I or K;
Wherein Xz, may be a neutral hydrophilic amino acid;
Wherein X3, may be an acidic amino acid;
Wherein X41 may be E, N or D, and;
Wherein X51 may be hydrophobic amino acid.
In an exemplary embodiment of the invention X21 may be T or S.
In another exemplary embodiment of the invention X3I may be D or E.
In yet another exemplary embodiment of the invention X41 may be N or E.
In a further exemplary embodiment of the invention X51 may be M, I or v.
Other specific embodiments of the invention include CDRH1 of SEQ ID NO. :82 where X21 is T; X31 is D; X41 is E; X5t is I or X51 is M.

In accordance with the present invention, the antibody may comprise a CDRH2 sequence comprising or consisting of formula:
X1jX21DPX3,TGX4,TX5) (SEQ ID NO. :83) Wherein Xii may be V or G
Wherein X2i may be a hydrophobic amino acid;
Wherein X3j may be A, G or E;
Wherein X4i may be R, G, D, A, S, N or V, and;
Wherein X51 may be a hydrophobic amino acid.
In an exemplary embodiment of the invention X2i may be I or L.
In another exemplary embodiment of the invention X5i may be A or V.
Other specific embodiments of the invention include CDRH2 of SEQ ID NO. :83 where X1, is V; X2i is X31 is E; X41 is D or X5, is A.
In accordance with the present invention, the antibody may comprise a CDRH2 sequence comprising or consisting of formula:
VX,I,DPX2kTGX3kTA (SEQ ID NO. :84) Wherein Xik may be an hydrophobic amino acid;
Wherein X2k may be A, E or G;
Wherein X3k may be R, G, A, S, N V or D.
In an exemplary embodiment of the invention Xik may be L or I.
Other specific embodiments of the invention include CDRH2 of SEQ ID NO.:84 where Xik is I, X2k is E, or X3k is D.
In accordance with the present invention, the antibody may comprise a CDRH2 sequence comprising or consisting of formula:
YIX,IX2IX3IGX41X51X61(SEQ ID NO. :85) Wherein X11 may be S or N;
Wherein X21 may be an aromatic amino acid Wherein X31 may be D, E or N;
Wherein X4 may be a D or H;
Wherein X51 may be Y, S or N;
Wherein X61 may be D, E or N.
In an exemplary embodiment of the invention X31may be D or N.

In another exemplary embodiment of the invention X61 may be D or N.
Other specific embodiments of the invention include CDRH2 of SEQ ID NO. :85 where X21 is F or Y, X1 is N, X41 is D or X61 is N.
In accordance with the present invention, the antibody may comprise a CDRH2 sequence comprising or consisting of formula:
X1mINPYNX2,VTE (SEQ ID NO. :86) wherein Xim may be N or Y, and;
wherein X2m may be E, D or N.
In an exemplary embodiment of the invention X2,, may be D or N.
Other specific embodiments of the invention include CDRH2 of SEQ ID NO. :86 where Xim is N or X2,, is D.
In accordance with the present invention, the antibody may comprise a CDRH2 sequence comprising or consisting of formula:
DINPX1pYGX2X3r,T (SEQ ID NO. :87) Wherein X1, may be N or Y, Wherein X2,, may be G or T and;
wherein X3n may be I or T.
In accordance with the present invention, the antibody may comprise a CDRH3 sequence comprising or consisting of formula:
MX1õX20,X3oDY (SEQ ID NO. :88) Wherein X,õ may be G or S;
Wherein X20 may be Y or H, and;
wherein X30 may be A or S.
Other specific embodiments of the invention include CDRH3 of SEQ ID NO. :88 where Xi is G; X20 is Y or X30 is S.
In accordance with the present invention, the antibody may comprise a CDRH3 sequence comprising or consisting of formula:
IX1pYAX2pDY (SEQ ID NO. :89) Wherein X1p may be G or S and;
Wherein X2p may be absent or M.

Other specific embodiments of the invention include CDRH3 of SEQ ID NO. :89 where Xlp is S or X2p is M.
In accordance with the present invention, the antibody may comprise a CDRH3 sequence comprising or consisting of formula:
AX1qX2qGLRX3q (SEQ ID NO. :90) Wherein Xiq may be R or W;
Wherein X2q may be an aromatic amino acid and;
wherein X3q may be a basic amino acid.
In an exemplary embodiment of the invention X2q may be W or F.
In another exemplary embodiment of the invention X3q may be Q or N.
Other specific embodiments of the invention include CDRH3 of SEQ ID NO. :90 where Xiq is R; X2q is W or X3q is N.
The framework region of the heavy and/or light chains described herein may be derived from one or more of the framework regions illustrated in Tables A and B. The antibody or antigen binding fragments may thus comprise one or more of the CDRs described herein (e.g., selected from the specific CDRs or consensus CDRs of SEQ
ID NO.:74 to 90) and framework regions originating from those illustrated in Tables A
and B. In Tables A and B, the expected CDRs are shown in bold, while the framework regions are not.
Table 2 describes the sequences of the nucleotides and the amino acids corresponding to the complete light and heavy chain immunoglobulins of specific examples of anti-KAAG1 antibodies.

TABLE 2 ¨ complete sequences of light and heavy chain immunoglobulins that bind to KAAG1 Antibody Chain type Nucleotide Amino acid designation sequence sequence (SEQ ID NO.:) (SEQ ID NO.:) 3D3 Light (L) 3 4 3D3 Heavy (H) 5 6 3G10 Light 7 8 3G10 Heavy 9 10 3C4 Light 11 12 3C4 Heavy 13 14 L chain with any one H chain immunoglobulin that is listed in Table 2. In certain embodiments, the light chain of antibody 3D3 may be combined with the heavy chain of 3D3 or the heavy chain of 3010 to form a complete antibody with KAAG1-binding activity. In an exemplary embodiment of the present invention, the 3D3 L chain may antibody 3D3 are shown in SEQ ID NOS:3 and 5, respectively, and the corresponding amino acid sequences of the light and heavy immunoglobulin chains of antibody are shown in SEQ ID NOS:4 and 6, respectively. Thus, in an exemplary embodiment, an antibody that binds to KAAG1 may comprise the light chain amino acid shown in The complete nucleotide sequences of the light and heavy immunoglobulin chains of antibody 3G10 are shown in SEQ ID NOS:8 and 10, respectively. Thus, in an exemplary embodiment, an antibody that binds to KAAG1 may comprise the light chain amino acid shown in SEQ ID NO. :8 combined with the heavy chain amino acid sequence shown in SEQ ID NO.:10. In another embodiment, the antibody may comprise two identical 3G10 light chains comprising SEQ ID NO.:8 and two identical 3G10 heavy chains comprising SEQ ID NO.:10.
The complete nucleotide sequences of the light and heavy immunoglobulin chains of antibody 304 are shown in SEQ ID NOS:11 and 13, respectively and the corresponding amino acid sequences of the light and heavy immunoglobulin chains of antibody 3C4 are shown in SEQ ID NOS:12 and 14, respectively. Thus, in an exemplary embodiment, an antibody that binds to KAAG1 may comprise the light chain amino acid shown in SEQ ID NO.:12 combined with the heavy chain amino acid sequence shown in SEQ ID NO. :14. In another embodiment, the antibody may comprise two identical 304 light chains comprising SEQ ID NO.:12 and two identical 3C4 heavy chains comprising SEQ ID NO.:14.
Variants of other anti-K1AAG1 antibodies or antigen binding fragments formed by the combination of light and/or heavy immunoglobulin chains may each independently have at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% identity to the amino acid sequences listed in Table 2 are also provided. In certain embodiments, the antibody variants may comprise at least one light chain and one heavy chain.
In other instances, the antibody variants may comprise two identical light chains and two identical heavy chains. In accordance with the present invention, the region of variation may be located in the constant region or in the variable region.
Also in accordance with the present invention, the region of variation may be located in the framework region.
Also encompassed by the present invention are antibodies comprising a light chain comprising one of the variable region illustrated in Table A and a heavy chain comprising one of the variable region illustrated in Table B. The light chain and heavy chain may comprise a constant domain. Combinations of light chains and heavy chains of Table 2, Table A and Table B are also encompassed by the present invention.
Antibodies or antigen binding fragments that contain the light chain and heavy chain variable regions are also provided in the present invention. Additionally, certain embodiments include antigen binding fragments, variants, and derivatives of these light and heavy chain variable regions.
Yet other exemplary embodiments of the invention includes an isolated antibody or antigen binding fragment capable of specific binding to SEQ ID NO.:2, to an extracellular portion of SEQ ID NO.:2, or to a secreted form of SEQ ID NO.:2 or to a variant thereof, the antibody comprising:
a. the light chain variable domain defined in SEQ ID NO.:16 and the heavy chain variable domain defined in SEQ ID NO.:18, b. the light chain variable domain defined in SEQ ID NO.:20 and the heavy chain variable domain defined in SEQ ID NO.:22;
c. the light chain variable domain defined in SEQ ID NO. :24 and the heavy chain variable domain defined in SEQ ID NO. :26;
d. the light chain variable domain defined in SEQ ID NO.:105 and the heavy chain variable domain defined in SEQ ID NO.:132, e. the light chain variable domain defined in SEQ ID NO.:106 and the heavy chain variable domain defined in SEQ ID NO.:133, f. the light chain variable domain defined in SEQ ID NO.:107 and the heavy chain variable domain defined in SEQ ID NO.:134, g. the light chain variable domain defined in SEQ ID NO.:108 and the heavy chain variable domain defined in SEQ ID NO.:154, h. the light chain variable domain defined in SEQ ID NO.:109 and the heavy chain variable domain defined in SEQ ID NO.:153, I. the light chain variable domain defined in SEQ ID NO.:110 and the heavy chain variable domain defined in SEQ ID NO.:135, j. the light chain variable domain defined in SEQ ID NO.:111 and the heavy chain variable domain defined in SEQ ID NO.:136, k. the light chain variable domain defined in SEQ ID NO.:112 and the heavy chain variable domain defined in SEQ ID NO. :149, I. the light chain variable domain defined in SEQ ID NO.:113 and the heavy chain variable domain defined in SEQ ID NO.:137, m. the light chain variable domain defined in SEQ ID NO.:114 and the heavy chain variable domain defined in SEQ ID NO.:140, n. the light chain variable domain defined in SEQ ID NO.:115 and the heavy chain variable domain defined in SEQ ID NO.:141, o. the light chain variable domain defined in SEQ ID NO.:116 and the heavy chain variable domain defined in SEQ ID NO.:142, p. the light chain variable domain defined in SEQ ID NO.:117 and the heavy chain variable domain defined in SEQ ID NO.:139, q. the light chain variable domain defined in SEQ ID NO.:119 and the heavy chain variable domain defined in SEQ ID NO.:143, r. the light chain variable domain defined in SEQ ID NO.:120 and the heavy chain variable domain defined in SEQ ID NO.:152, s. the light chain variable domain defined in SEQ ID NO.:121 and the heavy chain variable domain defined in SEQ ID NO.:146, t. the light chain variable domain defined in SEQ ID NO.:122 and the heavy chain variable domain defined in SEQ ID NO.:138, u. the light chain variable domain defined in SEQ ID NO.:123 and the heavy chain variable domain defined in SEQ ID NO.:150, v. the light chain variable domain defined in SEQ ID NO.:124 and the heavy chain variable domain defined in SEQ ID NO.:144, w. the light chain variable domain defined in SEQ ID NO.:126 and the heavy chain variable domain defined in SEQ ID NO.:145, x. the light chain variable domain defined in SEQ ID NO.:127 and the heavy chain variable domain defined in SEQ ID NO.:157, y. the light chain variable domain defined in SEQ ID NO.:128 and the heavy chain variable domain defined in SEQ ID NO.:155, z. the light chain variable domain defined in SEQ ID NO.:129 and the heavy chain variable domain defined in SEQ ID NO.:156, or;
aa. the light chain variable domain defined in SEQ ID NO.:130 and the heavy chain variable domain defined in SEQ ID NO.:151.
It is to be understood herein, that the light chain variable region of the specific combination provided above may be changed for any other light chain variable region.
Similarly, the heavy chain variable region of the specific combination provided above may be changed for any other heavy chain variable region.
Specific examples of sequences present in these light and heavy chain variable regions are disclosed in Table 3.

Table 3 ¨ Sequences of light and heavy chain variable regions that bind to Antibody Variable Nucleotide Amino acid designation region type sequence sequence (SEQ ID NO.:) (SEQ ID NO.:) 3D3 Light (VL) 15 16 3D3 Heavy (VH) 17 18 3G10 Light 19 20 3G10 Heavy 21 22 304 Light 23 24 304 Heavy 25 26 3z1A02 Light 105 3z1A02 Heavy 132 3z1E10 Light 109 3z1E10 Heavy 153 3z1G12L Light 126 3z1G12H Heavy 145 Therefore, antibodies and antigen binding fragments that bind to KAAG1 may Variants of other anti-KAAG1 antibodies formed by the combination of light and/or heavy chain variable regions that each have at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% identity to the amino acid sequences listed in Table 3 are also provided. Those skilled in the art will also recognize that the anti-KAAG1 antibody deletions, or additions in the amino acid sequences of the light and/or heavy chain variable regions listed in Table 3.
In accordance with the present invention, the region of variation may be located in the framework region of the variable region. Table 4 ¨ Sequences of the light and heavy chain CDRs Antibody Chain type CDR SEQ ID NO.: Amino acid sequence designation 3D3 Light (L) CDR L1 27 KSSQSLLNSNFQKNFLA
3D3 Light CDR L2 28 FASTRES
3D3 Light CDR L3 29 - QQHYSTPLT
3D3 Heavy (H) CDR H1 30 GYIFTDYEIH
3D3 Heavy CDR H2 31 VIDPETGNTA
3D3 Heavy CDR H3 32 MGYSDY
3G10 Light CDR L1 33 RSSQSLLHSNGNTYLE
3G10 Light CDR L2 ' 34 KVSNRFS
3G10 Light CDR L3 ' 35 FQGSHVPLT
3G10 Heavy CDR H1 ' 36 GYTFTDNYMN
3G10 Heavy CDR H2 37 DINPYYGTTT
3G10 Heavy CDR H3 38 ARDDWFDY
3C4 Light CDR L1 39 KASQDIHNFLN
' 3C4 ' Light CDR L2 40 RANRLVD
3C4 Light CDR L3 ' 41 LQYDEIPLT
' 3C4 Heavy CDR H1 42 ' GFSITSGYGWH
3C4 Heavy CDR H2 43 YINYDGHND
- 3C4 ' Heavy CDR H3 '44 ASSYDGLFAY
, - 3z1A02 Light CDR L1 158 KSSQSLLHSDGKTYLN
3z1A02 Light CDR L2 159 ' LVSKLDS
P-3z1A02 Light CDR L3 ' 160 WQGTHFPRT
, ___________ 3z1A02 Heavy CDR H1 161 GYTFTD YNMH
3z1A02 Heavy CDR H2 162 YINPYNDVTE
3z1A02 Heavy CDR H3 163 AWFGL RQ _____________ ;
________________________________________________________________________ i 3z1E10 Light CDR L1 '164 RSSKSLLHSNGN TYLY
3z1E10 Light CDR L2 ' 165 RMSNLAS
3z1E10 Light CDR L3 166 MQHLEYPYT

Antibody Chain type CDR SEQ ID NO.: Amino acid sequence designation 3z1E10 Heavy CDR H1 167 GDTFTD YYMN
3z1E10 Heavy CDR H2 168 DINPNYGGIT
3z1E10 Heavy CDR H3 169 QAYYRNS DY
3z1G12L Light CDR L1 170 KASQDVGTAVA
3z1G12L Light CDR L2 171 WTSTRHT
3z1G12L Light CDR L3 172 QQHYSIPLT
3z1G12H Heavy CDR H1 173 GYIFTDYEIH
3z1G12H Heavy CDR H2 174 VIDPETGNTA
3z1G12H Heavy CDR H3 175 MGYSDY
In certain embodiments of the present invention, the anti-KAAG1 antibodies or antigen binding fragments may comprise the CDR sequences shown in Table 4 or have substantial sequence identity to the CDR sequences of Table 4. In an exemplary embodiment, the 3D3 anti-KAAG1 antibody may comprise a light chain variable region containing CDR1, 2, and 3 that are encoded by SEQ ID NOS:27, 28, and 29, respectively, and/or a heavy chain variable region containing CDR1, 2, and 3 that are encoded by SEQ ID NOS:30, 31, and 32, respectively. In other embodiments the CDR3 region may be sufficient to provide antigen binding. As such polypeptides comprising the CDR3L or the CDR3H or both the CDR3L and the CDR3H are encompassed by the present invention.
Additionally, the anti-KAAG1 antibodies or antigen binding fragments may include any combination of the CDRs listed in Table 4. For example, the antibodies or antigen binding fragments may include the light chain CDR3 and the heavy chain CDR3.
It is understood that the CDRs that are contained in the anti-KAAG1 antibodies or antigen binding fragments may be variant CDRs with 80%, 85%, 90%, or 95% sequence identity to the CDR sequences presented in Table 4. Those skilled in the art will also recognize that the variants may include conservative amino acid changes, amino acid substitutions, deletions, or additions in the CDR sequences listed in Table 4.
Other exemplary embodiments of the invention includes an isolated antibody or antigen binding fragment capable of specific binding to SEQ ID NO. :2, to an extracellular portion of SEQ ID NO. :2 or to a secreted form of SEQ ID NO. :2 or to a variant thereof, the antibody comprising:

a. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:16 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:18, b. the 3CDRs of a light chain variable domain defined in SEQ ID NO. :20 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:22;
c. the 3CDRs of a light chain variable domain defined in SEQ ID NO. :24 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:26;
d. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:105 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:132, e. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:106 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:133, f. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:107 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:134, g. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:108 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:154, h. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:109 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:153, i. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:110 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:135, j. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:111 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:136, k. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:112 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:149, I. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:113 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:137, m. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:114 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:140, n. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:115 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:141, o. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:116 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:142, p. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:117 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:139, q. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:119 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:143, r. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:120 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:152, s. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:121 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:146, t. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:122 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:138, u. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:123 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:150, v. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:124 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:144, w. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:126 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:145, x. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:127 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:157, y. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:128 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:155, z. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:129 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:156, or;
aa. the 3CDRs of a light chain variable domain defined in SEQ ID NO.:130 and/or the 3CDRs of a heavy chain variable domain defined in SEQ ID
NO.:151.
Again, the light chain variable region of the specific combination provided above may be changed for any other light chain variable region described herein.
Similarly, the heavy chain variable region of the specific combination provided above may be changed for any other heavy chain variable region described herein.
Variant antibody and antigen binding fragments The present invention also encompasses variants of the antibodies or antigen binding fragments described herein. Variant antibodies or antigen binding fragments included are those having a variation in the amino acid sequence. For example, variant antibodies or antigen binding fragments included are those having at least one variant CDR (two, three, four, five or six variant CDRs or even twelve variant CDRs), a variant light chain variable domain, a variant heavy chain variable domain, a variant light chain and/or a variant heavy chain. Variant antibodies or antigen binding fragments included in the present invention are those having, for example, similar or improved binding affinity in comparison with the original antibody or antigen binding fragment.
As used herein the term "variant" applies to any of the sequence described herein and includes for example, a variant CDR (either CDRL1, CDRL2, CDRL3, CDRH1, CDRH2 and/or CDRH3), a variant light chain variable domain, a variant heavy chain variable domain, a variant light chain, a variant heavy chain, a variant antibody, a variant antigen binding fragment and a KAAG1 variant.
Variant antibodies or antigen binding fragments encompassed by the present invention are those which may comprise an insertion, a deletion or an amino acid substitution (conservative or non-conservative). These variants may have at least one amino acid residue in its amino acid sequence removed and a different residue inserted in its place.
Exemplary embodiments of variant antibodies and antigen binding fragments of the present invention are a group of antibodies and antigen binding fragments capable of binding to KAAG1 and characterized herein as being humanized.
The humanized antibodies and antigen binding fragments of the present invention includes more particularly, humanized 3D3 or 304 antibodies and antigen binding fragments.
In accordance with an embodiment of the invention, the humanized antibody may have a light chain variable region of formula:
DIVMTQSPXSLAVSXGXXXTXNCKSSQSLLNSNFQKNFLAWYQQKPGQXPKLLIYF
ASTRESSXPDRFXGSGSGTDFTLTISSXQAEDXAXYXCQQHYSTPLTFGXGTKLEX
K (SEQ ID NO.:184);
wherein at least one of the amino acid identified by X is an amino acid substitution (conservative or non-conservative) in comparison with a corresponding amino acid in the polypeptide set forth in SEQ ID NO.:16. The amino acid substitution may be, for example conservative.
In accordance with a more specific embodiment, the humanized antibody may have a light chain variable region of formula:

XA7PKLLIYFASTRESSXA8PDRFXA9GSGSGTDFTLTISSXA10QAEDXA11AX,m2YXA13CQ
QHYSTPLTFGXA14GTKLEXA15K (SEQ ID NO.:185);
Wherein Xpki may be, for example, D or S;
Wherein XA2 may be, for example, a hydrophobic amino acid or more particularly L or I;
Wherein XA3 may be, for example, E or Q;
Wherein XA4 may be, for example, a basic amino acid or more particularly R or K;
Wherein XA5 may be, for example, a hydrophobic amino acid or more particularly A or V;
Wherein XA6 may be, for example, a hydrophobic amino acid or more particularly I or M;
Wherein XA7 may be, for example, P or S;
Wherein XA8 may be, for example, a hydrophobic amino acid or more particularly V or I;

Wherein XAg may be, for example, S or I;
Wherein Xpog may be, for example, a hydrophobic amino acid or more particularly L or V;
Wherein XAii may be, for example, a hydrophobic amino acid or more particularly V
or L;
Wherein XA12 may be, for example, V or D;
Wherein XA13 may be, for example, an aromatic amino acid or more particularly Y or F;
Wherein Xpoil may be, for example, Q or A and;
Wherein XAig may be, for example, a hydrophobic amino acid or more particularly I or L.
In accordance with an even more specific embodiment, the humanized antibody may have a light chain variable region of formula:
DIVMTQSPXaiSLAVSXa2GXa3Xa4Xa5TXa6NCKSSQSLLNSNFQKNFLAVVYQQKP3QX
a7PKLLIYFASTRESSXa8PDRFXa9GSGSGTDFTLTI55Xalo0AEDXailAXai2YXai30QQH
YSTPLTFGXa14GTKLEX,15K (SEQ ID NO.:186);
Wherein Xal may be, for example, D or S;
Wherein Xa2 may be, for example, L or I;
Wherein Xa3 may be, for example, E or Q;
Wherein Xa4 may be, for example, R or K;
Wherein Xag may be, for example, A or V;
Wherein Xag may be, for example, I or M;
Wherein Xa, may be, for example, P or S;
Wherein Xag may be, for example, V or I;
Wherein Xag may be, for example, S or I;
Wherein Xaig may be, for example, L or V;
Wherein Xall may be, for example, V or L;
Wherein X812 may be, for example, V or D;
Wherein Xa13 may be, for example, Y or F;
Wherein X814 may be, for example, Q or A and;
Wherein X815 is for example, I or L.
In accordance with an embodiment of the present invention, the humanized antibody may have a heavy chain variable region of formula:
EVQLXQSXAEXXXPGASVXXSCKASGYIFTDYEIHVVVXQXPXXGLEWXGVIDPETG
NTAFNQKFKGXXTXTADXSXSTAYMELSSLTSEDXAVYYCMGYSDYVVGQGTXXTV

SS (SEQ ID NO.:187); wherein at least one of the amino acid identified by X is an amino acid substitution (conservative or non-conservative) in comparison with a corresponding amino acid in the polypeptide set forth in SEQ ID NO.:18. The amino acid substitution may be, for example conservative.
In accordance with a more specific embodiment, the humanized antibody may have a heavy chain variable region of formula:
EVQLXBiQSXB2AEXB3XB4XB5PGASVXB6X137SCKASGYIFTDYEIHWVXB8QX139PXBi oXBi iGLEWXBi2GVIDPETGNTAFNQKFKGXBi3XBi4TXBi5TADX1316SXB17STAYMELSSLTS
EDX1318AVYYCMGYSDYWGQGTXB19XB20TVSS (SEQ ID NO.:188), Wherein Xgi may be, for example, V or Q;
Wherein Xg2 may be, for example, G or V;
Wherein Xg3 may be, for example, a hydrophobic amino acid or more particularly V or L;
Wherein Xg4 may be, for example, K or V;
Wherein Xg5 may be, for example, a basic amino acid or more particularly K or R;
Wherein Xg6 may be, for example, K or T;
Wherein Xg7 may be, for example, a hydrophobic amino acid or more particularly V or L;
Wherein Xgg may be, for example, a basic amino acid or more particularly R or K;
Wherein Xgg may be, for example, A or T;
Wherein Xgi 0 may be, for example, G or V;
Wherein Xgi I may be, for example, Q or H;
Wherein Xg12 may be, for example, a hydrophobic amino acid or more particularly M
or I;
Wherein Xg13 may be, for example, a basic amino acid or more particularly R or K;
Wherein Xg14 may be, for example, a hydrophobic amino acid or more particularly V
or A;
Wherein Xg15 may be, for example, a hydrophobic amino acid or more particularly I or L;
Wherein Xgig may be, for example, T or I;
Wherein Xg17 may be, for example, a neutral hydrophilic amino acid or more particularly T or S;
Wherein X818 may be, for example, a neutral hydrophilic amino acid or more particularly T or S;
Wherein Xgig may be, for example, L or T and;

Wherein Xi3,20 may be, for example, a hydrophobic amino acid or more particularly V
or L.
In accordance with a more specific embodiment, the humanized antibody may have a heavy chain variable region of formula:
EVQLXbiQSXb2AEXb3XbaXb5PGASVXb6X,D7SCKASGYIFTDYEIHVVVX,D8QXb9PXbioXbil GLEWXbi2GVIDPETGNTAFNQKFKGXbi3X041-XbisTADXbi6SX07STAYMELSSLTSE
DXb18AVYYCMGYSDYWGQGTXb19Xb20TVSS (SEQ ID NO.:189);
Wherein Xbi may be, for example, V or Q;
Wherein Xb2 may be, for example, G or V;
Wherein Xbg may be, for example, V or L;
Wherein Xb4 may be, for example, K or V;
Wherein Xb5 may be, for example, K or R;
Wherein Xb6 may be, for example, K or T;
Wherein Xb7 may be, for example, V or L;
Wherein Xbg may be, for example, R or K;
Wherein Xbg may be, for example, A or T;
Wherein Xbio may be, for example, G or V;
Wherein Xbil may be, for example, Q or H;
Wherein Xb12 may be, for example, M or I;
Wherein Xb13 may be, for example, R or K;
Wherein Xbiet may be, for example, V or A;
Wherein Xb15 may be, for example, I or L;
Wherein Xbig may be, for example, T or I;
Wherein Xb17 may be, for example, T or S;
Wherein Xbig may be, for example, T or S;
Wherein Xbig may be, for example, L or T;
Wherein Xb20 may be, for example, V or L.
In accordance with an embodiment of the present invention, the humanized antibody may have a light chain variable region of formula:
DIVMXQSPSSXXASXGXRVTITCKASQDIHNFLNWFQQKPGKXPKTLIFRANRLVD
GVPSRFSGSGSGXDYXLTISSLXXEDXXXYSCLQYDEIPLTFGXGTKLEXX (SEQ ID
NO.:190); wherein at least one of the amino acid identified by X is an amino acid substitution (conservative or non-conservative) in comparison with a corresponding amino acid in the polypeptide set forth in SEQ ID NO.24. The amino acid substitution may be, for example conservative.

In accordance with a more specific embodiment, the humanized antibody may have a light chain variable region of formula:
DIVMXciQSPSSXc2Xc3ASXc4GXc5RVTITCKASQDIHNFLNWFQQKPGKXc6PKTLIF
RAN RLVOGVPSRFSGSGSGXc7DYXcaLTI SSLXc9Xci DEDXci 1Xc12Xc13YSCLQYDEIP
LTFGXc14GTKLEXc15Xc16(SEQ ID NO.:191);
Wherein Xcl may be, for example, a neutral hydrophilic amino acid or more particularly T or S;
Wherein Xc2 may be, for example, a hydrophobic amino acid or more particularly L or M;
Wherein Xcg may be, for example, S or Y;
Wherein Xc4 may be, for example, a hydrophobic amino acid or more particularly V or L;
Wherein Xc6 may be, for example, an acidic amino acid or more particularly D
or E;
Wherein Xcg may be, for example, A or S;
Wherein Xc7 may be, for example, T or Q;
Wherein Xcg may be, for example, a neutral hydrophilic amino acid or more particularly T or S;
Wherein Xcg may be, for example, Q or E;
Wherein Xclo may be, for example, P or F;
Wherein Xc11 may be, for example, F or L;
Wherein Xc12 may be, for example, A or G;
Wherein Xc13 may be, for example, T or I;
Wherein Xc14 may be, for example, Q or A;
Wherein Xc15 may be, for example, a hydrophobic amino acid or more particularly I or L, and; wherein Xc16 may be, for example, a basic amino acid or more particularly K
or R.
In accordance with a more specific embodiment, the humanized antibody may have a light chain variable region of formula:
DIVMXc1QSPSSX,2X,3ASX,4GX,5RVTITCKASQDIHNFLNWFQQKPGKXc6PKTLIFRA
NRLVDGVPSRFSGSGSGXc7DYXcaLTISSLXc9XcloEDXci 1Xc12X03YSCLQYDEIPLTF
GX04GTKLEXc15X06 (SEQ ID NO.:192);
Wherein Xc1 may be, for example, T or S;
Wherein X,2 may be, for example, L or M;
Wherein Xcg may be, for example, S or Y;
Wherein Xc4 may be, for example, V or L;

Wherein Xc5 may be, for example, D or E;
Wherein X,6 may be, for example, A or S;
Wherein Xc may be, for example, T or Q;
Wherein X,8 may be, for example, T or S;
Wherein Xc, may be, for example, Q or E;
Wherein X,10 may be, for example, P or F;
Wherein Xcl, may be, for example, F or L;
Wherein Xc12 may be, for example, A or G;
Wherein X03 may be, for example, T or I;
Wherein X,14 may be, for example, Q or A;
Wherein Xc15 may be, for example, I or L and;
wherein X016 may be, for example, K or R.
In accordance with an embodiment of the present invention, the humanized antibody may have a heavy chain variable region of formula:
EVQLQESGPXLVKPSQXLSLTCTVXGFSITSGYGWHWIRQXPGXXLEWXGYINYDG
HNDYNPSLKSRXXIXQDTSKNQFXLXLXSVTXXDTAXYYCASSYDGLFAYWGQGTL
VTVSX (SEQ ID NO.:193); wherein at least one of the amino acid identified by X
is an amino acid substitution (conservative or non-conservative) in comparison with a corresponding amino acid in the polypeptide set forth in SEQ ID NO. :26. The amino acid substitution may be, for example conservative.
In accordance with a more specific embodiment, the humanized antibody may have a heavy chain variable region of formula:

XD7GYINYDGHNDYNPSLKSRXD8XD9IXo10QDTSKNQFXD111-XD121-Xo13SVTXD14XD15D
TAXD16YYCASSYDGLFAYWGQGTLVTVSXD17(SEQ ID NO.:194);
Wherein XDi may be, for example, G or D;
Wherein XD2 may be, for example, a neutral hydrophilic amino acid or more particularly T or S;
Wherein X03 may be, for example, a neutral hydrophilic amino acid or more particularly S or T;
Wherein XD4 may be, for example, H or F;
Wherein XD5 may be, for example, K or N;
Wherein XD6 may be, for example, G or K;
Wherein XD7 may be, for example, a hydrophobic amino acid or more particularly I or M;

Wherein XD8 may be, for example, a hydrophobic amino acid or more particularly V or I;
Wherein XD9 may be, for example, a neutral hydrophilic amino acid or more particularly T or S;
Wherein )(Dip may be, for example, a neutral hydrophilic amino acid or more particularly S or T;
Wherein X011 may be, for example, a neutral hydrophilic amino acid or more particularly S or F;
Wherein X012 may be, for example, a basic amino acid or more particularly K or Q;
Wherein XD13 may be, for example, S or N;
Wherein XD14 may be, for example, A or T;
Wherein XD15 may be, for example, A or E;
Wherein XD16 may be, for example, V or T and;
Wherein XD17 may be any amino acid, A or absent.
In accordance with a more specific embodiment, the humanized antibody may have a heavy chain variable region of formula:
EVQLQESGPXdiLVKPSQXd2LSLTCTVXd3GFSITSGYGWHWIRQXd4PGXd5Xd6LEWXd 7GYINYDGHNDYNPSLKSRXdaXd9IXd100DTSKNQFXd11DQ121-Xd13SVTXd14Xd15DTAXd 16YYCASSYDGLFAYVVGQGTLVTVSX,117(SEQ ID NO.:195);
Wherein Xdi may be, for example, G or D;
Wherein Xd2 may be, for example, T or S;
Wherein Xd3 may be, for example, S or T;
Wherein Xd4 may be, for example, H or F;
Wherein Xd5 may be, for example, K or N;
Wherein Xd6 may be, for example, G or K;
Wherein Xd7 may be, for example, I or M;
Wherein Xd5 may be, for example, V or I;
Wherein Xdg may be, for example, T or S;
Wherein Xdio may be, for example, S or T;
Wherein Xdii may be, for example, S or F;
Wherein Xd12 may be, for example, K or Q;
Wherein Xd13 may be, for example, S or N;
Wherein Xd14 may be, for example, A or T;
Wherein Xd15 may be, for example, A or E;
Wherein Xd16 may be, for example, V or T and;

Wherein Xd17, A or absent.
Accordingly, the present invention provides in one aspect, an antibody or antigen binding fragment thereof capable of specific binding to Kidney associated antigen 1 (KAAG1) which may have a light chain variable region at least 80% identical to SEQ
ID NO.:16 and/or a heavy chain variable region at least 80% identical to SEQ
ID
NO.:18. The antibody or antigen binding fragment thereof may also comprise at least one amino acid substitution in comparison with SEQ ID NO.:16 or SEQ ID NO.:18.
The present invention also provides in another aspect, an antibody or antigen binding fragment thereof which may have a light chain variable region at least 80%
identical to SEQ ID NO.:24 and/or a heavy chain variable region at least 80% identical to SEQ
ID NO. :26. The antibody or antigen binding fragment thereof may also comprise at least one amino acid substitution in comparison with SEQ ID NO. :24 or SEQ ID
NO. :26.
In accordance with an embodiment of the invention, the amino acid substitution may be outside of a complementarity determining region (CDR). An antibody or antigen binding fragment having such an amino acid sequence is identified herein as a humanized antibody or antigen binding fragment.
In accordance with an embodiment of the invention, the antibody the amino acid substitution may be located, for example, in the light chain variable region.
In accordance with an additional embodiment of the invention, the antibody or antigen binding fragment thereof may comprise at least two or at least three amino acid substitutions. Such amino acid substitutions may be located in the same variable region or may be located in distinct variable regions.
Further in accordance with the present invention, the antibody or antigen binding fragment thereof may comprise for example, from one to twenty amino acid substitutions in the light chain variable region and/or heavy chain variable region.
As used herein the term "from one to twenty" includes every individual values and ranges such as for example, 1, 2, 3, and up to 20; 1 to 20; 1 to 19; 1 to 18;
1 to 17; 1 to 16; 1 to 15 and soon; 2 to 20; 2 to 19; 2 to 18; 2 to 17 and so on; 3 to 20; 3 to 19;
3 to 18 and so on; 4 to 20; 4 to 19; 4 to 18; 4 to 17; 4 to 16 and so on; 5 to 20; 5 to 19; 5 to 18; 5 to 17 and so on, etc.

Likewise, the term "from one to fifteen" includes every individual values and ranges such as for example, 1, 2, 3, and up to 15; 1 to 15; 1 to 14; 1 to 13; 1 to 12; 1 to 11; 1 to 10 and so on; 2 to 15; 2 to 14; 2 to 13; 2 to 12 and so on; 3 to 15; 3 to 14; 3 to 13 and so on; 4 to 15; 4 to 14; 4 to 13; 4 to 12; 4 to 11 and so on; 5 to 15; 5 to 14; 5 to 13; 5 to 12 and soon, etc.
In a more specific embodiment of the invention, the number of amino acid substitutions that may be accommodated in a humanized light chain variable region derived from SEQ ID NO.:16 may be for example, from 1 to 15 amino acid substitutions.
In yet a more specific embodiment of the invention, the number of amino acid substitutions that may be accommodated in a humanized heavy chain variable region derived from SEQ ID NO.:18 may be for example, from 1 to 20 amino acid substitutions. In some instances, when considering a humanized version of SEQ
ID
NO.:18, it may be useful to have at least three amino acid substitutions.
In a further more specific embodiment of the invention, the number of amino acid substitutions that may be accommodated in a humanized light chain variable region derived from SEQ ID NO.:24 may be for example, from 1 to 16 amino acid substitutions.
In yet a further more specific embodiment of the invention, the number of amino acid substitutions that may be accommodated in a humanized heavy chain variable region of SEQ ID NO.:26 may be for example, from 1 to 17 amino acid substitutions.
In accordance with an embodiment of the invention, the one to twenty amino acid substitutions may be for example, in the light chain variable region.
In accordance with an embodiment of the invention, the one to twenty amino acid substitutions may be for example, in the heavy chain variable region.
A humanized antibody or antigen binding fragment may therefore have a light chain variable region having up to twenty amino acid substitutions in comparison with SEQ
ID NO.:16 or SEQ ID NO.:24 and may have a heavy chain variable region having up to twenty amino acid substitutions in comparison with SEQ ID NO.:18 or SEQ ID
NO. :26, It is to be understood herein that when the humanized antibody or antigen binding fragment has two light chain variable regions and two heavy chain variable regions, each one of the light chain variable regions may independently have up to twenty amino acid substitutions and each one of the heavy chain variable regions may have up to twenty amino acid substitutions.
As discussed herein the amino acid substitutions may be conservative or non-conservative. In an exemplary embodiment the amino acid substitutions may be conservative.
It is to be understood herein that the humanized antibody or antigen binding fragment of the invention may also have a light chain variable region and/or heavy chain variable region showing a deletion in comparison with SEQ ID NO.:16, SEQ ID
NO.:18, SEQ ID NO.:24 and/or SEQ ID NO.:26. Such deletion may be found, for example, at an amino- or carboxy-terminus of the light chain variable region and/or heavy chain variable region.
Another exemplary embodiment of the humanized antibody or antigen binding fragment of the present invention includes for example, an antibody or antigen binding fragment having a light chain variable region which may comprise at least 90 consecutive amino acids of any of SEQ ID NO.:184, SEQ ID NO.:185, SEQ ID
NO.:186 or SEQ ID NO.:178.
As used herein the term "at least 90 consecutive amino acids of SEQ ID
NO.:184"
also includes the terms "at least 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112 or at least 113 consecutive amino acids". The term "at least 90 consecutive amino acids of SEQ ID NO.:184"
encompasses any possible sequence of at least 90 consecutive amino acids found in SEQ ID NO.:184 and especially those sequences which include the 3 CDRs of SEQ
ID NO.:184, such as, for example a sequence comprising amino acids 6 to 108, 5 to 109, 13 to 103, 14 to 111 of SEQ ID NO.:184 and so on.
As used herein the term "at least 90 consecutive amino acids of SEQ ID
NO.:185"
also includes the terms "at least 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112 or at least 113 consecutive amino acids". The term "at least 90 consecutive amino acids of SEQ ID NO.:185"
encompasses any possible sequence of at least 90 consecutive amino acids found in SEQ ID NO.:185 and especially those sequences which include the 3 CDRs of SEQ
ID NO.:185, such as, for example a sequence comprising amino acids 7 to 109, 12 to 104,22 to 113, 18 to 112 of SEQ ID NO.:185 and so on.

The terms "at least 90 consecutive amino acids of SEQ ID NO.:186" or "at least consecutive amino acids of SEQ ID NO.:178" has a similar meaning.
In accordance with the present invention, the antibody or antigen binding fragment of the present invention may have, for example, a light chain variable region as set forth in SEQ ID NO.:178.
The humanized antibody or antigen binding fragment of the invention includes (or further includes) for example, a heavy chain variable region which may comprise at least 90 consecutive amino acids of any of SEQ ID NOs.:187, 188, 189 or 179.
As used herein the term "at least 90 consecutive amino acids of SEQ ID
NO.:187"
also includes the terms "at least 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112 or at least 113 consecutive amino acids". The term "at least 90 consecutive amino acids of SEQ ID NO.:187"
encompasses any possible sequence of at least 90 consecutive amino acids found in SEQ ID NO.:187 and especially those sequences which include the 3 CDRs of SEQ
ID NO.:187, such as, for example a sequence comprising amino acids 1 to 106, 2 to 112, 11 to 113, 7 to 102 of SEQ ID NO.:187 and so on.
As used herein the term "at least 90 consecutive amino acids of SEQ ID
NO.:188"
also includes the terms "at least 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112 or at least 113 consecutive amino acids". The term "at least 90 consecutive amino acids of SEQ ID NO.:188"
encompasses any possible sequence of at least 90 consecutive amino acids found in SEQ ID NO.:188 and especially those sequences which include the 3 CDRs of SEQ
ID NO.:188, for example a sequence comprising amino acids 6 to 109, 8 to 113, 1 to 102, 2 to 105 of SEQ ID NO.:188 and so on.
The terms "at least 90 consecutive amino acids of SEQ ID NO.:189" or "at least consecutive amino acids of SEQ ID NO.:179" has a similar meaning.
In accordance with the present invention, the antibody or antigen binding fragment of the present invention may have, for example, a heavy chain variable region as set forth in SEQ ID NO.:179.
In accordance with the present invention the antibody or antigen binding fragment may comprise, for example, a) a light chain variable region which may comprise at least 90 consecutive amino acids of SEQ ID NO.:184 and a heavy chain variable region which may comprise at least 90 consecutive amino acids of any of SEQ ID NO.:187, SEQ ID NO.:188, SEQ ID
NO.:189 or SEQ ID NO.:179;
b) a light chain variable region which may comprise at least 90 consecutive amino acids of SEQ ID NO.:185 and a heavy chain variable region which may comprise at least 90 consecutive amino acids of any of SEQ ID NO.:187, SEQ ID NO.:188, SEQ ID
NO.:189 or SEQ ID NO.:179;
c) a light chain variable region which may comprise amino acids at least 90 consecutive amino acids of SEQ ID NO.:186 and a heavy chain variable region which may comprise at least 90 consecutive amino acids of any of SEQ ID NO.:187, SEQ ID NO.:188, SEQ ID
NO.:189 or SEQ ID NO.:179 or;
d) a light chain variable region which may comprise at least 90 consecutive amino acids of SEQ ID NO.:178 and a heavy chain variable region which may comprise at least 90 consecutive amino acids of any of SEQ ID NO.:187, SEQ ID NO.:188, SEQ ID
NO.:189 or SEQ ID NO.:179.
In accordance with a more specific embodiment of the invention, the light chain variable region may comprise at least 90 consecutive amino acids of SEQ ID
NO.:178 and the heavy chain variable region may comprise at least 90 consecutive amino acids of SEQ ID NO.:179.
In accordance with an even more specific embodiment of the invention, the light chain variable region may be as set forth in SEQ ID NO.:178 and the heavy chain variable region may be as set forth in SEQ ID NO.:179.
Other exemplary embodiments of the humanized antibodies or antigen binding fragments of the invention are those which may comprise a light chain variable region which may comprise at least 90 consecutive amino acids of any of SEQ ID Nos.
190, 191, 192 or 182.

As used herein the term "at least 90 consecutive amino acids of SEQ ID
NO.:190"
also includes the terms "at least 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106 or at least 107, consecutive amino acids". The term "at least 90 consecutive amino acids of SEQ ID NO.:190" encompasses any possible sequence of at least 90 consecutive amino acids found in SEQ ID NO.:190 and especially those sequences which include the 3 CDRs of SEQ ID NO.:190, for example a sequence comprising amino acids 6 to 102, 11 to 106, 1 to 106, 3 to 95, 5 to 95 of SEQ
ID
NO.:190 and so on.
also includes the terms "at least 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106 or at least 107, consecutive amino acids". The term "at least 90 consecutive amino acids of SEQ ID NO.:191" encompasses any possible sequence of at least 90 consecutive amino acids found in SEQ ID NO.:191 and especially those The terms "at least 90 consecutive amino acids of SEQ ID NO.:192" or "at least consecutive amino acids of SEQ ID NO.:182" has a similar meaning.
the present invention may have, for example, a light chain variable region as set forth in SEQ ID NO.:182.
The humanized antibody or antigen binding fragment of the invention includes (or further includes) for example, a heavy chain variable region which may comprise at As used herein the term "at least 90 consecutive amino acids of SEQ ID
NO.:193"
also includes the terms "at least 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115 or at least consecutive amino acids". The term "at least 90 consecutive amino acids of SEQ
ID

As used herein the term "at least 90 consecutive amino acids of SEQ ID
NO.:195"
also includes the terms "at least 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115 or at least consecutive amino acids". The term "at least 90 consecutive amino acids of SEQ
ID
NO.:195" encompasses any possible sequence of at least 90 consecutive amino acids found in SEQ ID NO.:195 and especially those sequences which include the CDRs of SEQ ID NO.:195, such as, for example a sequence comprising amino acids 3 to 107, 1 to 115, 1 to 110, 22 to 116, 20 to 115 of SEQ ID NO.:195 and so on.
The terms "at least 90 consecutive amino acids of SEQ ID NO.:194" or "at least consecutive amino acids of SEQ ID NO.:183" has a similar meaning.
In accordance with the present invention, the antibody or antigen binding fragment of the present invention may have, for example, a heavy chain variable region as set forth in SEQ ID NO.:183.
In accordance with the present invention the antibody or antigen binding fragment may comprise, for example, a) a light chain variable region which may comprise at least 90 consecutive amino acids of SEQ ID NO.:190 and a heavy chain variable region which may comprise at least 90 consecutive amino acids of any of SEQ ID NO.:193, SEQ ID NO.:194, SEQ ID
NO.:195 or SEQ ID NO.:183;
b) a light chain variable region which may comprise at least 90 consecutive amino acids of SEQ ID NO.:191and a heavy chain variable region which may comprise at least 90 consecutive amino acids of any of SEQ ID NO.:193, SEQ ID NO.:194, SEQ ID
NO.:195 or SEQ ID NO.:183;
c) a light chain variable region which may comprise amino acids at least 90 consecutive amino acids of SEQ ID NO.:192 and a heavy chain variable region which may comprise at least 90 consecutive amino acids of any of SEQ ID NO.:193, SEQ ID NO.:194, SEQ ID
NO.:195 or SEQ ID NO.:183 or;
d) a light chain variable region which may comprise at least 90 consecutive amino acids of SEQ ID NO.:182 and a heavy chain variable region which may comprise at least 90 consecutive amino acids of any of SEQ ID NO.:193, SEQ ID NO.:194, SEQ ID
NO.:195 or SEQ ID NO.:183.
In accordance with a more specific embodiment of the invention, the light chain variable region may have at least 90 consecutive amino acids of SEQ ID NO.:182 and the heavy chain variable region may have at least 90 consecutive amino acids of SEQ ID NO.:183.
In accordance with an even more specific embodiment of the invention, the light chain variable region may be as set forth in SEQ ID NO.:182 and the heavy chain variable region may be as set forth in SEQ ID NO.:183.
The antibody or antigen binding fragment of the present invention may have a light chain variable region and/or heavy chain variable region as described above and may further comprise amino acids of a constant region, such as, for example, amino acids of a constant region of a human antibody.
In an exemplary embodiment, the antibody or antigen binding fragment of the present invention may comprise, for example, a human IgG1 constant region.
In accordance with another exemplary embodiment of the invention, the antigen binding fragment may be, for example, a scFv, a Fab, a Fab' or a (Fab1)2.
In a further aspect, the present invention relates to a humanized antibody or antigen binding fragment described herein for use in the treatment of cancer comprising tumor cells expressing KAAG1 or a KAAG1 variant having at least 80% amino acid sequence identity with KAAG1.
In yet a further aspect, the present invention relates to a humanized antibody or antigen binding fragment described herein for use in the detection of cancer comprising tumor cells expressing KAAG1 or a KAAG1 variant having at least 80%

amino acid sequence identity with KAAG1.
In another aspect, the present invention relates to a humanized antibody or antigen binding fragment described herein for use in preventing or treating metastasis.
In yet another aspect, the present invention relates to an antibody or antigen binding fragment capable of competing with the humanized antibody or antigen binding fragment described herein.

The invention also provides in other aspects, nucleic acids encoding any of the humanized light chain variable region and/or heavy chain variable region of the humanized antibody or antigen binding fragment described herein as well as vectors (e.g., expression vectors) comprising these nucleic acid sequences.
The nucleic acid or vector may encode both the light chain variable region and the heavy chain variable region on the same molecule.
Nucleic acid fragments (of at least 10, 15, 20 etc.) or complements are also encompassed by the present invention. Such fragments or complements may be useful, for example, as probes or primers.
The present invention also relates to (isolated) cells which may comprise nucleic acids encoding any of the humanized light chain variable region and/or heavy chain variable region of the humanized antibody or antigen binding fragment described herein or nucleic acid fragments.
The isolated cell may comprise, for example, a nucleic acid encoding a light chain variable region and a nucleic acid encoding a heavy chain variable region.
The present invention also relates to (isolated) cells which may comprise the humanized light chain variable region and/or heavy chain variable region of the humanized antibody or antigen binding fragment described herein or which expresses the humanized antibody or antigen binding fragment.
The present invention also provides in an additional aspect, a composition (e.g., pharmaceutical composition) which may comprise the humanized antibody or antigen binding fragment described herein and a carrier. Pharmaceutical compositions may comprise a pharmaceutically acceptable carrier.
The present invention also provides in another aspect, a method of treating cancer or metastasis. The method may comprise administering the humanized antibody or antigen binding fragment described herein to a subject in need. The subject in need may have, for example, a cancer or tumor cells expressing KAAG-1 or a KAAG-1 variant.
The present invention further provides in another aspect, a method for reducing tumor spread, reducing tumor invasion, tumor formation or for inducing tumor lysis.
The method may comprise administering the humanized antibody or antigen binding fragment described herein to a subject in need.

In another aspect, the present invention relates to a method of detecting a tumor.
The method may comprise administering the humanized antibody or antigen binding fragment described herein to a subject in need. The tumor may comprise cells expressing KAAG-1 or a KAAG-1 variant.
In yet another aspect, the present invention relates to the use of the humanized antibody or antigen binding fragment described herein in the manufacture of a medicament for treatment of cancer, for reducing tumor spread, for reducing metastasis, for reducing invasion of tumor cells, for reducing formation tumor cells, for inducing tumor lysis of tumor cells and/or for reducing spread of tumor cells.
The present invention also relates to the use of the humanized antibody or antigen binding fragment described herein in the diagnosis of cancer or in the detection of tumor cells. The tumor or cancer may comprise cells expressing KAAG-1 or a KAAG-1 variant.
In accordance with the present invention the subject may have a cancer such as ovarian cancer, skin cancer, renal cancer, colorectal cancer, sarcoma, leukemia, brain tumor, thyroid tumor, breast cancer, prostate cancer, oesophageal tumor, bladder tumor, lung tumor or head and neck tumor.
In a particular embodiment the subject may have, for example, an ovarian cancer.
In a more particular embodiment the subject may have, for example, a recurrent ovarian cancer.
In yet another embodiment, the subject may have, for example, a metastatic cancer.
In accordance with the present invention the cancer or tumor may be, for example, ovarian cancer, skin cancer, renal cancer, colorectal cancer, sarcoma, leukemia, brain tumor, thyroid tumor, breast cancer, prostate cancer, oesophageal tumor, bladder tumor, lung tumor or head and neck tumor.
In a particular embodiment the cancer may be, for example, ovarian cancer.
In a more particular embodiment the cancer may be, for example, recurrent ovarian cancer.
In yet another embodiment, the cancer may be, for example, metastatic.

The present invention also relates in a further aspect to a method for detecting KAAG1 (SEQ ID NO. :2) or a KAAG1 variant having at least 80% sequence identity with SEQ ID NO. :2. The method may comprise, for example, contacting a cell expressing KAAG1 or the KAAG1 variant or a sample comprising or suspected of comprising KAAG1 or the KAAG1 variant with the humanized antibody or antigen binding fragment described herein and measuring binding.
The present invention further relates to a kit which may comprise the humanized antibody or antigen binding fragment described herein.
The sites of greatest interest for substitutional mutagenesis include the hypervariable regions (CDRs), but modifications in the framework region or even in the constant region are also contemplated. Conservative substitutions may be made by exchanging an amino acid (of a CDR, variable chain, antibody, etc.) from one of the groups listed below (group 1 to 6) for another amino acid of the same group.
Other exemplary embodiments of conservative substitutions are shown in Table under the heading of "preferred substitutions". If such substitutions result in a undesired property, then more substantial changes, denominated "exemplary substitutions" in Table 1A, or as further described below in reference to amino acid classes, may be introduced and the products screened.
It is known in the art that variants may be generated by substitutional mutagenesis and retain the biological activity of the polypeptides of the present invention. These variants have at least one amino acid residue in the amino acid sequence removed and a different residue inserted in its place. For example, one site of interest for substitutional mutagenesis may include a site in which particular residues obtained from various species are identical. Examples of substitutions identified as "conservative substitutions" are shown in Table 1A. If such substitutions result in a change not desired, then other type of substitutions, denominated "exemplary substitutions" in Table 1A, or as further described herein in reference to amino acid classes, are introduced and the products screened.
Substantial modifications in function or immunological identity are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation. (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. Naturally occurring residues are divided into groups based on common side chain properties:
(group 1) hydrophobic: norleucine, methionine (Met), Alanine (Ala), Valine (Val), Leucine (Leu), lsoleucine (Ile) (group 2) neutral hydrophilic: Cysteine (Cys), Serine (Ser), Threonine (Thr) (group 3) acidic: Aspartic acid (Asp), Glutamic acid (Glu) (group 4) basic: Asparagine (Asn), Glutamine (Gin), Histidine (His), Lysine (Lys), Arginine (Arg) (group 5) residues that influence chain orientation: Glycine (Gly), Proline (Pro); and (group 6) aromatic: Tryptophan (Trp), Tyrosine (Tyr), Phenylalanine (Phe) Non-conservative substitutions will entail exchanging a member of one of these classes for another.
Table 1A. Amino acid substitution Original residue Exemplary substitution Conservative substitution Ala (A) Val, Leu, Ile Val Arg (R) Lys, Gln, Asn Lys Asn (N) Gin, His, Lys, Arg, Asp Gin Asp (D) Glu, Asn Glu Cys (C) Ser, Ala Ser Gin (Q) Asn; Glu Asn Glu (E) Asp, Gin Asp Gly (G) Ala Ala His (H) Asn, Gin, Lys, Arg, Arg Ile (I) Leu, Val, Met, Ala, Phe, Leu norleucine Leu (L) Norleucine, Ile, Val, Met, Ile Ala, Phe Lys (K) Arg, Gin, Asn Arg Met (M) Leu, Phe, Ile Leu Phe (F) Leu, Val, Ile, Ala, Tyr Tyr Pro (P) Ala Ala Ser (S) Thr Thr Original residue Exemplary substitution Conservative substitution Thr (T) Ser Ser Trp (W) Tyr, Phe Tyr Tyr (Y) Trp, Phe, Thr, Ser Phe Val (V) Ile, Leu, Met, Phe, Ala, Leu norleucine Variation in the amino acid sequence of the variant antibody or antigen binding fragment may include an amino acid addition, deletion, insertion, substitution etc., one or more modification in the backbone or side-chain of one or more amino acid, or an addition of a group or another molecule to one or more amino acids (side-chains or backbone).
Variant antibody or antigen binding fragment may have substantial sequence similarity and/or sequence identity in its amino acid sequence in comparison with that the original antibody or antigen binding fragment amino acid sequence. The degree of similarity between two sequences is based upon the percentage of identities (identical amino acids) and of conservative substitution.
Generally, the degree of similarity and identity between variable chains has been determined herein using the Blast2 sequence program (Tatiana A. Tatusova, Thomas L. Madden (1999), "Blast 2 sequences - a new tool for comparing protein and nucleotide sequences", FEMS Microbiol Lett. 174:247-250) using default settings, i.e., blastp program, BLOSUM62 matrix (open gap 11 and extension gap penalty 1; gapx dropoff 50, expect 10.0, word size 3) and activated filters.
Percent identity will therefore be indicative of amino acids which are identical in comparison with the original peptide and which may occupy the same or similar position.
Percent similarity will be indicative of amino acids which are identical and those which are replaced with conservative amino acid substitution in comparison with the original peptide at the same or similar position.
Variants of the present invention therefore comprise those which may have at least 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity with an original sequence or a portion of an original sequence.

Exemplary embodiments of variants are those having at least 81% sequence identity to a sequence described herein and 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 98%, 97%, 98%, 99% or 100% sequence similarity with an original sequence or a portion of an original sequence.
Other exemplary embodiments of variants are those having at least 82% sequence identity to a sequence described herein and 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence similarity with an original sequence or a portion of an original sequence.
Further exemplary embodiments of variants are those having at least 85%
sequence identity to a sequence described herein and 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence similarity with an original sequence or a portion of an original sequence.
Other exemplary embodiments of variants are those having at least 90% sequence identity to a sequence described herein and 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence similarity with an original sequence or a portion of an original sequence.
Additional exemplary embodiments of variants are those having at least 95%
sequence identity to a sequence described herein and 95%, 96%, 97%, 98%, 99%
or 100% sequence similarity with an original sequence or a portion of an original sequence.
Yet additional exemplary embodiments of variants are those having at least 97%

sequence identity to a sequence described herein and 97%, 98%, 99% or 100%
sequence similarity with an original sequence or a portion of an original sequence.
For a purpose of concision the applicant provides herein a Table 1B
illustrating exemplary embodiments of individual variants encompassed by the present invention and comprising the specified % sequence identity and % sequence similarity.
Each "X" is to be construed as defining a given variant.

Table 1B
Percent (/0) sequence identity (f) (1) 87 X X X X X X X X

er 89 X X X X X X X X X X
a) u) 90 X X X X XXXX XXX

a) 93 X X X X X X X X X X X X X X
f-2 w 94 X X X X X X X XXX X X X X X

98 X X X X _X,X X XXX X X X X XXX X X
99 XXX X .X XXX X X XXX X X XXX X X

The present invention encompasses CDRs, light chain variable domains, heavy chain variable domains, light chains, heavy chains, antibodies and/or antigen binding fragments which comprise at least 80% identity with the sequence described herein.
Exemplary embodiments of the antibody or antigen binding fragment of the present invention are those comprising a light chain variable domain comprising a sequence selected from the group consisting of a sequence at least 70%, 75%, 80%
identical to SEQ ID NO.:16, a sequence at least 70%. 75%, 80% identical to SEQ ID NO.:20, a sequence at least 70%. 75%, 80% identical to SEQ ID NO.:24, a sequence at least 70%. 75%, 80% identical to SEQ ID NO.:105, a sequence at least 70%. 75%, 80%
identical to SEQ ID NO.:109 and a sequence at least 70%. 75%, 80% identical to SEQ ID NO.:126.
These light chain variable domain may comprise a CDRL1 sequence at least 80 %
identical to SEQ ID NO. :27, a CDRL2 sequence at least 80 % identical to SEQ
ID
NO.:28 and a CDRL3 sequence at least 80 % identical to SEQ ID NO.:29.
In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be at least 90 % identical to SEQ ID NO. :27.

In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be 100% identical to SEQ ID NO.:27.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence at least 90 `)/0 identical to SEQ ID
NO.:28.
In yet another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence which may be 100% identical to SEQ ID NO.:28.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be at least 90 %
identical to SEQ ID NO. :29.
In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be 100% identical to SEQ ID NO. :29.
The light chain variable domain listed above may comprise a CDRL1 sequence at least 80 % identical to SEQ ID NO.33, a CDRL2 sequence at least 80 % identical to SEQ ID NO.:34 and a CDRL3 sequence at least 80 % identical to SEQ ID NO.35.
In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be at least 90 % identical to SEQ ID NO.33.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be 100% identical to SEQ ID NO.:33.
In yet a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence which may be at least 90 %
identical to SEQ ID NO.:34.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence which may be 100% identical to SEQ ID NO.:34.

In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be at least 90 A) identical to SEQ ID NO.:35.
In yet another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be 100% identical to SEQ ID NO.35.
The light chain variable domain listed above may comprise a CDRL1 sequence at least 80 % identical to SEQ ID NO.39, a CDRL2 sequence at least 80 % identical to SEQ ID NO.:40 and a CDRL3 sequence at least 80% identical to SEQ ID NO.:41.
In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be at least 90 % identical to SEQ ID NO.:39.
In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be 100% identical to SEQ ID NO.:39.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence which may be at least 90 %
identical to SEQ ID NO.:40.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence which may be 100% identical to SEQ ID NO.:40.
In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be at least 90 %
identical to SEQ ID NO.:41.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be 100% identical to SEQ ID NO.:41.
The light chain variable domain listed above may comprise a CDRL1 sequence at least 80 % identical to SEQ ID NO.:158, a CDRL2 sequence at least 80 %
identical to SEQ ID NO.:159 and a CDRL3 sequence at least 80 % identical to SEQ ID NO.:160.

In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be at least 90 % identical to SEQ ID NO.:158.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be 100% identical to SEQ ID NO.:158.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence which may be at least 90 %
identical to SEQ ID NO.:159.
In yet a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence which may be 100% identical to SEQ ID NO.:159.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be at least 90 %
identical to SEQ ID NO.:160.
In yet another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be 100% identical to SEQ ID NO.:160.
The light chain variable domain listed above may comprise a CDRL1 sequence at least 80 % identical to SEQ ID NO.:164, a CDRL2 sequence at least 80 %
identical to SEQ ID NO.:165 and a CDRL3 sequence at least 80 % identical to SEQ ID NO.:166.
In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be at least 90 % identical to SEQ ID NO.:164.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be 100% identical to SEQ ID NO.:164.
In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence which may be at least 90 %
identical to SEQ ID NO.:165.

In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence which may be 100% identical to SEQ ID NO.:165.
In yet another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be at least 90 %
identical to SEQ ID NO.:166.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be 100% identical to SEQ ID NO.:166.
The light chain variable domain listed above may comprise a CDRL1 sequence at least 80 % identical to SEQ ID NO.:170, a CDRL2 sequence at least 80 %
identical to SEQ ID NO.:171 and a CDRL3 sequence at least 80% identical to SEQ ID NO.:172.
In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be at least 90 % identical to SEQ ID NO.:170.
In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL1 sequence which may be 100% identical to SEQ ID NO.: 170.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence which may be at least 90 %
identical to SEQ ID NO.: 171.
In yet another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL2 sequence which may be 100% identical to SEQ ID NO.: 171.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be at least 90 A
identical to SEQ ID NO.: 172.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRL3 sequence which may be 100% identical to SEQ ID NO.: 172.

An exemplary embodiment of a variant antibody light chain variable region encompasses a light chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:16 and having up to 22 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:16. A SEQ ID NO.:16 variant is provided in SEQ ID NO.:178.
An exemplary embodiment of a variant antibody light chain variable region encompasses a light chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:20 and having up to 22 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:20.
An exemplary embodiment of a variant antibody light chain variable region encompasses a light chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:24 and having up to 21 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:24. A SEQ ID NO.:24 variant is provided in SEQ ID NO.:182.
An exemplary embodiment of a variant antibody light chain variable region encompasses a light chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:105 and having up to 22 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:105.
An exemplary embodiment of a variant antibody light chain variable region encompasses a light chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:109 and having up to 22 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:109.
An exemplary embodiment of a variant antibody light chain variable region encompasses a light chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:126 and having up to 21 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:126.
In some instances, the variant antibody light chain variable region may comprise amino acid deletions or additions (in combination or not with amino acid substitutions). Often 1, 2, 3, 4 or 5 amino acid deletions or additions may be tolerated.
In an exemplary embodiment, the antibody or antigen binding fragment may comprise a heavy chain variable domain comprising a sequence selected from the group consisting of a sequence at least 80% identical to SEQ ID NO.:18, a sequence at least 70%. 75%, 80% identical to SEQ ID NO. :22, a sequence at least 70%. 75%, 80% identical to SEQ ID NO. :26, a sequence at least 70%. 75%, 80% identical to SEQ ID NO.:132, a sequence at least 70%. 75%, 80% identical to SEQ ID NO.: 145 and a sequence at least 70%. 75%, 80% identical to SEQ ID NO.:153.
These heavy chain variable domains may comprise a CDRH1 sequence at least 80 %
identical to SEQ ID NO.:30, a CDRH2 sequence at least 80 c1/0 identical to SEQ
ID
NO.:31 and a CDRH3 sequence at least 80 % identical to SEQ ID NO. :32.
In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be at least 90 % identical to SEQ ID NO.:30.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be 100% identical to SEQ ID NO.:30.
In yet another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be at least 90 %
identical to SEQ ID NO.:31.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be 100% identical to SEQ ID NO.:31.
In yet a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be at least 90 c1/0 identical to SEQ ID NO. :32.

In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be 100% identical to SEQ ID NO. :32.
The heavy chain variable domain listed above may comprise a CDRH1 sequence at least 80 A) identical to SEQ ID NO.:36, a CDRH2 sequence at least 80 A) identical to SEQ ID NO.:37 and a CDRH3 sequence at least 80 % identical to SEQ ID NO. :38.
In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be at least 90 % identical to SEQ ID NO.:36.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be 100% identical to SEQ ID NO. :36.
In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be at least 90 %
identical to SEQ ID NO.:37.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be 100% identical to SEQ ID NO.:37.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be at least 90 %
identical to SEQ ID NO. :38.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be 100% identical to SEQ ID NO. :38.
The heavy chain variable domain listed above may comprise a CDRH1 sequence at least 80 % identical to SEQ ID NO. :42, a CDRH2 sequence at least 80 %
identical to SEQ ID NO.:43 and a CDRH3 sequence at least 80 % identical to SEQ ID NO.44.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be at least 90 A
identical to SEQ ID NO.:42.

In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be 100% identical to SEQ ID NO.:42.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be at least 90 %
identical to SEQ ID NO.43.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be 100% identical to SEQ ID NO.:43.
In yet a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be at least 90 %
identical to SEQ ID NO.:44.
In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be 100% identical to SEQ ID NO.44.
The heavy chain variable domain listed above may comprise a CDRH1 sequence at least 80% identical to SEQ ID NO.:161, a CDRH2 sequence at least 80% identical to SEQ ID NO.:162 and a CDRH3 sequence at least 80 % identical to SEQ ID NO.:163.
In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be at least 90 % identical to SEQ ID NO.:161.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be 100% identical to SEQ ID NO.:161.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be at least 90 %
identical to SEQ ID NO.:162.
In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be 100% identical to SEQ ID NO.:162.

In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be at least 90 %
identical to SEQ ID NO.:163.
In yet another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be 100% identical to SEQ ID NO.:163.
The heavy chain variable domain listed above may comprise a CDRH1 sequence at least 80 % identical to SEQ ID NO.:167, a CDRH2 sequence at least 80 %
identical to SEQ ID NO.:168 and a CDRH3 sequence at least 80 % identical to SEQ ID NO.:169.
In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be at least 90 % identical to SEQ ID NO.:166.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be 100% identical to SEQ ID NO.:166.
In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be at least 90 %
identical to SEQ ID NO.:168.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be 100% identical to SEQ ID NO.:168.
In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be at least 90 A
identical to SEQ ID NO.:169.
In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be 100% identical to SEQ ID
NO.:169.
The heavy chain variable domain listed above may comprise a CDRH1 sequence at least 80 % identical to SEQ ID NO.:173, a CDRH2 sequence at least 80 %
identical to SEQ ID NO.:174 and a CDRH3 sequence at least 80 % identical to SEQ ID NO.:175.

In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be at least 90 % identical to SEQ ID NO.:173.
In an exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH1 sequence which may be 100% identical to SEQ ID
NO.: 173.
In an additional exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be at least 90 %
identical to SEQ ID NO.: 174.
In a further exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH2 sequence which may be 100% identical to SEQ ID NO.: 174.
In another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be at least 90 %
identical to SEQ ID NO.: 175.
In yet another exemplary embodiment of the present invention, any of the antibodies provided herein may comprise a CDRH3 sequence which may be 100% identical to SEQ ID NO.: 175.
An exemplary embodiment of a variant antibody heavy chain variable region encompasses a heavy chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:18 and having up to 22 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:18. A SEQ ID NO.:18 variant is provided in SEQ ID NO.:179.
An exemplary embodiment of a variant antibody heavy chain variable region encompasses a heavy chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:22 and having up to 23 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:22.
An exemplary embodiment of a variant antibody heavy chain variable region encompasses a heavy chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:26 and having up to 23 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:26. A SEQ ID NO.:26 variant is provided in SEQ ID NO.:183.
An exemplary embodiment of a variant antibody heavy chain variable region encompasses a heavy chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:132 and having up to 23 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:132.
An exemplary embodiment of a variant antibody heavy chain variable region encompasses a heavy chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:153 and having up to 23 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:153.
An exemplary embodiment of a variant antibody heavy chain variable region encompasses a heavy chain variable region having CDR amino acid sequences that are 100% identical to the CDR amino acid sequence of SEQ ID NO.:145 and having up to 22 amino acid modifications (e.g., conservative or non-conservative amino acid substitutions) in its framework region in comparison with the framework region of SEQ
ID NO.:145.
In some instances, the variant antibody heavy chain variable region may comprise amino acid deletions or additions (in combination or not with amino acid substitutions). Often 1, 2, 3, 4 or 5 amino acid deletions or additions may be tolerated.
Production of the antibodies in cells The anti-KAAG1 antibodies that are disclosed herein can be made by a variety of methods familiar to those skilled in the art, such as hybridoma methodology or by recombinant DNA methods.
In an exemplary embodiment of the invention, the anti-KAAG1 antibodies may be produced by the conventional hybridoma technology, where a mouse is immunized with an antigen, spleen cells isolated and fused with myeloma cells lacking HGPRT

expression and hybrid cells selected by hypoxanthine, aminopterin and thymine (HAT) containing media.
In an additional exemplary embodiment of the invention, the anti-KAAG1 antibodies may be produced by recombinant DNA methods.
In order to express the anti-KAAG1 antibodies, nucleotide sequences able to encode any one of a light and heavy immunoglobulin chains described herein or any other may be inserted into an expression vector, i.e., a vector that contains the elements for transcriptional and translational control of the inserted coding sequence in a particular host. These elements may include regulatory sequences, such as enhancers, constitutive and inducible promoters, and 5 and 3' un-translated regions.
Methods that are well known to those skilled in the art may be used to construct such expression vectors. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
A variety of expression vector/host cell systems known to those of skill in the art may be utilized to express a polypeptide or RNA derived from nucleotide sequences able to encode any one of a light and heavy immunoglobulin chains described herein.

These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors;
yeast transformed with yeast expression vectors; insect cell systems infected with baculovirus vectors; plant cell systems transformed with viral or bacterial expression vectors; or animal cell systems. For long-term production of recombinant proteins in mammalian systems, stable expression in cell lines may be effected. For example, nucleotide sequences able to encode any one of a light and heavy immunoglobulin chains described herein may be transformed into cell lines using expression vectors that may contain viral origins of replication and/or endogenous expression elements and a selectable or visible marker gene on the same or on a separate vector.
The invention is not to be limited by the vector or host cell employed. In certain embodiments of the present invention, the nucleotide sequences able to encode any one of a light and heavy immunoglobulin chains described herein may each be ligated into a separate expression vector and each chain expressed separately. In another embodiment, both the light and heavy chains able to encode any one of a light and heavy immunoglobulin chains described herein may be ligated into a single expression vector and expressed simultaneously.

Alternatively, RNA and/or polypeptide may be expressed from a vector comprising nucleotide sequences able to encode any one of a light and heavy immunoglobulin chains described herein using an in vitro transcription system or a coupled in vitro transcription/translation system respectively.
In general, host cells that contain nucleotide sequences able to encode any one of a light and heavy immunoglobulin chains described herein and/or that express a polypeptide encoded by the nucleotide sequences able to encode any one of a light and heavy immunoglobulin chains described herein, or a portion thereof, may be identified by a variety of procedures known to those of skill in the art.
These procedures include, but are not limited to, DNA/DNA or DNA/RNA hybridizations, PCR amplification, and protein bioassay or immunoassay techniques that include membrane, solution, or chip based technologies for the detection and/or quantification of nucleic acid or amino acid sequences. Immunological methods for detecting and measuring the expression of polypeptides using either specific polyclonal or monoclonal antibodies are known in the art. Examples of such techniques include enzyme-linked immunosorbent assays (ELISAs), radioimmunoassays (RIAs), and fluorescence activated cell sorting (FACS). Those of skill in the art may readily adapt these methodologies to the present invention.
Host cells comprising nucleotide sequences able to encode any one of a light and heavy immunoglobulin chains described herein may thus be cultured under conditions for the transcription of the corresponding RNA (mRNA, siRNA, shRNA etc.) and/or the expression of the polypeptide from cell culture. The polypeptide produced by a cell may be secreted or may be retained intracellularly depending on the sequence and/or the vector used. In an exemplary embodiment, expression vectors containing nucleotide sequences able to encode any one of a light and heavy immunoglobulin chains described herein may be designed to contain signal sequences that direct secretion of the polypeptide through a prokaryotic or eukaryotic cell membrane.
Due to the inherent degeneracy of the genetic code, other DNA sequences that encode the same, substantially the same or a functionally equivalent amino acid sequence may be produced and used, for example, to express a polypeptide encoded by nucleotide sequences able to encode any one of a light and heavy immunoglobulin chains described herein. The nucleotide sequences of the present invention may be engineered using methods generally known in the art in order to alter the nucleotide sequences for a variety of purposes including, but not limited to, modification of the cloning, processing, and/or expression of the gene product. DNA

shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides may be used to engineer the nucleotide sequences.
For example, oligonucleotide-mediated site-directed mutagenesis may be used to introduce mutations that create new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, and so forth.
In addition, a host cell strain may be chosen for its ability to modulate expression of the inserted sequences or to process the expressed polypeptide in the desired fashion. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation. In an exemplary embodiment, anti-KAAG1 antibodies that contain particular glycosylation structures or patterns may be desired. Post-translational processing, which cleaves a "prepro" form of the polypeptide, may also be used to specify protein targeting, folding, and/or activity. Different host cells that have specific cellular machinery and characteristic mechanisms for post-translational activities (e.g., CHO, HeLa, MDCK, HEK293, and W138) are available commercially and from the American Type Culture Collection (ATCC) and may be chosen to ensure the correct modification and processing of the expressed polypeptide.
Those of skill in the art will readily appreciate that natural, modified, or recombinant nucleic acid sequences may be ligated to a heterologous sequence resulting in translation of a fusion polypeptide containing heterologous polypeptide moieties in any of the aforementioned host systems. Such heterologous polypeptide moieties may facilitate purification of fusion polypeptides using commercially available affinity matrices. Such moieties include, but are not limited to, glutathione S-transferase (GST), maltose binding protein, thioredoxin, calmodulin binding peptide, 6-His (His), FLAG, c-myc, hemaglutinin (HA), and antibody epitopes such as monoclonal antibody epitopes.
In yet a further aspect, the present invention relates to a polynucleotide which may comprise a nucleotide sequence encoding a fusion protein. The fusion protein may comprise a fusion partner (e.g., HA, Fc, etc.) fused to the polypeptide (e.g., complete light chain, complete heavy chain, variable regions, CDRs etc.) described herein.
Those of skill in the art will also readily recognize that the nucleic acid and polypeptide sequences may be synthesized, in whole or in part, using chemical or enzymatic methods well known in the art. For example, peptide synthesis may be performed using various solid-phase techniques and machines such as the ABI

Peptide synthesizer (PE Biosystems) may be used to automate synthesis. If desired, the amino acid sequence may be altered during synthesis and/or combined with sequences from other proteins to produce a variant protein.
Antibody conjugates The antibody or antigen binding fragment of the present invention may be conjugated with a detectable moiety (i.e., for detection or diagnostic purposes) or with a therapeutic moiety (for therapeutic purposes) A "detectable moiety" is a moiety detectable by spectroscopic, photochemical, biochemical, immunochemical, chemical and/or other physical means. A
detectable moiety may be coupled either directly and/or indirectly (for example via a linkage, such as, without limitation, a DOTA or NHS linkage) to antibodies and antigen binding fragments thereof of the present invention using methods well known in the art. A
wide variety of detectable moieties may be used, with the choice depending on the sensitivity required, ease of conjugation, stability requirements and available instrumentation. A suitable detectable moiety include, but is not limited to, a fluorescent label, a radioactive label (for example, without limitation, 1251, ln111, Tc99, 113' and including positron emitting isotopes for PET scanner etc), a nuclear magnetic resonance active label, a luminiscent label, a chemiluminescent label, a chronnophore label, an enzyme label (for example and without limitation horseradish peroxidase, alkaline phosphatase, etc.), quantum dots and/or a nanoparticle. Detectable moiety may cause and/or produce a detectable signal thereby allowing for a signal from the detectable moiety to be detected.
In another exemplary embodiment of the invention, the antibody or antigen binding fragment thereof may be coupled (modified) with a therapeutic moiety (e.g., drug, cytotoxic moiety).
In an exemplary embodiment, the anti-KAAG1 antibodies and antigen binding fragments may comprise a chemotherapeutic or cytotoxic agent. For example, the antibody and antigen binding fragments may be conjugated to the chemotherapeutic or cytotoxic agent. Such chemotherapeutic or cytotoxic agents include, but are not limited to, Yttrium-90, Scandium-47, Rhenium-186, lodine-131, lodine-125, and many others recognized by those skilled in the art (e.g., lutetium (e.g., Lu177), bismuth (e.g., , Bi213s) copper (e.g., Cu67)). In other instances, the chemotherapeutic or cytotoxic agent may be comprised of, among others known to those skilled in the art, 5-fluorouracil, adriamycin, irinotecan, taxanes, pseudomonas endotoxin, ricin and other toxins.
Alternatively, in order to carry out the methods of the present invention and as known in the art, the antibody or antigen binding fragment of the present invention (conjugated or not) may be used in combination with a second molecule (e.g., a secondary antibody, etc.) which is able to specifically bind to the antibody or antigen binding fragment of the present invention and which may carry a desirable detectable, diagnostic or therapeutic moiety.
Pharmaceutical compositions of the antibodies and their use Pharmaceutical compositions of the anti-KAAG1 antibodies or antigen binding fragments (conjugated or not) are also encompassed by the present invention.
The pharmaceutical composition may comprise an anti-KAAG1 antibody or an antigen binding fragment and may also contain a pharmaceutically acceptable carrier.
Other aspects of the invention relate to a composition which may comprise the antibody or antigen binding fragment described herein and a carrier.
The present invention also relates to a pharmaceutical composition which may comprise the antibody or antigen binding fragment described herein and a pharmaceutically acceptable carrier.
Yet other aspects of the invention relate to the use of the isolated antibody or antigen binding fragment described herein in the treatment or diagnosis of ovarian cancer.
In addition to the active ingredients, a pharmaceutical composition may contain pharmaceutically acceptable carriers comprising water, PBS, salt solutions, gelatins, oils, alcohols, and other excipients and auxiliaries that facilitate processing of the active compounds into preparations that may be used pharmaceutically. In other instances, such preparations may be sterilized.
As used herein, "pharmaceutical composition" means therapeutically effective amounts of the agent together with pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvant and/or carriers. A
"therapeutically effective amount" as used herein refers to that amount which provides a therapeutic effect for a given condition and administration regimen. Such compositions are liquids or lyophilized or otherwise dried formulations and include diluents of various buffer content (e.g., Tris-HCI., acetate, phosphate), pH and ionic strength, additives such as albumin or gelatin to prevent absorption to surfaces, detergents (e.g., Tween 20, Tween 80, Pluronic F68, bile acid salts). Solubilizing agents (e.g., glycerol, polyethylene glycerol), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., thimerosal, benzyl alcohol, parabens), bulking substances or tonicity modifiers (e.g., lactose, mannitol), covalent attachment of polymers such as polyethylene glycol to the protein, complexation with metal ions, or incorporation of the material into or onto particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, hydrogels, etc, or onto liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts, or spheroplasts.
Such compositions will influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance. Controlled or sustained release compositions include formulation in lipophilic depots (e.g., fatty acids, waxes, oils).
Also comprehended by the invention are particulate compositions coated with polymers (e.g., poloxamers or poloxamines). Other embodiments of the compositions of the invention incorporate particulate forms protective coatings, protease inhibitors or permeation enhancers for various routes of administration, including parenteral, pulmonary, nasal, oral, vaginal, rectal routes. In one embodiment the pharmaceutical composition is administered parenterally, paracancerally, transmucosally, transdermally, intramuscularly, intravenously, intradermally, subcutaneously, intraperitonealy, intraventricularly, intracranially and intratumorally.
Further, as used herein "pharmaceutically acceptable carrier" or "pharmaceutical carrier" are known in the art and include, but are not limited to, 0.01-0.1 M
or 0.05 M
phosphate buffer or 0.8 % saline. Additionally, such pharmaceutically acceptable carriers may be aqueous or non-aqueous solutions, suspensions, and emulsions.
Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's orfixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers such as those based on Ringer's dextrose, and the like.
Preservatives and other additives may also be present, such as, for example, antimicrobials, antioxidants, collating agents, inert gases and the like.
For any compound, the therapeutically effective dose may be estimated initially either in cell culture assays or in animal models such as mice, rats, rabbits, dogs, or pigs.

An animal model may also be used to determine the concentration range and route of administration. Such information may then be used to determine useful doses and routes for administration in humans. These techniques are well known to one skilled in the art and a therapeutically effective dose refers to that amount of active ingredient that ameliorates the symptoms or condition. Therapeutic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or with experimental animals, such as by calculating and contrasting the ED50 (the dose therapeutically effective in 50% of the population) and LD50 (the dose lethal to 50% of the population) statistics. Any of the therapeutic compositions described above may be applied to any subject in need of such therapy, including, but not limited to, mammals such as dogs, cats, cows, horses, rabbits, monkeys, and humans.
The pharmaceutical compositions utilized in this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, or rectal means.
The term "treatment" for purposes of this disclosure refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) the targeted pathologic condition or disorder. Those in need of treatment include those already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented.
The anti-KAAG1 antibodies and antigen binding fragments thereof may have therapeutic uses in the treatment of various cancer types, such as ovarian cancer, renal cancer, colon cancer, lung cancer, melanoma, etc. In an exemplary embodiment, the antibodies and fragments have therapeutic uses in ovarian cancer.
In certain instances, the anti-KAAG1 antibodies and fragments may interact with cancer cells that express KAAG1 and induce an immunological reaction by mediating ADCC. In other instances, the anti-KAAG1 antibodies and fragments may block the interaction of KAAG1 with its protein partners.
The anti-KAAG1 antibodies and antigen binding fragments thereof may have therapeutic uses in the treatment of various types of ovarian cancer. Several different cell types may give rise to different ovarian cancer histotypes. The most common form of ovarian cancer is comprised of tumors that originate in the epithelial cell layer of the ovary or the fallopian tube. Such epithelial ovarian cancers include serous tumors, endonnetroid tumors, mucinous tumors, clear cell tumors, and borderline tumors. In other embodiments, the anti-KAAG1 antibodies and antigen binding fragments thereof have uses in the treatment of other types of ovarian cancer such as germ line and sex cord ovarian cancer.
In certain instances, the anti-KAAG1 antibodies and antigen binding fragments thereof may be administered concurrently in combination with other treatments given for the same condition. As such, the antibodies may be administered with anti-mitotics (eg., taxanes), platinum-based agents (eg., cisplatin), DNA damaging agents (eg.
Doxorubicin) and other anti-cancer therapies that are known to those skilled in the art.
In other instances, the anti-KAAG1 antibodies and antigen binding fragments thereof may be administered with other therapeutic antibodies. These include, but are not limited to, antibodies that target EGFR, CD-20, and Her2.
The present invention relates in a further aspect thereof to a method for inhibiting the growth of a KAAG1-expressing cell, the method which may comprise contacting the cell with an effective amount of the antibody or antigen binding fragment described herein.
The present invention also encompasses method of treating cancer or inhibiting the growth of a KAAG1 expressing cells in a mammal, the method may comprise administering the antibody or antigen binding fragment described herein to a mammal in need.
In further aspects, the present invention provides method of treatment, diagnostic methods and method of detection using the antibody or antigen binding fragment of the present invention and the use of these antibodies or antigen binding fragment in the manufacture of a pharmaceutical composition or drug for such purposes.
Method of treatment encompassed by the present invention includes administering an antibody or antigen binding fragment described herein to a mammal in need, and especially to a patient having or susceptible of having a cancer.
The invention also provides in further aspects, methods for reducing tumor spread, tumor invasion, tumor formation or for inducing tumor lysis, which may comprise administering an isolated antibody or antigen binding fragment to a mammal in need.
The invention therefore relates to the use of the isolated antibody or antigen binding fragment described herein in the (manufacture of a pharmaceutical composition for) treatment of cancer, reduction of tumor spread, tumor invasion, tumor formation or for inducing tumor lysis of KAAG1-expressing tumor cells.
The antibody or antigen binding fragment may more particularly be applicable for malignant tumor including, for example, a malignant tumor having the ability to metastasize and/or tumor cells characterized by anchorage-independent growth.The antibody or antigen binding fragment of the present invention may also be used in the diagnosis of cancer. The diagnosis of cancer may be performed in vivo by administering the antibody or antigen binding fragment of the present invention to a mammal having or suspected of having a cancer. The diagnosis may also be performed ex vivo by contacting a sample obtained from the mammal with the antibody or antigen binding fragment and determining the presence or absence of cells (tumor cells) expressing KAAG1 or a KAAG1 variant.
The present invention also encompasses method of detecting cancer or detecting a KAAG1 expressing cells in a mammal, the method may comprise administering the antibody or antigen binding fragment described herein to a mammal in need.
The present invention relates in another aspect thereof to a method for detecting a cell expressing KAAG1 or a KAAG1 variant, the method may comprise contacting the cell with an antibody or antigen binding fragment described herein and detecting a complex formed by the antibody and the KAAG1-or KAAG1 variant-expressing cell.
Exemplary embodiments of antibodies or antigen binding fragments used in detection methods are those which are capable of binding to the extracellular region of KAAG1.
Other exemplary embodiments of antibodies or antigen binding fragments used in detection methods are those which bind to KAAG1 or KAAG1 variant expressed at the surface of a tumor cells.
Patients which would benefit from treatment, detection or diagnostic methods described herein are those which have or are suspected of having ovarian cancer (e.g., serous, endometroid, clear cell or mucinous), skin cancer (e.g., melanomas, squamous cell carcinomas), renal cancer (e.g., papillary cell carcinomas, clear cell carcinomas), colorectal cancer (e.g., colorectal carcinomas), sarcoma, leukemia, brain tumor, thyroid tumor, breast cancer (e.g., mammary carcinomas), prostate cancer (e.g., prostatic carcinomas), oesophageal tumor, bladder tumor, lung tumor (e.g., lung carcinomas) or head and neck tumor and especially when the cancer is characterized as being malignant and/or when the cells expressing KAAG1 or a KAAG1 variant are characterized by anchorage-independent growth.
Especially encompassed by the present invention are patients having or susceptible of having ovarian cancer (e.g., serous, endometroid, clear cell or mucinous), skin cancer (e.g., melanomas, squamous cell carcinomas) or renal cancer (e.g., papillary cell carcinomas) and especially when the cancer is characterized as being malignant and/or when the cells expressing KAAG1 or a KAAG1 variant are characterized by anchorage-independent growth.
Another aspect of the invention relates a method for detecting KAAG1 (SEQ ID
NO.:2), a KAAG1 variant having at least 80% sequence identity with SEQ ID
NO.:2 or a secreted form of circulating form of KAAG1 or KAAG1 variant, the method may comprise contacting a cell expressing KAAG1 or the KAAG1 variant or a sample (biopsy, serum, plasma, urine etc.) comprising or suspected of comprising KAAG1 or the KAAG1 variant with the antibody or antigen binding fragments described herein and measuring binding. The sample may originate from a mammal (e.g., a human) which may have cancer (e.g., ovarian cancer, a metastatic cancer) or may be suspected of having cancer (e.g., ovarian cancer, a metastatic cancer). The sample may be a tissue sample obtained from the mammal or a cell culture supernatant.
In accordance with the invention the sample may be a serum sample, a plasma sample, a blood sample or ascitic fluid obtained from the mammal. The antibody or antigen binding fragment described herein may advantageously detect a secreted or circulating form (circulating in blood) of KAAG1.
The method may comprise quantifying the complex formed by the antibody or antigen binding fragment bound to KAAG1 or to the KAAG1 variant.
The binding of an antibody to an antigen will cause an increase in the expected molecular weight of the antigen. A physical change therefore occurs upon specific binding of the antibody or antigen binding fragment and the antigen.
Such changes may be detected using, for example, electrophoresis followed by Western blot and coloration of the gel or blot, mass spectrometry, HPLC
coupled with a computer or else. Apparatus capable of computing a shift in molecular weight are known in the art and include for example, PhosphorimagerTM.

When the antibody comprises for example a detectable label, the antigen-antibody complex may be detected by the fluorescence emitted by the label, radiation emission of the label, enzymatic activity of a label provided with its substrate or else.
Detection and/or measurement of binding between an antibody or antigen binding fragment and an antigen may be performed by various methods known in the art.
Binding between an antibody or antigen binding fragment and an antigen may be monitored with an apparatus capable of detecting the signal emitted by the detectable label (radiation emission, fluorescence, color change etc.). Such apparatus provides data which indicates that binding as occurred and may also provide indication as to the amount of antibody bound to the antigen. The apparatus (usually coupled with a computer) may also be capable of calculating the difference between a background signal (e.g., signal obtained in the absence of antigen-antibody binding) or background noise and the signal obtained upon specific antibody-antigen binding.
Such apparatuses may thus provide the user with indications and conclusions as to whether the antigen has been detected or not.
Additional aspects of the invention relates to kits which may include one or more container containing one or more antibodies or antigen binding fragments described herein.
Nucleic acids, vectors and cells Antibodies are usually made in cells allowing expression of the light chain and heavy chain expressed from a vector(s) comprising a nucleic acid sequence encoding the light chain and heavy chain.
The present therefore encompasses nucleic acids capable of encoding any of the CDRs, light chain variable domains, heavy chain variable domains, light chains, heavy chains described herein.
The present invention therefore relates in a further aspect to a nucleic acid encoding a light chain variable domain and/or a heavy chain variable domain of an antibody which is capable of specific binding to KAAG1.
In accordance with an embodiment of the invention, the nucleic acid may especially encode a light chain variable domain and/or heavy chain variable domain of an antibody which may be capable of inducing killing (elimination, destruction, lysis) of KAAG1- or KAAG1 variant-expressing tumor cells.

In accordance with another embodiment of the invention, the nucleic acid may especially encode a light chain variable domain and/or heavy chain variable domain of an antibody which may be capable of reducing spreading of KAAG1- or KAAG1 variant-expressing tumor cells.
In accordance with yet another embodiment of the invention, the nucleic acid may particularly encode a light chain variable domain and/or heavy chain variable domain of an antibody which may be capable of decreasing or impairing formation of or KAAG1 variant-expressing tumors.
Exemplary embodiments of nucleic acids of the present invention include nucleic acids encoding a light chain variable domain comprising:
a. a CDRL1 sequence selected from the group consisting of SEQ ID
NO.:74 and SEQ ID NO.:75;
b. a CDRL2 sequence selected from the group consisting of SEQ ID
NO.:76, SEQ ID NO.: 77 and SEQ ID NO.:78, or;
c. a CDRL3 sequence selected from the group consisting of SEQ ID
NO.:79, SEQ ID NO.:80 and SEQ ID NO.:81.
In accordance with the present invention, the nucleic acid may encode a light chain variable domain which may comprise at least two CDRs of a CDRL1, a CDRL2 or a CDRL3.
Also in accordance with the present invention, the nucleic acid may encode a light chain variable domain which may comprise one CDRL1, one CDRL2 and one CDRL3.
The present invention also relates to a nucleic acid encoding a heavy chain variable domain comprising:
a. a CDRH1 sequence comprising SEQ ID NO.:82;
b. a CDRH2 sequence selected from the group consisting of SEQ ID
NO.:83, SEQ ID NO.:84, SEQ ID NO.:85, SEQ ID NO.:86 and SEQ ID
NO. :87, or;
c. a CDRH3 sequence selected from the group consisting of SEQ ID
NO.:88, SEQ ID NO.:89 and SEQ ID NO.:90.
In accordance with the present invention, the nucleic acid may encode a heavy chain variable domain which may comprise at least two CDRs of a CDRH1, a CDRH2 or a CDRH3.

In accordance with the present invention, the nucleic acid may encode a heavy chain variable domain which may comprise one CDRH1, one CDRH2 and one CDRH3.
Also encompassed by the present invention are nucleic acids encoding antibody variants having at least one conservative amino acid substitution.
In accordance with the present invention, the nucleic acid may encode a CDR
comprising at least one conservative amino acid substitution.
In accordance with the present invention, the nucleic acid may encode a CDR
comprising at least one conservative amino acid substitution in at least two of the CDRs.
In accordance with the present invention, the nucleic acid may encode a CDR
comprising at least one conservative amino acid substitution in the 3 CDRs.
In accordance with the present invention, the nucleic acid may encode a CDR
comprising at least two conservative amino acid substitutions in at least one of the CDRs.
In accordance with the present invention, the nucleic acid may encode a CDR
comprising at least two conservative amino acid substitutions in at least two of the CDRs.
In accordance with the present invention, the nucleic acid may encode a CDR
comprising at least two conservative amino acid substitutions in the 3 CDRs.
Other aspects of the invention relate to a nucleic acid encoding a light chain variable domain having at least 70%. 75%, 80% sequence identity to a sequence selected from the group consisting of SEQ ID NO.:16, SEQ ID NO.:20, SEQ ID NO.:24, SEQ
ID NO.:105, SEQ ID NO.:106, SEQ ID NO.:107, SEQ ID NO.:108, SEQ ID NO.:109, SEQ ID NO.:110, SEQ ID NO.:111, SEQ ID NO.:112, SEQ ID NO.:113, SEQ ID
NO.:114, SEQ ID NO.:115, SEQ ID NO.:116, SEQ ID NO.:117, SEQ ID NO.:118, SEQ ID NO.:119, SEQ ID NO.:120, SEQ ID NO.:121, SEQ ID NO.:122, SEQ ID
NO.:123, SEQ ID NO.:124, SEQ ID NO.:125, SEQ ID NO.:126, SEQ ID NO.:127.
SEQ ID NO.:128, SEQ ID NO.:129, SEQ ID NO.:130 and SEQ ID NO.:131.
Yet other aspects of the invention relate to a nucleic acid encoding a heavy chain variable domain having at least 70%. 75%, 80% sequence identity to a sequence selected from the group consisting of SEQ ID NO.:18, SEQ ID NO.:22, SEQ ID

NO.:26, SEQ ID NO.:132, SEQ ID NO.:133, SEQ ID NO.:134, SEQ ID NO.:135, SEQ
ID NO.:136, SEQ ID NO.:137, SEQ ID NO.:138, SEQ ID NO.:139, SEQ ID NO.:140, SEQ ID NO.:141, SEQ ID NO.:142, SEQ ID NO.:143, SEQ ID NO.:144, SEQ ID
NO.:145, SEQ ID NO.:146, SEQ ID NO.:147, SEQ ID NO.:148, SEQ ID NO.:149, SEQ ID NO.:150, SEQ ID NO.:151, SEQ ID NO.:152, SEQ ID NO.:153, SEQ ID
NO.:154, SEQ ID NO.:155, SEQ ID NO.:156, SEQ ID NO.:157.0ther aspects of the invention relates to the use of a nucleic acid selected from the group consisting of SEQ ID NO.:1, a fragment of 10 to 884 nucleotides of SEQ ID NO.:1 and a complement of any of the preceding for impairing migration or survival of tumor cells expressing KAAG1 or a KAAG1 variant. Exemplary embodiments of such nucleic acid comprise siRNAs, antisense, ribozymes and the like.
In yet another aspect, the present invention relates to a vector comprising the nucleic acids described herein.
In accordance with the present invention, the vector may be an expression vector.
Vector that contains the elements for transcriptional and translational control of the inserted coding sequence in a particular host are known in the art. These elements may include regulatory sequences, such as enhancers, constitutive and inducible promoters, and 5' and 3' un-translated regions. Methods that are well known to those skilled in the art may be used to construct such expression vectors. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
In another aspect the present invention relates to an isolated cell which may comprise the nucleic acid described herein.
The isolated cell may comprise a nucleic acid encoding a light chain variable domain and a nucleic acid encoding a heavy chain variable domain either on separate vectors or on the same vector. The isolated cell may also comprise a nucleic acid encoding a light chain and a nucleic acid encoding a heavy chain either on separate vectors or on the same vector.
In accordance with the present invention, the cell may be capable of expressing, assembling and/or secreting an antibody or antigen binding fragment thereof.
In another aspect, the present invention provides a cell which may comprise and/or may express the antibody described herein.

In accordance with the invention, the cell may comprise a nucleic acid encoding a light chain variable domain and a nucleic acid encoding a heavy chain variable domain.
The cell may be capable of expressing, assembling and/or secreting an antibody or antigen binding fragment thereof.
The examples below are presented to further outline details of the present invention.
EXAMPLES
Example *1 This example describes the pattern of expression of the KAAG1 gene in ovarian tumors and ovarian cancer cell line.
PCR analysis was performed to verify the percentage of ovarian tumors that express the mRNA encoding KAAG1 (indicated as AB-0447 in the Figure). The results showed that the KAAG1 gene is expressed in greater than 85% of ovarian tumors from all stages of the disease and 100% of late stage tumors. The expression of KAAG1 is lower or undetectable in LMP samples (see Fig. 1A). For each sample, p.g of amplified RNA was reverse transcribed with random hexamers using Thermoscript RT (Invitrogen). The cDNA was diluted and 1/200th of the reaction was used as template for each FOR reaction with gene-specific primers as indicated. The primers used to amplify the KAAG1 mRNA contained the sequences shown in SEQ
ID NOS:45 and 46. FOR reactions were carried out in 96-well plates and half of the .1 reaction was electrophoresed on a 1% agarose gel. The gels were visualized and photographed with a gel documentation system (BioRad). The upper panel of Fig. 1A shows the results from 6 LMP samples (LMP) and 22 ovarian tumor and 6 25 ovarian cell line (last 6 lanes on the right, OVCa) samples. The lower panel of Fig. 1 shows the RNA samples from 30 normal tissues that were tested as indicated.
KAAG1 expression was weakly detected in a few normal tissues whereas the mRNA
was evident in the fallopian tube and the pancreas (see Fig. 1A). The amount of total RNA used in these reactions was controlled with parallel FOR amplifications of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a housekeeping gene, and the results showed that equivalent starting material was present in each sample (see Fig. 1A). The primers used to amplify the GAPDH gene contained the sequences shown in SEQ ID NOs: 47 and 48. Thus, the expression of the KAAG1 gene fulfills an important selection criteria: it is over-expressed in a large proportion of ovarian tumors and its expression is low or absent in most normal tissues. These data suggest that ovarian tumors may be specifically targeted with high affinity monoclonal antibodies against KAAG1.
Early stage cancer or tumors tend to be made up of cells that are in a high state of differentiation but as the tumor progresses to a more aggressive and invasive state, the cancer cells become increasingly undifferentiated. There are needs to identify factors that contribute to this transition and exploit these proteins as targets for the development of therapeutics. Several ovarian cancer cell lines are available that were derived from primary tumors and serve as excellent models for the functional studies.
The expression of KAAG1 was examined in these cell lines. Four lines termed TOV-21G, TOV-112D, TOV-1946, and TOV-2223G were established from primary tumors whereas OV-90 and OV-1946 are cell lines derived from cells contained in ascites fluid of patients with advanced ovarian cancer. Total RNA from cells established from primary tumors (see in Fig. 1B, lanes 1, TOV-21G; 2, TOV-112D; 5, TOV-1946; 6, TOV-2223G) and cells established from ascitic cells (lanes 3, OV-90; 4, OV-1946) was converted to cDNA with reverse transcriptase and used as template in PCR
reactions with KAAG1-specific primers( SEQ ID NOS:45 and 46). As a negative control, the reaction was carried out with total RNA from normal ovary. Equal amounts of starting material were utilized as evidenced by parallel PCR
reactions with GAPDH (SEQ ID NOS:47 and 48). A sample of the PCR reaction was electrophoresed on an agarose gel and visualized with ethidium bromide. As shown in Fig. 1B, KAAG1 was detectable but weakly expressed in the cell lines from the primary tumors and PCR reactions performed at a higher number of cycles revealed the KAAG1 transcript in all four of these cell lines. Conversely, both cell lines established from the ascitic fluid cells exhibited high level of the KAAG1 transcript.
The increased expression in cells from the ascitic fluid suggests that the environment of the cells influences the regulation of the KAAG1 gene.
Ascitic cells are associated with advanced disease and the pattern of expression disclosed in Fig. 1B implies that increased KAAG1 levels are associated with anchorage-independent growth. This question was addressed by culturing the cells in hanging droplets, a condition that prevents the cells from adhering to the petri dish, as is the case when they are grown as monolayers. These so called three-dimensional cultures allow the cells to associate and the formation of spheroids is observed (see Fig. 1C). Spheroids were cultures as follows: TOV-112D, OV-90, or TOV-21G
cells (4 000 in 15 1.1,1) were incubated for 4 days in medium in the absence (left panels, Fig.
10) or presence of 5% FBS (right panels, Fig. 1C, +5% serum). The magnification of the image was set to 100x. These spheroids have been extensively characterized and exhibit many of the properties found in primary tumors including morphological and functional properties as well as the molecular signature as measured by microarray-based expression profiling.
Total RNA was isolated from spheroid preparations and RT-PCR was performed as described for Fig. 1A. TOV-21G, TOV-112D, OV-90 cells were seeded as described in the legend for Fig. 10 under conditions to produce spheroids. After 4 days, total RNA was isolated and used to perform RT-PCR reactions with KAAG1-specific primers (SEQ ID NOS:45 and 46). PCR reactions were electrophoresed on agarose gels. Conducting parallel reactions to amplify GAPDH (SEQ ID NOS:47 and 48) demonstrated that equal amounts of starting material were present in each sample.
The following acronyms are used in Fig. 1D: Ce., cells grown as monolayers;
Sph., cells grown as spheroids. Strikingly, KAAG1 expression was up-regulated when TOV-21G and TOV-112D were grown as spheroids (see Fig. 1D). In the case of the OV-cells, the level of expression of the KAAG1 gene was unchanged and remained very high. Presumably, the level of expression attained in the cell lines derived from the ascitic fluid, as exemplified by the OV-90 cells and the OV-1946 cells (see Fig. 1A) has reached a maximum.
These results correlated with the previous data showing high expression in cell lines derived from ascitic fluid and confirm that expression of KAAG1 is influenced by the microenvironment of the cancer cells. Additionally, the up-regulation of KAAG1 transcription that was observed in spheroids implies that high levels of KAAG1 are present in malignant ovarian cancer.
Example 2 This example describes in vitro results that suggest a critical role for KAAG1 in the survival of ovarian cancer cells.
With the demonstration that KAAG1 expression is regulated in ovarian cancer cells, the function of this gene in these cells was examined. To address this question, in vitro assays were conducted to determine if this protein plays a role in cancer cell proliferation, migration, and/or survival. RNAi was used to knock down the expression of the endogenous KAAG1 gene in the TOV-21G ovarian cancer cell line. The design of two separate short-hairpin RNA (shRNA) sequences was performed using web-based software that is freely available to those skilled in the art (Qiagen for example).

These chosen sequences, usually 19-mers, were included in two complementary oligonucleotides that form the template for the shRNAs, i.e. the 19-nt sense sequence, a 9-nt linker region (loop), the 19-nt antisense sequence followed by a 5-6 poly-T tract for termination of the RNA polymerase III. The sequences of the 19-mers that were used to knock down the expression of KAAG1 are shown in SEQ ID
NOS:49 and 50. Appropriate restriction sites were inserted at the ends of these oligonucleotides to facilitate proper positioning of the inserts so that the transcriptional start point is at a precise location downstream of the hU6 promoter. The plasmid utilized in all RNA interference studies, pSilencer 2.0 (SEQ ID NO.:51), was purchase from a commercial supplier (Ambion, Austin, TX). Two different shRNA
expression vectors were constructed to increase the chance of observing RNAi effects and the specificity of phenotypic observations. TOV-21G cells were seeded in 6-well plates and transfected 24h later with 1 lig of pSil-shRNA vector. Sh.1 and sh.2 were used to designate 2 different shRNA sequences targeting the KAAG1 gene. Stable transfectants were selected for 5 ¨ 7 days, expanded, and grown to confluence.
All of the following in vitro cell-based assays were performed using these stably transfected cell lines that contain shRNAs specific for KAAG1.
The migration or mobility of the cells was measured in a standard cell motility assay.
This scratch assay, as it is called, measures the speed at which cells fill a denuded area in a confluent monolayer. As illustrated in Fig. 2A, TOV-21G cells containing the scrambled shRNA filled up the wound almost completely after 24h compared to the control untreated cells (compare middle-left panel with left panel). By contrast, the ability of TOV-21G cells expressing KAAG1 shRNAs to fill the denuded area was greatly reduced. In fact, the number of cells that filled the denuded area in the presence of the KAAG1 shRNA cells more closely resembled the number of cells at time Oh (compare the left panel with the right panels).
To examine the longer-term effects of reduced expression of KAAG1 in ovarian cancer cells, the cells were extensively diluted and cultured for 10 days in a colony survival assay. TOV-21G cells were seeded in 12-well plates at a density of 50 cells/well and transfected 24h later with 1 1_19 of pSil-shRNA vector. Sh-1 and sh-2 are used to designate 2 different shRNA sequences targeting the same gene. The next day, fresh medium was applied containing 2 g/m1 puromycin and the selection of the cells was carried out for 3 days. The cells were washed and fresh medium without puromycin was added and growth continued for another 5 days. To visualize the remaining colonies, the cells were washed in PBS and fixed and stained simultaneously in 1% crystal violet/10% ethanol in PBS for 15 minutes at room temperature. Following extensive washing in PBS, the dried plates were scanned for photographic analysis. A significant decrease in the survival of the cancer cell line was observed and a representative experiment is displayed in Fig. 2B.
Identical results were obtained when the shRNAs were transfected into another ovarian cancer cell line, TOV-112D.
Thus, taken together, the regulated expression of KAAG1 in detached cells coupled with the requirement of this gene in the migration and the survival of ovarian cancer cells supports an important role for KAAG1 in ovarian cancer cells.
Furthermore, these experiments suggest that an antagonist of KAAG1 protein, such as a monoclonal antibody, would result in reduced invasiveness and decreased tumor survival.
Example 3 This example provides details pertaining to the family of monoclonal antibodies that bind to KAAG1.
The antibodies that bind KAAG1 were generated using the Biosite phage display technology. A detailed description of the technology and the methods for generating these antibodies can be found in the U.S. Patent No. 6,057,098. Briefly, the technology utilizes stringent panning of phage libraries that display the antigen binding fragments (Fabs). After a several rounds of panning, a library, termed the Omniclonal, was obtained that was enriched for recombinant Fabs containing light and heavy chain variable regions that bound to KAAG1 with very high affinity and specificity. From this library, more precisely designated Omniclonal AL0003Z1, individual recombinant monoclonal Fabs were prepared from E. coli and tested for KAAG1 binding.
To measure the relative binding of each individual monoclonal antibody, recombinant human KAAG1 was produced in 293E cells using the large-scale transient transfection technology (Durocher et al., 2002; Durocher, 2004). The entire coding region of the KAAG1 cDNA was amplified by PCR using a forward primer that incorporated a BamHI restriction site (SEQ ID NO. :52) and a reverse primer that incorporated a HindlIl restriction site (SEQ ID NO.:53). The resulting PCR
product measured 276 base pairs and following digestion with BamHI and HindIII, the fragment was ligated into the expression vector pYD5 (SEQ ID NO.:54) that was similarly digested with the same restriction enzymes. The pYD5 expression plasmid contains the coding sequence for the human Fc domain that allows fusion proteins to be generated as well as the sequence encoding the IgG1 signal peptide to allow the secretion of the fusion protein into the culture medium. For each milliliter of cells, one microgram of the expression vector, called pYD5-0447, was transfected in 293E
cells grown in suspension to a density of 1.5 ¨ 2.0 million cells/ml. The transfection reagent used was polyethylenimine (PEI), (linear, MW 25,000, Cat# 23966 Polysciences, Inc., Warrington, PA) which was included at a DNA:PEI ratio of 1:3. Growth of the cells was continued for 5 days after which the culture medium was harvested for purification of the recombinant Fc-KAAG1 fusion protein. The protein was purified using Protein-A agarose as instructed by the manufacturer (Sigma-Aldrich Canada Ltd., Oakville, ON). A representative polyacrylamide gel showing a sample of the purified Fc-KAAG1 (indicated as Fc-0447) is shown in Fig. 3A.
The 96-well master plate of monoclonal preparations contained different concentrations of purified anti-KAAG1 Fabs in each well. A second stock master plate was prepared by diluting the Fabs to a final concentration of 10 jig/m1 from which all subsequent dilutions were performed for ELISA measurements. To carry out the binding of Fc-KAAG1 to the monoclonal preparations, the Fc-KAAG1 was biotinylated with NHS-biotin (Pierce, Rockford, IL) and 10 ng/well was coated in a streptavidin 96-well plate. One nanogram of each Fab monoclonal preparation was added to each well and incubated at room temperature for 30 minutes. Bound antibody was detected with HRP-conjugated mouse anti-kappa light chain antibody in the presence of TMB
liquid substrate (Sigma-Aldrich Canada Ltd., Oakville, ON) and readings were conducted at 450 nm in microtiter plate reader. As shown in Fig. 3B, a total of 48 (highlighted in grey) monoclonal antibodies displayed significant binding in this assay (>0.1 arbitrary 012150 units). The antibodies were purposely diluted to 1 ng/well to accentuate the binding of those antibodies with the most affinity for KAAG1.
As a control, the antibodies did not bind to biotinylated Fc domain. These data also revealed that the binding of the antibodies varied from well to well indicating that they exhibited different affinities for KAAG1.
Example 4 This example describes the epitope mapping studies to determine which region of KAAG1 the antibodies bind to.
To further delineate the regions of KAAG1 that are bound by the monoclonal antibodies, truncated mutants of KAAG1 were expressed and used in the ELISA.
As for the full length KAAG1, the truncated versions were amplified by PCR and ligated into BamHI/Hind111 digested pYD5. The primers that were used combined the forward oligonucleotide with the sequence shown in SEQ ID NO. :52 with primers of SEQ
ID
NOS:55 and 56, to produce Fc-fused fragments that ended at amino acid number and 35 of KAAG1, respectively. The expression of these mutants was conducted as was described above for the full length Fc-KAAG1 and purified with Protein-A
agarose. A representative gel of the protein preparations that were used in the ELISA
is shown in Fig. 4A and a schematic of the mutant proteins used for epitope mapping is depicted in Fig. 4B.
The results showed that the library was comprised of antibodies that could bind to each of the delineated KAAG1 regions. In particular, of the 48 mAbs that bound to KAAG1 in the first ELISA, nine (wells A2, Al2, C2, 04, D1, E10, Fl, H3, and H8) were found to interact with the first 35 amino acids of KAAG1 whereas five (D12, E8, F5, G10, and H5) were found to interact with the last 25 amino acids of KAAG1.
Thus, the remaining 34 antibodies interacted with a region of KAAG1 spanned by amino acids 36 ¨ 59. These results were in agreement with the sequence analysis of representative light and heavy chain variable regions. Indeed, alignment of these sequences revealed that the antibodies clustered into three groups based on the percentage identity in their respective CDRs. Antibodies contained in each cluster all interacted with the same region of KAAG1.
Therefore, based on the relative binding affinity of the mAb, differential epitope interaction characteristics, and the differences in variable domain sequences, three antibodies from the plate described in Example 3 were selected for further analysis as exemplary anti-KAAG1 monoclonal antibodies.
Example 5 This example discloses the methods used to convert the Fabs into full IgG1 chimeric monoclonal antibodies. A scheme of the methodology is presented in Fig. 5.
Aside from the possibility of conducting interaction studies between the Fab monoclonals and the KAAG1 protein, the use of Fabs is limited with respect to conducting meaningful in vitro and in vivo studies to validate the biological function of the antigen. Thus, it was necessary to transfer the light and heavy chain variable regions contained in the Fabs to full antibody scaffolds, to generate mouse-human chimeric IgG1s. The expression vectors for both the light and heavy immunoglobulin chains were constructed such that i) the original bacterial signal peptide sequences upstream of the Fab expression vectors were replaced by mammalian signal peptides and ii) the light and heavy chain constant regions in the mouse antibodies were replaced with human constant regions. The methods to accomplish this transfer utilized standard molecular biology techniques that are familiar to those skilled in the art. A brief overview of the methodology is described here (see Fig. 5).
Light chain expression vector ¨ an existing mammalian expression plasmid, called pTTVH8G (Durocher et al., 2002), designed to be used in the 293E transient transfection system was modified to accommodate the mouse light chain variable region. The resulting mouse-human chimeric light chain contained a mouse variable region followed by the human kappa constant domain. The cDNA sequence encoding the human kappa constant domain was amplified by PCR with primers 0GS1773 and 0GS1774 (SEQ ID NOS:57 and 58, respectively). The nucleotide sequence and the corresponding amino acid sequence for the human kappa constant region are shown in SEQ ID NOS:59 and 60, respectively. The resulting 321 base pair FOR product was ligated into pTTVH8G immediately downstream of the signal peptide sequence of human VEGF A (NM_003376). This cloning step also positioned unique restriction endonuclease sites that permitted the precise positioning of the cDNAs encoding the mouse light chain variable regions. The sequence of the final expression plasmid, called pTTVK1, is shown in SEQ ID NO.:61. Based on the sequences disclosed in Table 3, FOR primers specific for the light chain variable regions of antibodies 3D3, 3G10, and 3C4 (SEQ ID NOS:15, 19, and 23, respectively) were designed that incorporated, at their 5'-end, a sequence identical to the last 20 base pairs of the VEGF A signal peptide. The sequences of these primers are shown in SEQ ID
NOS:62, 63, and 64. The same reverse primer was used to amplify all three light chain variable regions since the extreme 3'-ends were identical. This primer (SEQ ID
NO. :65) incorporated, at its 3'-end, a sequence identical to the first 20 base pairs of the human kappa constant domain. Both the FOR fragments and the digested pTTVK1 were treated with the 3' ¨ 5' exonuclease activity of T4 DNA polymerase resulting in complimentary ends that were joined by annealing. The annealing reactions were transformed into competent E. coli and the expression plasm ids were verified by sequencing to ensure that the mouse light chain variable regions were properly inserted into the pTTVK1 expression vector. Those skilled in the art will readily recognize that the method used for construction of the light chain expression plasmids applies to all anti-KAAG1 antibodies contained in the original Fab library.
Heavy chain expression vector¨ the expression vector that produced the heavy chain immunoglobulins was designed in a similar manner to the pTTVK1 described above for production of the light chain immunoglobulins. Plasmid pYD11 (Durocher et al., 2002), which contains the human IgGK signal peptide sequence as well as the and CH3 regions of the human Fc domain of IgG1, was modified by ligating the cDNA
sequence encoding the human constant CH1 region. PCR primers 0GS1769 and 0GS1770 (SEQ ID NOS:66 and 67), designed to contain unique restriction endonuclease sites, were used to amplify the human IgG1 CH1 region containing the nucleotide sequence and corresponding amino acid sequence shown in SEQ ID
NOS:68 and 69. Following ligation of the 309 base pair fragment of human CH1 immediately downstream of the IgGK signal peptide sequence, the modified plasmid (SEQ ID NO.:70) was designated pYD15. When a selected heavy chain variable region is ligated into this vector, the resulting plasmid encodes a full IgG1 heavy chain immunoglobulin with human constant regions. Based on the sequences disclosed in Table 3, PCR primers specific for the heavy chain variable regions of antibodies 3D3, 3G10, and 304 (SEQ ID NOS:17, 21, and 25, respectively) were designed that incorporated, at their 5'-end, a sequence identical to the last 20 base pairs of the IgGK signal peptide. The sequences of these primers are shown in SEQ ID NOS:71 (3D3 and 3G10 have the same 5'-end sequence) and 72. The same reverse primer was used to amplify all three heavy chain variable regions since the extreme 3'-ends were identical. This primer (SEQ ID NO.:73) incorporated, at its 3'-end, a sequence identical to the first 20 base pairs of the human CH1 constant domain. Both the PCR
fragments and the digested pYD15 were treated with the 3' ¨ 5' exonuclease activity of T4 DNA polymerase resulting in complimentary ends that were joined by annealing. The annealing reactions were transformed into competent E. coil and the expression plasmids were verified by sequencing to ensure that the mouse heavy chain variable regions were properly inserted into the pYD15 expression vector.
Those skilled in the art will readily recognize that the method used for construction of the heavy chain expression plasmids applies to all anti-KAAG1 antibodies contained in the original Fab library.
Expression of human IgGIs in 293E cells ¨ The expression vectors prepared above that encoded the light and heavy chain immunoglobulins were expressed in 293E
cells using the transient transfection system (Durocher et al., 2002). The methods used for co-transfecting the light and heavy chain expression vectors were described in Example 3. The ratio of light to heavy chain was optimized in order to achieve the most yield of antibody in the tissue culture medium and it was found to be 9:1 (L:H).
The ability of the chimeric anti-KAAG1 monoclonal antibodies to bind to recombinant Fc-KAAG1 was measured in the ELISA and compared with the original mouse Fabs.

The method was described in Example 3. As depicted in Fig. 6, the binding of the 3D3, and 3G10 chimeric IgG1 monoclonal antibodies was very similar to the Fabs. In the case of the 3C4, the binding activity of the chimeric was slightly less than the Fab.
Despite this, this result shows that the transposition of the variable domains from the mouse Fabs into a human IgG1 backbone did not significantly affect the capacity of the light and heavy chain variable regions to confer KAAG1 binding.
Example 6 This example describes the use of anti-KAAG1 antibodies to block the activity of KAAG1 in ovarian cancer cell models.
Example 2 disclosed RNAi studies showing that KAAG1 played an important role in the behavior of ovarian cancer cells. The monoclonal antibodies described above were used to determine whether it was possible to reproduce these results by targeting KAAG1 at the cell surface. TOV-21G and OV-90 cells were cultured under conditions to produce spheroids and treated with 10 g/ml of 3D3, 3G10, or 304 anti-KAAG1 chimeric monoclonal antibody. As illustrated in Fig. 7, both cell lines efficiently formed spheroids when left untreated (parental) or when treated with antibody dilution buffer (control). In contrast, the presence of anti-KAAG1 antibodies resulted in loosely packed structures and in certain cases, the cells were unable to assemble into spheroids. These results confirm the earlier observations and suggest that the anti-KAAG1 monoclonal antibodies can modulate the activity of KAAG1 during the formation of spheroids. Since spheroid formation by cancer cell lines is an in vitro model for tumor formation, the results also suggest that blocking KAAG1 could lead to decreased tumor formation in vivo.
Example 7 This example describes the use of anti-KAAG1 antibodies for detecting the expression of KAAG1 in ovarian tumors.
As a means of confirming the expression of KAAG1 protein in ovarian cancer tumors and in order determine if expression of the gene correlated with the presence of the protein, immunohistochemistry was conducted. Tissue microarrays were obtained that contained dozens of ovarian tumor samples generated from patient biopsies.
Paraffin-embedded epithelial ovarian tumor samples were placed on glass slides and fixed for 15 min at 50 C. Deparaffinization was conducted by treating 2x with xylene followed by dehydration in successive 5 min washes in 100%, 80%, and 70%
ethanol.
The slides were washed 2x in PBS for 5 min and treated with antigen retrieval solution (citrate-EDTA) to unmask the antigen. Endogenous peroxide reactive species were removed by incubating slides with H202 in methanol and blocking was performed by incubating the slides with serum-free blocking solution (Dakocytomation) for 20 min at room temperature. The primary mAb (anti-KAAG1 3D3) was added for 1 h at room temperature. KAAG1-reactive antigen was detected by incubating with biotin-conjugated mouse anti-kappa followed by streptavidin-HRP
tertiary antibody. Positive staining was revealed by treating the slides with DAB-hydrogen peroxide substrate for less than 5 min and subsequently counterstained with hematoxylin. The KAAG1 protein was found to be expressed at very high levels in the vast majority of ovarian tumor samples. A representative array containing 70 tumors is depicted in Fig. 8A. As demonstrated by the expression profiling studies that were performed using RT-PCR, KAAG1 transcripts were present in greater than 85% of ovarian tumor samples analyzed. Clearly, there is an excellent correlation between the transcription of the KAAG1 gene and the presence of the protein in ovarian cancer. Some of the samples were inspected at a higher magnification to determine which cells were expressing the KAAG1 protein. As depicted in Fig.
8B, KAAG1 is predominantly expressed in the surface epithelium of ovarian tumors.
In addition, strong intensity was observed on the apical side of these epithelial cells (see arrows in Fig. 8B, magnification: 20x). Finally, immunohistochemistry was repeated on ovarian tumor samples that originated from different histotypes. As explained earlier, epithelial ovarian cancer can be classified into 4 major histotypes:
serous, endometroid, clear cell, and mucinous. The expression of KAAG1 was detected in all types of epithelial ovarian cancer, in particular serous and endometroid histotypes (see Fig. 80).
Taken together, these immunohistochemical studies illustrate the utility of detecting KAAG1 in ovarian cancer with the monoclonal antibodies.
Example 8 IgGi antibodies against KAAG1 can mediate ADCC
Antibody-Dependent Cell Cytotoxicity (ADCC) is a mechanism of cell-mediated immunity whereby effector cells, typically natural killer (NK) cells, of the immune system actively lyse target cells that have been bound by specific antibodies.
The interaction between the NK cells and the antibody occurs via the constant Fc domain of the antibody and high-affinity Fqy receptors on the surface of the NK
cells. IgGis have the highest affinity for the Fc receptors while IgG2 mAbs exhibit very poor affinity. For this reason the chimeric antibodies targeting KAAG1 were designed as IgGis. This type of effector function that is mediated in this manner can often lead to the selective killing of cancer cells that express high level of antigen on their cell surfaces.
An in vitro assay to measure ADCC activity of the anti-KAAG1 IgGi chimeric antibodies was adapted from a previously published method, which measured the ADCC activity of the anti-CD20 rituxan in the presence of a lymphoma cell line called WIL2-S (ldusogie et aL, (2000) J. Immunol. 164, 4178-4184). Human peripheral blood mononuclear cells (PBMNCs) were used as a source of NK cells which were activated in the presence of increasing concentration of the 3D3 chimeric IgGi antibody (see Fig. 13). The target cells were incubated with the activated PBMNCs at a ratio of 1 to 25. As shown, cell death increased in a dose-dependent manner both in the presence of OVCAR-3 and the lymphoma cell line, the latter of which was shown to express KAAG1 by RT-PCR (not shown). As a positive control, the results from the published method were reproduced where high level of ADCC was obtained for rituxan in the presence of WIL2-S cells.
ADCC was also observed with other ovarian cancer cell lines that express relatively high levels of KAAG1. These results demonstrate that IgGi antibodies that are specific for KAAG1, as exemplified by 3D3, can enhance the lysis of cancer cells which express the antigen on their cell surface.
Example 9 Antibodies against KAAG1 can reduce the invasion of ovarian tumors Patients that develop ovarian cancer have lesions that typically initiate by an uncontrolled growth of the cells in the epithelial layer of the ovary or, in some instances, the fallopian tube. If detected early, these primary tumors are surgically removed and first-line chemotherapy can result in very good response rates and improved overall survival. Unfortunately, 70% of the patients will suffer recurrent disease resulting in the spread of hundreds of micro-metastatic tumors throughout the abdominal cavity. Second-line therapies can be efficacious, but often patients either respond poorly or the tumors develop chemoresistance. Treatment options are limited and there are urgent needs for new therapies to circumvent resistance to cytotoxic drugs.
In order to test the efficacy of anti- KAAG1 antibodies in vivo, an animal model of ovarian cancer was used that is the closest representation of the clinical manifestation of the disease in humans. The TOV-112D cell line is of endometrioid RECTIFIED SHEET (RULE 91) origin and expresses the KAAG1 antigen as measured by RT-PCR. Previous IHC
studies showed that ovarian tumors of the endometrioid histotype contain strong expression of KAAG1 thus rendering the 112D cell line an appropriate selection for testing anti- KAAG1 antibodies.
The intra-peritoneal inoculation of the TOV-112D cell line in SCID mice resulted in the implantation of dozens of micro-metastatic tumors that closely resemble those that are observed in humans. Mice treated with PBS, the diluent for the antibodies, contained upon examination, an average of 25 ¨ 30 tumors per animal (Fig. 14A
and B). In some cases, the number of tumors was so high in the abdominal cavity of these mice that the number of tumors could not be easily determine; these mice were excluded from the statistical analysis. When the mice were treated with the 3C4 and 3D3 antibodies, the number of micro-metastatic tumors was drastically reduced.
In addition, there was at least one animal per group treated with anti- KAAG1 where no tumors were seen. A second experiment was conducted in mice containing a larger number of TOV-112D tumors (> 50/animal) and very similar results were obtained.
Moreover, there was very little difference between the groups treated with the compared to the 3D3 antibody. However, the tendency in these in vivo experiments as well as the results obtained in the cell-based assays show that the 3D3 antibody displayed slightly more efficacy. Whether, this is due to a more accessible epitope or a higher affinity of 3D3 compared to 3C4 for the antigen still remains to be established. The results from these two experiments demonstrated that targeting KAAG1 on the surface of ovarian cancer cells could lead to a significant reduction in the spread of the tumors in vivo.
Furthermore, these findings are in complete agreement with the observations that were made in the cell-based assays. For example, the increased expression of the KAAG1 mRNA in the spheroids compared to cell lines grown as monolayers; the reduction in cell migration in the presence of KAAG1 shRNAs, the reduction in the ability of cell lines to form spheroids when treated with KAAG1 antibodies;
and finally, enhancement of ADCC activity by anti- KAAG1 IgGIs. Taken together, the results strongly suggest that targeting KAAG1 with an antibody has great therapeutic potential in recurrent ovarian cancer.
Example 10 KAAG1 is expressed in skin tumors and renal cell carcinomas and is a therapeutic target in these indications.

RECTIFIED SHEET (RULE 91) The mRNA profiling studies that were conducted showed that the transcript encoding the KAAG1 antigen was highly expressed in cell lines derived from melanoma samples and renal carcinomas. These results were disclosed in Sooknanan et al., 2007. To confirm the transcriptional regulation of the KAAG1 gene in these cancer types, immunohistochemistry was performed with an anti-KAAG1 antibody on human skin tumor tissue microarrays (Pantomics Inc., Richmond, CA) containing several sections isolated from squamous cell carcinomas and melanomas. The analysis of this array showed that there was very strong staining in biopsies isolated from squamous cell carcinomas and melanomas (Figure 15, top panel). Both of these types are among the most common forms of skin cancers and interestingly, the squamous cell carcinomas are the most metastatic, a fact that again links the expression of KAAG1 to an invasive phenotype. As previously observed, the presence of KAAG1 was very weak or absent on the three normal skin samples that were contained on the array. Similarly, KAAG1 was detected in many of the samples contained in an array of renal cancer. Most of the positive samples were predominantly of the papillary cell carcinoma type and a few clear cell carcinomas expressed KAAG1 protein. Papillary carcinomas represent approximately 20% of renal cancer cases.
In order to test if the function of KAAG1 is the same in these types of cancer compared to its role in ovarian cancer, cell lines derived from melanoma and renal cell carcinomas were obtained and tested in the spheroid culture assay (see Example 1 and 6). For the melanoma model, A375 and SK-MEL5 cells, two malignant melanoma cell lines, were cultured under conditions that allowed them to form spheroids in the presence of 5% FBS. The cultures were incubated with or without the anti-KAAG1 chimeric 3D3 antibody at a concentration of 5 g/ml. As shown in Figure 16, inclusion of 3D3 antibody in the cultures prevented the proper assembly of spheroid structures in melanoma cell lines. This result suggested that KAAG1 plays a similar role in melanoma as it does in ovarian cancer. Cell lines derived from renal cell carcinoma were also tested. The A-498 cell line is a renal papillary cell carcinoma cell line whereas the 786-0 is a renal clear cell carcinoma.
As depicted in Figure 16, only the A-498 spheroids were affected by the presence of the 3D3 anti-KAAG1 antibody while the 786-0 cell line was unaffected in this assay.
These results parallel the immunohistochemistry results described above and indicate that the inhibition of spheroids formation is dependent on the presence of KAAG1 on the surface of renal cancer cells derived predominantly from papillary RECTIFIED SHEET (RULE 91) kidney cancers. It is possible however, that the anti-KAAG1 antibody may work in other types of assays for renal clear cell carcinoma.
Taken together, these data are strongly supportive of a critical function in role of KAAG1 in melanoma and kidney cancer and indicate that blocking KAAG1 with antibodies in these indications has therapeutic potential.
Example //
KAAG1 is expressed on the surface of ovarian cancer cells. The combined results from the bioinformatics analysis of the primary structure of the cDNA
encoding KAAG1, biochemical studies, and immunohistochemical detection of the protein in epithelial cells suggested that the KAAG1 antigen was located on the cell surface. However, more direct evidence was required to demonstrate that KAAG1 is indeed a membrane-bound protein. In one approach, ovarian cancer cell lines known to express KAAG1 were plated in micro-titer plates, fixed under conditions that do not permeate the cells, and incubated with increasing concentration of anti- KAAG1 chimeric antibodies. Following extensive washing of the cells, bound antibody was detected with HRP-conjugated anti-human IgG as a secondary antibody in a modified cell-based ELISA (see Figure 17A). The first observation that can be made from these experiments is that the antibodies could be specifically captured by the cells suggesting that the KAAG1 was present at the cell surface. Secondly, the amount of binding was strongest on SKOV-3 cells and the TOV-21G cells exhibited the weakest binding. This was in complete agreement with RT-PCR data which demonstrated that the KAAG1 mRNA was expressed in similar proportions in these cell lines (not shown). Additionally, the 3D3 antibody produced the strongest signal implying that the epitope targeted by this antibody was the most accessible in this assay.
The 3G10 could only detect KAAG1 in the cell line that expressed the highest level of AB-0447 (SKOV-3 cells, see right panel of Figure 17A). A second approach used was flow cytometry. In this case, a mouse 3D3 anti-KAAG1 antibody was incubated with SKOV-3 ovarian cancer cells at saturating conditions and following extensive washing, the bound 3D3 anti-KAAG1 antibody was detected with anti-mouse IgG
conjugated to FITC in a flow cytometer. As shown in Fig. 17B, the signal at the surface of SKOV-3 cells was much higher compared to same cells labeled with the negative control, an anti-KLH (Keyhole limpet hemocyanin) antibody, specific for a non-mammalian unrelated protein, which was at a fluorescence level the same as the background readings. Taken together, these results demonstrate that KAAG1 is located on the surface of cells.

RECTIFIED SHEET (RULE 91) Example /2 Methods for the use of humanized anti-KAAG1 antibodies.
On the basis of both the in vitro and preliminary in vivo results, two mouse anti-KAAG1 antibody candidates, designated 3D3 and 3C4, were selected for humanization using in silico modeling using methods familiar to those in the art. In brief, the variable regions of the murine antibodies were modeled in 3D based on available crystal structures of mouse, humanized, and fully human variable regions that displayed high sequence homology and similar CDR loop lengths. The CDRs are the amino acid sequences that contribute to antigen binding; there are 3 CDRs on each antibody chain. Additionally, the framework regions, the amino acid sequences that intervene between the CDRs, were modified by standard homology comparison between mouse and human antibody sequences resulting in the 'best-fit' human sequence. These modifications ensured that the proper positioning of the CDR
loops was maintained to ensure maximum antigen binding in the humanized structure as well as preserving the potential N- and 0-linked glycosylation sites. The sequence of both the heavy and light chain variable regions in the humanized (h) 3D3 and resulted in 96% and 94% humanization, respectively. The structure of the 3D3 and 3C4 models for each antibody is shown in Figure 18A and 18B, respectively. As illustrated in these structures, the 3D3 required the maintenance of 3 unusual amino acids (Figure 18A, Met93 and G1y94 on the heavy chain and Ser57 on the light chain) because of their proximity to the CDRs. Modeling predicted that replacement of these mouse amino acids with human equivalents might compromise binding of the antibody with the KAAG1 antigen. In the case of 3C4, 6 amino acids were considered unusual (Figure 18B, Glu1, GIn72 and Ser98 on the heavy chain and Thr46, Phe49 and Ser87 on the light chain). In both figures, the light chain CDRs are indicated by L1, L2, and L3 for CDR1, CDR2, and CDR3, respectively, whereas the heavy chain CDRs are indicated by H1, H2, and H3 for CDR1, CDR2, and CDR3, respectively.
The sequences that encode the complete anti-KAAG1 3D3 immunoglobulin light and heavy chains are shown in SEQ ID NO.:176 and 177, respectively. The variable region of the humanized 3D3 light chain is contained between amino acids 21-133 of SEQ ID N0.:176 and is shown in SEQ ID N0.:178. The variable region of the humanized 3D3 heavy chain is contained between amino acids 20-132 of SEQ ID
N0.:177 and is shown in SEQ ID N0.:179. The sequences that encode the complete anti-KAAG1 304 immunoglobulin light and heavy chains are shown in SEQ ID
N0.:180 and 181, respectively. The variable region of the humanized 3C4 light chain RECTIFIED SHEET (RULE 91) is contained between amino acids 21-127 of SEQ ID NO.:180 and is shown in SEQ
ID NO.:182. The variable region of the humanized 3C4 heavy chain is contained between amino acids 19-136 of SEQ ID NO.:181 and is shown in SEQ ID NO.:183.
Following assembly of expression vectors and production of the h3D3 in transfected mammalian cells (see Example 5), several assays were performed to demonstrate the bio-equivalence of the humanization process. Since an antibody harboring effector functions was required, the h3D3 was assembled as a human IgGi. ELISA-based assays were performed to directly compare the ability of the h3D3 to recombinant KAAG1. The methods used to perform these tests were as described in Example 3 using recombinant Fc-KAAG1. As shown in Figure 19A, the binding activity of the h3D3 was identical to that of the chimeric 3D3.
More precise measurements were conducted using Surface Plasmon Resonance (SPR) in a Biacore instrument. Kinetic analysis was used to compare the affinity of the chimeric 3D3 with the h3D3 as well as with hybrid antibodies encompassing different permutations of the light and heavy chains (see Figure 19B).
Briefly, anti-human Fc was immobilized on the Biacore sensor chip and chimeric or h3D3 was captured on the chip. Different concentrations of monomeric recombinant 1<AAG1 were injected and the data were globally fitted to a simple 1:1 model to determine the kinetic parameters of the interaction. The kinetic parameters of the chimeric were tabulated in Figure 19B (m3D3). The average KD of the chimeric 3D3 was 2.35 x 10-10 M. In comparison, all permutations of the chimeric(C)/humanized(H) displayed very similar kinetic parameters. The average KD of the chimeric light chain expressed with the chimeric heavy chain (indicated as 'CC' in Figure 19B) was 2.71 x 10-10 M, the average KD of the humanized light chain expressed with the chimeric heavy chain (indicated as 'HC' in Figure 19B) was 3.09 x 10-10 M, the average KD of the chimeric light chain expressed with the humanized heavy chain (indicated as 'CH' in Figure 19B) was 5.05 x 10-1 M, and the average KD of the humanized light chain expressed with the humanized heavy chain (indicated as in Figure 19B) was 4.39 x 10-10 M.
The analyses indicated that the humanization of 3D3 conserved the binding activity of the original mouse antibody.
The biological function of the h3D3 was evaluated in the spheroid culture assay (see Example 6). SKOV-3 ovarian cancer cells were cultured in the presence of 5%
FBS
in the presence of h3D3 or a non-KAAG1 binding isotype control antibody. The results (shown in Figure 19C), indicated that treatment with either the buffer or the non-related IgG did not inhibit the formation of the compact 3-D structures.
In RECTIFIED SHEET (RULE 91) contrast, both the chimeric 3D3 and the humanized 3D3 prevented the spheroids from forming. The results are shown in duplicate (left and right panels).
These results indicate that the biological activity of the chimeric 3D3 was conserved in the humanized 3D3 and suggests that the h3D3 will behave in an identical manner.
Example 13 Methods for use of anti-KAAG1 antibodies as antibody conjugates As demonstrated above, the KAAG1 antigen was detected on the surface of ovarian cancer cells using a cell-based ELISA method and flow cytometry. To further substantiate these findings, fluorescence microscopy was used to visualize the antigen-antibody complex on the surface of cells. OVCAR-3 cells were seeded on coverslips and grown 0/N. The chimeric 3D3 IgG1 anti-KAAG1 antibody was added to the coverslips and incubated at 37 C, 5% CO2 for no longer than 2h. The cells were fixed in para-formaldehyde and stained with human anti-IgG-FITC for 30 min at RT. After washing, the cells were observed using a confocal microscope. The antibody bound to the surface of cells was detected by incubating the coated cells in the presence of FITC-labeled anti-human IgG. As displayed in Figure 22, the pattern of staining is indicative of an enrichment at the cells surface (see upper right panel, KAAG1). Furthermore, staining with an antibody specific for E-cadherin, a known membrane protein, showed that the staining of E-cadherin co-localized with that of KAAG1 (see lower panels, E-cadherin and merge). Similar results were seen with SKOV-3 and TOV-112D cells, two other cell lines that are positive for KAAG1 expression. These data confirm that KAAG1 is detectable on the surface of ovarian cancer cells and that the 3D3 antibody can be used to bind to the antigen on these cells. Therefore, by several different methods, it was established that the protein encoded by the KAAG1 gene can be detected at the surface of ovarian cancer cells.
There are several different molecular events that can occur upon binding of an antibody to its target on the surface of cells. These include i) blocking accessibility to another cell-surface antigen/receptor or a ligand, ii) formation of a relatively stable antibody-antigen complex to allow cells to be targeted via ADCC or CDC, iii) signaling events can occur as exemplified by agonistic antibodies, iv) the complex can be internalized, or v) the complex can be shed from the cell surface. To address this question we wished to examine the behavior of the antibody-antigen complex on the surface of the cells as it pertains to KAAG1. SKOV-3 cells were plated, washed, and incubated with 5 pg/ml chimeric 3D3 antibody. After washing, complete OSE
medium was added and the cells placed at 37 C for up to 180 minutes. The cells were removed at the indicated times, rapidly cooled, and prepared for cytometry with FITC-conjugated anti-human IgG and the results were expressed as the percentage of mean fluorescence intensity (MFI, % surface binding) remaining. As illustrated in Fig. 23, the fluorescence signal decreases over a period of 30 - 45 minutes.
This result indicates that the complex between the 3D3 antibody and KAAG1 exhibits stability on the surface of SKOV-3 cells. However, it is evident from this experiment that the complex slowly disappeared from the cells which indicated that an internalization of the complex had occurred. Preliminary studies to elucidate the mechanism responsible for this decrease in cell-surface fluorescence have revealed that the complex appears to be internalized.
These findings were further confirmed by conducting immunofluorescence on live cells to see if this internalization could be microscopically observed. OVCAR-3 cells were seeded on cover slips in full medium (OSE medium (Wisent) containing 10%
FBS, 2 mM glutamine, 1 mM sodium-pyruvate, 1X non-essential amino acids, and antibiotics. Once the cells were properly adhered, the medium was removed and the cover slips washed twice gently with ice-cold PBS containing 1% FBS, 1 mM
MgC12, 1 mM CaCl2. Chimeric 3D3 (human IgG1) at 10 pg/ml was added to the cells and incubated on ice for 1h. The coverslips were incubated for 30 minutes full medium at 37 C and following washing in PBS, they were fixed at room temperature in 3.7%
formaldehyde (in PBS) containing saponin for 30 min. AB-3D3 was visualized with rabbit anti-human IgG-488 Dylight (Jackson ImmunoResearch) diluted 1:400 and mounted on microscope slides using Gold Antifade reagent with DAPI
(Invitrogen).
As seen in Figure 24, the 3D3 antibody is able to detect complexes predominantly on the cell surface at time 0 (Figure 24, left panel) but after 30 minutes incubation at 37 C, these complexes were detected inside the cells (see Fig. 24, right panel, arrows).
This data is in complete agreement with the flow cytometry data (see Fig. 24) and confirms that the KAAG1/3D3 complex is internalized in cells. Finally, in additional studies, it was found that the KAAG1/3D3 complexes co-localize with early endosome antigen 1 (EEA1) in SKOV-3 cells. EEA1 is a protein known to exist in vesicles that occur during endosomal trafficking.
Taken together, these studies demonstrated that antibodies specific for KAAG1 might have uses as an antibody-drug conjugate (ADC). Thus, the high level of ovarian cancer specificity of KAAG1 coupled with the capacity of this target to be internalized in cells would support the development of applications as an ADC.

CITED REFERENCES
Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, Feuer EJ and Thun MJ. Cancer statistics, 2005. CA Cancer J Clin 2005; 55: 10-30.
Menon U, Skates SJ, Lewis S, Rosenthal AN, Rufford B, Sibley K, Macdonald N, Dawnay A, Jeyarajah A, Bast RC Jr, Oram D and Jacobs IJ.
Prospective study using the risk of ovarian cancer algorithm to screen for ovarian cancer. J Clin Oncol. 2005; 23(31):7919-26.
- Bonome T, Lee JY, Park DC, Radonovich M, Pise-Masison C, Brady J, Gardner GJ, Hao K, Wong WH, Barrett JC, Lu KH, Sood AK, Gershenson DM, Mok SC and Birrer MJ. Expression profiling of serous low malignancy potential, low grade, and high-grade tumors of the ovary. Cancer Res 2005;
65: 10602-10612.
- Chambers, A and Vanderhyden, B. Ovarian Cancer Biomarkers in Urine. Clin Cancer Res 2006; 12(2): 323-327.
- Berek et al. Cancer Medicine. 5th ed. London: B.C. Decker, Inc.; 2000.
pp. 1687-1720.
- Bristow R.E. Surgical standards in the management of ovarian cancer.
Curr Opin Oncol 2000; 12: 474-480.
Brown E, Stewart M, Rye T, Al-Nafussi A, Williams AR, Bradburn M, Smyth J and Gabra H. Carcinosarcoma of the ovary: 19 years of prospective data from a single center. Cancer 2004; 100: 2148-2153.
- Shih L-M and Kurman RJ. Molecular Pathogenesis of Ovarian Borderline Tumors: New Insights and Old Challenges. Clin Cancer Res 2005;
11(20): 7273-7279.
- Seidman JD, Russell P, Kurman RJ. Surface epithelial tumors of the ovary. In: Kurman RJ, editor. Blaustein's pathology of the female genital tract.
5th ed. New York: Springer-Verlag; 2002. pp. 791-904.
- Cannistra SA and McGuire WP. Progress in the management of gynecologic cancer. J. Clin. Oncol. 2007; 25(20): 2865-2866.
- Oei AL, Sweep FC, Thomas CM, Boerman DC, Massuger LF. The use of monoclonal antibodies for the treatment of epithelial ovarian cancer. Int.
J.
Oncol. 2008; 32(6): 1145-1157.
12 Nicodemus CF and Berek JS. Monoclonal antibody therapy of ovarian cancer. Expert Rev. Anticancer Ther. 2005; 5(1): 87-96.
- Burger RA. Experience with bevacizumab in the management of epithelial ovarian cancer. J. Clin. Oncol. 2007; 25(20): 2902-2908.
Simon I, Zhuo S, Corral L, Diamandis EP, Sarno MJ, Wolfert RL, Kim NW. B7-H4 is a novel membrane-bound protein and a candidate serum and tissue biomerker for ovarian cancer. Cancer Res. 2006; 66(3): 1570-1575.
- Ebel W, Routhier EL, Foley B, Jacob S, McDonough JM, Patel RK, Turchin HA, Chao Q, Kline JB, Old LJ, Phillips MD, Nicolaides NC, Sass PM, Grasso L. Preclinical evaluation of MORab-003, a humanized monoclonal antibody antagonizing folate receptor-alpha. Cancer Immun. 2007; 7: 6-13.
- Van den Eynde BJ, Gaugler B, Probst-Kepper M, Michaux L, Devuyst 0, Lorge F, Weynants P, Boon T. A new antigen recognized by cytotoxic T
lymphocytes on a human kidney tumor results from reverse strand transcription. J. Exp. Med. 1999; 190(12): 1793-1799.
- Sooknanan R, Tremblay GB, Filion M. Polynucleotides and polypeptide sequences involved in cancer. 2007; PCT/CA2007/001134.
- Schumacher J, Anthoni H, Dahdouh F, Konig IR, Hillmer AM, Kluck N, Manthey M, Plume E, Warnke A, Remschmidt H, HOIsmann J, Cichon S, Lindgren CM, Propping P, Zucchelli M, Ziegler A, Peyrard-Janvid M, Schulte-Karne G, Nothen MM, Kere J. Strong genetic evidence of DCDC2 as a susceptibility gene for dyslexia. Am. J. Hum. Genet. 2006; 78: 52-62.
- Cope N, Harold D, Hill G, Moskvina V, Stevenson J, Holmans P, Owen MJ, O'Donovan MC, Williams J. Strong evidence that KIAA0319 on chromosome 6p is a susceptibility gene for developmental dyslexia. Am. J.
Hum. Genet. 2005; 76: 581-591.
- Mor G, Visintin I, Lai Y, Zhao H, Schwartz P, Rutherford T, Yue L, Bray-Ward P and Ward DC Serum protein markers for early detection of ovarian cancer. PNAS 2005; 102: 7677-7682.
- Kozak KR, Amneus MW, Pusey SM, Su F, Luong MN, Luong SA, Reddy ST and Farias-Eisner R. Identification of biomarkers for ovarian cancer using strong anion-exchange ProteinChips: potential use in diagnosis and prognosis. PNAS 2003; 100: 12343-12348.

- Benoit MH, Hudson TJ, Maire G, Squire JA, Arcand SL, Provencher D, Mes-Masson AM, Tonin PN. Global analysis of chromosome X gene expression in primary cultures of normal ovarian surface epithelial cells and epithelial ovarian cancer cell lines. Int. J. Oncol. 2007; 30(1): 5-17.
- Cody NA, Zietarska M, Filali-Mouhim A, Provencher DM, Mes-Masson AM, Tonin PN. Influence of monolayer, spheroid, and tumor growth conditions on chromosome 3 gene expression in tumorigenic epithelial ovarian cancer cell lines. BMC Med. Genomics 2008; 1(1) :34.
Buechler J, Valkirs G, Gray J. Polyvalent display libraries.2000; U.S.
6,057,098.
- Durocher Y, Kamen A, Perret S, Pham PL. Enhanced production of recombinant proteins by transient transfection of suspension-growing mammalian cells. 2002; Canadian patent application No. CA 2446185.
Durocher Y. Expression vectors for enhanced transient gene expression and mammalian cells expressing them.2004; U.S. patent application No. 60/662,392.

Sequences referred to in the description SEQ ID NO.:1 GAGGGGCATCAATCACACCGAGAAGTCACAGCCCCTCAACCACTGAGGTGTGGGGGGGTAGGG
ATCTGCATTTCTTCATATCAACCCCACACTATAGGGCACCTAAATGGGTGGGCGGTGGGGGAG
ACCGACTCACTTGAGTTTCTTGAAGGCTTCCTGGCCTCCAGCCACGTAATTGCCCCCGCTCTC
GATCTGGTCTAGCTTCCGGATTCGGTGGCCAGTCCGCGGGGTGTAGATGTTCCTGACGGCCCC
AAAGGGTGCCTGAACGCCGCCGGTCACCTCCTTCAGGAAGACTTCGAAGCTGGACACCTTCTT
CTCATGGATGACGACGCGGCGCCCCGCGTAGAAGGGGTCCCCGTTGCGGTACACAAGCACGCT
CT TCACGACGGGCTGAGACAGGTGGCTGGACCTGGCGCTGCTGCCGCTCATCTTCCCCGCTGG
CCGCCGCCTCAGCTCGCTGCTTCGCGTCGGGAGGCACCTCCGCTGTCCCAGCGGCCTCACCGC
ACCCAGGGCGCGGGATCGCCTCCTGAAACGAAC GAGAAACT GACGAATC CACAGGTGAAAGAG
AAGTAACGGCCGTGCGCCTAGGCGTCCACCCAGAGGAGACACTAGGAGCTTGCAGGACTCGGA
GTAGACGCTCAAGT TT TTCACCGTGGCGTGCACAGCCAATCAGGACCCGCAGTGCGCGCACCA
CACCAGGTTCACCTGCTACGGGCAGAATCAAGGTGGACAGCTTCTGAGCAGGAGCCGGAAACG
C GO GG GGC CT T CAAACAGGCAC GCCTAGT GAGGGCAGGAGAGAGGAGGACGCACACACACACA
CACACACAAATATGGTGAAAC CCAAT T TCT TACATCATATC TGTGCTACCC TT TCCAAACAGC
CTA
SEQ ID NO.:2 MDDDAAPRVEGVPVAVHKHALHDGLRQVAGPGAAAAHLPRWPPPQLAASRREAPPLSQRPHRT
QGAGSPPETNEKLTNPQVKEK
SEQ ID NO.:3 GACATTGTGATGACCCAGTCTCCATCCTCCCTGGCTGTGTCAATAGGACAGAAGGTCACTATG
AACTGCAAGTCCAGTCAGAGCCTTTTAAATAGTAACTTTCAAAAGAACTTTTTGGCCTGGTAC
CAGCAGAAACCAGGCCAGTCTCCTAAACTTCTGATATACTT TGCATCCACTCGGGAATCTAGT
ATCCCTGATCGCTTCATAGGCAGTGGATCTGGGACAGATTTCACTCT TACCATCAGCAGTGTG
CAGGCTGAAGACCTGGCAGATTACTTCTGTCAGCAACAT TATAGCACTCCGCTCACGT TCGGT
GCTGGGACCAAGCTGGAGCTGAAAGCTGTGGCTGCACCATCTGTOTTCATCTTCCCGCCATCT
GATGAGCAGTTGAAATCTGGAACTGCCTCTGT TGTGTGCCTGCTGAATAACTTCTATCCCAGA
GAGGCCAAAGTACAGTGGAAGGTGGATAACGCCCTCCAATCGGGTAACTCCCAGGAGAGTGTC
ACAGAGCAGGACAGCAAGGACAGCACCTACAGCC TCAGCAGCAC CC TGAC GC TGAGCAAAGCA
GACTACGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCAGGGCCTGAGCTCGCCCGTC
ACAAAGAGCTTCAACAGGGGAGAGTGT
SEQ ID NO.:4 DI VMTQS PS SLAVS I GQKVTMNCKS SQSLLNSNFQKNFLAWYQQKPGQS PKLL I YEAS TRE S S
I PDRFI GS GS GTDFTLT I SSVQAEDLADYFCQQHYST PLTFGAGTKLELKAVAAPSVFI FP PS
DEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQE SVTEQDSKDS T ySL SS TL TLSKA
DYEKHKVYACEVTHQGLSSPVTKS FNRGEC
SEQ ID NO.:5 GAGGTTCAGCTGCAGCAGTCTGTAGCTGAGCTGGTGAGGCCTGGGGCTTCAGTGACGCTGTCC
TGCAAGGCTTCGGGCTACATATT TACT GACTATGAGATACACT GGGTGAAGCAGACT CCTGT G
CATGGCCTGGAATGGATTGGGGTTATTGATCCTGAAACTGGTAATACTGCCTTCAATCAGAAG
TTCAAGGGCAAGGCCACACTGACTGCAGACATATCCT CCAGCACAGCCTACAT GGAACTCAGC
AGTT TGACATCTGAGGACTCTGCCGTCTAT TACTGTATGGGTTATTCTGATTATTGGGGCCAA
GGCACCACTCTCACAGTCTCCTCAGCCTCAACGAAGGGCCCATCTGTCTTTCCCCTGGCCCCC
TCCTCCAAGAGCACCTCTGGGGGCACAGCGGCCCTGGGCTGCCTGGTCAAGGACTACTTCCCC
GAACCGGTGACGGTGTCGTGGAACTCAGGCGCCCTGACCAGCGGCGTGCACACCTTCCCGGCT
GTCC TACAGT CC T CAGGACTC TACTCC CTCAGCAGC GTGGTGACC GT GCCC TCCAGCAGCTT G
GGCACCCAGACCTACATCTGCAACGTGAATCACAAGCCCAGCAACACCAAGGTGGACAAGAAA
GTTGAGCCCAAATCTTGTGAATTCACTCACACATGCCCACCGTGCCCAGCACCTGAACTCCTG
GGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACC

CCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGG
TACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGC
ACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTAC
AAGT GCAAGGTCTCCAACAAAGCCCTCCCAGCC CC CAT C GAGAAAAC CATCT C CAAAGCCAAA
GGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGATGAGCTGACCAAGAAC
CAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAG
AGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCC
TTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCA
TGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCC
G GGAAA
SEQ ID NO.:6 EVQLQQSVAELVRPGASVTLSCKASGYI FT DYE I HWVKQT PVHGLEW I GVI DPETGNTAFNQK
FKGKATLTADI SS STAYMELS SLTSEDSAVYYCMGyS DyWGQGT TL TVS SAS TKGPSVFPLAp SSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFpAVLQSSGLySLSSVVTVPSSSL
GTQTY I CNVNHKPSNTKVDKKVEPKS CE FTHTCppC PAPELLGGPSVFL FpPKPKDTLMI SRT
PEVTCVVVDVS HE DPEVKFNWYVDGVEVHNAKTKPREEQyNS TYRVVSVLTVLHQDWLNGKEY
KCKVSNKALPAP I EKT I SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFypSDIAVEWE
SNGQPENNYKTT PPVLDSDGSFELYSKLTVDKSRWQQGNVESCSVMHEALHNHYTQKSLSLS P
GK
SEQ ID NO.:7 GATGT TT TGATGACCCAAACTCCACGCTCCCTGTCTGTCAGTCT TGGAGATCAAGCCTCCATC
TC TT GTAGAT CGAGTCAGAGCCT TTTACATAGTAATGGAAACACC TATT TAGAAT GGTAT TT G
CAGAAACCAGGCCAGCCTCCAAAGGTCCTGATCTACAAAGTT TCCAACCGATT T TCTGGGGTC
CCAGACAGGTT CAGT GGCAGTGGATCAGGGACAGAT TT CACACTCAAGATCAGCGGAGTGGAG
GCTGAGGATCTGGGAGTTTATTACTGCTTTCAAGGTTCACATGTTCCTCTCACGTTCGGTGCT
GGGACCAAGCTGGAGCTGAAAGCTGTGGCTGCACCATCTGTCTTCATCTTCCCGCCATCTGAT
GAGCAGTTGAAATCTGGAACTGCCTCTGTTGTGTGCCTGCTGAATAACTTCTATCCCAGAGAG
GCCAAAGTACAGTGGAAGGTGGATAACGCCCTCCAATCGGGTAACTCCCAGGAGAGTGTCACA
GAGCAGGACAGCAAGGACAGCACCTACAGCCTCAGCAGCACCCTGACGCTGAGCAAAGCAGAC
TACGAGAAACACAAAGT CTAC GC C TGCGAAGTCACCCAT CAGGGC CT GAGC TC GCCCGTCACA
AAGAGCT TCAACAGGGGAGAGT GT
SEQ ID NO :8 DVLMTQTPRSLSVSLGDQAS I S CRS SQSLLHSNGNTYLEWYLQKPGQp pKVL I yKVSNRFSGV
PDRFS GS GS GT DFTLKI S GVEAEDLGVyyC FQGSHVPLT FGAGTKLELKAVAAPSVFI FP PS D
EQLKSGTASVVCLLNNEYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKAD
YEKHKVYACEVTHQGLSS PVTKSFNRGEC
SEQ ID NO.:9 GAGATCCAGCTGCAGCAGTCTGGACCTGAGT TGGTGAAGCCTGGGGCTTCAGTGAAGATATCC
TGTAAGGCT TCTGGATACACCT TCACTGACAACTACATGAACTGGGTGAAGCAGAGCCATGGA
AAGAGCCT TGAGT GGAT TGGAGATAT TAAT C C TTAC TAT GGTAC TACTAC CTACAACCAGAAG
TTCAAGGGCAAGGC CACATT GACTGTAGACAAGTC CT CC CGCACAGCC TACAT GGAGCTCCGC
GGCCTGACATCTGAGGACTCTGCAGTCTATTACTGTGCAAGAGATGACTGGTT TGAT TAT TGG
GGCCAAGGGACTCTGGTCACTGTCTCTGCAGCCTCAACGAAGGGCCCATCTGTCTTTCCCCTG
GCCCCCTCCTCCAAGAGCACCTCTGGGGGCACAGCGGCCCTGGGCTGCCTGGTCAAGGACTAC
TTCCCCGAACCGGTGACGGTGTCGTGGAACTCAGGCGCCCTGACCAGCGGCGTGCACACCTTC
CCGGCTGTCCTACAGTCCTCAGGACTCTACTCCCTCAGCAGCGTGGTGACCGTGCCCTCCAGC
AGCT TGGGCACC CAGACCTACATCTGCAAC GTGAATCACAAGCCCAGCAACACCAAGGT GGAC
AAGAAAGTT GAGC CCAAATCTT GT GAAT TCACT CACACATGCC CACCGT GCCCAGCACC TGAA
CTCCTGGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCC
CGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTC
AAC TGGTAC GT GGAC GGC GT GGAGGT GCATAAT GCCAAGACAAAGCCGCGGGAGGAGCAGTAC

AACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAG
GAGTACAAGTGCAAGGT C TCCAACAAAGC CC TC CCAGCC C C CATCGAGAAAACCATC TCCAAA
GCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGATGAGCTGACC
AAGAACCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAG
T GGGAGAGCAATGGGCAGCC GGAGAACAACTACAAGAC CAC CC CT C CC GTGCTGGACT CCGAC
GGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTC
TTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTG
T CT CCCGGGAAA
SEQ ID NO.:10 EIQLQQSGPELVKPGASVKI SCKASGYT FT DNYMNWVKQSHGKSLEW I GDINPYYGTTTYNQK
FKGKATLTVDKSSRTAYMELRGLTSEDSAVYYCARDDWFDYWGQGTLVTVSAASTKGPSVFPL
APS SKSTSGGTAALGCLVKDYFPE PVTVSWNSGAL TSGVHT FPAVLQS SGLYSLS SVVTVPS S
SLGTQTYI CNVNHKPSNTKVDKKVE PKS CEFTHTC P PC PAPELLGGPSVFL FP PKPKDTLMI S
RTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGK
EYKCKVSNKAL PAP I EKT I SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVE
WESNGQPENNYKTT P PVL DS DGS FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSL
S PGK
SEQ ID NO.:11 GACATC GT TAT GTCT CAGT CT CCATCTTCCATGTAT GCATCTCTAGGAGAGAGAGTCAC TAT C
ACTTGCAAGGCGAGTCAGGACATT CATAAC T TT TTAAACTGGT TCCAGCAGAAACCAGGAAAA
TCTCCAAAGACC CT GATCTT TCGT GCAAACAGATT GGTAGATGGGGT CC CAT CAAGGT TCAGT
GGCAGTGGATCTGGGCAAGATTATTCTCTCACCATCAGCAGCCTGGAGTTTGAAGATTTGGGA
ATTTATTCTTGTCTACAGTATGATGAGATTCCGCTCACGTTCGGTGCTGGGACCAAGCTGGAG
CTGAGAGCTGTGGCTGCACCATCTGTCTTCATCTTCCCGCCATCTGATGAGCAGTTGAAATCT
GGAACTGCCTCTGTTGTGTGCCTGCTGAATAACTTCTATCCCAGAGAGGCCAAAGTACAGTGG
AAGGT GGATAACGCC CT CCAATCGGGTAACTC CCAGGAGAGT GT CACAGAGCAGGACAGCAAG
GACAGCACCTACAGCCT CAGCAGCAC CC TGAC GCT GAGCAAAGCAGACTACGAGAAACACAAA
GT CTAC GC CT GCGAAGT CACCCATCAGGGCCTGAGCT CGCCCGTCACAAAGAGCT TCAACAGG
GGAGAGTGT
SEQ ID NO.:12 DIVMSQSPSSMYASLGERVT TCKAS QDI HNFLNWFQQKPGKS PKTL FRANRLVDGVPSRFS
GSGSGQDYSLT S SLEFEDLGI YSCLQYDE I PLT FGAGTKLELRAVAAPSVFI FP PS DEQLKS
GTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDS TYSLS S TLTLSKADYEKHK
VYACEVTHQGLSS PVTKS FNRGEC
SEQ ID NO..13 GAGGTGCAGCTTCAGGAGTCAGGACCTGACCTGGTGAAACCTTCTCAGTCACTTTCACTCACC
TGCACTGTCACTGGCTTCTCCATCACCAGTGGTTATGGCTGGCACTGGATCCGGCAGTTTCCA
GGAAACAAACTGGAGTGGAT GGGCTACATAAACTACGAT GGTCACAAT GACTACAACCCAT CT
CTCAAAAGT CGAAT CTC TAT CACT CAAGACACATCCAAGAACCAGTT CT TCC TGCAGTT GAAT
TCTGTGACTACTGAGGACACAGCCACATATTACTGTGCAAGCAGTTACGACGGCTTATTTGCT
TACTGGGGCCAAGGGACTCTGGTCACTGTCTCTGCAGCCTCAACGAAGGGCCCATCTGTCTTT
C CCCT GGCCCCCT CCTCCAAGAGCACCTC TGGGGGCACAGCGGC CC TGGGCT GC CT GGT CAAG
GACTACTTCCCCGAACCGGTGACGGTGTCGTGGAACTCAGGCGCCCTGACCAGCGGCGTGCAC
ACCTTCCCGGCTGTCCTACAGTCCTCAGGACTCTACTCCCTCAGCAGCGTGGTGACCGTGCCC
TCCAGCAGCTTGGGCACCCAGACCTACATCTGCAACGTGAATCACAAGCCCAGCAACACCAAG
GTGGACAAGAAAGTTGAGCCCAAATCTTGTGAATTCACTCACACATGCCCACCGTGCCCAGCA
CCTGAACTCCTGGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATG
ATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTC
AAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAG
CAGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAAT
GGCAAGGAGTACAAGT GCAAGGTCTCCAACAAAGC CCT C CCAGCCCC CATCGAGAAAACCATC

TCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGATGAG
CTGACCAAGAACCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCC
GT GGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGC CT C CCGTGCTGGAC
TCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGG
AACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTC
TCCCTGTCTCCCGGGAAA
SEQ ID NO.:14 EVQLQESGPDLVKPSQSLSLTCTVTGFS I TS GYGWHWIRQFPGNKLEWMGYI NYDGHNDYNPS
LKSRI S I TQDTSKNQFFLQLNSVTTEDTATYYCASSYDGLFAYWGQGTLVTVSAASTKGPSVF
PLAPSSKST SGGTAALGCLVKDYFPEPVTVSWNS GALTS GVHTFPAVLQSS GLYS LS SVVTVP
SSSLGTQT YICNVNHKPSNTKVDKKVEPKSCEFTHTC P PC PAPELLGGPSVFL FPPKPKDTLM
I SRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLN
GKEYKCKVSNKALPAP I EKT I SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPS DIA
VEWESNGQPENNYKTTPPVLDS DGSFELYSKLTVDKSRWQQGNVESCSVMHEALHNHYTQKSL
SLSPGK
SEQ ID NO.:15 GACATTGTGATGACCCAGTCTCCATCCTCCCTGGCTGTGTCAATAGGACAGAAGGTCACTATG
AACTGCAAGTCCAGTCAGAGCCTTTTAAATAGTAACTTTCAAAAGAACTTTTTGGCCTGGTAC
CAGCAGAAACCAGGCCAGT CT CCTAAACTTCTGATATACTTTGCATC CAC TCGGGAATCTAGT
ATCCCTGATCGCTTCATAGGCAGTGGATCTGGGACAGATTTCACTCTTACCATCAGCAGTGTG
CAGGCTGAAGACCTGGCAGATTACTTCTGTCAGCAACATTATAGCACTCCGCTCACGTTCGGT
GCTGGGACCAAGCTGGAGCTGAAA
SEQ ID NO.:16 DI VMTQS PS SLAVS IGQKVTMNCKS SQSLLNSNFQKNFLAWYQQKPGQS PKLL I YFASTRESS
I PDRFI GS GS GT DFTLT I SSVQAEDLADYFCQQHYST PLTFGAGTKLELK
SEQ ID NO.:17 GAGGT TCAGCTGCAGCAGTCTGTAGCTGAGCTGGTGAGGCCTGGGGCT TCAGTGACGCTGTCC
TGCAAGGCTTCGGGCTACATATTTACTGACTATGAGATACACTGGGTGAAGCAGACTCCTGTG
CATGGCCT GGAATGGATTGGGGT TAT T GATCCTGAAACT GGTAATAC TGCC TTCAATCAGAAG
TTCAAGGGCAAGGCCACACTGACTGCAGACATATCCTCCAGCACAGCCTACATGGAACTCA.GC
AGTTTGACATCTGAGGACTCTGCCGTCTATTACTGTATGGGTTATTCTGATTATTGGGGCCAA
GGCACCACTCTCACAGTCTCCTCA
SEQ ID NO.:18 EVQLQQSVAELVRPGASVTLSCKASGYI FT DYEI HWVKQT PVHGLEW I GVI DPETGNTAFNQK
FKGKATLTADI SSSTAYMELSSLTSEDSAVYYCMGYSDYWGQGTTLTVSS
SEQ ID NO.:19 GATGTTTTGATGACCCAAACTCCACGCTCCCTGTCTGTCAGTCTTGGAGATCAAGCCTCCATC
TC TTGTAGATCGAGTCAGAGCCT TT TACATAGTAATGGAAACACCTATT TAGAATGGTATTTG
CAGAAACCAGGCCAGCCTCCAAAGGTCCTGATCTACAAAGTTTCCAACCGATTTTCTGGGGTC
CCAGACAGGTTCAGTGGCAGTGGATCAGGGACAGATTTCACACTCAAGATCAGCGGAGTGGAG
GCTGAGGATCTGGGAGTT TAT TACT GCTT TCAAGGT TCACATGTTCCTCTCAC GTTCGGTGCT
GGGACCAAGCTGGAGCTGAAA
SEQ ID NO.:20 DVLMTQTPRSLSVSLGDQAS I S CRSSQSLLHSNGNT YLEWYLQKPGQP PKVL I YKVSNRFSGV
PDRFSGSGSGTDFTLKI SGVEAEDLGVYYCFQGSHVPLTFGAGTKLELK
SEQ ID NO.:21 GAGATCCAGCTGCAGCAGTCTGGACCTGAGTTGGTGAAGCCTGGGGCTTCAGTGAAGATATCC
TGTAAGGCT TCTGGATACACCTTCACTGACAAC TACATGAACTGGGTGAAGCAGAGCCATGGA
AAGAGCCTTGAGTGGATT GGAGATATTAATC CT TACTATGGTACTAC TACCTACAACCAGAAG
TTCAAGGGCAAGGCCACATTGACTGTAGACAAGTCCTCCCGCACAGCCTACATGGAGCTCCGC
GGCCTGACATCTGAGGACTCTGCAGTCTATTACTGTGCAAGAGATGACTGGTTTGATTATTGG
GGCCAAGGGACTCTGGTCACTGTCTCTGCA
SEQ ID NO.:22 EIQLQQSGPELVKPGASVKI SCKAS GYT FTDNYMNWVKQSHGKSLEW I GDINPYYGTTT YNQK
FKGKATLTVDKS SRTAYMELRGLTSEDSAVYYCARDDWFDYWGQGTLVTVSA
SEQ ID NO. 23 GACATCGT TAT GTCTCAGTCTCCATGT TCCATGTATGCATCT CTAGGAGAGAGAGT CAC TATC
ACTT GCAAGGC GAGTCAGGACATTCATAAC TT TTTAAACTGGT TCCAGCAGAAACCAGGAAAA
TCTCCAAAGACCCTGATCTTTCGTGCAAACAGAT TGGTAGATGGGGTCCCATCAAGGTTCAGT
GGCAGTG GAT CTGGGCAAGATTATTCT CT CAC CATCAGCAGCCTGGAGT TT GAAGAT T TGGGA
AT TTAT TCTTGTCTACAGTATGATGAGAT TCCGCTCACGTTCGGTGCTGGGACCAAGCTGGAG
CT GAGA
SEQ ID NO.:24 DIVMSQS PS SMYASLGERVTI TCKAS QDIHNFLNWFQQKPGKS PKTL I FRANRLVDGVPSRFS
GSGSGQDYSLT I SSLEFEDLGI YSCLQYDEI PLT FGAGTKLELR

GAGGTGCAGCTTCAGGAGTCAGGACCTGACCTGGTGAAACCTTCTCAGTCACTTTCACTCACC
TGCACTGTCACTGGCTTCTCCATCACCAGTGGTTATGGCTGGCACTGGATCCGGCAGTTTCCA
GGAAACAAACTGGAGTGGATGGGCTACATAAACTACGAT GGTCACAATGACTACAACCCAT CT
CTCAAAAGTCGAATCTCTATCACTCAAGACACATCCAAGAACCAGTTCTTCCTGCAGTTGAAT
TCTGTGACTACTGAGGACACAGCCAGATATTACTGTGCAAGCAGTTACGACGGCTTATTTGCT
TACTGGGGCCAAGGGACTCTGGTCACTGTCTCTGCA
SEQ ID NO.:26 EVQLQESGPDLVKPSQSLSLTCTVTGFS I T S GYGWHW I RQ FPGNKLEWMGYINYDGHNDYNPS
LKSRI SI TQDTSKNQFFLQLNSVTTEDTATYYCASSYDGLFAYWGQGTLVTVSA
SEQ ID NO.:27 KS S QS LLN SN FQKN FLA
SEQ ID NO.:28 FAST RE S
SEQ ID NO. :29 QQHYSTPLT
SEQ ID NO :30 GYIFTDYEIH
SEQ ID NO :31 VI DPETGNTA
SEQ ID NO.:32 MGYSDY
SEQ ID NO.:33 RS SQSLLHSNGNTYLE

SEQ ID NO.:34 KVSNRFS
SEQ ID NO.:35 FQGSHVPLT
SEQ ID NO.:36 GYTFTDNYMN
SEQ ID NO..37 DINPYYGTTT
SEQ ID NO.:38 ARDDWFDY
SEQ ID NO.:39 KASQDIHNFLN
SEQ ID NO. 40 RANRLVD
SEQ ID NO.:41 LQYDEI PLT
SEQ ID NO.:42 GFS I TSGYGWH

YINYDGHND
SEQ ID NO.:44 AS SYDGL FAY
SEQ ID NO.:45 GAGGGGCATCAATCACACCGAGAA
SEQ ID NO.:46 CCCCACCGCCCACCCATTTAGG
SEQ ID NO.:47 TGAAGGTCGGAGTCAACGGATTTGGT
SEQ ID NO.:48 CATGTGGGCCATGAGGTCCACCAC
SEQ ID NO..49 GGCCTCCAGCCACGTAATT
SEQ ID NO.:50 GGCGCTGCTGCCGCTCATC
SEQ ID NO :51 TCGCGCGTTTCGGTGATGACGGTGAAAACCTCTGACACATGCAGCTCCCGGAGACGGTCACAG
CTTGTCTGTAAGCGGATGCCGGGAGCAGACAAGCCCGTCAGGGCGCGTCAGCGGGTGTTGGCG

GGTGTCGGGGCTGGCTTAACTATGCGGCATCAGAGCAGATTGTACTGAGAGTGCACCATATGC
GGTGTGAAATACCGCACAGATGCGTAAGGAGAAAATACCGCATCAGGCGCCATTCGCCATTCA
GGCTGCGCAACTGTTGGGAAGGGCGATCGGTGCGGGCCTCTTCGCTATTACGCCAGCTGGCGA
AAGGGGGATGTGCTGCAAGGCGATTAAGTTGGGTAACGCCAGGGTTTTCCCAGTCACGACGTT
GTAAAACGACGGCCAGTGCCAAGCTTTTCCAAAAAACTACCGTTGTTATAGGTGTCTOTTGAA
CACCTATAACAACGGTAGTGGATCCCGCGTCCTTTCCACAAGATATATAAACCCAAGAAATCG
AAATACTTTCAAGTTACGGTAAGCATATGATAGTCCATTTTAAAACATAATTTTAAAACTGCA
AACTACCCAAGAAATTATTACTTTCTACGTCACGTATTTTGTACTAATATCTTTGTGTTTACA
GTCAAATTAATTCTAATTATCTCTCTAACAGCCTTGTATCGTATATGCAAATATGAAGGAATC
ATGGGAAATAGGCCCTCTTCCTGCCCGACCTTGGCGCGCGCTCGGCGCGCGGTCACGCTCCGT
CACGTGGTGCGTTTTGCCTGCGCGTCTTTCCACTGGGGAATTCATGCTTCTCCTCCCTTTAGT
GAGGGTAATTCTCTCTCTCTCCCTATAGTGAGTCGTATTAATTCCTTCTCTTCTATAGTGTCA
CCTAAATCGTTGCAATTCGTAATCATGTCATAGCTGTTTCCTGTGTGAAATTGTTATCCGCTC
ACAATTCCACACAACATACGAGCCGGAAGCATAAAGTGTAAAGCCTGGGGTGCCTAATGAGTG
AGCTAACTCACATTAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGAAACCTGTCGTGC
CAGCTGCATTAATGAATCGGCCAACGCGCGGGGAGAGGCGGTTTGCGTATTGGGCGCTCTTCC
GCTTCCTCGCTCACTGACTCGCTGCGCTCGGTCGTTCGGCTGCGGCGAGCGGTATCAGCTCAC
TCAAAGGCGGTAATACGGTTATCCACAGAATCAGGGGATAACGCAGGAAAGAACATGTGAGCA
AAAGGCCAGCAAAAGGCCAGGAACCGTAAAAAGGCCGCGTTGCTGGCGTTTTTCCATAGGCTC
CGOCCCCCTGACGAGCATCACAAAAATCGACGCTCAAGTCAGAGGTGGCGAAACCCGACAGGA
CTATAAAGATACCAGGCGTTTCCCCCTGGAAGCTCCCTCGTGCGCTCTCCTGITCCGACCCTG
CCGCTTACCGGATACCTGTCCGCCTTTCTCCCTTCGGGAAGCGTGGCGCTTTCTCATAGCTCA
CGCTGTAGGTATCTCAGTTCGGTGTAGGTCGTTCGCTCCAAGCTGGGCTGTGTGCACGAACCC
CCCGTTCAGCCCGACCGCTGCGCCTTATCCGGTAACTATCGTCTTGAGTCCAACCCGGTAAGA
CACGACTTATCGCCACTGGCAGCAGCCACTGGTAACAGGATTAGCAGAGCGAGGTATGTAGGC
GGTGCTACAGAGTTCTTGAAGTGGTGGCCTAACTACGGCTACACTAGAAGAACAGTATTTGGT
ATCTGCGCTCTGCTGAAGCCAGTTACCTTCGGAAAAAGAGTTGGTAGCTCTTGATCCGGCAAA
AAAACCACCGCTGGTAGCGGTGGTTTTTTTGTTTGCAAGCAGCAGATTACGCGCAGAAAAAAA
GGATCTCAAGAAGATCCTTTGATCTTTTCTACGGGGTCTGACGCTCAGTGGAACGAAAACTCA
CGTTAAGGGATTTTGGTCATGAGATTATCAAAAAGGATCTTCACCTAGATCCTTTTAAATTAA
AAATGAAGTTTTAAATCAATCTAAAGTATATATGAGTAAACTTGGTCTGACAGTTACCAATGC
TTAATCAGTGAGGCACCTATCTCAGCGATCTGTCTATTTCGTTCATCCATAGTTGCCTGACTC
CCCGTCGTGTAGATAACTACGATACGGGAGGGCTTACCATCTGGCCCCAGTGCTGCAATGATA
CCGCGAGACCCACGCTCACCGGCTCCAGATTTATCAGCAATAAACCAGCCAGCCGGAAGGGCC
GAGCGCAGAAGTGGTCCTGCAACTTTATCCGCCTCCATCCAGTCTATTAATTGTTGCCGGGAA
GCTAGAGTAAGTAGTTCGCCAGTTAATAGTTTGCGCAACGTTGTTGCCATTGCTACAGGCATC
GTGGTGTCACGCTCGTCGTTTGGTATGGCTTCATTCAGCTCCGGTTCCCAACGATCAAGGCGA
GTTACATGATCCCCCATGTTGTGCAAAAAAGCGGTTAGCTCCTTCGGTCCTCCGATCGTTGTC
AGAAGTAAGTTGGCCGCAGTGTTATCACTCATGGTTATGGCAGCACTGCATAATTCTCTTACT
GTCATGCCATCCGTAAGATGCTTTTCTGTGACTGGTGAGTACTCAACCAAGTCATTCTGAGAA
TAGTGTATGCGGCGACCGAGTTGCTCTTGCCCGGCGTCAATACGGGATAATACCGCGCCACAT
AGCAGAACTTTAAAAGTGCTCATCATTGGAAAACGTTCTTCGGGGCGAAAACTCTCAAGGATC
TTACCGCTGTTGAGATCCAGTTCGATGTAACCCACTCGTGCACCCAACTGATCTTCAGCATCT
TTTACTTTCACCAGCGTTTCTGGGTGAGCAAAAACAGGAAGGCAAAATGCCGCAAAAAAGGGA
ATAAGGGCGACACGGAAATGTTGAATACTCATACTCTTCCTTTTTCAATATTATTGAAGCATT
TATCAGGGTTATTGTCTCATGAGCGGATACATATTTGAATGTATTTAGAAAAATAAACAAATA
GGGGTTCCGCGCACATTTCCCCGAAAAGTGCCACCTATTGGTGTGGAAAGTCCCCAGGCTCCC
CAGCAGGCAGAAGTATGCAAAGCATGCATCTCAATTAGTCAGCAACCAGGTGTGGAAAGTCCC
CAGGCTCCCCAGCAGGCAGAAGTATGCAAAGCATGCATCTCAATTAGTCAGCAACCATAGTCC
CGCCCCTAACTCCGCCCATCCCGCCCCTAACTCCGCCCAGTTCCGCCCATTCTCCGCCCCATG
GCTGACTAATTTTTTTTATTTATGCAGAGGCCGAGGCCGCCTCGGCCTCTGAGCTATTCCAGA
AGTAGTGAGGAGGCTTTTTTGGAGGCCTAGGCTTTTGCAAAAAGCTAGCTTGCATGCCTGCAG
GTCGGCCGCCACGACCGGTGCCGCCACCATCCCCTGACCCACGCCCCTGACCCCTCACAAGGA
GACGACCTTCCATGACCGAGTACAAGCCCACGGTGCGCCTCGCCACCCGCGACGACGTCCCCC
GGGCCGTACGCACCCTCGCCGCCGCGTTCGCCGACTACCCCGCCACGCGCCACACCGTCGACC

CGGACCGCCACATCGAGCGGGTCACCGAGCTGCAAGAACTCTTCCTCACGCGCGTCGGGCTCG
ACATCGGCAAGGTGTGGGTCGCGGACGACGGCGCCGCGGTGGCGGTCTGGACCACGCCGGAGA
GCGTCGAAGCGGGGGCGGTGTTCGCCGAGATCGGCCCGCGCATGGCCGAGTTGAGCGGTTCCC
GGCTGGCCGCGCAGCAACAGATGGAAGGCCTCCTGGCGCCGCACCGGCCCAAGGAGCCCGCGT
GGTTCCTGGCCACCGTCGGCGTCTCGCCCGACCACCAGGGCAAGGGTCTGGGCAGCGCCGTCG
TGCTCCCCGGAGTGGAGGCGGCCGAGCGCGCCGGGGTGCCCGCCTTCCTGGAGACCTCCGCGC
CCCGCAACCTCCCCTTCTACGAGCGGCTCGGCTTCACCGTCACCGCCGACGTCGAGGTGCCCG
AAGGACCGCGCACCTGGTGCATGACCCGCAAGCCCGGTGCCTGACGCCCGCCCCACGACCCGC
AGCGCCCGACCGAAAGGAGCGCACGACCCCATGGCTCCGACCGAAGCCACCCGGGGCGGCCCC
GCCGACCCCGCACCCGCCCCCGAGGCCCACCGACTCTAGAGGATCATAATCAGCCATACCACA
TTTGTAGAGGTTTTACTTGCTTTAAAAAACCTCCCACACCTCCCCCTGAACCTGAAACATAA_A
ATGAATGCAATTGTTGTTGTTAACTTGTTTATTGCAGCTTATAATGGTTACAAATAAAGCAAT
AGCATCACAAATTTCACAAATAAAGCATTTTTTTCACTGCAATCTAAGAAACCATTATTATCA
TGACATTAACCTATAAAAATAGGCGTATCACGAGGCCCTTTCGTC
SEQ ID NO.-52 GTAAGCGGATCCATGGATGACGACGCGGCGCCC
SEQ ID NO. 53 GTAAGCAAGCTTCTTCTCTTTCACCTGTGGATT
SEQ ID NO.:54 GTACATTTATATTGGCTCATGTCCAATATGACCGCCATGTTGACATTGATTATTGACTAGTTA
TTAATAGTAATCAATTACGGGGTCATTAGTTCATAGCCCATATATGGAGTTCCGCGTTACATA
ACTTACCGTAAATGGCCCGCCTGGCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAAT
GACGTATGTTCCCATAGTAACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTT
ACGGTAAACTGCCCACTTGGCAGTACATCAAGTGTATCATATGCCAAGTCCGCCCCCTATTGA
CGTCAATGACGGTAAATGGCCCGCCTGGCATTATGCCCAGTACATGACCTTACGGGACTTTCC
TACTTGGCAGTACATCTACGTATTAGTCATCGCTATTACCATGGTGATGCGGTTTTGGCAGTA
CACCAATGGGCGTGGATAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCCATTGACGT
CAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCAAAATGTCGTAATAACCCCGC
CCCGTTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGAGGTCTATATAAGCAGAGCTCGTTT
AGTGAACCGTCAGATCCTCACTCTCTTCCGCATCGCTGTCTGCGAGGGCCAGCTGTTGGGCTC
GCGGTTGAGGACAAACTCTTCGCGGTCTTTCCAGTACTCTTGGATCGGAAACCCGTCGGCCTC
CGAACGGTACTCCGCCACCGAGGGACCTGAGCCAGTCCGCATCGACCGGATCGGAAAACCTCT
CGAGAAAGGCGTCTAACCAGTCACAGTCGCAAGGTAGGCTGAGCACCGTGGCGGGCGGCAGCG
GGTGGCGGTCGGGGTTGTTTCTGGCGGAGGTGCTGCTGATGATGTAATTAAAGTAGGCGGTCT
TGAGCCGGCGGATGGTCGAGGTGAGGTGTGGCAGGCTTGAGATCGAGCTGTTGGGGTGAGTAC
TCCCTCTCAAAAGCGGGCATGACTTCTGCGCTAAGATTGTCAGTTTCCAAAAACGAGGAGGAT
TTGATATTCACCTGGCCCGATCTGGCCATACACTTGAGTGACAATGACATCCACTTTGCCTTT
CTCTCCACAGGTGTCCACTCCCAGGTCCAAGTTTGCCGCCACCATGGAGACAGACACACTCCT
GCTATGGGTACTGCTGCTCTGGGTTCCAGGTTCCACTGGCGCCGGATCAACTCACACATGCCC
ACCGTGCCCAGCACCTGAACTCCTGGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAA
GGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGA
AGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAA
GCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCA
GGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCCCAGCCCCCAT
CGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCC
ATCCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCC
CAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCC
TCCCGTGTTGGACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAG
GTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACAC
GCAGAAGAGCCTCTCCCTGTCTCCCGGGAAAGCTAGCGGAGCCGGAAGCACAACCGAAAACCT
GTATTTTCAGGGCGGATCCGAATTCAAGCTTGATATCTGATCCCCCGACCTCGACCTCTGGCT
AATAAAGGAAATTTATTTTCATTGCAATAGTGTGTTGGAATTTTTTGTGTCTCTCACTCGGAA

GGACATATGGGAGGGCAAATCATTTGGTCGAGATCCCTCGGAGATCTCTAGCTAGAGCCCCGC
CGCCGGACGAACTAAACCTGACTACGGCATCTCTGCCCCTTCTTCGCGGGGCAGTGCATGTAA
TCCCTTCAGTTGGTTGGTACAACTTGCCAACTGAACCCTAAACGGGTAGCATATGCTTCCCGG
GTAGTAGTATATACTATCCAGACTAACCCTAATTCAATAGCATATGTTACCCAACGGGAAGCA
TATGCTATCGAATTAGGGTTAGTAAAAGGGTCCTAAGGAACAGCGATGTAGGTGGGCGGGCCA
AGATAGGGGCGCGATTGCTGCGATCTGGAGGACAAATTACACACACTTGCGCCTGAGCGCCAA
GCACAGGGTTGTTGGTCCTCATATTCACGAGGTCGCTGAGAGCACGGTGGGCTAATGTTGCCA
TGGGTAGCATATACTACCCAAATATCTGGATAGCATATGCTATCCTAATCTATATCTOGGTAG
CATAGGCTATCCTAATCTATATCTGGGTAGCATATGCTATCCTAATCTATATCTGGGTAGTAT
ATGCTATCCTAATTTATATCTGGGTAGCATAGGCTATCCTAATCTATATCTGGGTAGCATATG
CTATCCTAATCTATATCTGGGTAGTATATGCTATCCTAATCTGTATCCGGGTAGCATATGCTA
TCCTAATAGAGATTAGGGTAGTATATGCTATCCTAATTTATATCTOGOTAGCATATACTACCC
AAATATCTGGATAGCATATGCTATCCTAATCTATATCTGGGTAGCATATGCTATCCTAATCTA
TATCTGGGTAGCATAGGCTATCCTAATCTATATCTGGGTAGCATATGCTATCCTAATCTATAT
CTGGGTAGTATATGCTATCCTAATTTATATCTGGGTAGCATAGGCTATCCTAATCTATATCTG
GGTAGCATATGCTATCCTAATCTATATCTGGGTAGTATATGCTATCCTAATCTGTATCCGGGT
AGCATATGCTATCCTCACGATGATAAGCTGTCAAACATGAGAATTAATTCTTGAAGACGAAAG
GGCCTCGTGATACGCCTATTTTTATAGGTTAATGTCATGATAATAATGGTTTCTTAGACGTCA
GGTGGCACTTTTCGGGGAAATGTGCGCGGAACCCCTATTTGTTTATTTTTCTAAATACATTCA
AATATGTATCCGCTCATGAGACAATAACCCTGATAAATGCTTCAATAATATTGAAAAAGGAAG
AGTATGAGTATTCAACATTTCCGTGTCGCCCTTATTCCCTTTTTTGCGGCATTTTGCCTTCCT
GTTTTTGCTCACCCAGAAACGCTGGTGAAAGTAAAAGATGCTGAAGATCAGTTGGGTGCACGA
GTGGGTTACATCGAACTGGATCTCAACAGCGGTAAGATCCTTGAGAGTTTTCGCCCCGAAGAA
CGTTTTCCAATGATGAGCACTTTTAAAGTTCTGCTATGTGGCGCGGTATTATCCCGTGTTGAC
GCCGGGCAAGAGCAACTCGGTCGCCGCATACACTATTCTCAGAATGACTTGGTTGAGTACTCA
CCAGTCACAGAAAAGCATCTTACGGATGGCATGACAGTAAGAGAATTATGCAGTGCTGCCATA
ACCATGAGTGATAACACTGCGGCCAACTTACTTCTGACAACGATCGGAGGACCGAAGGAGCTA
ACCGCTTTTTTGCACAACATGGGGGATCATGTAACTCGCCTTGATCGTTGGGAACCGGAGCTG
AATGAAGCCATACCAAACGACGAGCGTGACACCACGATGCCTGCAGCAATGGCAACAACGTTG
CGCAAACTATTAACTGGCGAACTACTTACTCTAGCTTCCCGOCAACAATTAATAGACTGGATG
GAGGCGGATAAAGTTGCAGGACCACTTCTGCGCTCGGCCCTTCCGGCTGGCTGGTTTATTGCT
GATAAATCTGGAGCCGGTGAGCGTGGGTCTCGCGGTATCATTGCAGCACTGGGGCCAGATGGT
AAGCCCTCCCGTATCGTAGTTATCTACACGACGGGGAGTCAGGCAACTATGGATGAACGAAAT
AGACAGATCGCTGAGATAGGTGCCTCACTGATTAAGCATTGGTAACTGTCAGACCAAGTTTAC
TCATATATACTTTAGATTGATTTAAAACTTCATTTTTAATTTAAAAGGATCTAGGTGAAGATC
CTTTTTGATAATCTCATGACCAAAATCCCTTAACGTGAGTTTTCGTTCCACTGAGCGTCAGAC
CCCGTAGAAAAGATCAAAGGATCTTCTTGAGATCCTTTTTTTCTGCGCGTAATCTGCTGCTTG
CAAACAAAAAAACCACCGCTACCAGCGOTGGTTTGTTTGCCGGATCAAGAGCTACCAACTCTT
TTTCCGAAGGTAACTGGCTTCAGCAGAGCGCAGATACCAAATACTGTCCTTCTAGTGTAGCCG
TAGTTAGGCCACCACTTCAAGAACTCTGTAGCACCGCCTACATACCTCGCTCTGCTAATCCTG
TTACCAGTGGCTGCTGCCAGTGGCGATAAGTCGTGTCTTACCGGGTTGGACTCAAGACGATAG
TTACCGGATAAGGCGCAGCGGTCGGGCTGAACGGGGGGTTCGTGCACACAGCCCAGCTTGGAG
CGAACGACCTACACCGAACTGAGATACCTACAGCGTGAGCATTGAGAAAGCGCCAGGOTTCCC
GAAGGGAGAAAGGCGGACAGGTATCCGGTAAGCGGCAGGGTCGGAACAGGAGAGCGCACGAGG
GAGCTTCCAGGGGGAAACGCCTGGTATCTTTATAGTCCTGTCGGGTTTCGCCACCTCTGACTT
GAGCGTCGATTTTTGTGATGCTCGTCAGGGOGGCGGAGCCTATGGAAAAACGCCAGCAACGCG
GCCTTTTTACGGTTCCTGGCCTTTTGCTGGCCTTTTGCTCACATGTTCTTTCCTGCGTTATCC
CCTGATTCTGTGGATAACCGTATTACCGCCTTTGAGTGAGCTGATACCGCTCGCCGCAGCCGA
ACGACCGAGCGCAGCGAGTCAGTGAGCGAGGAAGC
SEQ ID NO.:55 GTAAGCAAGCTTAGGCCGCTGGGACAGCGGAGGTGC
SEQ ID NO :56 GTAAGCAAGCTTGGCAGCAGCGCCAGGTCCAGC

SEQ 0 NO :57 GTAAGCAGCGC T GT GGCTGCACCAT CT GT CT TC
SEQ ID NO.-58 GTAAGCGC TAGCC TAACACT CT CCC CT GTT GAAGC
SEQ ID NO..59 GCTGT GGCTGCACCATCTGTCTT CAT CTTCCCGCCATCTGATGAGCAGTT GAAATCTGGAACT
GCCTCTGTTGTGTGCCTGCTGAATAACTTCTATCCCAGAGAGGCCAAAGTACAGTGGAAGGTG
GATAACGCCCTCCAATCGGGTAACT CCCAGGAGAGTGTCACAGAGCAGGACAGCAAGGACAGC
ACCTACAGCCTCAGCAGCACCCTGACGCTGAGCAAAGCAGACTACGAGAAACACAAAGTCTAC
GCCTGCGAAGTCACCCATCAGGGCCTGAGCT CGCCCGTCACAAAGAGCTTCAACAGGGGAGAG
TGT TAG
SEQ ID NO.:60 AVAAPS VET EPPS DEQLKSGTASVVCLLNN FYPREAKVQWKVDNALQS GNSQESVTEQDS KDS
T YS LS STLTLSKADYEKHKVYACEVTHQGLS S PVT KS FNRGEC
SEQ ID NO :61 CTTGAGCCGGCGGATGGTCGAGGTGAGGTGTGGCAGGCT TGAGAT CCAGCTGTT GGGGTGAGT
ACTCCCTCTCAAAAGCGGGCATTACTTCTGCGCTAAGAT TGTCAGTTTCCAAAAACGAGGAGG
ATTTGATATTCACC TGGCCCGATCTGGCCATACACTTGAGTGACAATGACAT CCACTTTGCCT
TTCT CT CCACAGGTGTCCAC TCCCAGGTCCAAGT TTAAACGGAT CTCTAGCGAATT CATGAAC
TTTCTGCTGTCTTGGGTGCATTGGAGCCTTGCCTTGCTGCTCTACCTCCACCATGCCAAGTGG
TCCCAGGCTTGAGACGGAGCT TACAGCGCT GTGGCTGCACCATCTGTCT TCATCTTCCCGCCA
T C TGAT GAG CAGTT GAAAT CT GGAACTGCCT CTGTTGTGTGCCT GCT GAATAACT T C TAT CCC
AGAGAGGCGAAAGTACAGTGGAAGGTGGATAACGCCCTCCAATCGGGTAACTCCCAGGAGAGT
GTCACAGAGCAGGACAGCAAGGACAGCAC CTACACC CTCAGCAGCACCCTGACGCTGAGCAAA
GCAGACTACGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCAGGGCCTGAGCTCGCCC
GT CACAAAGAGCT T CAACAGGGGAGAGT GT TAGGGTACCGCGGCCGCT TCGAAT GAGAT CCCC
CGACCTCGACCTCTGGCTAATAAAGGAAATTTATTTT CATTGCAATAGTGTGTTGGAATTT TT
T GT GT CTCT CACT CGGAAGGACATAT GGGAGGGCAAAT CAT TT GGTC GAGATCCCT CGGAGAT
CTCTAGCTAGAGCCCCGCCGCCGGACGAACTAAACCTGACTACGGCATCTCTGCCCCTTCTTC
GCGGGGCAGT GCATGTAATCCCTT CAGTTGGTTGGTACAACTTGCCAACT GGGCCCT GT TCCA
CATGTGACACGGGGGGGGACCAAACACAAAGGGGTT CTCT GACTGTAGT TGACATC CT TATAA
AT GGAT GTGCACATT T GCCAACACT GAGT GGCTT T CATC CT GGAGCAGACT TT GCAGT CT GTG
GACTGCAACACAACATTGCCT TTAT GT GTAACTCTTGGCTGAAGCTCTTACACCAATGC TGGG
GGACATGTACCTCCCAGGGGCCCAGGAAGACTACGGGAGGCTACACCAACGTCAATCAGAGGG
GCCTGT GTAGC TACCGATAAGCGGACCCT CAAGAGGGCAT TAGCAATAGT GTT TATAAGGC CC
CC TT GT TAACCC TAAACGGGTAGCATAT GCTTCCCGGGTAGTAGTATATAC TAT CCAGAC TAA
CC C TAATT CAATAGCATAT GT TACCCAACGGGAAGCATAT GCTATC GAATTAGGGTTAGTAAA
AGGGTCC TAAGGAACAGCGATAT CT CCCACCCCAT GAGCT GT CAC GGTT TTAT TTACATGGGG
TCAGGATTCCACGAGGGTAGTGAACCATTTTAGTCACAAGGGCAGTGGCTGAAGATCAAGGAG
CGGGCAGTGAACTCTCCTGAATCTTCGCCTGCTTCTTCATTCTCCTTCGTTTAGCTAATAGAA
TAACT GC T GAGTT GT GAACAGTAAGGTG TATGT GAG CT GCT CGAAAACAAGGTT T CAGGT GAO
GCCCCCAGAATAAAATTTGGACGGGGGGTTCAGTGGTGGCATTGTGC TATGACACCAATATAA
CCCTCACAAACC CCTT GGGCAATAAATAC TAGTG TAGGAAT GAAACATT CT GAATAT OTT TAA
CRATAGAAATCCATGGGGTGGGGACAAGCCGTAAAGACTGGATGTCCATCTCACACGAATTTA
T GGC TAT GGGCAACACATAAT CCTAGT GCAATAT GATACTGGGGT TAT TAAGAT GT GT CCCAG
GCAGGGACCAAGACAGGT GAAC CAT GT T GT TACAC TC TAT TT GTAACAAGGGGAAAGAGAGTG
GACGCCGACAGCAGCGGACTCCACTGGTTGTCTCTAACACCCCCGAAAATTAAACGGGGCTCC
ACGCCAAT GGGGCCCATAAACAAAGACAAGT GGCCAC T CTTT TT T TT GAAATTGTGGAGTGGG
GGCACGCGTCAGCCCCCACACGCCGCCCTGCGGTTTTGGACTGTAAAATAAGGGTGTAATAAC
TTGGCTGATTGTAACCCCGCTAACCACTGCGGTCAAACCACTTGCCCACAAAACCACTAATGG

CACCCCGGGGAATACCTGCATAAGTAGGTGGGCGGGCCAAGATAGGGGCGCGATTGCTGCGAT
CTGGAGGACAAATTACACACACTTGCGCCTGAGCGCCAAGCACAGGGTTGTTGGTCCTCATAT
TCACGAGGTCGCTGAGAGCACGGTGGGCTAATGTTGCCATGGGTAGCATATACTACCCAAATA
TCTGGATAGCATATGCTATCCTAATCTATATCTGGGTAGCATAGGCTATCCTAATCTATATCT
GGGTAGCATATGCTATCCTAATCTATATCTGGGTAGTATATGCTATCCTAATTTATATCTGSG
TAGCATAGGCTATCCTAATCTATATCTGGGTAGCATATGCTATCCTAATCTATATCTGGGTAG
TATAT GCTATCCTAAT CTGTATCCGGGTAGCATATGCTATCCTAATAGAGAT TAGGGTAGTAT
ATGCTATCCTAATT TATATCTGGGTAGCATATACTACCCAAATATCTGGATAGCATAT GCTAT
CCTAATCTATATCTGGGTAGCATATGCTATCCTAATCTATATCTGGGTAGCATAGGCTATCCT
AATCTATATCTGGGTAGCATATGCTAT CCTAATCTATATCTGGGTAGTATATGCTAT CCTAAT
TTATAT CT GGGTAGCATAGGCTATCC TAATC TATAT CTGGGTAGCATATGCTAT CCTAATCTA
TATCTGGGTAGTATATGCTATCCTAATCTGTATCCGGGTAGCATATGCTATCCTCACGATGAT
AAGCT GTCAAACATGAGAAT TAATTCT TGAAGACGAAAGGGCCTCGTGATACGCCTATTTTTA
TAGGTTAATGTCATGATAATAATGGTTTCTTAGACGTCAGGTGGCACTTTTCGGGGAAATGTG
CGCGGAACCCCTATTTGTTTATTTTTCTAAATACATTCAAATATGTATCCGCTCATGAGACAA
TAACCCTGATAAATGCTTCAATAATATTGAAAAAGGAAGAGTATGAGTATTCAACATTTCCGT
GTCGCCCTTATTCCCTTTTTTGCGGCATTTTGCCTTCCTGTTTTTGCTCACCCAGAAACGCTG
GTGAAAGTAAAAGATGCTGAAGATCAGTTGGGTGCACGAGTGGGTTACATCGAACTGGATCTC
AACAGCGGTAAGATCCTTGAGAGTTTTCGCCCCGAAGAACGTTTTCCAATGATGAGCACTTTT
AAAGTTCTGCTATGTGGCGCGGTATTATCCCGTGTTGACGCCGGGCAAGAGCAACTCGGTCGC
CGCATACACTATTCTCAGAATGACTTGGTTGAGTACTCACCAGTCACAGAAAAGCATCTTACG
GAT GGCATGACAGTAAGAGAATTATGCAGT GC TGCCATAACCATGAGTGATAACACTGCGGCC
AACTTACT TCTGACAACGATCGGAGGACC GAAGGAGCTAACC GC TTTT TTGCACAACATGGGG
GAT CATGTAAC TC GCCT TGAT CGTT GGGAACCGGAGCTGAAT GAAGCCATACCAAAC GACGAG
CGTGACACCACGATGCCTGCAGCAATGGCAACAAC GTTGCGCAAACTAT TAACTGGCGAACTA
CT TACTCTAGCT TCCCGGCAACAAT TAATAGACTGGATGGAGGCGGATAAAGTTGCAGGACCA
CTTCTGCGCTCGGCCCTTCCGGCTGGCTGGTTTATTGCTGATAAATCTGGAGCCGGTGAGCGT
GGGTCTCGCGGTATCATTGCAGCACTGGGGCCAGATGGTAAGCCCTCCCGTATCGTAGTTATC
TACACGACGGGGAGTCAGGCAACTATGGATGAACGAAATAGACAGATCGCTGAGATAGGTGCC
TCACTGATTAAGCATTGGTAACTGTCAGACCAAGT TTACT CATATATACT T TAGAT TGAT TTA
AAACTT CAT TT T TAATT TAAAAGGATCTAGGTGAAGATCCT TT TT GATAATC TCATGACCAAA
ATCCCTTAACGTGAGTTTTCGTTCCACTGAGCGTCAGACCCCGTAGAAAAGATCAAAGGATCT
TCTTGAGATCCTTTTTTTCTGCGCGTAATCTGCTGCTTGCAAACAAAAAAACCACCGCTACCA
GCGGTGGTTTGTTTGCCGGATCAAGAGCTACCAACTCTTTTTCCGAAGGTAACTOGCTTCAGG
AGAGCGCAGATACCAAATACTGTCCTTCTAGTGTAGCCGTAGTTAGGCCACCACTTCAAGAAC
TCTGTAGCACCGCCTACATACCTCGCTCTGCTAATCCTGTTACCAGTGGCTGCTGCCAGTGGC
GATAAGTCGTGTCTTACCGGGTTGGACTCAAGACGATAGTTACCGGATAAGGCGCAGCGGTCG
GGC TGAACGGGGGGTTCGTGCAGACAGCCCAGCT TGGAGCGAACGACCTACACCGAACT GAGA
TACCTACAGCGTGAGCATTGAGAAAGCGCCACGCT TCCCGAAGGGAGAAAGGCGGACAGGTAT
CCGGTAAGCGGCAGGGTCGGAACAGGAGAGCGCACGAGGGAGCTTCCAGGGGGAAACGCCTGG
TATCTTTATAGTCCTGTCGGGTTTCGCCACCTCTGACTTGAGCGTCGATTTTTGTGATGCTCG
TCAGGGGGGCGGAGCCTATGGAAAAACGCCAGCAACGCGGCCTTTTTACGGTTCCTGGCCTTT
TGCTGGCCTTTTGCTCACATGTTCTTTCCTGCGTTATCCCCTGATTCTGTGGATAACCGTATT
ACCGCCTTTGAGTGAGCTGATACCGCTCGCCGCAGCCGAACGACCGAGCGCAGCGAGTCAGTG
AGCGAGGAAGCGGAAGAGCGCCCAATACGCAAACCGCCTCT CCCCGC GC GTTGGCCGAT T CAT
TAATGCAGCTGGCACGACAGGTTTCCCGACTGGAAAGCGGGCAGTGAGCGCAACGCAATTAAT
GTGAGTTAGCTCACTCATTAGGCACCCCAGGCTTTACACTTTATGCTTCCGGCTCGTATGTTG
TGTGGAAT TGT GAGCGGATAACAAT TT CACACAGGAAACAGC TAT GAC CAT GAT TACGCCAAG
CTCTAGCTAGAGGTCGACCAATTCTCATGTTTGACAGCTTATCATCGCAGATCCGGGCAACGT
TGTTGCATTGCTGCAGGCGCAGAACTGGTAGGTATGGCAGATCTATACATTGAATCAATATTG
GCAATTAGCCATATTAGTCATTGGTTATATAGCATAAATCAATATTGGCTATTGGCCATTGCA
TACGT T GTATCTATATCATAATATGTACAT T TATAT T GGCT CAT GT CCAATATGACCGCCATG
TTGACATTGATTATTGACTAGTTATTAATAGTAATCAATTACGGGGTCATTAGTTCATAGCCC
ATATATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGACCGCCCAACGA
CCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATAGGGACTTTCCA

TTGACGTCAATGGGTGGAGTATTTACGGTAAACTGCCCACTTGGCAGTACATCAAGTGTATCA
TATGCCAAGTCCGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGGCATTATGCCCA
GTACATGACCTTACGGGACTTTCCTACTTGGCAGTACATCTACGTATTAGTCATCGCTATTAC
CATGGTGATGCGGTTTTGGCAGTACACCAATCGGCGTGGATAGCGGTTTGACTCACGGGGATT
TCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGACTT
TCCAAAATGTCGTAATAACCCCGCCCCGTTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGA
GGTCTATATAAGCAGAGCTCGTTTAGTGAACCGTCAGATCCTCACTCTCTTCCGCATCGCTGT
CTGCGAGGGCCAGCTGTTGGGCTCGCGGTTGAGGACAAACTCTTCGCGGTCTTTCCAGTACTC
TTGGATCGGAAACCCGTCGGCCTCCGAACGGTACTCCGCCACCGAGGGACCTGAGCGAGTCCG
CATCGACCGGATCGGAAAACCTCTCGAGAAAGGCGTCTAACCAGTCACAGTCGCAAGGTAGGC
TGAGCACCGTGGCGGGCGGCAGCGGGTGGCGGTCGGGGTTGTTTCTGGCGGAGGTGCTGCTGA
TGATGTAATTAAAGTAGGCGGT
SEQ DI NO.:62 ATGCCAAGTGGTCCCAGGCTGACATTGTGATGACCCAGTCTGC
SEQ ID NO.:63 ATGCCAAGTGGTCCCAGGCTGATGTTTTGATGACCCAAACTCC
SEQ ID NO :64 ATGCCAAGTGGTCCCAGGCTGACATCGTTATGTCTCAGTCTCC
SEQ ID NO.:65 GGGAAGATGAAGACAGATGGTGCAGCCACAGC
SEQ ID NO..66 GTAAGCGCTAGCGCCTCAACGAAGGGCCCATCTGTCTTTCCCCTGGCcCC
SEQ ID NO.:67 GTAAGCGAATTCACAAGATTTGGGCTCAACTTTCTTG
SEQ ID NO. 68 GCCTCCACCAAGGGCCCATCGGTCTTCCCCCTGGCACCCTCCTCCAAGAGCACCTCTGGGGGC
ACAGCAGCCCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAACCGGTGACGGTGTCGTGGAAC
TCAGGCGCCCTGACCAGCGGCGTGCACACCTTCCCGGCTGTCCTACAGTCCTCAGGACTCTAC
TCCCTCAGCAGCGTGGTGACCGTGCCCTCCAGCAGCTTGGGCACCCAGACCTACATCTGCAAC
GTGAATCACAAGCCCAGCAACACCAAGGTGGACAAGAAAGTTGAGCCCAAATCTTGT
SEQ ID NO :69 ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLY
SLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSC
SEQ ID NO.:70 CTTGAGCCGGCGGATGGTCGAGGTGAGGTGTGGCAGGCTTGAGATCCAGCTGTTGGGGTGAGT
ACTCCCTCTCAAAAGCGGGCATTACTTCTOCGCTAAGATTGTCAGTTTCCAAAAACGAGGAGG
ATTTGATATTCACCTGGCCCGATCTGGCCATACACTTGAGTGACAATGACATCCACTTTGCCT
TTCTCTCCACAGGTGTCCACTCCCAGGTCCAAGTTTGCCGCCACCATGGAGACAGACACACTC
CTGCTATGGGTACTGCTGCTCTGGGTTCCAGGTTCCACTGGCGGAGACGGAGCTTACGGGCCC
ATCTGTCTTTCCCCTGGCCCCCTCCTCCAAGAGCACCTCTGGGGGCACAGCGGCCCTGGGCTG
CCTGGTCAAGGACTACTTCCCCGAACCGGTGACGGTGTCGTGGAACTCAGGCGCCCTGACCAG
CGGCGTGCACACCTTCCCGGCTGTCCTACAGTCCTCAGGACTCTACTCCCTCAGCAGCGTGGT
GACCGTGCCCTCCAGCAGCTTGGGCACCCAGACCTACATCTGCAACGTGAATCACAAGCCCAG
CAACACCAAGGTGGACAAGAAAGTTGAGCCCAAATCTTGTGAATTCACTCACACATGCCCACC
GTGCCCAGCACCTGAACTCCTGGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGA
CACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGA

CCCTGAGGTCAAGTTCAACTGGTACGTGGPiCGGCGTGGAGGTGCATAATGCCAAGACAAAGCC
GCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGA
CTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCT CCAACAAAGCCCTCCCAGCCCCCATCGA
GAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATC
CCGGGATGAGCTGACCAAGAACCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCCAG
CGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCC
CGTGCTGGACTCCGACGGCTCCTTCTTCCTCTACACCAAGCTCACCGTGGACAAGAGCAGGTG
GCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCA
GAAGAGCCTCTCCCTGTCTCCCGGGAAATGATCCCCCGACCTCGACCTCTGGCTAATAAAGGA
AATTTATTTTCATTGCAATAGTGTGTTGGAATTTTTTGTGTCTCTCACTCGGAAGGACATATG
GGAGGGCAAATCATTTGGTCGAGATCCCTCGGAGATCTCTAGCTAGAGCCCCGCCGCCGGACG
AACTAAACCTGACTACGGCATCTCTGCCCCTTCTTCGCGGGGCAGTGCATGTAATCCCTTCAG
TTGGTTGGTACAACTTGCCAACTGAACCCTAAACGGGTAGCATATGCTTCCCGGGTAGTAGTA
TATACTATCCAGACTAACCCTAATTCAATAGCATATGTTACCCAACGGGAAGCATATGCTATC
GAATTAGGGTTAGTAAAAGGGTCC TAAGGAACAGCGATGTAGGT GGGCGGGCCAAGATAGGGG
CGCGATTGCTGCGATCTGGAGGACAAATTACACACACTTGCGCCTGAGCGCCAAGCACAGGGT
TGTTGGTCCTCATATTCACGAGGTCGCTGAGAGCACGGTGGGCTAATGTTGCCATGGGTAGCA
TATACTACCCAAATATCTGGATAGCATATGCTATCCTAATCTATATCTGGGTAGCATAGGCTA
TCCTAATCTATATCTGGGTAGCATATGCTATCCTAATCTATATCTGGGTAGTATATGCTATCC
TAATTTATATCTGGGTAGCATAGGCTATCCTAATCTATATCTGGGTAGCATATGCTATCCTAA
TCTATATCTGGGTAGTATATGCTATCCTAATCTGTATCCGGGTAGCATATGCTATCCTAATAG
AGATTAGGGTAGTATATGCTATCCTAATT TATAT CTGGGTAGCATATACTACCCAAATATCTG
GATAGCATATGCTATCCTAATCTATATCTGGGTAGCATATGCTATCCTAATCTATATCTGGGT
AGCATAGGCTATCCTAATCTATATCTGGGTAGCATATGCTATCCTAATCTATATCTGGGTAGT
ATATGCTAT CCTAAT TTATAT CT GGGTAGCATAGGC TATCCTAATCTATATCT GGGTAGCATA
TGCTAT CC TAAT CTATATCTGGGTAGTATATGCTATCCTAATCTGTATCCGGGTAGCATATGC
TATCCTCACGATGATAAGCTGT CAAACAT GAGAATTAATT CT TGAAGACGAAAGGGCCTCGTG
ATACGCCTATT TT TATAGGT TAATGT CAT GATAATAATGGTT T CTTAGACGTCAGGTGGCACT
TTTCGGGGAAATGTGCGCGGAACCCCTATTTGTTTATTTTTCTAAATACATTCAAATATGTAT
CCGCTCATGAGACAATAACCCTGATAAATGCTTCAATAATATTGAAAAAGGAAGAGTATGAGT
ATTCAACATTTCCGTGTCGCCCTTATTCCCTTTTTTGCGGCATTTTGCCTTCCTGTTTTTGCT
CACCCAGAAACGCTGGTGAAAGTAAAAGATGCTGAAGATCAGTTGGGTGCACGAGTGGGTTAC
ATCGAACTGGATCTCAACAGCGGTAAGATCCTTGAGAGTTTTCGCCCCGAAGAACGTTTTCCA
ATGATGAGCACTTTTAAAGTTCTGCTATGTGGCGCGGTATTATCCCGTGTTGACGCCGGGCAA
GAGCAAC TCGGTCGCCGCATACACTAT TC TCAGAAT GAC TT GGTTGAGTACTCACCAGTCACA
GAAAAGCATCTTAGGGATGGCATGACAGTAAGAGAATTATGCAGTGCTGCCATAACCATGAGT
GATAACACTGCGGCCAACTTACTTCTGACAACGATCGGAGGACCGAAGGAGCTAACCGCTTTT
TTGCACAACATGGGGGATCATGTAACTCGCCTTGATCGTTGGGAACCGGAGCTGAATGAAGGC
ATACCAAACGACGAGCGTGACACCACGATGCCTGCAGCAATGGCAACAACGTTGCGCAAACTA
T TAACTGGCGAACTACT TACT CTAGCTTCCCGGCAACAATTAATAGACTGGATGGAGGCGGAT
AAAGTTGCAGGACCACTTCTGCGCTCGGCCCTTGCGGCTGGCTGGTTTATTGCTGATAAATCT
GGAGCCGGTGACCGTGGGTCTCGCGGTATCATTGCAGCACTGGGGCCAGATGGTAAGCCCTCC
CGTATCGTAGT TAT CTACACGACGGGGAGT CAGGCAAC TAT GGATGAACGAAATAGACAGATC
GCTGAGATAGGTGCCTCACTGATTAAGCATTGGTAACTGTCAGACCAAGTTTACTCATATATA
CTTTAGAT TGATTTAAAAC TT CATTTT TAATTTAAAAGGATCTAGGTGAAGATCCTT TT TGAT
AAT CT CATGACCAAAATCCC TTAACGTGAGTTTTCGTTCCACTGAGCGT CAGACCCCGTAGAA
AAGATCAAAGGATCTTCTTGAGATCCTTTTTTTCTGCGCGTAATCTGCTGCTTCCAAACAAAA
AAACCACCGCTACCAGCGGTGGTTTGTTTGCCGGATCAAGAGCTACCAACTCTTTTTCCGAAG
GTAACTGGCTTCAGCAGAGCGCAGATACCAAATACTGTCCTTCTAGTGTAGCCGTAGTTAGGC
CACCACTTCAAGAACTCTGTAGCACCGCCTACATACCTCGCTCTGCTAATCCTGTTACCAGTG
GCTGCTGCCAGTGGCGATAAGTCGTGTCTTACCGGGTTGGACTCAAGACGATAGTTACCGGAT
AAGGCGCAGCGGTCGGGCTGAACGGGGGGTTCGTGCACACAGCCCAGCTTGGAGCCAACGACC
TACACCGAACT GAGATACCTACAGCGT GAGCATT GAGAAAGCGCCACGCTTCCCGAAGGGAGA
AAGGCGGACAGGTATCCGGTAAGCGGCAGGGTCGGAACAGGAGAGCGCACGAGGGAGCTTCCA
GGGGGAAACGCCTCGTATCTTTATAGTCCTGTCGGGTTTCGCCACCTCTGACTTGAGCGTCGA

TTTTTGTGATGCTCGTCAGGGGGGCGGAGCCTATGGAAAAACGCCAGCAACGCGGCCTTTTTA
CGGTTCCTGGCCTTTTGCTGGCCTTTTGCTCACATGTTCTTTCCTGCGTTATCCCCTGATTCT
GTGGATAACCGTATTACCGCCTTTGAGTGAGCTGATACCGCTCGCCGCAGCCGAACGACCGAG
CGCAGCGAGTCAGTGAGCGAGGAAGCGTACATTTATATTGGCTCATGTCCAATATGACCGCCA
TGTTGACATTGATTATTGACTAGTTATTAATAGTAATCAATTACGGGGTCATTAGTTCATAGC
CCATATATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGACCGCCCAAC
GACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATAGGGACTTTC
CATTGACGTCAATGGGTGGAGTATTTACGGTAAACTGCCCACTTGGCAGTACATCAAGTGTAT
CATATGCCAAGTCCGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGGCATTATGCC
CAGTACATGACCTTACGGGACTTTCCTACTTGGCAGTACATCTACGTATTAGTCATCGCTATT
ACCATGGTGATGCGGTTTTGGCAGTACACCAATGGGCGTGGATAGCGGTTTGACTCACGGGGA
TTTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGAC
TTTCCAAAATGTCGTAATAACCCCGCCCCGTTGACGCAAATGGGCGGTAGGCGTGTACGGTGG
GAGGTCTATATAAGCAGAGCTCGTTTAGTGAACCGTCAGATCCTCACTCTCTTCCGCATCGCT
GTCTGCGAGGGCCAGCTGTTGGGCTCGCGGTTGAGGACAAACTCTTCGCGGTCTTTCCAGTAC
TCTTGGATCGGAAACCCGTCGGCCTCCGAACGGTACTCCGCCACCGAGGGACCTGAGCGAGTC
CGCATCGACCGGATCGGAAAACCTCTCGAGAAAGGCGTCTAACCAGTCACAGTCGCAAGGTAG
GCTGAGCACCGTGGCGGGCGGCAGCGGGTGGCGGTCGGGGTTGTTTCTGGCGGAGGTGCTGCT
GATGATGTAATTAAAGTAGGCGGT
SEQ ID NO.:71 GGGTTCCAGGTTCCACTGGCGAGGTTCAGCTGCAGCAGTCTGT
SEQ ID NO.:72 GGGTTCCAGGTTCCACTGGCGAGGTGCAGCTTCAGGAGTCAGG
SEQ ID NO.:73 GGGGCCAGGGGAAAGACAGATGGGCCCTTCGTTGAGGC
SEQ ID NO.: 91: Exemplary embodiment of CDRL1 K-S-S-Q-S-L-L-N/H-SIT-S/N/D-N/G-Q/N/K-K/L-N-Y-L-A
SEQ ID NO. :92: Exemplary embodiment of CDRL1 K-A-S-Q-D-I-H-NIT-Y/F-L-N
SEQ ID N093: Exemplary embodiment of CDRL2 F-A-S-T-R-E-S
SEQ ID NO.: 94: Exemplary embodiment of CDRL2 L-V-S-K-L-D-S
SEQ ID NO.:95: Exemplary embodiment of CDRL2 R-A-N-R-L-V-D
SEQ ID NO. :96: Exemplary embodiment of CDRL3 Q-Q-H-Y-S-T-P-L-T
SEQ ID NO.:97: Exemplary embodiment of CDRL3 VV/L-Q-Y/G-D/T-A/E/H-F-P-R-T
SEQ ID NO. :98: Exemplary embodiment of CDRH1 1 SEQ ID NO.:99: Exemplary embodiment of CDRH1 G-F-T/S-I-T-S-G-Y-G-W-H

SEQ ID NO. :100: Exemplary embodiment of CDRH2 V/N/G-I/L-D-P-E/NG-T/Y-G-X-T-A
SEQ ID NO.:101: Exemplary embodiment of CDRH2 Y-I-N/S-F/Y-N/D-G
SEQ ID NO.:102: Exemplary embodiment of CDRH3 M-G-Y-S/A-D-Y
SEQ ID NO.:103: Exemplary embodiment of CDRH3 A-S-S-Y-D-G-F-L-A-Y
SEQ ID NO.:104: Exemplary embodiment of CDRH3 3 A-RNV-W/F-G-L-R-Q/N
SEQ ID NO.:158 KSSQSLLHSDGKTYLN
SEQ ID NO.:159 LVSKLDS
SEQ ID NO.:160 SEQ ID NO.:161 GYTFTD YNMH
30 SEQ ID NO.:162 YINPYNDVTE
SEQ ID NO.:163 AWFGL RQ
SEQ ID NO.:164 RSSKSLLHSNGN TYLY
SEQ ID NO.:165 RMSNLAS
SEQ ID NO.:166 MQHLEYPYT
SEQ ID NO.:167 GDTFTD YYMN
SEQ ID NO.:168 DINPNYGGIT
SEQ ID NO.:169 QAYYRNS DY
SEQ ID NO.:170 KASQDVGTAVA

SEQ ID NO.:171 WTSTRHT
SEQ ID NO.:172 QQHYSIPLT
SEQ ID NO.:173 GYIFTDYEIH
SEQ ID NO.:174 VIDPETGNTA
SEQ ID NO.:175 MGYSDY
SEQ ID NO.:176 MVLQTQVFISLLLWISGAYGDIVMTQSPDSLAVSLGERATINCKSSQSLLNSNFQKN
FLAWYQQKPGQPPKLLIYFASTRESSVPDRFSGSGSGTDFTLTISSLQAEDVAVYYC
QQHYSTPLTFGQGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAK
VQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG
LSSPVTKSFNRGEC
SEQ ID NO.:177 M DWTVVRI LFLVAAATGTHAEVQLVQSGAEVKKPGASVKVSC KASGYI FTDYE I HVVV
RQAPGQGLEWMGVIDPETGNTAFNQKFKGRVTITADTSTSTAYMELSSLTSEDTAV
YYCMGYSDYVVGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPE
PVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSN
TKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVD
VSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEY
KCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDI
AVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEA
LHNHYTQKSLSLSPGK
SEQ ID N:178 DIVMTQSPDSLAVSLGERATINCKSSQSLLNSNFQKNFLAWYQQKPGQPPKLLIYFA
STRESSVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQHYSTPLTFGQGTKLEIK
SEQ ID NO.:179 EVQLVQSGAEVKKPGASVKVSCKASGYI FTDYE I HVVVRQAPGQGLEWMGVI DPET
GNTAFNQKFKGRVTITADTSTSTAYMELSSLTSEDTAVYYCMGYSDYWGQGTLVTV
SS
SEQ ID NO.:180 MVLQTQVFISLLLWISGAYGDIVMTQSPSSLSASVGDRVTITCKASQDIHNFLNWFQ
QKPGKAPKTL I FRANRLVDGVPSRFSGSGSGTDYTLTISSLQPEDFATYSCLQYDEI
PLTFGQGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVD
NALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTK
SFNRGEC
SEQ ID NO.:181 MDWTVVRILFLVAAATGTHAEVQLQESGPGLVKPSQTLSLTCTVSGFSITSGYGWH
WIRQHPGKGLEWIGYINYDGHNDYNPSLKSRVTISQDTSKNQFSLKLSSVTAADTAV
YYCASSYDGLFAYVVGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKD
YFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNH
KPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTC

VVVDVSHEDPEVKFNVVYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLN
GKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGF
YPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSV
MHEALHNHYTQKSLSLSPGK
SEQ ID No.:182 DIVMTQSPSSLSASVGDRVTITCKASQDIHNFLNWFQQKPGKAPKTLIFRANRLVDG
VPSRFSGSGSGTDYTLTISSLOPEDFATYSCLQYDEIPLTFGQGTKLEIK
SEQ ID NO.:183 EVQLQESGPGLVKPSQTLSLTCTVSGFSITSGYGWHWI RQ H PG KGLEWI GYI NYDG
HNDYNPSLKSRVTISQDTSKNQFSLKLSSVTAADTAVYYCASSYDGLFAYVVGQGTL
VTVS

Table A: Light chains variable region of selected antibodies SEQID
NO:
3z1A02L 105 DAVMTQIPLTLSVTIGQPASLSC KSSQSLLHSDGK TYLN
WLLQRPGQSPKRLIS LVSKLDS GVPDRFTGSGSGTDFTLKISRVEAEDLGLYYC WQGTHFPRT FAGGTNLEIK

N
3z1F06L 106 SIVMTQTPLTLSVTIGQPASITC KSSQSLLYSDGK TYLN WLLQRPGQSPKRLIS
LVSKLDS GVPDGFTGSGSGTDFTLKISRVEAEDLGVYYC WQGTHFPRT FGGGTKLEIK
1-, 1-, 3z1E08L 107 DAVMTQIPLTLSVTIGQPASISC KSSQSLLHSDGK TYLN
WLLQRPGQSPKRLIY LVSKLDS GVPDRFTGSGSGTDFTLKISRVEAEDLGVYYC WQGTHFPRT FGGGTKLEIK
'a CA
3z1G1OL 108 DVLMTQTPRSLSVSLGDQASISC RSSQSLLHSNGN TYLE
WYLQKPGQPPKVLIY KVSNRFS GVPDRFSGSGSGTDFTLKISGVEAEDLGVYYC FQGSHVPLT FGAGTKLELK
.6.
1-, 1-, 3z1E1OL 109 DIVMTQAAPSVPVTPGESVSISC RSSKSLLHSNGN TYLY
WFLQRPGQSPQLLIY RMSNLAS GVPDRFSGSGSGTAFTLRISRVEAEDVGVYYC MQHLEYPYT FGGGTKLEIK
N
3z1A09L 110 DIVMTQSPSSLAMSLGQKVTMSC KSSQSLLNSNNQLNYLA WYQQKPGQSPKLLVY
FASTRKS GVPDRFIGSGSGTDFTLTITSVQAEDLADYFC QQHFNTPLT FGAGTKLELK
3z1B01L 111 DIVMTQSPSSLAISVGQKVTMSC KSSQSLLNSSNQKNYLA WYQQKPGQSPKLLVF
FASTRES GVPDRFIGSGSGTDFTLTISSVQAEDLADYFC QQHYSIPLT FGAGTKLELK
3z1G05L 112 DIVMTQSPSSLAMSVGQKVTMSC KSSQSLLNSSNQKNYLA WYQQKPGQSPKLLVF
FASTRES GVPDRFIGSGSGTDFTLTITSVQAEDLADYFC QQHYSIPLT FGSGTKLELK
3z1B02L 113 DIVMTQSPSSLAMSVGQKVTMSC KSSQSLLNSSNQKNYLA WYQQKPGQSPKLLVY
FASTRES GVPDRFIGSGSGTDFTLTISSVQAEDLADYFC QQHYSIPLT FGAGTKLELK
3z1508L 114 DIVMTQSPSSLAMSVGQKVTMSC KSSQSLLNSSNQKNYLA WYQQKPGQSPKLLVY
FASTRES GVPDRFIGSGSGTDFTLTISSVQAEDLADYFC QQHYSTPLT FGAGTKLELK
g3z1G08L 115 DIVMTQSPSSLAMSVGQKVTMSC KSSQSLLNSSNQKNYLA
WYQQKPGQSPKLLVY FASTRES GVPDRFIGSGSGTDFTLTISSVQAEDLADYFC QQHYSTPLT FGAGTKLELK
n r) 3z1F07L 116 DIVMTQSPSSLAMSVGQKVTMSC
KSSQSLLNSSNQKNYLA WYQQKPGQSPKLLIY FASTRES GVPDRFIGSGSGTDFTLTISSVQAEDLADYFC
QQHYSTPLT FGAGTKLELK o 1-3iv 3z1E09L 117 DIVMTQSPSSLAMSVGQKVTMSC KSSQSLLNSSNQKNYLA
WYQQKPGQSPKLLVY FASTRES GVPDRFIGSGSGTEFTLTITSVQAEDLADYFC QQHYSTPLT FGAGTKLELK
co 1-eilH
M 3z1003L 118 DIVMTQSPSSLAMSVGQKVTMSC
KSSQSLLNSSNQKNYLA WYQQKPGQSPKLLVY FGSTRES GVPDRFIGSGSGTDFTLTISGVQAEDLADYFC
QQHYSTPLT FGAGTKLELK m Fl.
^ N'w 3z1E12L 119 DIVMTQSPSSLAMSVGQKVTMNC KSSQSLLNRSNQKNYLA WYQQKPGQSPKLLVY
FASTRES GVPDRFIGSGSGTDFTLTISSVQAEDLADYFC QQHYSIPLT FGAGTKLELK
cAn VI
: 421A02L 120 DIVMTQSPSSLAMSVGQKVTMNC
KSSQSLLNNSNQKNYLA WYQQKPGQSPKLLLY FASTRES GVPDRFIGSGSGTYFTLTISSVQAEDLADYFC
QQHYSTPLT FGAGTKLDLK iv M

H
M 3z1F1OL 121 DIVMTQSPSSLTMSVGQKVTMSC
KSSQSLLNTSNQLNYLA WYQQKPGQSPKLLVY FASTTES GVPDRFIGSGSGTDFTLTISSVQAEDLADYFC
QQHYSTPLT FGAGTKLELK w 3z1F04L 122 DIVMTQSPSSLTVTAGEKVTMSC KSSQSLLNTSNQKNYLA
WYQQKPGQSPKLLVY FASTRAS GVPDRFIGSGSGTDFTLTISSVQAEDLADYFC QQHYSTPLT FGAGTKLELK

II.
PZ
I
C71, 3z1B11L 123 DIVMTQSPSSLAMSVGQKVTMSC
KSSQSLLNSSNQKNYLA WYQQKPGQSPKLLVY FASTRES GVPDRFIGSGSGTDFTLTISSVQAEDLADYFC

M 3z1D03L 124 DIVMTQSPSSLAVSIGQKVTMNC KSSQSLLNSNFQKNFLA
WYQQKPGQSPKLLIY FASTRES SIPDRFIGSGSGTDFTLTISSVQAEDLADYFC QQHYSTPLT FGAGTKLELK
1-, 3z1003L 125 DIVMTQSPSSLAMSVGQKVTMSC KSSQSLLNSSNQKNYLA
WYQQKPGQSPKLLVY FGSTRES GVPDRFIGSGSGTDFTLTISGVQAEDLADYFC QQHYSTPLT FGAGTKLELK
....., 3z1G12L 126 DIVMTQSPKFMSTSVGDRVSITC KASQDVG TAVA
WYQQKPGQSPELLIY WTSTRHT GVPDRFSGSGSGTDFTLTISSVQAEDLADYFC QQHYSIPLT FGAGTKLELR
3z1C04L 127 DIVMSQSPSSMYASLGERVTITC KASQDIH NFLN
WFQQKPGKSPKTLIF RANRLVD GVPSRFSGSGSGQDYSLTISSLEFEDLGIYSC LQYDEIPLT FGAGTKLELR
3z1DO1L 128 DIKMTQSPSSMYASLGERVTITC KASQDIH TYLN
WFQQKPGKSPETLIY RANRLVD GVPSRFSGSGSGQDYSLTISSLEYEDMGIYYC LQYDEFPLT FGAGTKLELK
IV
3z1002L 129 DIQMTQSPSSMYASLGERVTLTC KASQDIH
NYLN WFQQKPGKSPKTLIH RANRLVA
GVPSRFSGSGSGQDYSLTISSLEYEDLGIYYC LQYDAFPLT FGAGTKLELK r) 1-i 3z1E06L 130 DIQMTQSPSSMYASLGERVTLTC KASQDIH NYLN
WFQQKPGKSPKTLIH RANRLVA GVPSRFSGSGSGQDYSLTISSLEYEDLGIYYC LQYDAFPLT FGAGTKLELK
r) 3z1H03L 131 DIVMSQSPSSMYASLGERVTITC KASQDIH RFLN
WFQQKPGKSPKTLIF HANRLVD GVPSRFSGSGSGLDYSLTISSLEYEDMGIYFC LQYDAFPLT FGAGTKLELK
N
1-, 'a 1-, -.1 CA

Table B: Heavy chains variable region of selected antibodies SEQID
NO:
3z1A02H 132 HEIQLQQSGPELVKPGASVKMSCKTS GYTFTD YNME WVKQKPGQGLEWIG
YINPYNDVTE YNEKFKGRATLTSDKSSSTAYMDLSSLTSDDSAVYFC AWFGL RQ WGQGTLVTVST
3z1F06H 133 HEVQLQQSGPELVKPGASVKMSCKAS GYIFTE YNIH WVKQKPGQGPEWIG
NINPYNDVTE YNEKFKGKATLTSDKASSTAYMDLSSLTSEDSAVYYC ARWGL RN WGQGTLVTVSA
3z1E08H 134 HEVQLQQSVPELVKPGASVKMSCKTS GYTFTE YNNE WVKQKPGQGPEWIG
NINPYNNVTE YNEKFKGKATLTSDKSSSTAYLDLSSLTSEDSAVYYC ARWGL RN WGQGTLVTVSA
3z1A09H 135 HQVQVQQPGAELVRPGASVTLSCKAS GYIFTD YEVE WVRQRPVHGLEWIG
VIDPETGDTA YNQKFKGKATLTADKSSSTAYMELSSLTAEDSAVYYC IGYA DY WGQGTTLTVSS
3z1B01H 136 HQVQLQQPGAELVRPGASVTLSCKAS GYTFTD YEIH WVKQTPVHGLEWIG
VIDPETGGTA YNQKFKGKATLTTDKSSSTAYMELRSLTSEDSAVYYC MGYS DY WGQGTTLTVSS
3z1B02H 137 HEVQLQQSGAELVRPGASVTLSCKAS GYTFTD YEIH WVKQTPVHGLEWIG
VIDPETGATA YNQKFKGKATLTADKSSSTAYMELSSLTSEDSAVYYC MGYS DY WGQGTTLTVSS
3z1F04H 138 HEVQLQQSGAELVRPGASVTLSCKAS GYTFTD YEIH WVKQTPVHGLEWIG
VIDPETGSTA YNQKFKGKATLTADKASSTAYMELSSLTSEDSAVYYC MGYS DY WGQGTTLTVSS

VIDPETGSTA YNQKFKGKATLTADKSSSTAYMELSSLTSEDSAVYYC MGYA DY WGQGTTLTVSS
3z1B08H 140 HEVQLQQSGAELVRPGASVTLSCKAS GYTFTD YEIH WVKQTPVHGLEWIG
VIDPETGDTA YNQNFTGKATLTADKSSSTAYMELSSLTSEDSAVYYC MGYA DY WGQGTTLTVSS
r) 3z1G08H 141 HQVQLKQSGAELVRPGASVTLSCKAS GYTFTD YEW WVKQTPVHGLEWIG
VIDPATGDTA YNQKFKGKATLTADKSSSTAYMEVSSLTSEDSAVYYC MGYS DY WGQGTTLTVSS
1-3 3z1F07H 142 HQAYLQQSGAELVRPGASVTLSCKAS GYTFTD YEIH WVKQTPVHGLEWIG
VIDPETGDTA YNQKFKDKATLTADKASSTAYMELSSLTSEDSAVYYC MGYS DY WGQGTTLTVSS
3z1E12H 143 HQVQLQQSEAELVKPGASVKLSCKAS GYTFTD YEIH WVKQTPVHGLEWIG
VIDPETGDTA YNQKFKGKATLTADKSSSTAYMELSRLTSEDSAVYYC MGHS DY WGQGTTLTVSS

VIDPETGNTA FNQKFKGKATLTADISSSTAYMELSSLTSEDSAVYYC MGYS DY WGQGTTLTVSS
cp 17' 3z1G12H 145 HEVQLQQSVAELVRPGASVTVSCKAS GYIFTD YEIH WVKQTPAHGLEWIG
VIDPETGNTA FNQKFKGKATLTADISSSTAYMELSSLTSEDSAVYYC MGYS DY WGQGTTLTVSS
3z1F1OH 146 HEVQLQQSVAELVRPGAPVTLSCKAS GYTFTD YEVH WVKQTPVHGLEWIG
VIDPETGATA YNQKFKGKATLTADKSSSAAYMELSRLTSEDSAVYYC MSYS DY WGQGTTLTVSS
3z1CO3H 147 HEVQLQQSVAEVVRPGASVTLSCKAS GYTFTD YEIH WVKQTPVHGLEWIG
VIDPETGVTA YNQRFRDKATLTTDKSSSTAYMELSSLTSEDSAVYFC MGYS DY WGQGTTLTVSS
1) C71, 3z1CO3H 148 HEVQLQQSVAEVVRPGASVTLSCKAS GYTFTD YEIH WVKQTPVHGLEWIG
VIDPETGVTA YNQRFRDKATLTTDKSSSTAYMELSSLTSEDSAVYFC MGYS DY WGQGTTLTVSS ( 3z1005H 149 HQVQLQQPGAELVRPGASVTLSCKAS GYTFTD YEIH WVKQTPVHGLEWIG
VLDPGTGRTA YNQKFKDKATLSADKSSSTAYMELSSLTSEDSAVYYC MSYS DY WGPGTTLTVSS
3z1B11H 150 HEVQLQQSVAELVRPGASVTLSCKAS GYTFTD YEMH WVKQTPVRGLEWIG
VIDPATGDTA YNQKFKGKATLTADKSSSAAFMELSSLTSEDSAVYYC MGYS DY WGQGTTLTVSS
3z1E06H 151 HQVQLQQSGAELVRPGASVTLSCKAS GYTFSD YEMH WVKQTPVHGLEWIG
GIDPETGDTV YNQKFKGKATLTADKSSSTAYMELSSLTSEDSAVYYC ISYAM DY WGQGTSVTVSS
4z1A02H 152 HQVKLQQSGTELVRPGASVTLSCKAS GYKFTD YEMH WVKQTPVHGLEWIG
GIDPETGGTA YNQKFKGKAILTADKSSTTAYMELRSLTSEDSAVYYC ISYAM DY WGQGTSVTVSS
3z1E1OH 153 HEVQLQQSGPELVKPGASVKISCKAS GDTFTD YYMN WVKQSHGKSLEWIG
DINPNYGGIT YNQKFKGKATLTVDTSSSTAYMELRGLTSEDSAVYYC QAYYRNS DY WGQGTTLTVSS

DINPYYGTTT YNQKFKGKATLTVDKSSRTAYMELRGLTSEDSAVYYC ARDDWF DY WGQGTLVTVSA

3z1D01H 155 HEVQLQESGPDLVKPSQSLSLTCTVT GFSITSGYGWH WIRQFPGDKLEWMG
YIS FNGDYN YNPSLKSRISITRDTSKNQFFLQLSSVTTEDTATYYC ASSYDGLFAY WGQGTLVTVSA
r) 3z1CO2H 156 HDVQLQESGPDLVKPSQSLSLTCTVT GFSITSGYGWH WIRQFPGNKLEWMG
YIS FNGDSN YNPSLKSRISITRDTSKNQFFLQLNSVTSEDTATYYC ASSYDGLFAY WGQGPLVTVSA
3z1C04H 157 HEVQLQESGPDLVKPSQSLSLTCTVT GFSITSGYGWH WIRQFPGNKLEWMG
YIN YDGHND YNPSLKSRISITQDTSKNQFFLQLNSVTTEDTATYYC ASSYDGLFAY WGQGTLVTVSA

Claims (88)

1. An antibody or antigen binding fragment thereof capable of specific binding to Kidney associated antigen 1 (KAAG1) and selected from the group consisting of:
a. an antibody or antigen binding fragment thereof having a light chain variable region at least 80% identical to SEQ ID NO.:16 and/or a heavy chain variable region at least 80% identical to SEQ ID NO.:18 wherein said antibody or antigen binding fragment thereof comprises at least one amino acid substitution in comparison with SEQ ID
NO.:16 or SEQ ID NO.:18 and wherein said amino acid substitution is outside of a complementarity determining region (CDR) and;
b. an antibody or antigen binding fragment thereof having a light chain variable region at least 80% identical to SEQ ID NO.:24 and/or a heavy chain variable region at least 80% identical to SEQ ID NO.:26 wherein said antibody or antigen binding fragment thereof comprises at least one amino acid substitution in comparison with SEQ ID
NO.:24 or SEQ ID NO.:26 and wherein said amino acid substitution is outside of a complementarity determining region (CDR).
2. The antibody or antigen binding fragment of claim 1, wherein the at least one amino acid substitution is in the light chain variable region.
3. The antibody or antigen binding fragment of claim 1, wherein said antibody or antigen binding fragment thereof comprises at least three amino acid substitutions.
4. The antibody or antigen binding fragment of claim 1, wherein said antibody or antigen binding fragment thereof comprises from one to twenty amino acid substitutions in the light chain variable region and/or heavy chain variable region.
5. The antibody or antigen binding fragment of claim 4, wherein the amino acid substitution is in the light chain variable region.
6. The antibody or antigen binding fragment of claim 4, wherein the amino acid substitution is in the heavy chain variable region.
7. The antibody or antigen binding fragment of any one of claims 1 to 6, wherein said amino acid substitution is conservative.
8. The antibody or antigen binding fragment of any one of claims 1 to 7, wherein the light chain variable region comprises amino acids 6 to 108 of SEQ ID NO.:184.
9. The antibody or antigen binding fragment of claim 8, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:185.
10. The antibody or antigen binding fragment of claim 9, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:186.
11. The antibody or antigen binding fragment of claim 10, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:178.
12. The antibody or antigen binding fragment of claim 11, wherein the light chain variable region is as set forth in SEQ ID NO.:178.
13. The antibody or antigen binding fragment of any one of claims 1 to 12 wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:187.
14. The antibody or antigen binding fragment of claim 13, wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:188.
15. The antibody or antigen binding fragment of claim 14, wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:189.
16. The antibody or antigen binding fragment of claim 15, wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:179.
17. The antibody or antigen binding fragment of claim 15, wherein the heavy chain variable region is as set forth in SEQ ID NO.:179.
18. The antibody or antigen binding fragment of claim 1, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:184 and wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:187.
19. The antibody or antigen binding fragment of claim 18, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:185 and wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:188.
20. The antibody or antigen binding fragment of claim 19, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:186 and wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:189.
21. The antibody or antigen binding fragment of claim 20, wherein the light chain variable region comprises at least 90 consecutive amino acids SEQ
ID NO.:178 and wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:179.
22. The antibody or antigen binding fragment of claim 21, wherein the light chain variable region is as set forth in SEQ ID NO.:178 and wherein the heavy chain variable region is as set forth in SEQ ID NO.:179.
23. The antibody or antigen binding fragment of any one of claims 1 to 7, wherein the light chain variable region comprises amino acids 6 to 102 of SEQ ID NO.:190.
24. The antibody or antigen binding fragment of claim 23, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:191.
25. The antibody or antigen binding fragment of claim 24, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:192.
26. The antibody or antigen binding fragment of claim 25, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:182.
27. The antibody or antigen binding fragment of claim 26, wherein the light chain variable region is as set forth in SEQ ID NO.:182.
28. The antibody or antigen binding fragment of any one of claims 1 to 7 or to 27, wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO:193.
29. The antibody or antigen binding fragment of claim 28, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:194.
30. The antibody or antigen binding fragment of claim 29, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:195.
31. The antibody or antigen binding fragment of claim 30, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:183.
32. The antibody or antigen binding fragment of claim 31, wherein the light chain variable region is as set forth in SEQ ID NO.:183.
33. The antibody or antigen binding fragment of claim 1, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:190 and wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:193.
34. The antibody or antigen binding fragment of claim 33, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:191 and wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:194.
35. The antibody or antigen binding fragment of claim 34, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:192 and wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:195.
36. The antibody or antigen binding fragment of claim 35, wherein the light chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:182 and wherein the heavy chain variable region comprises at least 90 consecutive amino acids of SEQ ID NO.:183.
37. The antibody or antigen binding fragment of claim 36, wherein the light chain variable region is as set forth in SEQ ID NO.:182 and wherein the heavy chain variable region is as set forth in SEQ ID NO:183.
38. The antibody or antigen binding fragment of any one of claims 1 to 37, wherein said antibody comprises amino acids of a constant region.
39. The antibody or antigen binding fragment of claim 38, wherein the amino acids of the constant region are from a human antibody.
40. The antibody or antigen binding fragment of any one of claims 1 to 40, comprising a human IgG1 constant region.
41. The antibody or antigen binding fragment of any one of claims 1 to 40, wherein the antigen binding fragment is a scFv, a Fab, a Fab' or a (Fab)2.
42. The antibody or antigen binding fragment of any one of claims 1 to 41, conjugated with a cytotoxic moiety.
43. The antibody or antigen binding fragment of any one of claims 1 to 41, conjugated with a detectable moiety.
44. The antibody or antigen binding fragment of any one of claims 1 to 43, for use in the treatment of cancer comprising tumor cells expressing KAAG1 or a KAAG1 variant.
45. The antibody or antigen binding fragment of any one of claims 1 to 43, for use in the detection of cancer comprising tumor cells expressing KAAG1 or a KAAG1 variant.
46. The antibody or antigen binding fragment of any one of claims 1 to 43, for use in preventing or treating metastasis.
47. An isolated antibody or antigen binding fragment capable of competing with the antibody or antigen binding fragment of any one of claims 1 to 43.
48. A nucleic acid encoding a light chain variable domain and/or a heavy chain variable domain of the antibody or antigen binding fragment of any one of claims 1 to 41.
49. A vector comprising the nucleic acid of claim 48.
50. The vector of claim 49, wherein said vector is an expression vector.
51. An isolated cell comprising the nucleic acid of claim 48.
52. The isolated cell of claim 51, wherein said cell comprises a nucleic acid encoding a light chain variable domain and a nucleic acid encoding a heavy chain variable domain.
53. The isolated cell of claim 52, wherein said cell is capable of expressing, assembling and/or secreting an antibody or antigen binding fragment thereof.
54. An isolated cell comprising or expressing the antibody or antigen binding fragment of any one of claims 1 to 41.
55. The isolated cell of claim 54, wherein said cell comprises a nucleic acid encoding a light chain variable domain and a nucleic acid encoding a heavy chain variable domain.
56. The isolated cell of claim 55, wherein said cell is capable of expressing, assembling and/or secreting an antibody or antigen binding fragment thereof.
57. A pharmaceutical composition comprising the antibody or antigen binding fragment of any one of claims 1 to 43, and a pharmaceutically acceptable carrier.
58. A composition comprising the antibody or antigen binding fragment of any one of claims 1 to 43, and a carrier.
59. A method of treating cancer comprising cells expressing kidney associated antigen 1 (KAAG1) or a KAAG1 variant or of treating metastasis, the method comprising administering the antibody or antigen binding fragment of any one of claims 1 to 43 to a subject in need.
60. The method of claim 59, wherein the cancer is selected from the group consisting of ovarian cancer, skin cancer, renal cancer, colorectal cancer, sarcoma, leukemia, brain tumor, thyroid tumor, breast cancer, prostate cancer, oesophageal tumor, bladder tumor, lung tumor and head and neck tumor.
61. The method of claim 60, wherein the cancer is ovarian cancer.
62. The method of claim 61, wherein the cancer is recurrent ovarian cancer.
63. A method for reducing tumor spread, reducing tumor invasion, tumor formation or for inducing tumor lysis, the method comprising administering the antibody or antigen binding fragment of any one of claims 1 to 43 to a subject in need.
64. The method of claim 63, wherein the subject in need has a cancer selected from the group consisting of ovarian cancer, skin cancer, renal cancer, colorectal cancer, sarcoma, leukemia, brain tumor, thyroid tumor, breast cancer, prostate cancer, oesophageal tumor, bladder tumor, lung tumor and head and neck tumor.
65. The method of claim 64, wherein the cancer is ovarian cancer.
66. The method of claim 65, wherein the cancer is recurrent ovarian cancer.
67. The method of any one of claims 63 to 66, wherein the cancer is metastatic.
68. A method of detecting a tumor comprising cells expressing kidney associated antigen 1 (KAAG1) or a KAAG1 variant, the method comprising administering the antibody or antigen binding fragment of any one of claims 1 to 43, to a subject in need.
69. The method of claim 68, wherein the subject in need has a cancer selected from the group consisting of ovarian cancer, skin cancer, renal cancer, colorectal cancer, sarcoma, leukemia, brain tumor, thyroid tumor, breast cancer, prostate cancer, oesophageal tumor, bladder tumor, lung tumor and head and neck tumor.
70. A method for detecting kidney associated antigen 1 (KAAG1: SEQ ID
NO.:2) or a KAAG1 variant having at least 80% sequence identity with SEQ ID NO.:2, the method comprising contacting a cell expressing KAAG1 or the KAAG1 variant or a sample comprising or suspected of comprising KAAG1 or the KAAG1 variant with the antibody of any one of claims 1 to 43 and measuring binding.
71. The method of claim 70, wherein the sample is from a mammal.
72. The method of claim 71, wherein the mammal has or is suspected of having cancer.
73. The method of claim 72, wherein the cancer is metastatic.
74. The method of any one of claims 70 to 73, wherein the sample is a serum sample, a plasma sample or a blood sample obtained from the mammal.
75. The method of any one of claims 70 to 73, wherein the sample is a tissue sample obtained from the mammal.
76. The method of claim 70 to 73, wherein the sample is a cell culture or a supernatant.
77. The method of any one of claims 70 to 76, comprising quantifying the amount of antibody bound to KAAG1 or the KAAG1 variant.
78. A kit comprising the antibody or antigen binding fragment of any one of claims 1 to 43.
79. Use of the antibody or antigen binding fragment of any one of claims 1 to 43 in the manufacture of a medicament for treatment of cancer comprising cells expressing kidney associated antigen 1 (KAAG1) or a KAAG1 variant, reducing tumor spread, reducing metastasis, reducing invasion of tumor cells expressing KAAG1 or a KAAG1 variant, reducing formation of tumor, inducing tumor lysis of tumor cells expressing KAAG1 or a KAAG1 variant and/or reducing spread of tumor cells expressing KAAG1 or a KAAG1 variant.
80. The use as defined in claim 79, wherein the cancer or tumor is selected from the group consisting of ovarian cancer, skin cancer, renal cancer, colorectal cancer, sarcoma, leukemia, brain tumor, thyroid tumor, breast cancer, prostate cancer, oesophageal tumor, bladder tumor, lung tumor and head and neck tumor.
81. The use as defined in claim 80, wherein the cancer is ovarian cancer.
82. The use as defined in claim 81, wherein the cancer is recurrent ovarian cancer.
83. The use as defined in any one of claims 79 to 82, wherein the cancer is metastatic.
84. Use of the antibody or antigen binding fragment of any one of claims 1 to 43 in the diagnosis of cancer comprising cells expressing kidney associated antigen 1 (KAAG1) or a KAAG1 variant or in the detection of cells expressing KAAG1 or a KAAG1 variant.
85. The use as defined in claim 84, wherein the cancer or tumor is selected from the group consisting of ovarian cancer, skin cancer, renal cancer, colorectal cancer, sarcoma, leukemia, brain tumor, thyroid tumor, breast cancer, prostate cancer, oesophageal tumor, bladder tumor, lung tumor and head and neck tumor.
86. The use as defined in claim 85, wherein the cancer is ovarian cancer.
87. The use as defined in claim 86, wherein the cancer is recurrent ovarian cancer.
88. The use as defined in any one of claims 84 to 87, wherein the cancer is metastatic.
CA2816437A 2009-11-03 2010-11-03 Antibodies that specifically block the biological activity of kidney associated antigen 1 Abandoned CA2816437A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA2816437A CA2816437A1 (en) 2009-11-03 2010-11-03 Antibodies that specifically block the biological activity of kidney associated antigen 1

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
PCT/CA2009/001586 WO2010060186A1 (en) 2008-11-03 2009-11-03 Antibodies that specifically block the biological activity of a tumor antigen
CAPCT/CA2009/001586 2009-11-03
PCT/CA2010/001795 WO2011054112A1 (en) 2009-11-03 2010-11-03 Antibodies that specifically block the biological activity of kidney associated antigen 1
CA2816437A CA2816437A1 (en) 2009-11-03 2010-11-03 Antibodies that specifically block the biological activity of kidney associated antigen 1

Publications (1)

Publication Number Publication Date
CA2816437A1 true CA2816437A1 (en) 2011-05-12

Family

ID=48570547

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2816437A Abandoned CA2816437A1 (en) 2009-11-03 2010-11-03 Antibodies that specifically block the biological activity of kidney associated antigen 1

Country Status (1)

Country Link
CA (1) CA2816437A1 (en)

Similar Documents

Publication Publication Date Title
US20180369269A1 (en) Antibodies that specifically block the biological activity of a tumor antigen
US10597450B2 (en) Antibodies against kidney associated antigen 1 and antigen binding fragments thereof
EA035947B1 (en) Anti-human trop-2 antibody exhibiting antitumor activity in vivo
WO2011054112A1 (en) Antibodies that specifically block the biological activity of kidney associated antigen 1
CA2816437A1 (en) Antibodies that specifically block the biological activity of kidney associated antigen 1
NZ615694B2 (en) Antibodies against kidney associated antigen 1 and antigen binding fragments thereof

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20130404

FZDE Dead

Effective date: 20170720