TLE3 AS A MARKER FOR CHEMOTHERAPY

Information

  • Patent Application
  • 20150198599
  • Publication Number
    20150198599
  • Date Filed
    March 27, 2015
    9 years ago
  • Date Published
    July 16, 2015
    9 years ago
Abstract
Methods of using TLE3 as a marker for predicting the likelihood that a patient's cancer will respond to chemotherapy. Methods of using TLE3 as a marker for selecting a chemotherapy for a cancer.
Description
SEQUENCE LISTING

In accordance with 37 C.F.R. §1.52(e)(5), a Sequence Listing in the form of a text file (entitled “Sequence Listing.txt,” created on Nov. 5, 2012, and 90 kilobytes) is incorporated herein by reference in its entirety.


BACKGROUND OF THE INVENTION

A major challenge of cancer treatment is the selection of chemotherapies that maximize efficacy and minimize toxicity for a given patient. Assays for cell surface markers, e.g., using immunohistochemistry (IHC), have provided means for dividing certain cancers into subclasses. For example, one factor considered in prognosis and treatment decisions for breast cancer is the presence or absence of the estrogen receptor (ER). ER-positive breast cancers typically respond much more readily to hormonal therapies such as tamoxifen, which acts as an anti-estrogen in breast tissue, than ER-negative cancers. Though useful, these analyses only in part predict the clinical behavior of breast cancers. There is phenotypic diversity present in cancers that current diagnostic tools fail to detect. As a consequence, there is still much controversy over how to stratify patients amongst potential treatments in order to optimize outcome (e.g., for breast cancer see “NIH Consensus Development Conference Statement: Adjuvant Therapy for Breast Cancer, Nov. 1-3, 2000”, J. Nat. Cancer Inst. Monographs, 30:5-15, 2001 and Di Leo et al., Int. J. Clin. Oncol. 7:245-253, 2002). In particular, there is currently no tool for predicting a patient's likely response to treatment with paclitaxel, a chemotherapeutic with particularly adverse side-effects. There clearly exists a need for improved methods and reagents for classifying cancers and thereby selecting therapeutic regimens that maximize efficacy and minimize toxicity for a given patient.


SUMMARY OF THE INVENTION

We have identified a correlation between the expression of TLE3 (transducin-like enhancer of split 3, Entrez Gene ID 7090) and a cancer's response to chemotherapy. This correlation has been demonstrated using TLE3 antibodies and samples from breast cancer cohorts which include both treated and untreated patients with known outcome. The inventors have also observed that binding of TLE3 antibodies in samples from treated ovarian cancer patients correlates with improved prognosis. In one aspect, the present invention therefore provides methods of using TLE3 as a marker for predicting the likelihood that a patient's cancer will respond to chemotherapy. In another aspect, the present invention provides methods of using TLE3 as a marker for deciding whether to administer chemotherapy to a cancer patient. In yet another aspect, the present invention provides methods of using TLE3 as a marker for selecting a chemotherapy for a cancer patient.


Expression of TLE3 can be detected using any known method. Thus, while the inventive methods have been exemplified by detecting TLE3 polypeptides using antibodies, in certain embodiments TLE3 polynucleotides may be detected using one or more primers as is well known in the art.


In general, TLE3 can be used in conjunction with other markers or clinical factors (e.g., stage, tumor size, node characteristics, age, etc.) to further improve the predictive power of the inventive methods.


BRIEF DESCRIPTION OF THE APPENDIX

This patent application refers to material comprising a table and data presented as Appendix A immediately after the section entitled “Exemplification” and immediately before the section entitled “Other Embodiments.” Specifically, Appendix A is a table that lists a variety of markers that could be used in a panel in conjunction with the TLE3 marker in an inventive method. The table includes the antibody ID, parent gene name, Entrez Gene ID, known aliases for the parent gene, peptides that may be used in preparing antibodies and exemplary antibody titers for staining Using the parent gene name, Entrez Gene ID and/or known aliases for the parent gene, a skilled person can readily obtain the nucleotide (and corresponding amino acid) sequences for each and every one of the parent genes that are listed in Appendix A from a public database (e.g., GenBank, Swiss-Prot or any future derivative of these). The nucleotide and corresponding amino acid sequences for each and every one of the parent genes that are listed in Appendix A are hereby incorporated by reference from these public databases. Antibodies with IDs that begin with S5 or S6 may be obtained from commercial sources as indicated.





BRIEF DESCRIPTION OF THE DRAWING


FIG. 1 compares IHC images of TLE3-negative (S0643-) and TLE3-positive (S0643+) samples from breast cancer patients.



FIG. 2 shows Kaplan-Meier recurrence curves that were generated using all patients in the Huntsville Hospital (HH) breast cancer cohort after classification based on staining with an antibody raised against the TLE3 marker. Recurrence data from TLE3-positive and TLE3-negative patients were used to generate the top and bottom curves, respectively. As shown in the Figure, antibody binding to the TLE3 marker correlates with improved prognosis across this breast cancer cohort (HR=0.573, p<0.004).



FIG. 3 shows Kaplan-Meier recurrence curves that were generated using all patients in the Roswell Park Cancer Institute (RP) breast cancer cohort after classification based on staining with an antibody raised against the TLE3 marker. The selected patients in the RP cohort were all triple negative for the ER (estrogen receptor, Entrez GeneID 2099), PR (progesterone receptor, Entrez GeneID 5241) and HER-2 markers (v-erb-b2 erythroblastic leukemia viral oncogene homolog 2, Entrez GeneID 2064). Recurrence data from TLE3-positive and TLE3-negative patients were used to generate the top and bottom curves, respectively. As shown in the Figure, antibody binding to the TLE3 marker correlates with improved prognosis across this breast cancer cohort (HR=0.24, p<0.011).



FIG. 4 shows Kaplan-Meier recurrence curves that were generated using patients in the HH breast cancer cohort of FIG. 1 that did not receive chemotherapy. Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, antibody binding to the TLE3 marker loses its correlation with prognosis in breast cancer patients that did not receive chemotherapy (HR=0.788, p=0.49).



FIG. 5 shows Kaplan-Meier recurrence curves that were generated using patients in the HH breast cancer cohort of FIG. 1 that did receive chemotherapy. Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, the correlation between antibody binding to the TLE3 marker and prognosis was restored in patients that did receive chemotherapy (HR=0.539, p<0.013).



FIG. 6 shows Kaplan-Meier recurrence curves that were generated using patients in the RP breast cancer cohort of FIG. 2 that did receive chemotherapy. Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, antibody binding to the TLE3 marker correlates with improved prognosis across this subset of breast cancer patients (HR=0.194, p=0.010). These results parallel those obtained in FIG. 5 with the HH cohort.



FIG. 7 shows Kaplan-Meier recurrence curves that were generated using patients in the HH breast cancer cohort of FIG. 5 that received CMF (cyclophosphamide, methotrexate and 5-fluorouracil) chemotherapy. Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, antibody binding to the TLE3 marker correlates with improved prognosis across this subset of treated patients (HR=0.398, p<0.019).



FIG. 8 shows Kaplan-Meier recurrence curves that were generated using patients in the HH breast cancer cohort of FIG. 5 that received CA (cyclophosphamide and adriamycin) or CAF (cyclophosphamide, adriamycin and 5-fluorouracil) chemotherapy (with or without a taxane). Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, the correlation between antibody binding to the TLE3 marker and prognosis loses significance in this subset of treated patients (HR=0.666, p=0.22).



FIG. 9 shows Kaplan-Meier recurrence curves that were generated using patients in the HH breast cancer cohort of FIG. 8 that received CA or CAF chemotherapy only (i.e., without a taxane). Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, there is no correlation between antibody binding to the TLE3 marker and prognosis in this subset of treated patients (HR=1.03, p=0.95).



FIG. 10 shows Kaplan-Meier recurrence curves that were generated using patients in the HH breast cancer cohort of FIG. 8 that received CA or CAF in combination with a taxane. Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, the correlation between antibody binding to the TLE3 marker and prognosis was restored in this subset of treated patients (HR=0.114, p=0.038).



FIG. 11 shows Kaplan-Meier recurrence curves that were generated using patients in the RP breast cancer cohort of FIG. 6 that received CA chemotherapy only (i.e., without a taxane). Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, there is no correlation between antibody binding to the TLE3 marker and prognosis in this subset of treated patients (HR=0.759, p=0.81).



FIG. 12 shows Kaplan-Meier recurrence curves that were generated using patients in the RP breast cancer cohort of FIG. 6 that received CA in combination with a taxane. Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, antibody binding to the TLE3 marker correlates with improved prognosis across this subset of treated patients (HR=0.142, p=0.011).



FIG. 13 shows Kaplan-Meier recurrence curves that were generated using patients in the RP breast cancer cohort of FIG. 6 that received a taxane or CMF. Some of the patients receiving a taxane also received CA. Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, antibody binding to the TLE3 marker correlates with improved prognosis across this subset of treated patients (HR=0.137, p=0.011).



FIG. 14 shows Kaplan-Meier recurrence curves that were generated using patients in the RP breast cancer cohort of FIG. 6 that received neoadjuvant chemotherapy. Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. The sample size was small (N=12); however, as shown in the Figure, antibody binding to the TLE3 marker showed significant correlation with improved prognosis across this subset of treated patients when measured using the Fisher Exact Test (p=0.005).



FIGS. 15-17 show Kaplan-Meier recurrence curves that were generated using patients in the RP breast cancer cohort of FIG. 6 that received chemotherapy. Recurrence data from TLE3-positive and TLE3-negative patients with stage II+ (FIG. 15), stage IIb+ (FIG. 16) and stage III+ (FIG. 17) cancers were used to generate the top and bottom curves, respectively. In each case, antibody binding to the TLE3 marker correlated with improved prognosis across these subsets of treated patients. The sample size was small in the subset of FIG. 17 (N=19); however significance was obtained when measured using the Fisher Exact Test (p=0.020).



FIG. 18 shows Kaplan-Meier recurrence curves that were generated using patients in the University of Alabama at Birmingham (UAB) ovarian cancer cohort. All patients received paclitaxel. Most patients also received platinum chemotherapy (carboplatin or cisplatin). Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, antibody binding to the TLE3 marker correlated with prognosis in these treated patients (HR=0.64, p<0.049).





DEFINITIONS

Binds—When an interaction partner “binds” a marker they are linked by direct non-covalent interactions.


Cancer markers—“Cancer markers” or “markers” are molecular entities that are detectable in cancer samples. Generally, markers may be polypeptides (e.g., TLE3 protein) or polynucleotides (e.g., TLE3 mRNA) that are indicative of the expression of a gene (e.g., TLE3 gene) and present within the cancer sample, e.g., within the cytoplasm or membranes of cancerous cells and/or secreted from such cells.


Cancer sample—As used herein, the term “cancer sample” or “sample” is taken broadly to include cell or tissue samples removed from a cancer patient (e.g., from a tumor, from the bloodstream, etc.), cells derived from a tumor that may be located elsewhere in the body (e.g., cells in the bloodstream or at a site of metastasis), or any material derived from such a sample. Derived material may include, for example, nucleic acids or proteins extracted from the sample, cell progeny, etc. In one embodiment, a cancer sample may be a tumor sample.


Correlation—“Correlation” refers to the degree to which one variable can be predicted from another variable, e.g., the degree to which a cancer's response to therapy can be predicted from the expression of a marker in a cancer sample. A variety of statistical methods may be used to measure correlation between two variables, e.g., without limitation the student t-test, the Fisher exact test, the Pearson correlation coefficient, the Spearman correlation coefficient, the Chi squared test, etc. Results are traditionally given as a measured correlation coefficient with a p-value that provides a measure of the likelihood that the correlation arose by chance. A correlation with a p-value that is less than 0.05 is generally considered to be statistically significant. Preferred correlations have p-values that are less than 0.01, especially less than 0.001.


Hybridized—When a primer and a marker are physically “hybridized” with one another as described herein, they are non-covalently linked by base pair interactions.


Interaction partner—An “interaction partner” is an entity that binds a polypeptide marker. For example and without limitation, an interaction partner may be an antibody or a fragment thereof that binds a marker. In general, an interaction partner is said to “bind specifically” with a marker if it binds at a detectable level with the marker and does not bind detectably with unrelated molecular entities (e.g., other markers) under similar conditions. Specific association between a marker and an interaction partner will typically be dependent upon the presence of a particular structural feature of the target marker such as an antigenic determinant or epitope recognized by the interaction partner. In general, it is to be understood that specificity need not be absolute. For example, it is well known in the art that antibodies frequently cross-react with other epitopes in addition to the target epitope. Such cross-reactivity may be acceptable depending upon the application for which the interaction partner is to be used. Thus the degree of specificity of an interaction partner will depend on the context in which it is being used. In general, an interaction partner exhibits specificity for a particular marker if it favors binding with that partner above binding with other potential partners, e.g., other markers. One of ordinary skill in the art will be able to select interaction partners having a sufficient degree of specificity to perform appropriately in any given application (e.g., for detection of a target marker, for therapeutic purposes, etc.). It is also to be understood that specificity may be evaluated in the context of additional factors such as the affinity of the interaction partner for the target marker versus the affinity of the interaction partner for other potential partners, e.g., other markers. If an interaction partner exhibits a high affinity for a target marker and low affinity for non-target molecules, the interaction partner will likely be an acceptable reagent for diagnostic purposes even if it lacks specificity.


Primer—A “primer” is an oligonucleotide entity that physically hybridizes with a polynucleotide marker. In general, a primer is said to “hybridize specifically” with a marker if it hybridizes at a detectable level with the marker and does not hybridize detectably with unrelated molecular entities (e.g., other markers) under similar conditions. Specific hybridization between a marker and a primer will typically be dependent upon the presence of a particular nucleotide sequence of the target marker which is complementary to the nucleotide sequence of the primer. In general, it is to be understood that specificity need not be absolute. The degree of specificity of a primer will depend on the context in which it is being used. In general, a primer exhibits specificity for a particular marker if it favors hybridization with that partner above hybridization with other potential partners, e.g., other markers. One of ordinary skill in the art will be able to select primers having a sufficient degree of specificity to perform appropriately in any given application. It is also to be understood that specificity may be evaluated in the context of additional factors such as the affinity of the primer for the target marker versus the affinity of the primer for other potential partners, e.g., other markers. If a primer exhibits a high affinity for a target marker and low affinity for non-target molecules, the primer will likely be an acceptable reagent for diagnostic purposes even if it lacks specificity.


Response—The “response” of a cancer to therapy may represent any detectable change, for example at the molecular, cellular, organellar, or organismal level. For instance, tumor size, patient life expectancy, recurrence, or the length of time the patient survives, etc., are all responses. Responses can be measured in any of a variety of ways, including for example non-invasive measuring of tumor size (e.g., CT scan, image-enhanced visualization, etc.), invasive measuring of tumor size (e.g., residual tumor resection, etc.), surrogate marker measurement (e.g., serum PSA, etc.), clinical course variance (e.g., measurement of patient quality of life, time to relapse, survival time, etc.).


Small molecule—A “small molecule” is a non-polymeric molecule. A small molecule can be synthesized in a laboratory (e.g., by combinatorial synthesis) or found in nature (e.g., a natural product). A small molecule is typically characterized in that it contains several carbon-carbon bonds and has a molecular weight of less than about 1500 Da, although this characterization is not intended to be limiting for the purposes of the present invention.


DETAILED DESCRIPTION OF CERTAIN PREFERRED EMBODIMENTS OF THE INVENTION

As noted above, we have identified a correlation between the expression of TLE3 (transducin-like enhancer of split 3, Entrez Gene ID 7090) in a cancer sample and a cancer's response to chemotherapy. As described in the Examples, this correlation has been demonstrated using TLE3 antibodies and samples from two breast cancer cohorts which include both treated and untreated patients with known outcome. We have also shown that this predictive model is consistent when applied to samples from a cohort of treated ovarian cancer patients. We have also demonstrated the utility of TLE3 for predicting response to specific types of chemotherapies including treatments which involve the administration of cell cycle specific chemotherapeutics, e.g., methotrexate and taxanes. Since these chemotherapeutics have known utility across different cancer types, these results suggest that the inventive methods will also be useful in predicting their efficacy across different cancer types.


Predicting Response to Chemotherapy and Selecting Chemotherapy

In one aspect, the present invention provides methods of using TLE3 as a marker for predicting the likelihood that a patient's cancer will respond to chemotherapy. In general, these methods involve providing a cancer sample from a cancer patient, determining whether TLE3 is expressed in the cancer sample, and predicting the likelihood that the patient's cancer will respond to chemotherapy based upon a result of the step of determining. In one embodiment, the step of predicting comprises predicting that the patient's cancer is likely to respond to chemotherapy based upon the presence of TLE3 expression in the cancer sample. In one embodiment, the step of predicting comprises predicting that the patient's cancer is unlikely to respond to chemotherapy based upon the absence of TLE3 expression in the cancer sample.


In certain embodiments, a negative control sample is provided and the step of determining comprises detecting a level of TLE3 expression in the cancer sample and the negative control sample and comparing the level of expression of TLE3 in the cancer sample and the negative control sample. In general, the negative control sample can be any sample that does not reproducibly express TLE3. In one embodiment, the negative control sample can be a sample that does not reproducibly bind TLE3 antibodies. In one embodiment, the negative control sample can be a sample that does not reproducibly produce a detectable level of TLE3 mRNA. In one embodiment, the negative control sample can be from a patient with a TLE3-negative cancer. In one embodiment, the negative control sample can be from a patient without cancer. In certain embodiments the negative control sample may originate from the same tissue type as the cancer in question (e.g., breast tissue when considering breast cancer). In other embodiments, the negative control sample may originate from a different tissue type or even a different organism, or a cell line.


Additionally or alternatively, in certain embodiments, a positive control sample is provided and the step of determining comprises detecting a level of TLE3 expression in the cancer sample and the positive control sample and comparing the level of expression of TLE3 in the cancer sample and the positive control sample. In general, the positive control sample can be any sample that reproducibly expresses TLE3. In one embodiment, the negative control sample can be a sample that reproducibly bind TLE3 antibodies. In one embodiment, the negative control sample can be a sample that reproducibly produces a detectable level of TLE3 mRNA. In one embodiment, the positive control sample can be from a patient with a TLE3-positive cancer. In certain embodiments the positive control sample may originate from the same tissue type as the cancer in question (e.g., breast tissue when considering breast cancer). In other embodiments, the positive control sample may originate from a different tissue type or even a different organism, or cell line.


Expression of TLE3 can be determined using any known method.


In one embodiment, TLE3 polypeptides may be detected using an interaction partner that binds a TLE3 polypeptide (e.g., TLE3 protein or an antigenic fragment thereof). For example, as described below one may use a TLE3 antibody as an interaction partner and detect TLE3 expression by contacting the cancer sample with the TLE3 antibody. In such embodiments, the inventive methods may involve providing a cancer sample from a cancer patient, contacting the cancer sample with an antibody directed to TLE3, and predicting the likelihood that the patient's cancer will respond to chemotherapy based upon binding of the antibody to the cancer sample. In one embodiment, the step of predicting may comprise predicting that the patient's cancer is likely to respond to chemotherapy based upon binding of the antibody to the cancer sample. In another embodiment, the step of predicting may comprise predicting that the patient's cancer is unlikely to respond to chemotherapy based upon lack of binding of the antibody to the cancer sample.


In another embodiment, TLE3 polynucleotides may be detected using one or more primers that hybridize with a TLE3 polynucleotide (e.g., a TLE3 mRNA, cDNA or RNA). In such embodiments, the inventive methods may involve providing a cancer sample from a cancer patient, contacting the cancer sample with one or more primers that hybridize with TLE3, and predicting the likelihood that the patient's cancer will respond to chemotherapy based upon hybridization of the one or more primers to the cancer sample. In one embodiment, the step of predicting may comprise predicting that the patient's cancer is likely to respond to chemotherapy based upon hybridization of the one or more primers to the cancer sample. In another embodiment, the step of predicting may comprise predicting that the patient's cancer is unlikely to respond to chemotherapy based upon lack of hybridization of the one or more primers to the cancer sample.


In another aspect, the present invention provides methods for deciding whether to administer chemotherapy to the cancer patient based upon the likelihood that the patient's cancer will respond to chemotherapy. In one embodiment, the step of deciding comprises deciding to administer chemotherapy to the cancer patient based upon the presence of TLE3 expression in the cancer sample. In one embodiment, the step of deciding comprises deciding not to administer chemotherapy to the cancer patient based upon the absence of TLE3 expression in the cancer sample.


In yet another aspect, the present invention provides methods for selecting a chemotherapy for a cancer patient. In general, these methods comprise providing a cancer sample from a cancer patient, determining whether TLE3 is expressed in the cancer sample, and selecting a chemotherapy for the cancer patient based upon the results of the step of determining. In one embodiment, the step of selecting comprises selecting a chemotherapy based upon the presence of TLE3 expression in the cancer sample.


As described in the Examples, we have demonstrated that TLE3 expression correlates with response to chemotherapy with methotrexate (see FIG. 7) and taxanes (see FIGS. 10, 12 and 13). Methotrexate and taxanes are thought to be cell cycle specific chemotherapeutics (e.g., see Goodman & Gilman's The Pharmacological Basis of Therapeutics, IX. Chemotherapy of Neoplastic Diseases Chapter 51. Antineoplastic Agents, 11 th Edition, Laurence L. Brunton, editor-in-chief, John S. Lazo and Keith L. Parker, Associate Editors). Cell cycle specific chemotherapeutics exhibit their mechanism of action within a specific phase of the cell cycle in contrast to non-cell cycle specific chemotherapeutics that work equally with all phases including the resting phase (GO). Other plant alkaloids besides the taxanes have also been classified in the literature as cell cycle specific chemotherapeutics as have other antimetabolites besides methotrexate. In contrast, many alkylating agents such as cisplatin and cyclophosamide have been classified as non-cell cycle specific chemotherapeutics. Our results suggest that the predictive power of TLE3 may extend to other cell cycle specific chemotherapeutics besides methotrexate and taxanes.


In some embodiments, the inventive methods may therefore be used to select, or decide whether to administer, a cell cycle specific chemotherapeutic. In one embodiment, the inventive methods may be used to select, or decide whether to administer, an antimetabolite. In one embodiment, the inventive methods may be used to select, or decide whether to administer, a plant alkaloid. In one embodiment, the inventive methods may be used to select, or decide whether to administer, methotrexate. In another embodiment, the inventive methods may be used to select, or decide whether to administer, a taxane. In one embodiment the taxane is paclitaxel. In one embodiment the taxane is docetaxel.


In each case it will be appreciated that these chemotherapeutics may be administered alone or in combination with other chemotherapeutics as is known in the art and discussed below. It will also be appreciated that the present invention encompasses methods in which the selected chemotherapeutic is a methotrexate or taxane derivative, i.e., a compound with a structure which is derived from methotrexate or a taxane. Derivatives will typically share most of the structure of the parent compound but may include different substituents, heteroatoms, ring fusions, levels of saturation, isomerism, stereoisomerism, etc. at one or more positions within the parent compound. Without limitation, the following U.S. Patents describe the preparation of exemplary methotrexate derivatives that could be employed according to an inventive method: U.S. Pat. Nos. 6,559,149 and 4,374,987. Without limitation, the following U.S. Patents describe the preparation of exemplary taxane derivatives that could be employed according to an inventive method: U.S. Pat. Nos. 7,074,945; 7,063,977; 6,906,101; 6,649,778; 6,596,880; 6,552,205; 6,531,611; 6,482,963; 6,482,850; 6,462,208; 6,455,575; 6,441,026; 6,433,180; 6,392,063; 6,369,244; 6,339,164; 6,291,690; 6,268,381; 6,239,167; 6,218,553; 6,214,863; 6,201,140; 6,191,290; 6,187,916; 6,162,920; 6,147,234; 6,136,808; 6,114,550; 6,107,332; 6,051,600; 6,025,385; 6,011,056; 5,955,489; 5,939,567; 5,912,263; 5,908,835; 5,869,680; 5,861,515; 5,821,263; 5,763,477; 5,750,561; 5,728,687; 5,726,346; 5,726,318; 5,721,268; 5,719,177; 5,714,513; 5,714,512; 5,703,117; 5,698,582; 5,686,623; 5,677,462; 5,646,176; 5,637,723; 5,621,121; 5,616,739; 5,606,083; 5,580,899; 5,476,954; 5,403,858; 5,380,916; 5,254,703; and 5,250,722. The entire contents of each of the aforementioned patents and any other reference which is cited herein is hereby incorporated by reference.


Methotrexate acts by inhibiting the metabolism of folic acid and has been approved for the treatment of bladder cancer, breast cancer, gastric cancer, choriocarcinoma, head and neck cancer, leptomeningeal cancer, leukemia (acute meningeal, acute lymphoblastic, acute lymphocytic), lymphoma (Burkitt's, childhood, non-Hodgkin's), mycosis fungoides, primary unknown cancer and lymphatic sarcoma (Methotrexate in BC Cancer Agency Cancer Drug Manual, 2007). Methotrexate has also been shown to be useful for treating esophageal cancer, lung cancer and testicular cancer (Methotrexate in UpToDate, 2007). In certain embodiments, the inventive methods comprise a step of selecting, or deciding whether to administer, methotrexate in combination with one or more additional chemotherapeutics. For example, methotrexate is commonly administered to cancer patients as a combination called CMF which also includes cyclophosphamide and 5-fluorouracil.


Taxanes are diterpenes produced by the plants of the genus Taxus. Taxanes can be obtained from natural sources or produced synthetically. Taxanes include paclitaxel (TAXOL™) and docetaxel (TAXOTERE™). Taxanes work by interfering with normal microtubule growth during cell division. In certain embodiments, the inventive methods comprise a step of selecting, or deciding whether to administer, a taxane (e.g., paclitaxel or docetaxel) in combination with one or more additional chemotherapeutics. For example, taxanes are commonly administered to cancer patients in combination with cyclophosphamide and adriamycin (doxorubicin) and optionally 5-fluorouracil (i.e., with CA or CAF).


Paclitaxel has been approved for the treatment of breast cancer, Kaposi's sarcoma, lung cancer and ovarian cancer (Paclitaxel in BC Cancer Agency Cancer Drug Manual, 2007 and Mekhail and Markman, Expert Opin. Pharmacother. 3:755-66, 2002). Paclitaxel has also been shown to be useful in treating cervical cancer (pp. 1124-34 in AHFS 2005 Drug Information. Bethesda, Md.: American Society of Health-System Pharmacists, 2005), endometrial cancer (Paclitaxel in BC Cancer Agency Cancer Drug Manual, 2007), bladder cancer (Paclitaxel in UpToDate, 2007), head and neck cancer (Paclitaxel in UpToDate, 2007), leukemia (Paclitaxel in UpToDate, 2007) and malignant melanoma (Paclitaxel in UpToDate, 2007). Side effects of paclitaxel include hypersensitivity reactions such as flushing of the face, skin rash, or shortness of breath. Patients often receive medication to prevent hypersensitivity reactions before they take paclitaxel. Paclitaxel can also cause temporary damage to the bone marrow. Bone marrow damage can cause a person to be more susceptible to infection, anemia, and bruise or bleed easily. Other side effects may include joint or muscle pain in the arms or legs; diarrhea; nausea and vomiting; numbness, burning, or tingling in the hands or feet; and loss of hair.


Docetaxel has been approved for the treatment of breast cancer (Aapro, Seminars in Oncology 25(5 Suppl 12):7-11, 1998; Nabholtz et al., Journal of Clinical Oncology 17(5):1413-24, 1999; Sjostrom et al., European Journal of Cancer 35(8):1194-201, 1999; and Burstein et al., Journal of Clinical Oncology 18(6):1212-9, 2000), non-small cell lung cancer (Fossella et al., Journal of Clinical Oncology 18(12):2354-62, 2000 and Hainsworth et al., Cancer 89(2):328-33, 2000) and ovarian cancer (Kaye et al., European Journal of Cancer 33(13):2167-70, 1997). Docetaxel has also been shown to be useful in treating esothelioma (Vorobiof et al., Proc Am Soc Clin Oncol 19:578a, 2000), prostate cancer (Picus et al., Seminars in Oncology 26(5 Suppl 17):14-8, 1999 and Petrylak et al., Journal of Clinical Oncology 17(3):958-67, 1999), urothelial transitional cell cancer (Dimopoulos et al., Annals of Oncology 10(11):1385-8, 1999 and Pectasides et al., European Journal of Cancer 36(1):74-9, 2000), head and neck cancer (Docetaxel in USP DI, 2000 and Couteau et al., British Journal of Cancer 81(3):457-62, 1999) and small cell lung cancer (Smyth et al., European Journal of Cancer 30A(8):1058-60, 1994).


Our observation that improved response to chemotherapy is observed for both breast and ovarian cancer patients that are TLE3-positive suggests that the inventive methods may be useful across different cancer types. Our observation that TLE3 expression is associated with improved response to treatment with methotrexate and taxanes further suggest that the inventive methods may be applicable across cancers that respond to these chemotherapeutics. As discussed above, this includes without limitation breast cancer, ovarian cancer, lung cancer, bladder cancer, gastric cancer, head and neck cancer, and leukemia.


In one embodiment, the inventive methods may be used with a cancer patient that has breast cancer. In one embodiment, the inventive methods may be used with a cancer patient that has ovarian cancer. In one embodiment, the inventive methods may be used with a cancer patient that has lung cancer. In one embodiment, the inventive methods may be used with a cancer patient that has bladder cancer. In one embodiment, the inventive methods may be used with a cancer patient that has gastric cancer. In one embodiment, the inventive methods may be used with a cancer patient that has head and neck cancer. In one embodiment, the inventive methods may be used with a cancer patient that has leukemia.


As demonstrated in the Examples, in one embodiment, the correlation between TLE3 expression and response to chemotherapy was observed with breast cancer patients that are triple negative for the ER (estrogen receptor, Entrez GeneID 2099), PR (progesterone receptor, Entrez GeneID 5241) and HER-2 markers (v-erb-b2 erythroblastic leukemia viral oncogene homolog 2, Entrez GeneID 2064). In certain embodiments, the inventive methods may therefore be used with breast cancer patients that belong to this class.


As demonstrated in the Examples, the correlation between TLE3 expression and response to chemotherapy was found to also exist when treatment was administered in a neoadjuvant setting. Thus, in certain embodiments, the inventive methods may be used with patients receiving chemotherapy in a neoadjuvant setting. In other embodiments, the chemotherapy may be administered in an adjuvant setting.


As demonstrated in the Examples, the correlation between TLE3 expression and response to chemotherapy was also found to be independent of stage. Thus, in certain embodiments, the inventive methods may be used with patients with a stage II+ (i.e., stage II or greater) cancer. In certain embodiments, the inventive methods may be used with patients with a stage IIb+ or a stage III+ cancer.


Detecting TLE3 Expression

As mentioned above, expression of TLE3 can be determined using any known method. In one embodiment, TLE3 expression may be determined by detecting TLE3 polypeptide markers using interaction partners (e.g., antibodies). In another embodiment, TLE3 expression may be determined by detecting TLE3 polynucleotide markers using primers.


Detecting TLE3 Polypeptide Markers

TLE3 polypeptide markers may be detected using any interaction partner that binds a TLE3 polypeptide marker (which could be a TLE3 protein or an antigenic fragment thereof). Thus, any entity that binds detectably to the TLE3 marker may be utilized as an interaction partner in accordance with the present invention, so long as it binds the marker with an appropriate combination of affinity and specificity.


Particularly preferred interaction partners are antibodies, or fragments (e.g., F(ab) fragments, F(ab′)2 fragments, Fv fragments, or sFv fragments, etc.; see, for example, Inbar et al., Proc. Nat. Acad. Sci. USA 69:2659, 1972; Hochman et al., Biochem. 15:2706, 1976; and Ehrlich et al., Biochem. 19:4091, 1980; Huston et al., Proc. Nat. Acad. Sci. USA 85:5879, 1998; U.S. Pat. Nos. 5,091,513 and 5,132,405 to Huston et al.; and U.S. Pat. No. 4,946,778 to Ladner et al., each of which is incorporated herein by reference). In certain embodiments, interaction partners may be selected from libraries of mutant antibodies (or fragments thereof). For example, collections of antibodies that each include different point mutations may be screened for their association with a marker of interest. Yet further, chimeric antibodies may be used as interaction partners, e.g., “humanized” or “veneered” antibodies as described in greater detail below.


When antibodies are used as interaction partners, these may be prepared by any of a variety of techniques known to those of ordinary skill in the art (e.g., see Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988, see also the Examples). For example, antibodies can be produced by cell culture techniques, including the generation of monoclonal antibodies, or via transfection of antibody genes into suitable bacterial or mammalian cell hosts, in order to allow for the production of recombinant antibodies. In one technique, an “immunogen” comprising an antigenic portion of a marker of interest (or the marker itself) is initially injected into any of a wide variety of mammals (e.g., mice, rats, rabbits, sheep or goats). In this step, a marker (or an antigenic portion thereof) may serve as the immunogen without modification. Alternatively, particularly for relatively short markers, a superior immune response may be elicited if the marker is joined to a carrier protein, such as bovine serum albumin or keyhole limpet hemocyanin (KLH). The immunogen is injected into the animal host, preferably according to a predetermined schedule incorporating one or more booster immunizations and the animals are bled periodically. Polyclonal antibodies specific for the marker may then be purified from such antisera by, for example, affinity chromatography using the marker (or an antigenic portion thereof) coupled to a suitable solid support. An exemplary method is described in the Examples.


If desired for diagnostic or therapeutic purposes, monoclonal antibodies specific for TLE3 may be prepared, for example, using the technique of Kohler and Milstein, Eur. J. Immunol. 6:511, 1976 and improvements thereto. Briefly, these methods involve the preparation of immortal cell lines capable of producing antibodies having the desired specificity (i.e., reactivity with the marker of interest). Such cell lines may be produced, for example, from spleen cells obtained from an animal immunized as described above. The spleen cells are then immortalized by, for example, fusion with a myeloma cell fusion partner, preferably one that is syngeneic with the immunized animal. A variety of fusion techniques may be employed. For example, the spleen cells and myeloma cells may be combined with a nonionic detergent for a few minutes and then plated at low density on a selective medium that supports the growth of hybrid cells, but not myeloma cells. A preferred selection technique uses HAT (hypoxanthine, aminopterin, thymidine) selection. After a sufficient time, usually about 1 to 2 weeks, colonies of hybrids are observed. Single colonies are selected and their culture supernatants tested for binding activity against the marker. Hybridomas having high reactivity and specificity are preferred.


Monoclonal antibodies may be isolated from the supernatants of growing hybridoma colonies. In addition, various techniques may be employed to enhance the yield, such as injection of the hybridoma cell line into the peritoneal cavity of a suitable vertebrate host, such as a mouse. Monoclonal antibodies may then be harvested from the ascites fluid or the blood. Contaminants may be removed from the antibodies by conventional techniques, such as chromatography, gel filtration, precipitation and extraction. TLE3 may be used in the purification process in, for example, an affinity chromatography step.


It is to be understood that the present invention is not limited to using antibodies or antibody fragments as interaction partners. In particular, the present invention also encompasses the use of synthetic interaction partners that mimic the functions of antibodies. Several approaches to designing and/or identifying antibody mimics have been proposed and demonstrated (e.g., see the reviews by Hsieh-Wilson et al., Acc. Chem. Res. 29:164, 2000 and Peczuh and Hamilton, Chem. Rev. 100:2479, 2000). For example, small molecules that bind protein surfaces in a fashion similar to that of natural proteins have been identified by screening synthetic libraries of small molecules or natural product isolates (e.g., see Gallop et al., J. Med. Chem. 37:1233, 1994; Gordon et al., J. Med. Chem. 37:1385, 1994; DeWitt et al., Proc. Natl. Acad. Sci. U.S.A. 90:6909, 1993; Bunin et al., Proc. Natl. Acad. Sci. U.S.A. 91:4708, 1994; Virgilio and Ellman, J. Am. Chem. Soc. 116:11580, 1994; Wang et al., J. Med. Chem. 38:2995, 1995; and Kick and Ellman, J. Med. Chem. 38:1427, 1995). Similarly, combinatorial approaches have been successfully applied to screen libraries of peptides and proteins for their ability to bind a range of proteins (e.g., see Cull et al., Proc. Natl. Acad. Sci. U.S.A. 89:1865, 1992; Mattheakis et al., Proc. Natl. Acad. Sci. U.S.A. 91:9022, 1994; Scott and Smith, Science 249:386, 1990; Devlin et al., Science 249:404, 1990; Corey et al., Gene 128:129, 1993; Bray et al., Tetrahedron Lett. 31:5811, 1990; Fodor et al., Science 251:767, 1991; Houghten et al., Nature 354:84, 1991; Lam et al., Nature 354:82, 1991; Blake and Litzi-Davis, Bioconjugate Chem. 3:510, 1992; Needels et al., Proc. Natl. Acad. Sci. U.S.A. 90:10700, 1993; and Ohlmeyer et al., Proc. Natl. Acad. Sci. U.S.A. 90:10922, 1993). Similar approaches have also been used to study carbohydrate-protein interactions (e.g., see Oldenburg et al., Proc. Natl. Acad. Sci. U.S.A. 89:5393, 1992) and polynucleotide-protein interactions (e.g., see Ellington and Szostak, Nature 346:818, 1990 and Tuerk and Gold, Science 249:505, 1990). These approaches have also been extended to study interactions between proteins and unnatural biopolymers such as oligocarbamates, oligoureas, oligosulfones, etc. (e.g., see Zuckermann et al., J. Am. Chem. Soc. 114:10646, 1992; Simon et al., Proc. Natl. Acad. Sci. U.S.A. 89:9367, 1992; Zuckermann et al., J. Med. Chem. 37:2678, 1994; Burgess et al., Angew. Chem., Int. Ed. Engl. 34:907, 1995; and Cho et al., Science 261:1303, 1993). Yet further, alternative protein scaffolds that are loosely based around the basic fold of antibody molecules have been suggested and may be used in the preparation of inventive interaction partners (e.g., see Ku and Schultz Proc. Natl. Acad. Sci. U.S.A. 92:6552, 1995). Antibody mimics comprising a scaffold of a small molecule such as 3-aminomethylbenzoic acid and a substituent consisting of a single peptide loop have also been constructed. The peptide loop performs the binding function in these mimics (e.g., see Smythe et al., J. Am. Chem. Soc. 116:2725, 1994). A synthetic antibody mimic comprising multiple peptide loops built around a calixarene unit has also been described (e.g., see U.S. Pat. No. 5,770,380 to Hamilton et al.).


Any available strategy or system may be utilized to detect association between an interaction partner and the TLE3 marker. In certain embodiments, association can be detected by adding a detectable label to the interaction partner. In other embodiments, association can be detected by using a labeled secondary interaction partner that binds specifically with the primary interaction partner, e.g., as is well known in the art of antigen/antibody detection. The detectable label may be directly detectable or indirectly detectable, e.g., through combined action with one or more additional members of a signal producing system. Examples of directly detectable labels include radioactive, paramagnetic, fluorescent, light scattering, absorptive and colorimetric labels. Examples of indirectly detectable include chemiluminescent labels, e.g., enzymes that are capable of converting a substrate to a chromogenic product such as alkaline phosphatase, horseradish peroxidase and the like.


Once a labeled interaction partner has bound the TLE3 marker, the complex may be visualized or detected in a variety of ways, with the particular manner of detection being chosen based on the particular detectable label, where representative detection means include, e.g., scintillation counting, autoradiography, measurement of paramagnetism, fluorescence measurement, light absorption measurement, measurement of light scattering and the like.


In general, association between an interaction partner and the TLE3 marker may be assayed by contacting the interaction partner with a cancer sample that includes the marker. Depending upon the nature of the sample, appropriate methods include, but are not limited to, immunohistochemistry (IHC), radioimmunoassay, ELISA, immunoblotting and fluorescence activates cell sorting (FACS). In the case where the protein is to be detected in a tissue sample, e.g., a biopsy sample, IHC is a particularly appropriate detection method. Techniques for obtaining tissue and cell samples and performing IHC and FACS are well known in the art.


Where large numbers of samples are to be handled (e.g., when simultaneously analyzing several samples from the same patient or samples from different patients), it may be desirable to utilize arrayed and/or automated formats. In certain embodiments, tissue arrays as described in the Examples may be used. Tissue arrays may be constructed according to a variety of techniques. According to one procedure, a commercially-available mechanical device (e.g., the manual tissue arrayer MTA1 from Beecher Instruments of Sun Prairie, Wis.) is used to remove an 0.6-micron-diameter, full thickness “core” from a paraffin block (the donor block) prepared from each patient, and to insert the core into a separate paraffin block (the recipient block) in a designated location on a grid. In preferred embodiments, cores from as many as about 400 patients (or multiple cores from the same patient) can be inserted into a single recipient block; preferably, core-to-core spacing is approximately 1 mm. The resulting tissue array may be processed into thin sections for staining with interaction partners according to standard methods applicable to paraffin embedded material.


Whatever the format, and whatever the detection strategy, identification of a discriminating titer can simplify binding studies to assess the desirability of using an interaction partner. In such studies, the interaction partner is contacted with a plurality of different samples that preferably have at least one common trait (e.g., tissue of origin), and often have multiple common traits (e.g., tissue of origin, stage, microscopic characteristics, etc.). In some cases, it will be desirable to select a group of samples with at least one common trait and at least one different trait, so that a titer is determined that distinguishes the different trait. In other cases, it will be desirable to select a group of samples with no detectable different traits, so that a titer is determined that distinguishes among previously indistinguishable samples. Those of ordinary skill in the art will understand, however, that the present invention often will allow both of these goals to be accomplished even in studies of sample collections with varying degrees of similarity and difference.


As discussed above and in the Examples, the inventors have applied these techniques to samples from breast and ovarian cancer patients. The invention also encompasses the recognition that markers that are secreted from the cells in which they are produced may be present in serum, enabling their detection through a blood test rather than requiring a biopsy specimen. An interaction partner that binds to such markers represents a particularly preferred embodiment of the invention.


In general, the results of such an assay can be presented in any of a variety of formats. The results can be presented in a qualitative fashion. For example, the test report may indicate only whether or not the TLE3 marker was detected, perhaps also with an indication of the limits of detection. Additionally the test report may indicate the subcellular location of binding, e.g., nuclear versus cytoplasmic and/or the relative levels of binding in these different subcellular locations. The results may be presented in a semi-quantitative fashion. For example, various ranges may be defined and the ranges may be assigned a score (e.g., 0 to 5) that provides a certain degree of quantitative information. Such a score may reflect various factors, e.g., the number of cells in which the marker is detected, the intensity of the signal (which may indicate the level of expression of the marker), etc. The results may be presented in a quantitative fashion, e.g., as a percentage of cells in which the marker is detected, as a concentration, etc. As will be appreciated by one of ordinary skill in the art, the type of output provided by a test will vary depending upon the technical limitations of the test and the biological significance associated with detection of the marker. For example, in certain circumstances a purely qualitative output (e.g., whether or not the marker is detected at a certain detection level) provides significant information. In other cases a more quantitative output (e.g., a ratio of the level of expression of the marker in two samples) is necessary.


Detecting TLE3 Polynucleotide Markers

Although in many cases detection of polypeptide markers using interaction partners such as antibodies may represent the most convenient means of determining whether TLE3 is expressed in a particular sample, the inventive methods also encompass the use of primers for the detection of polynucleotide markers. A variety of methods for detecting the presence of a particular polynucleotide marker are known in the art and may be used in the inventive methods. In general, these methods rely on hybridization between one or more primers and the polynucleotide marker.


Any available strategy or system may be utilized to detect hybridization between primers and the TLE3 polynucleotides (which could be a TLE3 mRNA, a cDNA produced by RT-PCR from mRNA, RNA produced from such cDNA, etc.). In certain embodiments, hybridization can be detected by simply adding a detectable label to the primer. In other embodiments, hybridization can be detected by using a labeled secondary primer that hybridizes specifically with the primary primer (e.g., a region of the primary primer that does not hybridize with the TLE3 marker). In yet other embodiments it may be advantageous to amplify the TLE3 marker within the cancer sample by PCR using a set of primers designed to amplify a region of the TLE3 gene. The resulting product can then be detected, e.g., using a labeled secondary primer that hybridizes with the amplified product. Those skilled in the art will appreciate variations on these embodiments.


Considerations for primer design are well known in the art and are described, for example, in Newton, et al. (eds.) PCR: Essential data Series, John Wiley & Sons; PCR Primer: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1995; White, et al. (eds.) PCR Protocols: Current methods and Applications, Methods in Molecular Biology, The Humana Press, Totowa, N.J., 1993. In addition, a variety of computer programs known in the art may be used to select appropriate primers.


In general, a detectable label may be directly detectable or indirectly detectable, e.g., through combined action with one or more additional members of a signal producing system. Examples of directly detectable labels include radioactive, paramagnetic, fluorescent, light scattering, absorptive and colorimetric labels. Examples of indirectly detectable include chemiluminescent labels, e.g., enzymes that are capable of converting a substrate to a chromogenic product such as alkaline phosphatase, horseradish peroxidase and the like.


Once a labeled primer has hybridized with the TLE3 marker, the complex may be visualized or detected in a variety of ways, with the particular manner of detection being chosen based on the particular detectable label, where representative detection means include, e.g., scintillation counting, autoradiography, measurement of paramagnetism, fluorescence measurement, light absorption measurement, measurement of light scattering and the like.


In general, hybridization between a primer and the TLE3 marker may be assayed by contacting the primer with a cancer sample that includes the marker. Depending upon the nature of the cancer sample, appropriate methods include, but are not limited to, microarray analysis, in situ hybridization, Northern blot, and various nucleic acid amplification techniques such as PCR, RT-PCR, quantitative PCR, the ligase chain reaction, etc.


Identification of Novel Therapies

The predictive power of TLE3 is useful according to the present invention not only to classify cancers with respect to their likely responsiveness to known therapies, but also to identify potential new therapies or therapeutic agents that could be useful in the treatment of cancer.


Indeed, TLE3 represents an attractive candidate for identification of new therapeutic agents (e.g., via screens to detect compounds or entities that bind or hybridize to the marker, preferably with at least a specified affinity and/or specificity, and/or via screens to detect compounds or entities that modulate (i.e., increase or decrease) expression, localization, modification, or activity of the marker. Thus, in one embodiment the present invention provides methods comprising steps of contacting a test compound with a cell expressing the TLE3 marker (e.g., individual engineered cells or in the context of a tissue, etc.); and determining whether the test compound modulates the expression, localization, modification, or activity of the TLE3 marker. In many instances, interaction partners or primers (e.g., antisense or RNAi primers) themselves may prove to be useful therapeutics.


Thus the present invention provides interaction partners and primers that are themselves useful therapeutic agents. For example, binding by an antibody raised against TLE3 to cancerous cells might inhibit growth of those cells. Alternatively or additionally, interaction partners defined or prepared according to the present invention could be used to deliver a therapeutic agent to a cancer cell. In particular, interaction partners (e.g., an antibody raised against TLE3) may be coupled to one or more therapeutic agents. Suitable agents in this regard include radionuclides and drugs. Preferred radionuclides include 90Y, 123I, 125I, 131I, 186Re, 188Re, 211At and 212Bi. Preferred drugs include chlorambucil, ifosphamide, meclorethamine, cyclophosphamide, carboplatin, cisplatin, procarbazine, decarbazine, carmustine, cytarabine, hydroxyurea, mercaptopurine, methotrexate, paclitaxel, docetaxel, thioguanine, 5-fluorouracil, actinomycin D, bleomycin, daunorubicin, doxorubicin, etoposide, vinblastine, vincristine, L-asparginase, adrenocorticosteroids, canciclovir triphosphate, adenine arabinonucleoside triphosphate, 5-aziridinyl-4-hydroxylamino-2-nitrobenzamide, acrolein, phosphoramide mustard, 6-methylpurine, etoposide, benzoic acid mustard, cyanide and nitrogen mustard.


According to such embodiments, the therapeutic agent may be coupled with an interaction partner by direct or indirect covalent or non-covalent interactions. A direct interaction between a therapeutic agent and an interaction partner is possible when each possesses a substituent capable of reacting with the other. For example, a nucleophilic group, such as an amino or sulfhydryl group, on one may be capable of reacting with a carbonyl-containing group, such as an anhydride or an acid halide, or with an alkyl group containing a good leaving group (e.g., a halide) on the other. Indirect interactions might involve a linker group that is itself non-covalently bound to both the therapeutic agent and the interaction partner. A linker group can function as a spacer to distance an interaction partner from an agent in order to avoid interference with association capabilities. A linker group can also serve to increase the chemical reactivity of a substituent on an agent or an interaction partner and thus increase the coupling efficiency. An increase in chemical reactivity may also facilitate the use of agents, or functional groups on agents, which otherwise would not be possible.


It will be evident to those skilled in the art that a variety of bifunctional or polyfunctional reagents, both homo- and hetero-functional (such as those described in the catalog of the Pierce Chemical Co., Rockford, Ill.), may be employed as the linker group. Coupling may be effected, for example, through amino groups, carboxyl groups, sulfydryl groups or oxidized carbohydrate residues. There are numerous references describing such methodology, e.g., U.S. Pat. No. 4,671,958, to Rodwell et al. It will further be appreciated that a therapeutic agent and an interaction partner may be coupled via non-covalent interactions, e.g., ligand/receptor type interactions. Any ligand/receptor pair with a sufficient stability and specificity to operate in the context of the invention may be employed to couple a therapeutic agent and an interaction partner. To give but an example, a therapeutic agent may be covalently linked with biotin and an interaction partner with avidin. The strong non-covalent binding of biotin to avidin would then allow for coupling of the therapeutic agent and the interaction partner. Typical ligand/receptor pairs include protein/co-factor and enzyme/substrate pairs. Besides the commonly used biotin/avidin pair, these include without limitation, biotin/streptavidin, digoxigenin/anti-digoxigenin, FK506/FK506-binding protein (FKBP), rapamycin/FKBP, cyclophilin/cyclosporin and glutathione/glutathione transferase pairs. Other suitable ligand/receptor pairs would be recognized by those skilled in the art, e.g., monoclonal antibodies paired with a epitope tag such as, without limitation, glutathione-S-transferase (GST), c-myc, FLAG® and maltose binding protein (MBP) and further those described in Kessler pp. 105-152 of Advances in Mutagenesis” Ed. by Kessler, Springer-Verlag, 1990; “Affinity Chromatography: Methods and Protocols (Methods in Molecular Biology)” Ed. by Pascal Baillon, Humana Press, 2000; and “Immobilized Affinity Ligand Techniques” by Hermanson et al., Academic Press, 1992.


Where a therapeutic agent is more potent when free from the interaction partner, it may be desirable to use a linker group which is cleavable during or upon internalization into a cell. A number of different cleavable linker groups have been described. The mechanisms for the intracellular release of an agent from these linker groups include cleavage by reduction of a disulfide bond (e.g., U.S. Pat. No. 4,489,710 to Spitler), by irradiation of a photolabile bond (e.g., U.S. Pat. No. 4,625,014 to Senter et al.), by hydrolysis of derivatized amino acid side chains (e.g., U.S. Pat. No. 4,638,045 to Kohn et al.), by serum complement-mediated hydrolysis (e.g., U.S. Pat. No. 4,671,958 to Rodwell et al.) and by acid-catalyzed hydrolysis (e.g., U.S. Pat. No. 4,569,789 to Blattler et al.).


In certain embodiments, it may be desirable to couple more than one therapeutic agent to an interaction partner. In one embodiment, multiple molecules of an agent are coupled to one interaction partner molecule. In another embodiment, more than one type of therapeutic agent may be coupled to one interaction partner molecule. Regardless of the particular embodiment, preparations with more than one agent may be prepared in a variety of ways. For example, more than one agent may be coupled directly to an interaction partner molecule, or linkers that provide multiple sites for attachment can be used.


Alternatively, a carrier can be used. A carrier may bear the agents in a variety of ways, including covalent bonding either directly or via a linker group. Suitable carriers include proteins such as albumins (e.g., U.S. Pat. No. 4,507,234 to Kato et al.), peptides, and polysaccharides such as aminodextran (e.g., U.S. Pat. No. 4,699,784 to Shih et al.). A carrier may also bear an agent by non-covalent bonding or by encapsulation, such as within a liposome vesicle (e.g., U.S. Pat. No. 4,429,008 to Martin et al. and 4,873,088 to Mayhew et al.). Carriers specific for radionuclide agents include radiohalogenated small molecules and chelating compounds. For example, U.S. Pat. No. 4,735,792 to Srivastava discloses representative radiohalogenated small molecules and their synthesis. A radionuclide chelate may be formed from chelating compounds that include those containing nitrogen and sulfur atoms as the donor atoms for binding the metal, or metal oxide, radionuclide. For example, U.S. Pat. No. 4,673,562 to Davison et al. discloses representative chelating compounds and their synthesis.


When interaction partners are themselves therapeutics, it will be understood that, in many cases, any interaction partner that binds the same marker may be so used.


In one preferred embodiment of the invention, the therapeutic agents (whether interaction partners or otherwise) are antibodies, e.g., an antibody against the TLE3 marker. As is well known in the art, when using an antibody or fragment thereof for therapeutic purposes it may prove advantageous to use a “humanized” or “veneered” version of an antibody of interest to reduce any potential immunogenic reaction. In general, “humanized” or “veneered” antibody molecules and fragments thereof minimize unwanted immunological responses toward antihuman antibody molecules which can limit the duration and effectiveness of therapeutic applications of those moieties in human recipients.


A number of “humanized” antibody molecules comprising an antigen binding portion derived from a non-human immunoglobulin have been described in the art, including chimeric antibodies having rodent variable regions and their associated complementarity-determining regions (CDRs) fused to human constant domains (e.g., see Winter et al., Nature 349:293, 1991; Lobuglio et al., Proc. Nat. Acad. Sci. USA 86:4220, 1989; Shaw et al., J. Immunol. 138:4534, 1987; and Brown et al., Cancer Res. 47:3577, 1987), rodent CDRs grafted into a human supporting framework region (FR) prior to fusion with an appropriate human antibody constant domain (e.g., see Riechmann et al., Nature 332:323, 1988; Verhoeyen et al., Science 239:1534, 1988; and Jones et al. Nature 321:522, 1986) and rodent CDRs supported by recombinantly veneered rodent FRs (e.g., see European Patent Publication No. 519,596, published Dec. 23, 1992). It is to be understood that the invention also encompasses “fully human” antibodies produced using the XenoMouse™ technology (AbGenix Corp., Fremont, Calif.) according to the techniques described in U.S. Pat. No. 6,075,181.


Yet further, so-called “veneered” antibodies may be used that include “veneered FRs”. The process of veneering involves selectively replacing FR residues from, e.g., a murine heavy or light chain variable region, with human FR residues in order to provide a xenogeneic molecule comprising an antigen binding portion which retains substantially all of the native FR protein folding structure. Veneering techniques are based on the understanding that the antigen binding characteristics of an antigen binding portion are determined primarily by the structure and relative disposition of the heavy and light chain CDR sets within the antigen-association surface (e.g., see Davies et al., Ann. Rev. Biochem. 59:439, 1990). Thus, antigen association specificity can be preserved in a humanized antibody only wherein the CDR structures, their interaction with each other and their interaction with the rest of the variable region domains are carefully maintained. By using veneering techniques, exterior (e.g., solvent-accessible) FR residues which are readily encountered by the immune system are selectively replaced with human residues to provide a hybrid molecule that comprises either a weakly immunogenic, or substantially non-immunogenic veneered surface.


Preferably, interaction partners suitable for use as therapeutics (or therapeutic agent carriers) exhibit high specificity for the target marker (e.g., TLE3) and low background binding to other markers. In certain embodiments, monoclonal antibodies are preferred for therapeutic purposes.


Pharmaceutical Compositions

As mentioned above, the present invention provides new therapies and methods for identifying these. In certain embodiments, an interaction partner or primer may be a useful therapeutic agent. Alternatively or additionally, interaction partners defined or prepared according to the present invention bind to markers (e.g., TLE3) that serve as targets for therapeutic agents. Also, inventive interaction partners may be used to deliver a therapeutic agent to a cancer cell. For example, interaction partners provided in accordance with the present invention may be coupled to one or more therapeutic agents.


The invention includes pharmaceutical compositions comprising these inventive therapeutic agents. In general, a pharmaceutical composition will include a therapeutic agent in addition to one or more inactive agents such as a sterile, biocompatible carrier including, but not limited to, sterile water, saline, buffered saline, or dextrose solution. The pharmaceutical compositions may be administered either alone or in combination with other therapeutic agents including other chemotherapeutic agents, hormones, vaccines and/or radiation therapy. By “in combination with”, here and elsewhere in the specification, it is not intended to imply that the agents must be administered at the same time or formulated for delivery together, although these methods of delivery are within the scope of the invention. In general, each agent will be administered at a dose and on a time schedule determined for that agent. Additionally, the invention encompasses the delivery of the inventive pharmaceutical compositions in combination with agents that may improve their bioavailability, reduce or modify their metabolism, inhibit their excretion, or modify their distribution within the body. Although the pharmaceutical compositions of the present invention can be used for treatment of any subject (e.g., any animal) in need thereof, they are most preferably used in the treatment of humans.


The pharmaceutical compositions of this invention can be administered to humans and other animals by a variety of routes including oral, intravenous, intramuscular, intra-arterial, subcutaneous, intraventricular, transdermal, rectal, intravaginal, intraperitoneal, topical (as by powders, ointments, or drops), bucal, or as an oral or nasal spray or aerosol. In general the most appropriate route of administration will depend upon a variety of factors including the nature of the agent (e.g., its stability in the environment of the gastrointestinal tract), the condition of the patient (e.g., whether the patient is able to tolerate oral administration), etc. At present the intravenous route is most commonly used to deliver therapeutic antibodies. However, the invention encompasses the delivery of the inventive pharmaceutical composition by any appropriate route taking into consideration likely advances in the sciences of drug delivery.


General considerations in the formulation and manufacture of pharmaceutical agents may be found, for example, in Remington's Pharmaceutical Sciences, 19th ed., Mack Publishing Co., Easton, Pa., 1995.


EXEMPLIFICATION
Example 1
Raising Antibodies

This example describes a method that was employed to generate the TLE3 antibodies used in these Examples. Similar methods may be used to generate an antibody that binds to any marker of interest (e.g., to proteins that are or are derived from other markers listed in Appendix A). In some cases, antibodies may be obtained from commercial sources (e.g., Chemicon, Dako, Oncogene Research Products, NeoMarkers, etc.) or other publicly available sources (e.g., Imperial Cancer Research Technology, etc.).


Materials and Solutions





    • Anisole (Cat. No. A4405, Sigma, St. Louis, Mo.)

    • 2,2′-azino-di-(3-ethyl-benzthiazoline-sulfonic acid) (ABTS) (Cat. No. A6499, Molecular Probes, Eugene, Oreg.)

    • Activated maleimide Keyhole Limpet Hemocyanin (Cat. No. 77106, Pierce, Rockford, Ill.)

    • Keyhole Limpet Hemocyanin (Cat. No. 77600, Pierce, Rockford, Ill.)

    • Phosphoric Acid (H3PO4) (Cat. No. P6560, Sigma)

    • Glacial Acetic Acid (Cat No. BP1185-500, Fisher)

    • EDC (EDAC) (Cat No. 341006, Calbiochem)

    • 25% Glutaraldehyde (Cat No. G-5882, Sigma)

    • Glycine (Cat No. G-8898, Sigma)

    • Biotin (Cat. No. B2643, Sigma)

    • Boric acid (Cat. No. B0252, Sigma)

    • Sepharose 4B (Cat. No. 17-0120-01, LKB/Pharmacia, Uppsala, Sweden)

    • Bovine Serum Albumin (LP) (Cat. No. 100 350, Boehringer Mannheim, Indianapolis, Ind.)

    • Cyanogen bromide (Cat. No. C6388, Sigma)

    • Dialysis tubing Spectra/Por Membrane MWCO: 6-8,000 (Cat. No. 132 665, Spectrum Industries, Laguna Hills, Calif.)

    • Dimethyl formamide (DMF) (Cat. No. 22705-6, Aldrich, Milwaukee, Wis.)

    • DIC (Cat. No. BP 592-500, Fisher)

    • Ethanedithiol (Cat. No. 39,802-0, Aldrich)

    • Ether (Cat. No. TX 1275-3, EM Sciences)

    • Ethylenediaminetetraacetatic acid (EDTA) (Cat. No. BP 120-1, Fisher, Springfield, N.J.)

    • 1-ethyl-3-(3′dimethylaminopropyl)-carbodiimide, HCL (EDC) (Cat. no. 341-006, Calbiochem, San Diego, Calif.)

    • Freund's Adjuvant, complete (Cat. No. M-0638-50B, Lee Laboratories, Grayson, Ga.)

    • Freund's Adjuvant, incomplete (Cat. No. M-0639-50B, Lee Laboratories)

    • Fritted chromatography columns (Column part No. 12131011; Frit Part No. 12131029, Varian Sample Preparation Products, Harbor City, Calif.)

    • Gelatin from Bovine Skin (Cat. No. G9382, Sigma)

    • Goat anti-rabbit IgG, biotinylated (Cat. No. A 0418, Sigma)

    • HOBt (Cat. No. 01-62-0008, Calbiochem)

    • Horseradish peroxidase (HRP) (Cat. No. 814 393, Boehringer Mannheim)

    • HRP-Streptavidin (Cat. No. S 5512, Sigma)

    • Hydrochloric Acid (Cat. No. 71445-500, Fisher)

    • Hydrogen Peroxide 30% w/w (Cat. No. H1009, Sigma)

    • Methanol (Cat. No. A412-20, Fisher)

    • Microtiter plates, 96 well (Cat. No. 2595, Corning-Costar, Pleasanton, Calif.)

    • N- -Fmoc protected amino acids from Calbiochem. See '97-'98 Catalog pp. 1-45.

    • N- -Fmoc protected amino acids attached to Wang Resin from Calbiochem. See '97-'98 Catalog pp. 161-164.

    • NMP (Cat. No. CAS 872-50-4, Burdick and Jackson, Muskegon, Mich.)

    • Peptide (Synthesized by Research Genetics. Details given below)

    • Piperidine (Cat. No. 80640, Fluka, available through Sigma)

    • Sodium Bicarbonate (Cat. No. BP328-1, Fisher)

    • Sodium Borate (Cat. No. B9876, Sigma)

    • Sodium Carbonate (Cat. No. BP357-1, Fisher)

    • Sodium Chloride (Cat. No. BP 358-10, Fisher)

    • Sodium Hydroxide (Cat. No. SS 255-1, Fisher)

    • Streptavidin (Cat. No. 1 520, Boehringer Mannheim)

    • Thioanisole (Cat. No. T-2765, Sigma)

    • Trifluoroacetic acid (Cat. No. TX 1275-3, EM Sciences)

    • Tween-20 (Cat. No. BP 337-500, Fisher)

    • Wetbox (Rectangular Servin' Saver™ Part No. 3862, Rubbermaid, Wooster, Ohio)

    • BBS—Borate Buffered Saline with EDTA dissolved in distilled water (pH 8.2 to 8.4 with HCl or NaOH), 25 mM Sodium borate (Borax), 100 mM Boric Acid, 75 mM NaCl and 5 mM EDTA.

    • 0.1 N HCl in saline as follows: concentrated HCl (8.3 ml/0.917 liter distilled water) and 0.154 M NaCl

    • Glycine (pH 2.0 and pH 3.0) dissolved in distilled water and adjusted to the desired pH, 0.1 M glycine and 0.154 M NaCl.

    • 5× Borate 1× Sodium Chloride dissolved in distilled water, 0.11 M NaCl, 60 mM Sodium Borate and 250 mM Boric Acid.

    • Substrate Buffer in distilled water adjusted to pH 4.0 with sodium hydroxide, 50 to 100 mM Citric Acid.

    • AA solution: HOBt is dissolved in NMP (8.8 grams HOBt to 1 liter NMP). Fmoc-N-a-amino at a concentration at 0.53 M.

    • DIC solution: 1 part DIC to 3 parts NMP.

    • Deprotecting solution: 1 part Piperidine to 3 parts DMF.

    • Reagent R: 2 parts anisole, 3 parts ethanedithiol, 5 parts thioanisole and 90 parts trifluoroacetic acid.





Equipment





    • MRX Plate Reader (Dynatech, Chantilly, Va.)

    • Hamilton Eclipse (Hamilton Instruments, Reno, Nev.)

    • Beckman TJ-6 Centrifuge (Model No. TJ-6, Beckman Instruments, Fullerton, Calif.)

    • Chart Recorder (Recorder 1 Part No. 18-1001-40, Pharmacia LKB Biotechnology)

    • UV Monitor (Uvicord SII Part No. 18-1004-50, Pharmacia LKB Biotechnology)

    • Amicon Stirred Cell Concentrator (Model 8400, Amicon, Beverly, Mass.)

    • 30 kD MW cut-off filter (Cat. No. YM-30 Membranes Cat. No. 13742, Amicon)

    • Multi-channel Automated Pipettor (Cat. No. 4880, Corning Costar, Cambridge, Mass.)

    • pH Meter Corning 240 (Corning Science Products, Corning Glassworks, Corning, N.Y.)

    • ACT396 peptide synthesizer (Advanced ChemTech, Louisville, Ky.)

    • Vacuum dryer (Box from Labconco, Kansas City, Mo. and Pump from Alcatel, Laurel, Md.).

    • Lyophilizer (Unitop 600 sl in tandem with Freezemobile 12, both from Virtis, Gardiner, N.Y.)





Peptide Selection

Peptide or peptides against which antibodies would be raised were selected from within the protein sequence of interest using a program that uses the Hopp/Woods method (described in Hopp and Woods, Mol. Immunol. 20:483, 1983 and Hopp and Woods, Proc. Nat. Acad. Sci. U.S.A. 78:3824, 1981). The program uses a scanning window that identifies peptide sequences of 15-20 amino acids containing several putative antigenic epitopes as predicted by low solvent accessibility. This is in contrast to most implementations of the Hopp/Woods method, which identify single short (˜6 amino acids) presumptive antigenic epitopes. Occasionally the predicted solvent accessibility was further assessed by PHD prediction of loop structures (described in Rost and Sander, Proteins 20:216, 1994). Preferred peptide sequences display minimal similarity with additional known human proteins. Similarity was determined by performing BLASTP alignments, using a wordsize of 2 (described in Altschul et al., J. Mol. Biol. 215:403, 1990). All alignments given an EXPECT value less than 1000 were examined and alignments with similarities of greater than 60% or more than four residues in an exact contiguous non-gapped alignment forced those peptides to be rejected. When it was desired to target regions of proteins exposed outside the cell membrane, extracellular regions of the protein of interest were determined from the literature or as defined by predicted transmembrane domains using a hidden Markov model (described in Krogh et al., J. Mol. Biol. 305:567, 2001). When the peptide sequence was in an extracellular domain, peptides were rejected if they contained N-linked glycosylation sites. As shown in Appendix A, for the preparation of TLE3 antibodies a single peptide was used having the amino acid sequence KNHHELDHRERESSAN (SEQ ID NO. 383). Appendix A provides one to three peptide sequences that can be used in preparing antibodies against other markers.


Peptide Synthesis

The sequence of the desired peptide was provided to the peptide synthesizer. The C-terminal residue was determined and the appropriate Wang Resin was attached to the reaction vessel. The peptide or peptides were synthesized C-terminus to N-terminus by adding one amino acid at a time using a synthesis cycle. Which amino acid is added was controlled by the peptide synthesizer, which looks to the sequence of the peptide that was entered into its database. The synthesis steps were performed as follows:

    • Step 1—Resin Swelling: Added 2 ml DMF, incubated 30 minutes, drained DMF.
    • Step 2—Synthesis cycle (repeated over the length of the peptide)
      • 2a—Deprotection: 1 ml deprotecting solution was added to the reaction vessel and incubated for 20 minutes.
      • 2b—Wash Cycle
      • 2c—Coupling: 750 ml of amino acid solution (changed as the sequence listed in the peptide synthesizer dictated) and 250 ml of DIC solution were added to the reaction vessel. The reaction vessel was incubated for thirty minutes and washed once. The coupling step was repeated once.
      • 2d—Wash Cycle
    • Step 3—Final Deprotection: Steps 2a and 2b were performed one last time.


Resins were deswelled in methanol (rinsed twice in 5 ml methanol, incubated 5 minutes in 5 ml methanol, rinsed in 5 ml methanol) and then vacuum dried.


Peptide was removed from the resin by incubating 2 hours in reagent R and then precipitated into ether. Peptide was washed in ether and then vacuum dried. Peptide was resolubilized in diH2O, frozen and lyophilized overnight.


Conjugation of Peptide with Keyhole Limpet Hemocyanin


Peptide (6 mg) was conjugated with Keyhole Limpet Hemocyanin (KLH). If the selected peptide includes at least one cysteine, three aliquots (2 mg) can be dissolved in PBS (2 ml) and coupled to KLH via glutaraldehyde, EDC or maleimide activated KLH (2 mg) in 2 ml of PBS for a total volume of 4 ml. When the peptide lacks cysteine (as in the TLE3 peptide), two aliquots (3 mg) can be coupled via glutaraldehyde and EDC methods.


Maleimide coupling can be accomplished by mixing 2 mg of peptide with 2 mg of maleimide-activated KLH dissolved in PBS (4 ml) and incubating 4 hr.


EDC coupling can be accomplished by mixing 2 mg of peptide, 2 mg unmodified KLH, and 20 mg of EDC in 4 ml PBS (lowered to pH 5 by the addition of phosphoric acid), and incubating for 4 hours. The reaction is then stopped by the slow addition of 1.33 ml acetic acid (pH 4.2). When using EDC to couple 3 mg of peptide, the amounts listed above are increased by a factor of 1.5.


Glutaraldehyde coupling occurs when 2 mg of peptide are mixed with 2 mg of KLH in 0.9 ml of PBS. 0.9 ml of 0.2% glutaraldehyde in PBS is added and mixed for one hour. 0.46 ml of 1 M glycine in PBS is added and mixed for one hour. When using glutaraldehyde to couple 3 mg of peptide, the above amounts are increased by a factor of 1.5.


The conjugated aliquots were subsequently repooled, mixed for two hours, dialyzed in 1 liter PBS and lyophilized.


Immunization of Rabbits

Two New Zealand White Rabbits were injected with 250 μg (total) KLH conjugated peptide in an equal volume of complete Freund's adjuvant and saline in a total volume of 1 ml. 100 μg KLH conjugated peptide in an equal volume of incomplete Freund's Adjuvant and saline were then injected into three to four subcutaneous dorsal sites for a total volume of 1 ml two, six, eight and twelve weeks after the first immunization. The immunization schedule was as follows:


















Day 0
Pre-immune bleed, primary immunization



Day 15
1st boost



Day 27
1st bleed



Day 44
2nd boost



Day 57
2nd bleed and 3rd boost



Day 69
3rd bleed



Day 84
4th boost



Day 98
4th bleed










Collection of Rabbit Serum

The rabbits were bled (30 to 50 ml) from the auricular artery. The blood was allowed to clot at room temperature for 15 minutes and the serum was separated from the clot using an IEC DPR-6000 centrifuge at 5000 g. Cell-free serum was decanted gently into a clean test tube and stored at −20° C. for affinity purification.


Determination of Antibody Titer

All solutions with the exception of wash solution were added by the Hamilton Eclipse, a liquid handling dispenser. The antibody titer was determined in the rabbits using an ELISA assay with peptide on the solid phase. Flexible high binding ELISA plates were passively coated with peptide diluted in BBS (100 μl, 1 μg/well) and the plate was incubated at 4° C. in a wetbox overnight (air-tight container with moistened cotton balls). The plates were emptied and then washed three times with BBS containing 0.1% Tween-20 (BBS-TW) by repeated filling and emptying using a semi-automated plate washer. The plates were blocked by completely filling each well with BBS-TW containing 1% BSA and 0.1% gelatin (BBS-TW-BG) and incubating for 2 hours at room temperature. The plates were emptied and sera of both pre- and post-immune serum were added to wells. The first well contained sera at 1:50 in BBS. The sera were then serially titrated eleven more times across the plate at a ratio of 1:1 for a final (twelfth) dilution of 1:204,800. The plates were incubated overnight at 4° C. The plates were emptied and washed three times as described.


Biotinylated goat anti-rabbit IgG (100 μl) was added to each microtiter plate test well and incubated for four hours at room temperature. The plates were emptied and washed three times. Horseradish peroxidase-conjugated Streptavidin (100 μl diluted 1:10,000 in BBS-TW-BG) was added to each well and incubated for two hours at room temperature. The plates were emptied and washed three times. The ABTS was prepared fresh from stock by combining 10 ml of citrate buffer (0.1 M at pH 4.0), 0.2 ml of the stock solution (15 mg/ml in water) and 10 μl of 30% hydrogen peroxide. The ABTS solution (100 μl) was added to each well and incubated at room temperature. The plates were read at 414 nm, 20 minutes following the addition of substrate.


Preparation of Peptide Affinity Purification Column:

The affinity column was prepared by conjugating 5 mg of peptide to 10 ml of cyanogen bromide-activated Sepharose 4B and 5 mg of peptide to hydrazine-Sepharose 4B. Briefly, 100 μl of DMF was added to peptide (5 mg) and the mixture was vortexed until the contents were completely wetted. Water was then added (900 μl) and the contents were vortexed until the peptide dissolved. Half of the dissolved peptide (500 μl) was added to separate tubes containing 10 ml of cyanogen-bromide activated Sepharose 4B in 0.1 ml of borate buffered saline at pH 8.4 (BBS) and 10 ml of hydrazine-Sepharose 4B in 0.1 M carbonate buffer adjusted to pH 4.5 using excess EDC in citrate buffer pH 6.0. The conjugation reactions were allowed to proceed overnight at room temperature. The conjugated Sepharose was pooled and loaded onto fitted columns, washed with 10 ml of BBS, blocked with 10 ml of 1 M glycine and washed with 10 ml 0.1 M glycine adjusted to pH 2.5 with HCl and re-neutralized in BBS. The column was washed with enough volume for the optical density at 280 nm to reach baseline.


Affinity Purification of Antibodies

The peptide affinity column was attached to a UV monitor and chart recorder. The titered rabbit antiserum was thawed and pooled. The serum was diluted with one volume of BBS and allowed to flow through the columns at 10 ml per minute. The non-peptide immunoglobulins and other proteins were washed from the column with excess BBS until the optical density at 280 nm reached baseline. The columns were disconnected and the affinity purified column was eluted using a stepwise pH gradient from pH 7.0 to 1.0. The elution was monitored at 280 nm and fractions containing antibody (pH 3.0 to 1.0) were collected directly into excess 0.5 M BBS. Excess buffer (0.5 M BBS) in the collection tubes served to neutralize the antibodies collected in the acidic fractions of the pH gradient.


The entire procedure was repeated with “depleted” serum to ensure maximal recovery of antibodies. The eluted material was concentrated using a stirred cell apparatus and a membrane with a molecular weight cutoff of 30 kD. The concentration of the final preparation was determined using an optical density reading at 280 nm. The concentration was determined using the following formula: mg/ml=OD280/1.4.


It will be appreciated that in certain embodiments, additional steps may be used to purify antibodies of the invention. In particular, it may prove advantageous to repurify antibodies, e.g., against one of the peptides that was used in generating the antibodies. It is to be understood that the present invention encompasses antibodies that have been prepared with such additional purification or repurification steps. It will also be appreciated that the purification process may affect the binding between samples and the inventive antibodies.


Example 2
Preparing and Staining Tissue Arrays

This example describes a method that was employed to prepare the tissue arrays that were used in the Examples. This example also describes how the antibody staining was performed.


Tissue arrays were prepared by inserting full-thickness cores from a large number of paraffin blocks (donor blocks) that contain fragments of tissue derived from many different patients and/or different tissues or fragments of tissues from a single patient, into a virgin paraffin block (recipient block) in a grid pattern at designated locations in a grid. A standard slide of the paraffin embedded tissue (donor block) was then made which contained a thin section of the specimen amenable to H & E staining A trained pathologist, or the equivalent versed in evaluating tumor and normal tissue, designated the region of interest for sampling on the tissue array (e.g., a tumor area as opposed to stroma). A commercially available tissue arrayer from Beecher Instruments was then used to remove a core from the donor block which was then inserted into the recipient block at a designated location. The process was repeated until all donor blocks had been inserted into the recipient block. The recipient block was then thin-sectioned to yield 50-300 slides containing cores from all cases inserted into the block.


The selected antibodies were then used to perform immunohistochemical staining using the DAKO Envision+, Peroxidase IHC kit (DAKO Corp., Carpenteria, Calif.) with DAB substrate according to the manufacturer's instructions. FIG. 1 shows exemplary IHC staining images of samples that are TLE3-negative (S0643-) and TLE3-positive (S0643+).


Example 3
TLE3 Expression Correlates with Response to Chemotherapy in Cancer Patients

Tumor samples from two different breast cancer cohorts—Huntsville Hospital (HH) and Roswell Park Cancer Institute (RP)—were stained with the TLE3 antibody of Example 1. Treatment and recurrence data were available for all patients in both cohorts. FIG. 2 shows Kaplan-Meier recurrence curves that were generated using all patients in the HH cohort after classification based on staining with the TLE3 antibody. Recurrence data from TLE3-positive and TLE3-negative patients were used to generate the top and bottom curves, respectively. As shown in the Figure, antibody binding to the TLE3 marker correlates with improved prognosis across this breast cancer cohort (HR=0.573, p=0.004). FIG. 3 shows Kaplan-Meier recurrence curves that were generated in a similar fashion using all patients in the RP cohort. As with the HH cohort, antibody binding to the TLE3 marker was found to correlate with improved prognosis (HR=0.239, p=0.011).


In order to determine whether TLE3 expression is correlated with response to chemotherapy, separate Kaplan-Meier recurrence curves were generated using HH cohort patients that did or did not receive chemotherapy (FIGS. 4 and 5, respectively). As shown in FIG. 4, antibody binding to the TLE3 marker lost its correlation with prognosis in patients that did not receive chemotherapy (HR=0.788, p=0.490). However, as shown in FIG. 5, the correlation was restored in patients that did receive chemotherapy (HR=0.539, p=0.013). These results demonstrate that TLE3 expression is correlated with improved response to chemotherapy (i.e., TLE3-positive cancers are more likely to respond to chemotherapy than TLE-3 negative cancers). Kaplan-Meier recurrence curves that were generated using patients in the RP breast cancer cohort that received chemotherapy are consistent with this prediction model (see FIG. 6, HR=0.194, p=0.010). Kaplan-Meier recurrence curves that were generated using patients in the UAB ovarian cancer cohort that received chemotherapy are also consistent with this prediction model (see FIG. 18, HR=0.64, p=0.049).


Example 4
Specific Chemotherapeutic Correlations

Since different patients in the HH and RP cohorts received different types of chemotherapy we were also able to determine whether TLE3 expression correlates with response to specific types of chemotherapy.



FIG. 7 shows Kaplan-Meier recurrence curves that were generated using patients in the HH breast cancer cohort of FIG. 5 that received CMF (cyclophosphamide, methotrexate and 5-fluorouracil) chemotherapy. Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in FIG. 7, antibody binding to the TLE3 marker correlates with improved prognosis across this subset of treated patients (HR=0.398, p=0.019). Based on the results below which demonstrated a loss of correlation for patients in the HH cohort that were treated with CA (cyclophosphamide and adriamycin, HR=1.000) or CAF (cyclophosphamide, adriamycin and 5-fluorouracil, HR=1.000) we were able to establish that the predictive correlation in FIG. 7 is between TLE3 binding and treatment with methotrexate (see also FIG. 9 which combines the CA and CAF treated subsets, HR=1.030).



FIG. 8 shows Kaplan-Meier recurrence curves that were generated using patients in the HH breast cancer cohort of FIG. 5 that received CA or CAF chemotherapy (with or without a taxane). As shown in the Figure, the correlation between antibody binding to the TLE3 marker and prognosis loses significance in this subset of treated patients (HR=0.666, p=0.22). When the curves were generated using patients that received CA or CAF chemotherapy only (i.e., without a taxane) the significance was further reduced (see FIG. 9, HR=1.030, p=0.95). However, the correlation was restored in patients that received CA or CAF in combination with a taxane (see FIG. 10, HR=0.114, p=0.038). These results demonstrate a correlation between TLE3 binding and treatment with a taxane.



FIG. 11 shows Kaplan-Meier recurrence curves that were generated using patients in the RP breast cancer cohort of FIG. 6 that received CA chemotherapy only (i.e., without a taxane). Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, there is no correlation between antibody binding to the TLE3 marker and prognosis in this subset of treated patients (HR=0.759, p=0.81). The correlation was restored when the curves were generated using patients that received CA chemotherapy in combination with a taxane (see FIG. 12, HR=0.153, p=0.018). These results support the results of FIGS. 8 and 9 that were obtained using samples from the HH cohort.



FIG. 13 shows Kaplan-Meier recurrence curves that were generated using patients in the RP breast cancer cohort of FIG. 6 that received a taxane or CMF. Some of the patients receiving a taxane also received CA. Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. As shown in the Figure, antibody binding to the TLE3 marker correlates with improved prognosis across this subset of treated patients (HR=0.137, p=0.011).



FIG. 14 shows Kaplan-Meier recurrence curves that were generated using patients in the RP breast cancer cohort of FIG. 6 that received neoadjuvant chemotherapy. Recurrence data from TLE3-positive and TLE3-negative patients in this subset were used to generate the top and bottom curves, respectively. The sample size was small (N=12); however, as shown in the Figure, antibody binding to the TLE3 marker showed significant correlation with improved prognosis across this subset of treated patients when measured using the Fisher Exact Test (p=0.005). In addition, of the 12 patients receiving neoadjuvant chemotherapy, two received CA (both showed recurrence) while ten received CA with a taxane (seven showed recurrence, three did not). Notably, the three patients that did not show any recurrence were the only patients with TLE3-positive samples. These results are significant since they show that the correlation between TLE3 binding and response to chemotherapy applies irrespective of whether treatment is administered in an adjuvant or neoadjuvant setting.



FIGS. 15-17 show Kaplan-Meier recurrence curves that were generated using patients in the RP breast cancer cohort of FIG. 6 that received chemotherapy. Recurrence data from TLE3-positive and TLE3-negative patients with stage II+ (FIG. 15), stage IIb+ (FIG. 16) and stage III+ (FIG. 17) cancers were used to generate the top and bottom curves, respectively. In each case, antibody binding to the TLE3 marker correlated with improved prognosis across these subsets of treated patients. The sample size was small in the subset of FIG. 17 (N=19); however significance was obtained when measured using the Fisher Exact Test (p=0.020). These results are of clinical importance since they demonstrate that the predictive power of the TLE3 marker is independent of stage and remains significant even in patients with the worst prognosis (e.g., stage III+ patients).


Example 5
Bivariate Analysis

In order to confirm that the predictive power of TLE3 is independent of other clinical factors (e.g., age, tumor size, nodes status, necrosis, etc.) we performed bivariate statistical analysis using results from the RP breast cohort. The results are summarized in Table 1 below. As shown in the Table, prediction using TLE3 remained significant in all bivariate analyses demonstrating its independence of other clinical factors.









TABLE 1







Bivariate Analysis
















HR for
p for




Factor 1
Factor 2
N
TLE3
TLE3
HR
P





TLE3

81
0.239
0.0110




TLE3
Age
81
0.223
0.0082
0.967
0.1200


TLE3
Tumor Size
78
0.219
0.0077
1.292
0.0002


TLE3
Nodes Met Ca1
79
0.252
0.0150
1.066
0.0086


TLE3
Necrosis
72
0.232
0.0100
1.903
0.2600


TLE3
Vasc. Lymph Inv.2
74
0.205
0.0071
0.412
0.0790


TLE3
Stage
80
0.284
0.0280
2.063
0.0130


TLE3
Contains Tax3
70
0.168
0.0061
2.749
0.0980






1Nodes found with metastatic cancer.




2Vascular lymphatic invasion.




3Taxane containing regimens.





















APPENDIX A







ENTREZ

PEPTIDE 1
PEPTIDE 2
PEPTIDE 3



AGI

GENE

(SEQ ID
(SEQ ID
(SEQ ID



ID
GENE NAME
ID
ALIASES
NO.)
NO.)
NO.)
TITER






















S0011
vav 3 oncogene
10451
VAV3; VAV3 ONCOGENE;
TEESINDED
EKRTNGL
DYISKSKE
1:90-





ONCOGENE VAV3; vav 3 oncogene
IYKGLPDLI
RRTPKQV
DVKLK (3)
1:300






DE (1)
D (2)







S0017
WAP four-disulfide
10406
WFDC2; WAP5; dJ461P17.6; major
EKTGVCPE
PNDKEGS
RDQCQVD
1:25-



core domain 2

epididymis-specific protein E4;
LQADQNCT
CPQVNIN
TQCPGQM
1:500





epididymal secretory protein E4; WAP
QE (4)
(5)
K (6)






four-disulfide core domain 2; WAP









domain containing protein HE4-V4;









epididymis-specific, whey-acidic









protein type, four-disulfide core; WAP









four-disulfide









S0018
secretoglobin, family
4250
UGB2; MGB1; SCGB2A2;
SKTINPQVS
DDNATTN
NQTDETL
1:300-



2A, member 2

mammaglobin 1; secretoglobin, family
KTEYKELL
AIDELKEC
SNVEVFM
1:1000





2A, member 2
QE (7)
(8)
Q (9)






S0020
PPAR binding protein
5469
RB18A; TRIP2; PPARGBP; PBP;
SSDDGIRPL
DGKSKDK
NKTKKKK
1:100





CRSP1; PPARBP; CRSP200;
PEYSTEKH
PPKRKKA
SSRLPPEK






DRIP230; PPAR-BINDING
KK (10)
DTE (11)
(12)






PROTEIN; PPARG binding protein;









PPAR binding protein; CRSP, 200-KD









SUBUNIT; PEROXISOME









PROLIFERATOR-ACTIVATED









RECEPTOR-BINDING PROTEIN;









THYROID HORMONE RECEPTOR









INTERACTOR 2; RECOGN









S0021
hypothetical protein
222256
FLJ23834; hypothetical protein
KNKEPLTK
KLTCTDL
EVDYENP
1:200-



FLJ23834

FLJ23834
KGETKTAE
DSSPRSFR
SNLAAGN
1:2500






RD (13)
YS (14)
KYT (15)






S0022
cytochrome P450 4Z1
199974
CYP4Z1; cytochrome P450 4Z1;
KTLQVFNP
QHFAIIEC
RKFLAPD
1:50-





cytochrome P450, family 4, subfamily
LRFSRENSE
KVAVALT
HSRPPQPV
1:500





Z, polypeptide 1
KIH (16)
(17)
RQ (18)






S0024
RAS-like, estrogen-
85004
RERG; RAS-like, estrogen-regulated,
MAKSAEV
VLPLKNIL
YELCREV
1:900-



regulated, growth-

growth-inhibitor
KLAIFGRA
DEIKKPK
RRRRMVQ
1:2700



inhibitor


GVGK (19)
N (20)
GKT (21)






S0032
fatty acid binding
2170
MDGI; O-FABP; FABP3; FABP11;
TKPTTIIEK
KNTEISFK
HLQKWD
1:225



protein 3, muscle and

H-FABP; FATTY ACID-BINDING
NGDILTLK
LGVEFDE
GQETTLV




heart (mammary-

PROTEIN, SKELETAL MUSCLE;
TH (22)
(23)
RE (24)




derived growth

Fatty acid-binding protein 3, muscle;







inhibitor)

fatty acid binding protein 11; FATTY









ACID-BINDING PROTEIN,









MUSCLE AND HEART; fatty acid









binding protein 3, muscle and heart









(mammary-de









S0036
gamma-aminobutyric
2568
GABRP; GAMMA-
DGNDVEFT
LQQMAA
KRKISFAS
1:250-



acid (GABA) A

AMINOBUTYRIC ACID
WLRGNDS
KDRGTTK
IEISSDNV
1:500



receptor, pi

RECEPTOR, PI; GABA-A
VRGLEH
EVEEVS
DYSD (27)






RECEPTOR, PI POLYPEPTIDE;
(25)
(26)







gamma-aminobutyric acid (GABA) A









receptor, pi









S0037
annexin A8
244
ANX8; ANXA8; annexin VIII;
QRQQIAKS
REIMKAY
EEYEKIAN
1:30-





annexin A8
FKAQFGKD
EEDYGSS
KSIEDSIK
1:40






LTE (28)
LEEDIQ
SE (30)








(29)







S0039
CDNA FLJ25076 fis,
134111
similar to 3110006E14Rik protein;
EGGSLVPA
RKAGKSK
KTHEKYG
1:50-



clone CBL06117

CDNA FLJ25076 fis, clone CBL06117
ARQQHCTQ
KSFSRKE
WVTPPVS
1:30000






VRSRR (31)
AE (32)
DG (33)






S0040
ATP-binding cassette,
5243
P-gp; PGY1; CLCS; ABCB1; ABC20;
MDLEGDR
NLEDLMS
RGSQAQD
1:200-



sub-family B

CD243; GP170; MDR1; doxorubicin
NGGAKKK
NITNRSDI
RKLSTKE
1:400



(MDR/TAP), member

resistance; colchicin sensitivity; P-
N (34)
NDTG (35)
A (36)




1

GLYCOPROTEIN 1; multidrug









resistance 1; P glycoprotein 1; ATP-









binding cassette sub-family B member









1; ATP-BINDING CASSETTE,









SUBFAMILY B, MEMBER 1; ATP-









bin









S0041
ATP-binding cassette,
5244
MDR3; PGY3; PFIC-3; ABCB4;
MDLEAAK
NFSFPVNF
KNSQMCQ
1:60-



sub-family B

ABC21; MDR2/3; P-
NGTAWRPT
SLSLLNPG
KSLDVET
1:300



(MDR/TAP), member

GLYCOPROTEIN 3; MULTIDRUG
SAE (37)
K (38)
DG (39)




4

RESISTANCE 3; P-glycoprotein-









3/multiple drug resistance-3; P









glycoprotein 3/multiple drug resistance









3; ATP-binding cassette, sub-family B









(MDR/TAP), member 4; ATP-binding









cassette, sub









S0042
ATP-binding cassette,
4363
ABCC1; MRP1; GS-X; ABC29;
MALRGFCS
KNWKKE
DSIERRPV
1:40-



sub-family C

multidrug resistance protein;
ADGSD (40)
CAKTRKQ
KDGGGTN
1:500



(CFTR/MRP),

MULTIDRUG RESISTANCE-

PVK (41)
S (42)




member 1

ASSOCIATED PROTEIN 1; multiple









drug resistance-associated protein;









multiple drug resistance protein 1;









ATP-BINDING CASSETTE,









SUBFAMILY C, MEMBER 1; ATP-









binding cassette, sub-fami









S0043
ATP-binding cassette,
1244
MRP2; cMRP; CMOAT; ABCC2;
MLEKFCNS
SILCGTFQ
ENNESSN
1:50-



sub-family C

ABC30; DJS; MULTIDRUG
TFWNSSFL
FQTLIRT
NPSSIAS
1:333



(CFTR/MRP),

RESISTANCE-ASSOCIATED
DSPE (43)
(44)
(45)




member 2

PROTEIN 2; canalicular multispecific









organic anion transporter;









MULTISPECIFIC ORGANIC ANION









TRANSPORTER, CANALICULAR;









ATP-BINDING CASSETTE,









SUBFAMILY C, MEMBER 2; ATP-









binding cassette,









S0044
ATP-binding cassette,
10257
MOAT-B; MRP4; MOATB; ABCC4;
QEVKPNPL
DEISQRNR
VQDFTAF
1:20-



sub-family C

EST170205; MULTIDRUG
QDANICSR
QLPSDGK
WDKASET
1:100



(CFTR/MRP),

RESISTANCE-ASSOCIATED
(46)
K (47)
PTLQ (48)




member 4

PROTEIN 4; MULTISPECIFIC









ORGANIC ANION TRANSPORTER









B; ATP-binding cassette, sub-family









C, member 4; ATP-BINDING









CASSETTE, SUBFAMILY C,









MEMBER 4; ATP-binding cassette,









sub-family C (CFT









S0045
ATP-binding cassette,
8714
MOAT-D; ABC31; MLP2; ABCC3;
MDALCGSG
RKQEKQT
DPQSVER
1:2000



sub-family C

EST90757; cMOAT2; MULTIDRUG
ELGSKFWD
ARHKASA
KTISPG




(CFTR/MRP),

RESISTANCE-ASSOCIATED
SN (49)
A (50)
(51)




member 3

PROTEIN 3; canicular multispecific









organic anion transporter;









CANALICULAR MULTISPECIFIC









ORGANIC ANION TRANSPORTER









2; ATP-BINDING CASSETTE,









SUBFAMILY C, MEMBER 3; ATP-









binding cas









S0046
ATP-binding cassette,
10057
MOAT-C; ABCC5; MRP5;
MKDIDIGK
RDREDSK
SKHESSD
1:100-



sub-family C

EST277145; ABC33; SMRP;
EYIIPSPGY
FRRTRPLE
VNCRRLE
1:450



(CFTR/MRP),

pABC11; MOATC; MULTIDRUG
RS (52)
CQD (53)
R (54)




member 5

RESISTANCE-ASSOCIATED









PROTEIN 5; canalicular multispecific









organic anion transporter C; ATP-









binding cassette, sub-family C,









member 5; ATP-BINDING









CASSETTE, SUBFAMILY C,









MEMBER 5; ATP-bi









S0047
ATP-binding cassette,
368
MRP6; ARA; EST349056; ABCC6;
MAAPAEPC
DPGVVDS
HTLVAEN
1:50



sub-family C

MOATE; PXE; MLP1; ABC34;
AGQGVWN
SSSGSAA
AMNAEK




(CFTR/MRP),

ANTHRACYCLINE RESISTANCE-
QTEPE (55)
GKD (56)
(57)




member 6

ASSOCIATED PROTEIN;









MULTIDRUG RESISTANCE-









ASSOCIATED PROTEIN 6; ATP-









binding cassette, sub-family C,









member 6; ATP-BINDING









CASSETTE, SUBFAMILY C,









MEMBER 6; ATP-binding cassette,









S0048
ATP-binding cassette,
8647
BSEP; ABCB11; PFIC-2; SPGP;
MSDSVILRS
TNSSLNQ
QEVLSKIQ
1:600



sub-family B

PGY4; PFIC2; ABC16; SISTER OF P-
IKKFGEEN
NMTNGTR
HGHTIIS




(MDR/TAP), member

GLYCOPROTEIN; bile salt export
D (58)
(59)
(60)




11

pump; progressive familial intrahepatic









cholestasis 2; ABC member 16,









MDR/TAP subfamily; ATP-BINDING









CASSETTE, SUBFAMILY B,









MEMBER 11; ATP-binding cassette,









sub-fam









S0049
ATP-binding cassette,
23456
MTABC2; EST20237; MABC2; M-
GADDPSSV
NAVASPE
KPNGIYR
1:10-



sub-family B

ABC2; ABCB10; MITOCHONDRIAL
TAEEIQR
FPPRFNT
KLMNKQS
1:25



(MDR/TAP), member

ABC PROTEIN 2; ATP-BINDING
(61)
(62)
FISA (63)




10

CASSETTE, SUBFAMILY B,









MEMBER 10; ATP-binding cassette,









sub-family B, member 10; ATP-









binding cassette, sub-family B









(MDR/TAP), member 10









S0050
transporter 1, ATP-
6890
RING4; ABC17; D6S114E; ABCB2;
MASSRCPA
QGGSGNP
EFVGDGI
1:80



binding cassette, sub-

TAP1; APT1; PEPTIDE
PRGCR (64)
VRR (65)
YNNTMG




family B (MDR/TAP)

TRANSPORTER PSF1;


HVHS (66)






TRANSPORTER, ABC, MHC, 1;









ABC transporter, MHC 1; antigen









peptide transporter 1; peptide supply









factor 1; ABC TRANSPORTER,









MHC, 1; ATP-BINDING CASSETTE,









SUBFAMILY B, MEMBER 2;









TRANSPORTER









S0052
ATP-binding cassette,
6833
SUR1; MRP8; PHHI; ABC36;
MPLAFCGS
DHLGKEN
EIREEQCA
1:25-



sub-family C

ABCC8; HRINS; sulfonylurea
ENHSAAYR
DVFQPKT
PHEPTPQG
1:150



(CFTR/MRP),

receptor (hyperinsulinemia);
(67)
QFLG (68)
(69)




member 8

SULFONYLUREA RECEPTOR,









BETA-CELL HIGH-AFFINITY;









ATP-binding cassette, sub-family C,









member 8; ATP-BINDING









CASSETTE, SUBFAMILY C,









MEMBER 8; ATP-binding cassette,









sub-family C









S0053
ATP-binding cassette,
10060
ABCC9; ABC37; sulfonylurea
MSLSFCGN
QRVNETQ
DEIGDDS
1:25-



sub-family C

receptor 2A; ATP-BINDING
NISS (70)
NGTNNTT
WRTGESS
1:50



(CFTR/MRP),

CASSETTE, SUBFAMILY C,

GISE (71)
LPFE (72)




member 9

MEMBER 9; ATP-binding cassette,









sub-family C (CFTR/MRP), member









9; ATP-binding cassette, sub-family C,









member 9 isoform SUR2B; ATP-









binding cassette, sub-family C,









member 9 isoform









S0055
integral membrane
9445
E25B; ABRI; E3-16; FBD; BRI2;
MVKVTFNS
QTIEENIKI
HDKETYK
1:450-



protein 2B

BRICD2B; ITM2B; BRI GENE;
ALAQKEAK
FEEEEVE
LQRRETIK
1:500





BRICHOS domain containing 2B;
KDEPK (73)
(74)
GIQKRE






integral membrane protein 2B


(75)






S0057
ankyrin 3, node of
288
ankyrin-G; ANK3; ankyrin-3, node of
MAHAASQ
HKKETES
EGFKVKT
1:750



Ranvier (ankyrin G)

Ranvier; ankyrin 3 isoform 1; ankyrin
LKKNRDLE
DQDDEIE
KKEIRHV






3 isoform 2; ankyrin 3, node of
INAEE (76)
KTDRRQ
EKKSHS






Ranvier (ankyrin G)

(77)
(78)






S0058
hypothetical protein
80004
FLJ21918; hypothetical protein
ERALAAAQ
TAGMKDL
DPPRTVL
1:20



FLJ21918

FLJ21918
RCHKKVM
LSVFQAY
QAPKEWV







KER (79)
Q (80)
CL (81)






S0059
tripartite motif-
23650
ATDC; TRIM29; tripartite motif-
MEAADASR
ELHLKPH
EGEGLGQ
1:50-



containing 29

containing 29; ataxia-telangiectasia
SNGSSPEA
LEGAAFR
SLGNFKD
1:3000





group D-associated protein; tripartite
RDAR (82)
DHQ (83)
DLLN (84)






motif protein TRIM29 isoform alpha;









tripartite motif protein TRIM29









isoform beta









S0059
tripartite motif-
23650
ATDC; TRIM29; tripartite motif-
ELHLKPHL
N/A
N/A
1:30-


P2
containing 29

containing 29; ataxia-telangiectasia
EGAAFRDH


1:90





group D-associated protein; tripartite
Q (85)








motif protein TRIM29 isoform alpha;









tripartite motif protein TRIM29









isoform beta









S0063
iroquois homeobox
79191
IRX3; iroquois homeobox protein 3
GSEERGAG
KIWSLAE
KKLLKTA
1:200-



protein 3


RGSSGGRE
TATSPDNP
FQPVPRRP
1:1200






E (86)
RRS (87)
QNHLD









(88)






S0068
RAS-like, estrogen-
85004
RERG; RAS-like, estrogen-regulated,
RRSSTTHV
N/A
N/A
1:500-



regulated, growth-

growth-inhibitor
KQAINKML


1:40000



inhibitor


TKISS (89)








S0070
G protein-coupled
26996
GPCR150; GPR160; putative G
MRRKNTC
NETILYFP
KVQIPAYI
1:10-



receptor 160

protein-coupled receptor; G protein-
QNFMEYFC
FSSHSSYT
EMNIPLVI
1:100





coupled receptor 160
ISLAF (90)
VRSKK
LCQ (92)








(91)







S0072
S100 calcium binding
6279
CP-10; L1Ag; CALPROTECTIN;
MLTELEKA
RDDLKKL
KMGVAA
1:6500-



protein A8

60B8AG; S100A8; MIF; CAGA; NIF;
LNSIIDVYH
LETECPQ
HKKSHEE
1:10000



(calgranulin A)

MRP8; MA387; CFAG; CGLA
K (93)
YIRKKGA
SHKE (95)






S100A8/S100A9 COMPLEX; cystic

D (94)







fibrosis antigen; S100 calcium-binding









protein A8; S100 calcium binding









protein A8 (calgranulin A)









S0073
forkhead box A1
3169
HNF3A; MGC33105; TCF3A;
PESRKDPS
HGLAPHE
EQQHKLD
1:100-





FOXA1; forkhead box A1;
GASNPSAD
SQLHLKG
FKAYEQA
1:2700





HEPATOCYTE NUCLEAR FACTOR
S (96)
D (97)
LQYS (98)






3-ALPHA; hepatocyte nuclear factor 3,









alpha









S0073
forkhead box A1
3169
HNF3A; MGC33105; TCF3A;
HGLAPHES
N/A
N/A
1:50-


P2


FOXA1; forkhead box A1;
QLHLKGD


1:450





HEPATOCYTE NUCLEAR FACTOR
(99)








3-ALPHA; hepatocyte nuclear factor 3,









alpha









S0074
trefoil factor 3
7033
TFF3; trefoil factor 3 (intestinal);
EEYVGLSA
RVDCGYP
VPWCFKP
1:2500-



(intestinal)

trefoil factor 3, HITF, human intestinal
NQCAVPAK
HVTPKEC
LQEAECT
1:30000





trefoil factor
DRVD (100)
N (101)
F (102)






S0074
trefoil factor 3
7033
TFF3; trefoil factor 3 (intestinal);
VPWCFKPL
N/A
N/A
1:400-


P3
(intestinal)

trefoil factor 3, HITF, human intestinal
QEAECTF


1:810





trefoil factor
(103)








S0076
keratin 17
3872
PC2; PCHC1; KRT17; K17;
KKEPVTTR
QDGKVISS
SSSIKGSS
1:200


x1


CYTOKERATIN 17; keratin 17
QVRTIVEE
REQVHQT
GLGGGSS







(104)
TR (105)
(106)






S0078
kynureninase (L-
8942
3.7.1.3; XANTHURENICACIDURIA;
DEEDKLRH
KPREGEE
EERGCQL
1:180-



kynurenine hydrolase)

KYNU;
FRECFYIPK
TLRIEDIL
TITFSVPN
1:200





HYDROXYKYNURENINURIA;
IQD (107)
EVIEKE
KDVFQE






KYNURENINASE DEFICIENCY;

(108)
(109)






XANTHURENIC ACIDURIA;









kynureninase (L-kynurenine









hydrolase)









S0079
solute carrier family
25800
SLC39A6; LIV-1 protein, estrogen
DHNHAASG
EEPAMEM
QRYSREE
1:200-



39 (zinc transporter),

regulated; solute carrier family 39
KNKRKALC
KRGPLFS
LKDAGVA
1:800



member 6

(zinc transporter), member 6; solute
PDHD (110)
HLSSQNI
TL (112)






carrier family 39 (metal ion

(111)







transporter), member 6









S0081
N-acetyltransferase 1
9
AAC1; 2.3.1.5; NAT1; arylamine N-
MDIEAYLE
QMWQPLE
FNISLQRK
1:10-



(arylamine N-

acetyltransferase-1; ACETYL-
RIGYKKSR
LISGKDQP
LVPKHGD
1:240



acetyltransferase)

CoA:ARYLAMINE N-
NKLDLE
QVPCVFR
RFFTI






ACETYLTRANSFERASE;
(113)
(114)
(115)






ARYLAMINE N-









ACETYLTRANSFERASE 1; N-









acetyltransferase 1 (arylamine N-









acetyltransferase); arylamide acetylase









1 (N-acetyltransferase 1)









S0086
X-box binding protein
7494
XBP2; TREB5; XBP1; X-box-binding
RQRLTHLS
EKTHGLV
QPPFLCQ
1:180-



1

protein-1; X BOX-BINDING
PEEKALRR
VENQELR
WGRHQPS
1:400





PROTEIN 1; X BOX-BINDING
KLKNR
QRLGMD
WKPLMN






PROTEIN 2; X-box binding protein 1
(116)
(117)
(118)






S0088
claudin 10
9071
CPETRL3; OSP-L; CLDN10; claudin
NKITTEFFD
FSISDNNK
EDFKTTN
1:333-





10; claudin 10 isoform a; claudin 10
PLFVEQK
TPRYTYN
PSKQFDK
1:1000





isoform b
(119)
GAT (120)
NAYV









(121)






S0090
sparc/osteonectin,
9806
KIAA0275; testican-2; SPOCK2;
EGDAKGLK
EWCFCFW
EEEGEAG
1:100-



cwcv and kazal-like

TESTICAN 2;
EGETPGNF
REKPPCL
EADDGGY
1:800



domains proteoglycan

SPARC/OSTEONECTIN, CWCV,
MEDE (122)
AELER
IW (124)




(testican) 2

AND KAZAL-LIKE DOMAINS

(123)







PROTEOGLYCAN 2;









sparc/osteonectin, cwcv and kazal-like









domains proteoglycan (testican) 2









S0091
lipocalin 2 (oncogene
3934
UTEROCALIN; NGAL; LCN2;
DKDPQKM
KKCDYWI
ENFIRFSK
1:100



24p3)

NEUTROPHIL GELATINASE-
YATIYE
RTFVPGC
YLGLPEN






ASSOCIATED LIPOCALIN;
(125)
Q (126)
(127)






ONCOGENIC LIPOCALIN 24P3;









lipocalin 2 (oncogene 24p3)









S0092
paired box gene 8
7849
PAX8; paired box gene 8; paired box
DDSDQDSC
RQHYPEA
NTPLGRN
1:30-





gene 8 isoform PAX8C; paired box
RLSIDSQ
YASPSHT
LSTHQTY
1:100





gene 8 isoform PAX8D; paired box
(128)
K (129)
PVVAD






gene 8 isoform PAX8E; paired box


(130)






gene 8 isoform PAX8A; paired box









gene 8 isoform PAX8B; PAIRED









DOMAIN GENE 8 PAX8/PPARG









FUSION GENE









S0093
mesothelin
10232
CAK1; SMR; MSLN; mesothelin;
RLVSCPGP
KMSPEDIR
SPEELSSV
1:500





MEGAKARYOCYTE-
LDQDQQE
KWNVTSL
PPSSIWAV






POTENTIATING FACTOR;
(131)
ETLK (132)
RPQD (133)






SOLUBLE MPF/MESOTHELIN-









RELATED PROTEIN; mesothelin









isoform 2 precursor; mesothelin









isoform 1 precursor; megakaryocyte









potentiating factor precursor;









ANTIGEN RECOGNIZED BY









MONOCLONAL ANTIBODY









S0094
kallikrein 6 (neurosin,
5653
Bssp; PRSS18; KLK6; Klk7; SP59;
EEQNKLVH
ELIQPLPL
GKTADGD
1:150-



zyme)

PRSS9; MGC9355; protease M;
GGPCDKTS
ERDCSAN
FPDTIQC
1:300





kallikrein 6 preproprotein; protease,
H (134)
T (135)
(136)






serine, 18; protease, serine, 9;









kallikrein 6 (neurosin, zyme)









S0095
Rap guanine
10411
bcm910; MGC21410;
REQWPERR
KVNSAGD
QQLKVID
1:250-



nucleotide exchange

9330170P05Rik; EPAC; RAPGEF3;
RCHRLENG
AIGLQPD
NQRELSR
1:1000



factor (GEF) 3

cAMP-GEFI; RAP guanine-
CGNA (137)
AR (139)
LSRELE






nucleotide-exchange factor 3;


(140)






EXCHANGE PROTEIN









ACTIVATED BY cAMP; RAP









guanine-nucleotide-exchange factor









(GEF) 3; cAMP-REGULATED









GUANINE NUCLEOTIDE









EXCHANGE FACTOR I; RAP









GUANINE NUCLE









S0096
ATPase, H+
525
Vma2; VPP3; ATP6V1B1; RTA1B;
REHMQAV
KKSKAVL
DEFYSRE
1:100-



transporting,

3.6.3.14; VATB; ATP6B1; V-ATPase
TRNYITHPR
DYHDDN
GRLQDLA
1:800



lysosomal 56/58 kDa,

B1 subunit; H+-ATPase beta 1
(141)
(142)
PDTAL




V1 subunit B, isoform

subunit; H(+)-transporting two-sector


(143)




1 (Renal tubular

ATPase, 58 kD subunit; vacuolar







acidosis with

proton pump, subunit 3;







deafness)

endomembrane proton pump 58 kDa









subunit; ATPase, H+ transporting,









lysos









S0097
frizzled homolog 8
8325
FZ-8; hFZ8; FZD8; frizzled 8; frizzled
KQQDGPTK
ELRVLSK
RRGGEGG
1:100-



(Drosophila)

homolog 8 (Drosophila); FRIZZLED,
THKLEKLM
ANAIVPG
EENPSAA
1:500





DROSOPHILA, HOMOLOG OF, 8
IR (144)
LSGGE
KGHLMG








(145)
(146)






S0099
histone 1, H2ba
255626
HIST1H2BA; histone 1, H2ba
MPEVSSKG
GFKKAVV
KEGKKRK
1:333-






ATISKK
KTQK
RTRKE
1:500






(147)
(148)
(149)






S0110
hypothetical protein
84259
MGC2714; hypothetical protein
RYAFDFAR
SVFYQYL
EDGAWPV
1:500-



MGC2714

MGC2714
DKDQRSLD
EQSKYRV
LLDEFVE
1:2500






ID (150)
MNKDQ
WQKVRQ








(151)
TS (152)






S0117
reproduction 8
7993
D8S2298E; REP8; reproduction 8;
SFKSPQVY
RKKQQEA
EDIGITVD
1:200-





Reproduction/chromosome 8
LKEEEEKN
QGEKASR
TVLILEEK
1:375






EKR (153)
YIE (154)
EQTN (155)






S0119
slit homolog 1
6585
SLIT3; MEGF4; SLIL1; Slit-1; SLIT1;
KAFRGATD
DFRCEEG
DGTSFAE
1:900



(Drosophila)

slit homolog 1 (Drosophila); SLIT,
LKNLRLDK
QEEGGCL
EVEKPTK






DROSOPHILA, HOMOLOG OF, 1;
NQ (156)
PRPQ (157)
CGCALCA






MULTIPLE EPIDERMAL GROWTH


(158)






FACTOR-LIKE DOMAINS 4









S0122
leucyl-tRNA
23395
6.1.1.4; MGC26121; KIAA0028;
QRIKEQAS
HAKTKEK
KSPQPQLL
1:150



synthetase 2,

LEURS; LARS2; leucine translase;
KISEADKS
LEVTWEK
SNKEKAE




mitochondrial

leucine-tRNA ligase; LEUCYL-tRNA
KPKF (159)
MSKSKHN
ARK (161)






SYNTHETASE, MITOCHONDRIAL;

(160)







leucyl-tRNA synthetase 2,









mitochondrial; leucyl-tRNA synthetase









2, mitochondrial precursor









S0123
homeo box D4
3233
HOX4B; HOXD4; HHO.C13; HOX-
MLFEQGQQ
KDQKAKG
HSSQGRL
1:100-





5.1; HOMEOBOX D4; HOMEOBOX
ALELPECT
ILHSPASQ
PEAPKLT
1:500





4B; HOMEOBOX X; homeo box D4;
(162)
SPERS
HL (164)






homeobox protein Hox-D4; Hox-4.2,

(163)







mouse, homolog of homeo box X









S0124
sphingosine-1-
8879
KIAA1252; SPL; SGPL1;
KRGARRGG
KIVRVPLT
QFLKDIRE
1:990-



phosphate lyase 1

sphingosine-1-phosphate lyase 1
WKRKMPS
KMMEVD
SVTQIMK
1:1500






TDL (165)
VR (166)
NPKA









(167)






S0126
HBxAg transactivated
55789
XTP1; HBxAg transactivated protein 1
SKQGVVIL
VQTFSRCI
LKKPFQPF
1:450-



protein 1


DDKSKELP
LCSKDEV
QRTRSFR
1:1600






HW (168)
DLDEL
M (170)








(169)







S0132
SRY (sex determining
6662
SRA1; CMD1; CMPD1; SOX9; SRY-
MNLLDPFM
NTFPKGEP
KNGQAEA
1:100-



region Y)-box 9

BOX 9; transcription factor SOX9;
KMTDEQEK
DLKKESE
EEATEQT
1:500



(campomelic

SRY-RELATED HMG-BOX GENE
GLS (171)
EDK (172)
HISPN




dysplasia, autosomal

9; SEX REVERSAL, AUTOSOMAL,


(173)




sex-reversal)

1; SRY (sex-determining region Y)-









box 9 protein; SRY (sex-determining









region Y)-box 9 (campomelic









dysplasia, autosomal sex-reversal);









SRY (









S0137
cadherin, EGF LAG
1952
Flamingo1; CELSR2; EGFL2;
QASSLRLEP
ELKGFAE
RSGKSQPS
1:1800-



seven-pass G-type

KIAA0279; MEGF3; CDHF10; EGF-
GRANDGD
RLQRNES
YIPFLLRE
1:5000



receptor 2 (flamingo

like-domain, multiple 2; epidermal
WH (174)
GLDSGR
E (176)




homolog, Drosophila)

growth factor-like 2; multiple

(175)







epidermal growth factor-like domains









3; cadherin EGF LAG seven-pass G-









type receptor 2; cadherin, EGF LAG









seven-pass G-type receptor 2









S0139
gamma-glutamyl
8836
3.4.19.9; GGH; gamma-glutamyl
RRSDYAKV
KNFTMNE
EFFVNEA
1:2500-



hydrolase (conjugase,

hydrolase precursor; gamma-glutamyl
AKIFYNLSI
KLKKFFN
RKNNHHF
1:30000



folylpolygammagluta

hydrolase (conjugase,
QSFDD
VLTTN
KSESEE




myl hydrolase)

folylpolygammaglutamyl hydrolase)
(177)
(178)
(179)






S0140
bullous pemphigoid
667
BP240; FLJ13425; FLJ32235;
KNTQAAEA
QENQPEN
KQMEKDL
1:250-



antigen 1,

FLJ21489; FLJ30627; CATX-15;
LVKLYETK
SKTLATQ
AFQKQVA
1:20000



230/240 kDa

KIAA0728; BPAG1; dystonin;
LCE (180)
LNQ (181)
EKQLK






hemidesmosomal plaque protein;


(182)






bullous pemphigoid antigen 1,









230/240 kDa; bullous pemphigoid









antigen 1 (230/240 kD); bullous









pemphigoid antigen 1 isoform 1 eA









precursor; bullo









S0143
fatty acid synthase
2194
2.3.1.85; OA-519; FASN; MGC14367;
EFVEQLRK
DRHPQAL
REVRQLT
1:5000-





MGC15706; fatty acid synthase
EGVFAKEV
EAAQAEL
LRKLQEL
1:30000






R (183)
QQHD
SSKADE








(184)
(185)






S0143
fatty acid synthase
2194
2.3.1.85; OA-519; FASN; MGC14367;
REVRQLTL
N/A
N/A
1:200-


P3


MGC15706; fatty acid synthase
RKLQELSS


1:630






KADE (186)








S0144
matrix
4323
MMP-X1; 3.4.24.-; MMP14;
AYIREGHE
DEASLEP
RGSFMGS
1:500-



metalloproteinase 14

MTMMP1; MT1-MMP ; membrane-
KQADIMIFF
GYPKHIK
DEVFTYF
1:20000



(membrane-inserted)

type-1 matrix metalloproteinase;
AE (187)
ELGR (188)
YK (189)






matrix metalloproteinase 14









preproprotein; MATRIX









METALLOPROTEINASE 14,









MEMBRANE-TYPE; matrix









metalloproteinase 14 (membrane-









inserted); membrane-type matrix









metalloprotein









S0147
cystatin A (stefin A)
1475
STF1; CSTA; STFA; cystatin AS;
MIPGGLSE
NETYGKL
DLVLTGY
1:100-





cystatin A (stefin A)
AKPATPEIQ
EAVQYKT
QVDKNKD
1:5000






EIV (190)
Q (191)
DELTGF









(192)






S0149
transient receptor
55503
TRPV6; ECAC2; CAT1; CATL;
RQEHCMSE
QGHKWG
RACGKRV
1:400-



potential cation

CALCIUM TRANSPORTER 1;
HFKNRPAC
ESPSQGTQ
SEGDRNG
1:20000



channel, subfamily V,

CALCIUM TRANSPORTER-LIKE
LGAR (193)
AGAGK
SGGGKW




member 6

PROTEIN; EPITHELIAL CALCIUM

(194)
G (195)






CHANNEL 2; transient receptor









potential cation channel, subfamily V,









member 6









S0156
fatty acid binding
2173
B-FABP; FABP7; FABPB; MRG;
MVEAFCAT
QVGNVTK
KVVIRTLS
1:100-



protein 7, brain

mammary-derived growth inhibitor-
WKLTNSQN
PTVIISQE
TFKNTE
1:20000





related; FATTY ACID-BINDING
(196)
(197)
(198)






PROTEIN 7; FATTY ACID-









BINDING PROTEIN, BRAIN; fatty









acid binding protein 7, brain









S0158
cadherin 3, type 1, P-
1001
CDHP; HJMD; PCAD; CDH3;
RAVFREAE
QEPALFST
QKYEAHV
1:150-



cadherin (placental)

placental cadherin; CADHERIN,
VTLEAGGA
DNDDFTV
PENAVGH
1:2000





PLACENTAL; cadherin 3, P-cadherin
EQE (199)
RN (200)
E (201)






(placental); calcium-dependent









adhesion protein, placental; cadherin 3,









type 1 preproprotein; cadherin 3, type









1, P-cadherin (placental)









S0165
chemokine (C-X-C
2919
MGSA-a; NAP-3; CXCL1; SCYB1;
KKIIEKML
N/A
N/A
1:100-



motif) ligand 1

GROa; GRO1, FORMERLY; GRO
NSDKSN


1:500



(melanoma growth

PROTEIN, ALPHA; GRO1
(202)






stimulating activity,

ONCOGENE, FORMERLY;







alpha)

MELANOMA GROWTH









STIMULATORY ACTIVITY,









ALPHA; GRO1 oncogene (melanoma









growth-stimulating activity);









CHEMOKINE, CXC MOTIF,









LIGAND 1; GRO1 oncogene









(melanoma grow









S0171
baculoviral IAP
null
BIRC5; baculoviral IAP repeat-
GKPGNQNS
QAEAPLV
NCFLTER
1:22500-



repeat-containing 5

containing 5 (survivin)
KNEPPKKR
PLSRQNK
KAQPDE
1:30000



(survivin)


ERER (203)
(204)
(205)






S0193
procollagen-lysine, 2-
5352
PLOD2; LYSYL HYDROXYLASE 2;
EFDTVDLS
NKEVYHE
KQVDLEN
1:20000



oxoglutarate 5-

LYSINE HYDROXYLASE 2;
AVDVHPN
KDIKVFFD
VWLDFIR




dioxygenase (lysine

PROCOLLAGEN-LYSINE, 2-
(206)
KAK (207)
E (208)




hydroxylase) 2

OXOGLUTARATE 5-









DIOXYGENASE 2; procollagen-









lysine, 2-oxoglutarate 5-dioxygenase









(lysine hydroxylase) 2; procollagen-









lysine, 2-oxoglutarate 5-dioxygenase









(lysine hydroxylase) 2 isoform









S0202
PTK7 protein tyrosine
5754
PTK7; CCK4; protein-tyrosine kinase
LKKPQDSQ
KAKRLQK
KDRPSFSE
1:500-



kinase 7

PTK7; colon carcinoma kinase-4;
LEEGKPGY
QPEGEEPE
IASALGDS
1:800





PTK7 protein tyrosine kinase 7; PTK7
LD (209)
ME (210)
TVDSKP






protein tyrosine kinase 7 isoform e


(211)






precursor; PTK7 protein tyrosine









kinase 7 isoform a precursor; PTK7









protein tyrosine kinase 7 isoform d









precursor;









S0211
cytochrome P450,
1549
CYPIIA7; P450-IIA4; 1.14.14.1;
KRGIEERIQ
DRVIGKN
NPQHFLD
1:500-



family 2, subfamily A,

CPA7; CYP2A7; CPAD;
EESGFLIE
RQPKFED
DKGQFKK
1:2500



polypeptide 7

CYTOCHROME P450, SUBFAMILY
(212)
RTK (213)
SD (214)






IIA, POLYPEPTIDE 7; cytochrome









P450, subfamily IIA (phenobarbital-









inducible), polypeptide 7; cytochrome









P450, family 2, subfamily A,









polypeptide 7; cytochrome P450,









family 2, su









S0218
solute carrier family
222962
SLC29A4; solute carrier family 29
RHCILGEW
KQRELAG
RNAHGSC
1:20-



29 (nucleoside

(nucleoside transporters), member 4
LPILIMAVF
NTMTVSY
LHASTAN
1:50



transporters), member


N (215)
MS (216)
GSILAGL




4




(217)






S0221
solute carrier family
9153
HCNT2; SLC28A2; HsT17153;
ELMEKEVE
KARSFCK
KNKRLSG
1:500-



28 (sodium-coupled

SPNT1; CONCENTRATIVE
PEGSKRTD
THARLFK
MEEWIEG
1:1200



nucleoside

NUCLEOSIDE TRANSPORTER 2;
(218)
K (219)
EK (220)




transporter), member

SODIUM-DEPENDENT PURINE







2

NUCLEOSIDE TRANSPORTER 1;









solute carrier family 28 (sodium-









coupled nucleoside transporter),









member 2









S0223
angiopoietin-like 4
51129
HFARP; FIAF; ANGPTL4; PGAR;
EGSTDLPL
KVAQQQR
DHKHLDH
1:30-





angiopoietin-like 4; FASTING-
APESRVDP
HLEKQHL
EVAKPAR
1:10000





INDUCED ADIPOSE FACTOR;
E (221)
R (222)
RKRLPE






PPARG ANGIOPOIETIN-RELATED


(223)






PROTEIN; HEPATIC









FIBRINOGEN/ANGIOPOIETIN-









RELATED PROTEIN









S0235
carcinoembryonic
1048
CEACAM5; CD66e;
KLTIESTPF
KSDLVNE
KPVEDKD
1:500-



antigen-related cell

carcinoembryonic antigen-related cell
NVAEGKEC
EATGQFR
AVAFTCE
1:4500



adhesion molecule 5

adhesion molecule 5
(224)
VYPELPK
PEAQ (226)








(225)







S0237
podocalyxin-like
5420
podocalyxin-like; Gp200; PCLP;
DEKLISLIC
KDKWDEL
DSWIVPL
1:1000-





PODXL; PODOCALYXIN-LIKE
RAVKATFN
KEAGVSD
DNLTKDD
1:2000





PROTEIN; podocalyxin-like precursor
PAQDK
MKLGD
LDEEEDT







(227)
(228)
HL (229)






S0238
xenotropic and
9213
X3; XPR1; X RECEPTOR; SYG1,
EAVVTNEL
RRYRDTK
KARDTKV
1:100-



polytropic retrovirus

YEAST, HOMOLOG OF; xenotropic
EDGDRQKA
RAFPHLV
LIEDTDDE
1:500



receptor

and polytropic retrovirus receptor
MKRLR
NAGK
ANT (232)







(230)
(231)







S0241
glycyl-tRNA
2617
GlyRS; GARS; CMT2D; 6.1.1.14;
RKRVLEAK
RHGVSHK
EARYPLFE
1:500-



synthetase

SMAD1; GLYCYL-tRNA
ELALQPKD
VDDSSGSI
GQETGKK
1:7500





SYNTHETASE; glycine tRNA ligase;
DIVD (233)
GRRYAR
ETIEE






Charcot-Marie-Tooth neuropathy,

(234)
(235)






neuronal type, D









S0244
dachshund homolog 1
1602
DACH1; FLJ10138; dachshund
DLAGHDM
EKQVQLE
EADRSGG
1:100-



(Drosophila)

homolog (Drosophila);
GHESKRMH
KTELKMD
RTDAERTI
1:3000





DACHSHUND, DROSOPHILA,
IEKDE (236)
FLRERE
QDGR






HOMOLOG OF; dachshund homolog

(237)
(238)






1 (Drosophila); dachshund homolog 1









isoform a; dachshund homolog 1









isoform b; dachshund homolog 1









isoform c









S0251
transcription factor
29841
TFCP2L2; LBP-32; MGR; GRHL1;
EALYPQRR
DYYKVPR
DKYDVPH
1:5400



CP2-like 2

mammalian grainyhead; LBP protein
SYTSEDEA
ERRSSTA
DKIGKIFK






32; transcription factor CP2-like 2;
WK (239)
KPEVE
KCKK






leader-binding protein 32 isoform 2;

(240)
(241)






leader-binding protein 32 isoform 1









S0253
lysosomal associated
55353
LAPTM4B; lysosomal associated
DPDQYNFS
EYIRQLPP
DTTVLLPP
1:500-



protein

protein transmembrane 4 beta
SSELGGDF
NFPYRDD
YDDATVN
1:2000



transmembrane 4 beta


EFMDD
(243)
GAAKE







(242)

(244)






S0255
cyclin E2
9134
CYCE2; CCNE2; cyclin E2; G1/S-
RREEVTKK
KESRYVH
DFFDRFM
1:1000-





specific cyclin E2; cyclin E2 isoform
HQYEIR
DKHFEVL
LTQKDIN
1:2000





2; cyclin E2 isoform 3; cyclin E2
(245)
HSDLE
K (247)






isoform 1

(246)







S0260
nicastrin
23385
KIAA0253; nicastrin; NCSTN; APH2;
ESKHFTRD
ETDRLPR
ESRWKDI
1:2400-





ANTERIOR PHARYNX
LMEKLKGR
CVRSTAR
RARIFLIA
1:5400





DEFECTIVE 2, C. ELEGANS,
TSR (248)
LAR (249)
SKELE






HOMOLOG OF


(250)






S0265
FXYD domain
5349
MAT-8; MAT8; PLML; FXYD3;
KVTLGLLV
SEWRSSG
KCKCKFG
1:400-



containing ion

phospholemman-like protein;
FLAGFPVL
EQAGR
QKSGHHP
1:1200



transport regulator 3

MAMMARY TUMOR, 8-KD; FXYD
DANDLED
(252)
GE (253)






domain-containing ion transport
(251)








regulator 3; FXYD domain containing









ion transport regulator 3; FXYD









domain containing ion transport









regulator 3 isoform 2 precursor; FXYD









domai









S0267
immunoglobulin
3321
EWI-3; V8; IGSF3; immunoglobin
KVAKESDS
EREKTVT
KRAEDTA
1:200-



superfamily, member

superfamily, member 3;
VFVLKIYH
GEFIDKES
GQTALTV
1:250



3

immunoglobulin superfamily, member
LRQED
KRPK (255)
MRPD






3
(254)

(256)






S0270
signal transducing
10254
STAM2B; STAM2; DKFZp564C047;
KVARKVR
ETEVAAV
EIKKSEPE
1:1000-



adaptor molecule

Hbp; STAM2A; SIGNAL-
ALYDFEAV
DKLNVID
PVYIDED
1:9000



(SH3 domain and

TRANSDUCING ADAPTOR
EDNE (257)
DDVE
KMDR




ITAM motif) 2

MOLECULE 2; signal transducing

(258)
(259)






adaptor molecule 2; STAM-like









protein containing SH3 and ITAM









domains 2; signal transducing adaptor









molecule (SH3 domain and ITAM









motif) 2









S0273
dickkopf homolog 1
22943
DKK1; DKK-1; SK; dickkopf-1 like;
DEECGTDE
RGEIEETI
N/A
1:400-



(Xenopus laevis)

dickkopf (Xenopus laevis) homolog 1;
YCASPTRG
TESFGND

1:500





dickkopf homolog 1 (Xenopus laevis);
GD (260)
HSTLD







DICKKOPF, XENOPUS, HOMOLOG

(261)







OF, 1









S0280
solute carrier family
65010
SLC26A6; solute carrier family 26,
MDLRRRD
DTDIYRD
EFYSDAL
1:1800-



26, member 6

member 6
YHMERPLL
VAEYSEA
KQRCGVD
1:2400






NQEHLEE
KE (263)
VDFLISQK







(262)

KK (264)






S0286
WNT inhibitory factor
11197
WIF1; WIF-1; WNT inhibitory factor
DAHQARVL
ERRICECP
KRYEASLI
1:90



1

1; Wnt inhibitory factor-1 precursor
IGFEEDILIV
DGFHGPH
HALRPAG







SE (265)
CEK (266)
AQLR









(267)






S0288
preferentially
23532
MADE; PRAME; OPA-
KRKVDGLS
KEGACDE
DIKMILK
1:1200



expressed antigen in

INTERACTING PROTEIN 4; Opa-
TEAEQPFIP
LFSYLIEK
MVQLDSI




melanoma

interacting protein OIP4; preferentially
VE (268)
VKRKK
EDLE (270)






expressed antigen in melanoma;

(269)







melanoma antigen preferentially









expressed in tumors









S0295
prostaglandin E
9536
PGES; TP53112; MGST1L1; PP1294;
RLRKKAFA
RSDPDVE
RVAHTVA
1:100-



synthase

PP102; PTGES; MGC10317; PIG12;
NPEDALR
RCLRAHR
YLGKLRA
1:2400





MGST1-L1; MGST-IV; MGST1-like
(271)
ND (272)
PIR (273)






1; p53-INDUCED GENE 12;









prostaglandin E synthase; p53-induced









apoptosis protein 12; prostaglandin E









synthase isoform 2; prostaglandin E









synthase isoform 1; micros









S0296
solute carrier family 7
8140
SLC7A5; MPE16; D16S469E; CD98;
KRRALAAP
EAREKML
MIWLRHR
1:300-



(cationic amino acid

LAT1; 4F2 light chain; Membrane
AAEEKEEA
AAKSADG
KPELERPI
1:5000



transporter, y+

protein E16; L-TYPE AMINO ACID
R (274)
SAPAGE
K (276)




system), member 5

TRANSPORTER 1; Solute carrier

(275)







family 7, member 5; solute carrier









family 7 (cationic amino acid









transporter, y+ system), member 5









S0296
solute carrier family 7
8140
SLC7A5; MPE16; D16S469E; CD98;
KRRALAAP
N/A
N/A
1:225-


P1
(cationic amino acid

LAT1; 4F2 light chain; Membrane
AAEEKEEA


1:3150



transporter, y+

protein E16; L-TYPE AMINO ACID
R (277)






system), member 5

TRANSPORTER 1; Solute carrier









family 7, member 5; solute carrier









family 7 (cationic amino acid









transporter, y+ system), member 5









S0297
v-maf
7975
FLJ32205; NFE2U; MAFK; NFE2,
KPNKALKV
KRVTQKE
RLELDAL
1:333-



musculoaponeurotic

18-KD SUBUNIT; nuclear factor
KKEAGE
ELERQRV
RSKYE
1:800



fibrosarcoma

erythroid-2, ubiquitous (p18);
(278)
ELQQEVE
(280)




oncogene homolog K

NUCLEAR FACTOR ERYTHROID

K (279)





(avian)

2, UBIQUITOUS SUBUNIT; v-maf









musculoaponeurotic fibrosarcoma









oncogene homolog K (avian); v-maf









avian musculoaponeurotic









fibrosarcoma oncogen









S0301
signal peptide, CUB
57758
SCUBE2; signal peptide, CUB
KMHTDGRS
KKGFKLL
KRTEKRL
1:3500-



domain, EGF-like 2

domain, EGF-like 2
CLEREDTV
TDEKSCQ
RKAIRTLR
1:5400






LEVTE (281)
DVDE
KAVHRE








(282)
(283)






S0303
gamma-aminobutyric
2564
GABRE; GABA-A RECEPTOR,
RVEGPQTE
EETKSTET
KWENFKL
1:300-



acid (GABA) A

EPSILON POLYPEPTIDE; GAMMA-
SKNEASSR
ETGSRVG
EINEKNS
1:500



receptor, epsilon

AMINOBUTYRIC ACID
D (284)
KLPE (285)
WKLFQFD






RECEPTOR, EPSILON; gamma-


(286)






aminobutyric acid (GABA) A receptor,









epsilon; gamma-aminobutyric acid









(GABA) A receptor, epsilon isoform 2;









gamma-aminobutyric acid (GABA) A









receptor, epsilon is









S0305
S100 calcium binding
6281
CAL1L; GP11; p10; 42C; S100A10;
DKGYLTKE
KDPLAVD
N/A
1:8332-



protein A10 (annexin

ANX2LG; CLP11; Ca[1];
DLRVLMEK
KIMKDLD

1:24996



II ligand, calpactin I,

CALPACTIN I, p11 SUBUNIT;
E (287)
QCRDGK





light polypeptide

ANNEXIN II, LIGHT CHAIN;

(288)





(p 11))

CALPACTIN I, LIGHT CHAIN; S100









calcium-binding protein A10 (annexin









II ligand, calpactin I, light 









polypeptide (p11)); S100 calcium 









binding protein A10









S0311
v-myb myeloblastosis
4605
MYBL2; MGC15600; MYB-
MSRRTRCE
EEDLKEV
RRSPIKKV
1:750-



viral oncogene

RELATED GENE BMYB; MYB-
DLDELHYQ
LRSEAGIE
RKSLALDI
1:5000



homolog (avian)-like

related protein B; v-myb
DTDSD
LIIEDDIR
VDED




2

myeloblastosis viral oncogene
(289)
(290)
(291)






homolog (avian)-like 2; V-MYB









AVIAN MYELOBLASTOSIS VIRAL









ONCOGENE HOMOLOG-LIKE 2









S0312
nucleoside
4860
NP; 2.4.2.1; nucleoside phosphorylase;
EDYKNTAE
DERFGDR
KVIMDYE
1:1000-



phosphorylase

PURINE-
WLLSHTKH
FPAMSDA
SLEKANH
1:3600





NUCLEOSIDE:ORTHOPHOSPHATE
R (292)
YDRTMRQ
EE (294)






RIBOSYLTRANSFERASE; purine

R (293)







nucleoside phosphorylase; PNP









NUCLEOSIDE PHOSPHORYLASE









DEFICIENCY; ATAXIA WITH









DEFICIENT CELLULAR









IMMUNITY









S0314
chaperonin containing
22948
KIAA0098; CCT5; chaperonin
DQDRKSRL
KGVIVDK
RMILKIDD
1:6000-



TCP1, subunit 5

containing TCP1, subunit 5 (epsilon)
MGLEALKS
DFSHPQM
IRKPGESE
1:30000



(epsilon)


HIMAAK
PKKVED
E (297)







(295)
(296)







S0315
non-metastatic cells 1,
4830
GAAD; NME1; NDPKA; 2.7.4.6;
RLQPEFKP
KFMQASE
DSVESAE
1:9000-



protein (NM23A)

NM23-H1; AWD NM23H1B; GZMA-
KQLEGTMA
DLLKEHY
KEIGLWF
1:18000



expressed in

ACTIVATED DNase; NUCLEOSIDE
NCER (298)
VDLKDR
HPEELVD






DIPHOSPHATE KINASE-A; AWD,

(299)
(300)






DROSOPHILA, HOMOLOG OF;









METASTASIS INHIBITION









FACTOR NM23; nucleoside-









diphosphate kinase 1 isoform b;









NONMETASTATIC PROTEIN 23,









HOMOLOG 1; nucleo









S0316
squalene epoxidase
6713
SQLE; 1.14.99.7; squalene epoxidase;
KSPPESENK
RDGRKVT
DHLKEPF
1:1000-





squalene monooxygenase
EQLEARRR
VIERDLKE
LEATDNS
1:10000






R (301)
PDR (302)
HLR (303)






S0319
pregnancy-induced
29948
OKL38; pregnancy-induced growth
DLEVKDW
EYHKVHQ
RHQLLCF
1:900



growth inhibitor

inhibitor; PREGNANCY-INDUCED
MQKKRRG
MMREQSI
KEDCQAV






GROWTH INHIBITOR OKL38
LRNSR (304)
LSPSPYEG
FQDLEGV








YR (305)
EK (306)






S0326
mal, T-cell
114569
MAL2; mal, T-cell differentiation
GPDILRTYS
CSLGLAL
N/A
1:120-



differentiation protein 2

protein 2
GAFVCLE
RRWRP

1:1200






(307)
(308)







S0330
aldo-keto reductase
1645
1.1.1.213; 2-ALPHA-HSD; 1.3.1.20;
RYLTLDIFA
N/A
N/A
1:2500-



family 1, member

20-ALPHA-HSD; MGC8954; H-37;
GPPNYPFS


1:75000



C1/2 (dihydrodiol

HAKRC; MBAB; C9; DDH1;
DEY (309)






dehydrogenase 1; 20-

AKR1C1; trans-1,2-dihydrobenzene-







alpha (3-alpha)-

1,2-diol dehydrogenase; chlordecone







hydroxysteroid

reductase homolog; aldo-keto







dehydrogenase)

reductase C; 20 alpha-hydroxysteroid









dehydrogenase; hepatic dihydrodiol









S0330-
aldo-keto reductase
1645
1.1.1.213; 2-ALPHA-HSD; 1.3.1.20;
RYLTLDIFA
N/A
N/A
1:600


x1
family 1, member

20-ALPHA-HSD; MGC8954; H-37;
GPPNYPFS






C1/2 (dihydrodiol

HAKRC; MBAB; C9; DDH1;
DEY (310)






dehydrogenase 1; 20-

AKR1C1; trans-1,2-dihydrobenzene-







alpha (3-alpha)-

1,2-diol dehydrogenase; chlordecone







hydroxysteroid

reductase homolog; aldo-keto







dehydrogenase)

reductase C; 20 alpha-hydroxysteroid









dehydrogenase; hepatic dihydrodiol









S0331
aldo-keto reductase
8644
HA1753; 1.1.1.188; DD3; hluPGFS;
HYFNSDSF
N/A
N/A
1:300-



family 1, member C3

HSD17B5; 1.3.1.20; 1.1.1.213;
ASHPNYPY


1:999



(3-alpha

AKR1C3; KIAA0119; HAKRB;
SDEY (311)






hydroxysteroid

HAKRe; trans-1,2-dihydrobenzene-







dehydrogenase, type

1,2-diol dehydrogenase; chlordecone







II)

reductase homolog; dihydrodiol









dehydrogenase 3; prostaglandin F









synthase; ALDO-KETO









REDUCTASE B; 3-









S0331-
aldo-keto reductase
8644
HA1753; 1.1.1.188; DD3; hluPGFS;
HYFNSDSF
N/A
N/A
1:150-


x1
family 1, member C3

HSD17B5; 1.3.1.20; 1.1.1.213;
ASHPNYPY


1:300



(3-alpha

AKR1C3; KIAA0119; HAKRB;
SDEY (312)






hydroxysteroid

HAKRe; trans-1,2-dihydrobenzene-







dehydrogenase, type

1,2-diol dehydrogenase; chlordecone







II)

reductase homolog; dihydrodiol









dehydrogenase 3; prostaglandin F









synthase; ALDO-KETO









REDUCTASE B; 3-









S0332
aldo-keto reductase
1645
1.1.1.213; 2-ALPHA-HSD; 1.3.1.20;
RYVVMDFL
N/A
N/A
1:300-



family 1, member C4

20-ALPHA-HSD; MGC8954; H-37;
MDHPDYPF


1:400



(dihydrodiol

HAKRC; MBAB; C9; DDH1;
SDEY (313)






dehydrogenase 1; 20-

AKR1C1; trans-1,2-dihydrobenzene-







alpha (3-alpha)-

1,2-diol dehydrogenase; chlordecone







hydroxysteroid

reductase homolog; aldo-keto







dehydrogenase)

reductase C; 20 alpha-hydroxysteroid









dehydrogenase; hepatic dihydrodiol









S0332-
aldo-keto reductase
1645
1.1.1.213; 2-ALPHA-HSD; 1.3.1.20;
RYVVMDFL
N/A
N/A
1:75-


x1
family 1, member C4

20-ALPHA-HSD; MGC8954; H-37;
MDHPDYPF


1:150



(dihydrodiol

HAKRC; MBAB; C9; DDH1;
SDEY (314)






dehydrogenase 1; 20-

AKR1C1; trans-1,2-dihydrobenzene-







alpha (3-alpha)-

1,2-diol dehydrogenase; chlordecone







hydroxysteroid

reductase homolog; aldo-keto







dehydrogenase)

reductase C; 20 alpha-hydroxysteroid









dehydrogenase; hepatic dihydrodiol









S0336
chromosome 20 open
140809
C20orf139; chromosome 20 open
DPAKVQSL
RETIPAKL
N/A
1:1600-



reading frame 139

reading frame 139
VDTIREDP
VQSTLSD

1:2400






D (315)
LR (316)







S0342
solute carrier family
2154091
SLC2A12; solute carrier family 2
SDTTEELT
N/A
N/A
1:400-



(facilitated glucose

(facilitated glucose transporter),
VIKSSLKDE


1:1250



transporter), member 12

member 12
(317)








S0343
solute carrier family
2154091
SLC2A12; solute carrier family 2
HSRSSLMP
N/A
N/A
1:50-



(facilitated glucose

(facilitated glucose transporter),
LRNDVDKR


1:125



transporter), member 12

member 12
(318)








S0357
HTPAP protein
84513
HTPAP; HTPAP protein
YRNPYVEA
N/A
N/A
1:100-






EYFPTKPM


1:300






FVIA (319)








S0364
KIAA0746 protein
23231
KIAA0746; KIAA0746 protein
KKFPRFRN
N/A
N/A
1:200-






RELEATRR


1:300






QRMD (320)








S0367
peroxisomal acyl-CoA
122970
PTE2B; peroxisomal acyl-CoA
SGNTAINY
N/A
N/A
1:200-



thioesterase 2B

thioesterase 2B
KHSSIP


1:600






(321)








S0374
chloride intracellular
53405
CLIC5; chloride intracellular channel 5
DANTCGED
N/A
N/A
1:5000-



channel 5


KGSRRKFL


1:9000






DGDE (322)








S0380
keratinocyte
200634
KRTCAP3; keratinocyte associated
QLEEMTEL
N/A
N/A
1:2000-



associated protein 3

protein 3
ESPKCKRQ


1:9000






ENEQ (323)








S0384
FERM, RhoGEF
10160
p63RhoGEF; CDEP; FARP1;
QADGAASA
N/A
N/A
1:100



(ARHGEF) and

chondrocyte-derived ezrin-like protein;
PTEEEEEV






pleckstrin domain

FERM, RhoGEF, and pleckstrin
VKDR (324)






protein 1

domain protein 1; FERM, ARHGEF,







(chondrocyte-derived)

AND PLECKSTRIN DOMAIN-









CONTAINING PROTEIN 1; FERM,









RhoGEF (ARHGEF) and pleckstrin









domain protein 1 (chondrocyte-









derived)









S0388
trichorhinophalangeal
7227
GC79; TRPS1; TRPS1 GENE;
SGDSLETK
N/A
N/A
1:600



syndrome I

trichorhinophalangeal syndrome I; zinc
EDQKMSPK








finger transcription factor TRPS1
ATEE (325)








S0396
cytochrome P450,
1576
1.14.14.1; HLP; CYP3A3; CYP3A4;
RKSVKRM
N/A
N/A
1:15



family 3, subfamily A,

P450C3; NF-25; CP33; CP34; P450-
KESRLEDT






polypeptide 4

III, STEROID-INDUCIBLE;
QKHRV








nifedipine oxidase; glucocorticoid-
(326)








inducible P450; CYTOCHROME









P450PCN1; P450, FAMILY III; P450-









III, steroid inducible; cytochrome









P450, subfamily IIIA, polypeptide 4;









S0398
FAT tumor suppressor
2195
CDHF7; FAT; cadherin ME5; FAT
KIRLPEREK
N/A
N/A
1:45-



homolog 1

tumor suppressor precursor; cadherin-
PDRERNAR


1:200



(Drosophila)

related tumor suppressor homolog
REP (327)








precursor; cadherin family member 7









precursor; homolog of Drosophila Fat









protein precursor; FAT tumor









suppressor homolog 1 (Drosophila);









FAT TUMOR SUPPRESS









S0401
granulin
2896
ACROGRANIN; PROEPITHELIN;
RGTKCLRR
N/A
N/A
1:600-





PROGRANULIN; PEPI; PCDGF;
EAPRWDAP


1:3000





granulin; GRN; EPITHELIN
LRDP (328)








PRECURSOR









S0404
N-myc downstream
10397
HMSNL; TARG1; CMT4D; RTP;
GTRSRSHT
N/A
N/A
1:100-



regulated gene 1

PROXY1; NDRG1; GC4; NMSL;
SEGTRSRS


1:900





TDD5; RIT42; NDR1; differentiation-
HTSE (329)








related gene 1 protein; nickel-specific









induction protein Cap43; protein









regulated by oxygen-1; NMYC









DOWNSTREAM-REGULATED









GENE 1; reducing agents and









tunicamycin-respon









S0411
fatty acid binding
2171
PAFABP; EFABP; E-FABP; FABP5;
EETTADGR
N/A
N/A
1:1800



protein 5 (psoriasis-

PA-FABP; FATTY ACID-BINDING
KTQTVCNF






associated)

PROTEIN, EPIDERMAL; FATTY
TD (330)








ACID-BINDING PROTEIN 5;









FATTY ACID-BINDING PROTEIN,









PSORIASIS-ASSOCIATED; fatty









acid binding protein 5 (psoriasis-









associated)









S0413
cyclin-dependent
1028
WBS; p57(KIP2); BWCR; CDKN1C;
AKRKRSAP
N/A
N/A
1:2700



kinase inhibitor 1C

BWS; Beckwith-Wiedemann
EKSSGDVP






(p57, Kip2)

syndrome; cyclin-dependent kinase
(331)








inhibitor 1C (p57, Kip2)









S0414
alpha-methylacyl-
23600
AMACR; 5.1.99.4; ALPHA-
RVDRPGSR
N/A
N/A
1:100



CoA racemase

METHYLACYL-CoA RACEMASE;
YDVSRLGR








AMACR DEFICIENCY; AMACR
GKRS (332)








ALPHA-METHYLACYL-CoA









RACEMASE DEFICIENCY; alpha-









methylacyl-CoA racemase isoform 1;









alpha-methylacyl-CoA racemase









isoform 2









S0415
gamma-aminobutyric
2562
MGC9051; GABRB3; GABA-A
ETVDKLLK
N/A
N/A
1:600-



acid (GABA) A

RECEPTOR, BETA-3
GYDIRLRP


1:1800



receptor, beta 3

POLYPEPTIDE; GAMMA-
D (333)








AMINOBUTYRIC ACID









RECEPTOR, BETA-3; gamma-









aminobutyric acid (GABA) A receptor,









beta 3; gamma-aminobutyric acid









(GABA) A receptor, beta 3 isoform 2









precursor; gamma-aminobutyric acid









(GABA) A rece









S0417
HSV-1 stimulation-
22879
HSRG1; KIAA0872; HSV-1
APGGAEDL
N/A
N/A
1:9000



related gene 1

stimulation-related 1; HSV-1
EDTQFPSEE








stimulation-related gene 1
ARE (334)








S0425
tumor necrosis factor
27242
TNFRSF21; DR6; BM-018; TNFR-
RKSSRTLK
N/A
N/A
1:9000



receptor superfamily,

related death receptor 6; tumor
KGPRQDPS






member 21

necrosis factor receptor superfamily,
AIVE (335)








member 21; tumor necrosis factor









receptor superfamily, member 21









precursor









S0429
jumonji domain
221037
JMJD1C; TRIP8; jumonji domain
GSESGDSD
N/A
N/A
1:1200



containing 1C

containing 1C; THYROID
ESESKSEQR








HORMONE RECEPTOR
TKR (336)








INTERACTOR 8









S0432
chromosome 9 open
null
C9orf140; chromosome 9 open reading
EADSGDAR
N/A
N/A
1:90-



reading frame 140

frame 140
RLPRARGE


1:300






RRRH (337)








S0440
cell division cycle
994
3.1.3.48; CDC25B; cell division cycle
RLERPQDR
N/A
N/A
1:350-



25B

25B; cell division cycle 25B isoform 4;
DTPVQNKR


1:3600





cell division cycle 25B isoform 5; cell
RRS (338)








division cycle 25B isoform 1; cell









division cycle 25B isoform 2; cell









division cycle 25B isoform 3









S0445
laminin, beta 1
3912
LAMB1; LAMININ, BETA-1; CUTIS
DRVEDVM
N/A
N/A
1:600-





LAXA-MARFANOID SYNDROME;
MERESQFK


1:1800





laminin, beta 1; laminin, beta 1
EKQE (339)








precursor; LAMB1 NEONATAL









CUTIS LAXA WITH MARFANOID









PHENOTYPE









S0447
papillary renal cell
5546
TPRC; MGC17178; MGC4723;
DEAFKRLQ
N/A
N/A
1:4000-



carcinoma

PRCC; proline-rich protein PRCC;
GKRNRGRE


1:6000



(translocation-

RCCP1 PRCC/TFE3 FUSION GENE;
E (340)






associated)

papillary renal cell carcinoma









(translocation-associated); RENAL









CELL CARCINOMA, PAPILLARY,









1 GENE; papillary renal cell









carcinoma translocation-associated









gene product









S0455
tumor necrosis factor
8743
APO2L; TL2; Apo-2L; TNFSF10;
RFQEEIKEN
N/A
N/A
1:900



(ligand) superfamily,

Apo-2 ligand; APO2 LIGAND; TNF-
TKNDKQ






member 10

RELATED APOPTOSIS-INDUCING
(341)








LIGAND; TNF-related apoptosis









inducing ligand TRAIL; tumor









necrosis factor (ligand) superfamily,









member 10; TUMOR NECROSIS









FACTOR LIGAND SUPERFAMILY,









MEMBER 10









S0459
titin
7273
connectin; TMD; titin; CMD1G;
KRDKEGVR
N/A
N/A
1:2700-





CMPD4; TTN; FLJ32040; CMH9,
WTKCNKK


1:8100





included; titin isoform N2-A; titin
TLTD (342)








isoform N2-B; titin isoform novex-1;









titin isoform novex-2; titin isoform









novex-3; cardiomyopathy, dilated 1G









(autosomal dominant); TTN









CARDIOMYOPATHY, FAMILIAL









S0469
DNA fragmentation
1676
DFF45; DFF1; DFFA; ICAD; DFF-45;
KEGSLLSK
N/A
N/A
1:600



factor, 45 kDa, alpha

INHIBITOR OF CASPASE-
QEESKAAF






polypeptide

ACTIVATED DNase; DNA
GEE (343)








FRAGMENTATION FACTOR, 45-









KD, ALPHA SUBUNIT; DNA









fragmentation factor, 45 kDa, alpha









polypeptide; DNA fragmentation









factor, 45 kD, alpha subunit; DNA









fragmentation factor, 45 kD, alp









S0494
caspase 2, apoptosis-
835
ICH-1L/1S; CASP2; ICH1; CASP-2;
ESDAGKEK
N/A
N/A
1:2000



related cysteine

ICH-1 protease; caspase 2 isoform 3;
LPKMRLPT






protease (neural

caspase 2 isoform 4; NEDD2 apoptosis
RSD (344)






precursor cell

regulatory gene; caspase 2 isoform 2







expressed,

precursor; caspase 2 isoform 1







developmentally

preproprotein; NEURAL







down-regulated 2)

PRECURSOR CELL EXPRESSED,









DEVELOPMENTALLY









DOWNREGULATED 2;









S0501
G1 to S phase
2935
GSPT1; eRF3a; ETF3A; GST1,
ERDKGKTV
N/A
N/A
1:15000



transition 1

YEAST, HOMOLOG OF; PEPTIDE
EVGRAYFE








CHAIN RELEASE FACTOR 3A; G1-
TEK (345)








TO S-PHASE TRANSITION 1; G1 to









S phase transition 1









S0502
GCN5 general control
2648
hGCN5; GCN5L2; GCN5 (general
EKFRVEKD
N/A
N/A
1:9000



of amino-acid

control of amino-acid synthesis, yeast,
KLVPEKR






synthesis 5-like 2

homolog)-like 2; GCN5 general
(346)






(yeast)

control of amino-acid synthesis 5-like









2 (yeast); General control of amino









acid synthesis, yeast, homolog-like 2









S0503
geminin, DNA
51053
GMNN; geminin, DNA replication
EVAEKRRK
N/A
N/A
1:333



replication inhibitor

inhibitor
ALYEALKE









NEK (347)








S0507
ADP-ribosylation
64225
ARL6IP2; ADP-ribosylation factor-
ENYEDDDL
N/A
N/A
1:8000-



factor-like 6

like 6 interacting protein 2
VNSDEVM


1:9000



interacting protein 2


KKP (348)








50511
DNA replication
51659
Pfs2; DNA replication complex GINS
PKADEIRTL
N/A
N/A
1:2000



complex GINS

protein PSF2
VKDMWDT






protein PSF2


R (349)








S0524
ankyrin repeat domain 10
55608
ANKRD10; ankyrin repeat domain 10
RKRCLEDS
N/A
N/A
1:4500






EDFGVKKA









RTE (350)








S0527
potassium channel
null
KCTD2; potassium channel
EPKSFLCRL
N/A
N/A
1:900-



tetramerisation

tetramerisation domain containing 2
CCQEDPEL


1:1500



domain containing 2


DS (351)








S0528
rabconnectin-3
23312
RC3; KIAA0856; rabconnectin-3
EEYDRESK
N/A
N/A
1:350-






SSDDVDYR


1:1200






GS (352)








S0538
acidic (leucine-rich)
81611
ANP32E; acidic (leucine-rich) nuclear
CVNGEIEG
N/A
N/A
1:1200



nuclear

phosphoprotein 32 family, member E
LNDTFKEL






phosphoprotein 32


EF (353)






family, member E











S0544
chromosome 9 open
84904
C9orf100; chromosome 9 open reading
EQRARWER
N/A
N/A
1:40-



reading frame 100

frame 100
KRACTARE


1:240






(354)








S0545
HpaII tiny fragments
27037
D22S1733E; HTF9C; HpaII tiny
ERKQLECE
N/A
N/A
1:900-



locus 9C

fragments locus 9C; HpaII tiny
QVLQKLAK


1:5400





fragments locus 9C isoform2; HpaII
E (355)








tiny fragments locus 9C isoform 1









S0546
cell division cycle
157313
CDCA2; cell division cycle associated 2
RNSETKVR
N/A
N/A
1:1200



associated 2


RSTRLQKD









LEN (356)








S0553
mitotic
129401
MP44; NUP35; LOC129401;
SDYQVISD
N/A
N/A
1:3000-



phosphoprotein 44

NUCLEOPORIN, 35-KD; mitotic
RQTPKKDE


1:5400





phosphoprotein 44
(357)








S0557
SMC4 structural
10051
SMC4L1; CAPC; hCAP-C;
DIEGKLPQT
N/A
N/A
1:200



maintenance of

chromosome-associated polypeptide C;
EQELKE






chromosomes 4-like 1

SMC4 (structural maintenance of
(358)






(yeast)

chromosomes 4, yeast)-like 1; SMC4









structural maintenance of









chromosomes 4-like 1 (yeast);









structural maintenance of









chromosomes (SMC) family member,









chromosome-ass









S0564
phosphatidylserine
9791
KIAA0024; PSSA; PTDSS1;
DDVNYKM
N/A
N/A
1:1000-



synthase 1

phosphatidylserine synthase 1
HFRMINEQ


1:8000






QVED (359)








S0565
polo-like kinase 1
5347
2.7.1.-; PLK1; STPK13; polo-like
ENPLPERPR
N/A
N/A
1:10-



(Drosophila)

kinase (Drosophila); polo (Drosophia)-
EKEEPVVR


1:100





like kinase; SERINE/THREONINE
(360)








PROTEIN KINASE 13; polo-like









kinase 1 (Drosophila)









S0567
Pirin
8544
Pirin; PIR
REQSEGVG
N/A
N/A
1:240






ARVRRSIG









RPE (361)








S0578
ATP-binding cassette,
21
ABCA3; ABC3; LBM180; ABC-C;
PRAVAGKE
N/A
N/A
1:1500



sub-family A (ABC1),

EST111653; ABC transporter 3; ATP- 
EEDSDPEK






member 3

binding cassette 3; ATP-BINDING
ALR (362)








CASSETTE TRANSPORTER 3;









ATP-BINDING CASSETTE,









SUBFAMILY A, MEMBER 3; ATP-









binding cassette, sub-family A member









3; ATP-binding cassette, sub-family A









(ABC1), memb









S0579
ATP-binding cassette,
10347
ABCX; ABCA7; ABCA-SSN;
EKADTDME
N/A
N/A
1:300-



sub-family A (ABC1),

autoantigen SS-N; macrophage ABC
GSVDTRQE


1:400



member 7

transporter; SJOGREN SYNDROME
K (363)








ANTIGEN SS-N; ATP-BINDING









CASSETTE, SUBFAMILY A,









MEMBER 7; ATP-binding cassette,









sub-family A (ABC1), member 7;









ATP-binding cassette, sub-family A,









member 7 isoform a; A









S0581
ATP-binding cassette,
22
ABCB7; Atm1p; ASAT; ABC7;
RVQNHDNP
N/A
N/A
1:4000-



sub-family B

EST140535; ABC TRANSPORTER 7;
KWEAKKE


1:10000



(MDR/TAP), member

ATP-binding cassette 7; ATP-
NISK (364)






7

BINDING CASSETTE









TRANSPORTER 7; Anemia,









sideroblastic, with spinocerebellar









ataxia; ATP-BINDING CASSETTE,









SUBFAMILY B, MEMBER 7; ATP-









binding cassette, sub-family B,









member









S0585
ATP-binding cassette,
94160
MRP9; ABCC12; MULTIDRUG
RSPPAKGA
N/A
N/A
1:500



sub-family C

RESISTANCE-ASSOCIATED
TGPEEQSD






(CFTR/MRP),

PROTEIN 9; ATP-BINDING
SLK (365)






member 12

CASSETTE, SUBFAMILY C,









MEMBER 12; ATP-binding cassette,









sub-family C (CFTR/MRP), member









12









S0586
ATP-binding cassette,
9429
ABC15; MXR1; ABCP; EST157481;
REEDFKAT
N/A
N/A
1:333-



sub-family G

MRX; ABCG2; BCRP1; BMDP;
EIIEPSKQD


1:400



(WHITE), member 2

MITOXANTRONE-RESISTANCE
KP (366)








PROTEIN; mitoxantrone resistance









protein; placenta specific MDR









protein; ATP-BINDING CASSETTE









TRANSPORTER, PLACENTA-









SPECIFIC; breast cancer resistance









protein; ATP-BINDING CASS









S0593
solute carrier organic
28234
OATP1B3; SLC21A8; OATP8;
DKTCMKW
N/A
N/A
1:500-



anion transporter

SLCO1B3; ORGANIC ANION
STNSCGAQ


1:2400



family, member 1B3

TRANSPORTER 8; solute carrier
(367)








organic anion transporter family,









member 1B3; SOLUTE CARRIER









FAMILY 21, MEMBER 8









(ORGANIC ANION









TRANSPORTER); solute carrier









family 21 (organic anion transporter),









member 8









S0597
solute carrier family
9356
ROAT1; MGC45260; HOAT1; PAHT;
DANLSKNG
N/A
N/A
1:3000



22 (organic anion

SLC22A6; PAH TRANSPORTER;
GLEVWL






transporter), member

para-aminohippurate transporter; renal
(368)






6

organic anion transporter 1; solute









carrier family 22 member 6 isoform b;









solute carrier family 22 member 6









isoform c; solute carrier family 22









member 6 isoform









S0604
solute carrier family
7355
UGT2; UGTL; UGAT; SLC35A2;
EPFLPKLLT
N/A
N/A
1:2400



35 (UDP-galactose

UGT1; UDP-galactose translocator;
K (369)






transporter), member

UDP-GALACTOSE







A2

TRANSPORTER, ISOFORM 2;









UGALT UDP-GALACTOSE









TRANSPORTER, ISOFORM 1; solute









carrier family 35 (UDP-galactose









transporter), member A2; solute carrier









family 35 (UDP-galactose transpo









S0607
cell division cycle
994
3.1.3.48; CDC25B; cell division cycle
RKSEAGSG
N/A
N/A
1:1800



25B

25B; cell division cycle 25B isoform 4;
AASSSGED








cell division cycle 25B isoform 5; cell
KEN (370)








division cycle 25B isoform 1; cell









division cycle 25B isoform 2; cell









division cycle 25B isoform 3









S0609
stearoyl-CoA
6319
SCD; acyl-CoA desaturase; stearoyl-
DDIYDPTY
N/A
N/A
1:2000-



desaturase (delta-9-

CoA desaturase (delta-9-desaturase);
KDKEGPSP


1:5000



desaturase)

fatty acid desaturase
KVE (371)








S0611
mitogen-activated
6300
SAPK3; p38gamma; SAPK-3; p38-
QSDEAKNN
N/A
N/A
1:100



protein kinase 12

GAMMA; PRKM12; MAPK12;
MKGLPELE








ERK3; ERK6; EXTRACELLULAR
KKD (372)








SIGNAL-REGULATED KINASE 6;









mitogen-activated protein kinase 3;









stress-activated protein kinase 3;









mitogen-activated protein kinase 12









S0612
nuclear factor of
4791
LYT-10; LYT10; NFKB2;
SRPQGLTE
N/A
N/A
1:4500



kappa light

ONCOGENE LYT 10;
AEQRELEQ






polypeptide gene

TRANSCRIPTION FACTOR NFKB2;
EAK (373)






enhancer in B-cells 2

NFKB, p52/p100 SUBUNIT;







(p49/p100)

LYMPHOCYTE TRANSLOCATION









CHROMOSOME 10; NUCLEAR









FACTOR KAPPA-B, SUBUNIT 2;









Nuclear factor of kappa light chain









gene enhancer in B-cells 2; nuclear









factor of kappa 1









S0613
tumor necrosis factor
958
Bp50; TNFRSF5; MGC9013;
RVQQKGTS
N/A
N/A
1:250-



receptor superfamily,

CDW40; CD40 antigen; CD40L
ETDTIC


1:270



member 5

receptor; B CELL-ASSOCIATED
(374)








MOLECULE CD40; CD40 type II









isoform; B cell surface antigen CD40;









nerve growth factor receptor-related B-









lymphocyte activation molecule; tumor









necrosis factor receptor superfam









S0614
Epstein-Barr virus
10148
EBI3; IL27, EBI3 SUBUNIT;
VRLSPLAE
N/A
N/A
1:1200-



induced gene 3

EPSTEIN-BARR VIRUS-INDUCED
RQLQVQW


1:3000





GENE 3; INTERLEUKIN 27, EBI3
E (375)








SUBUNIT; Epstein-Barr virus induced









gene 3; Epstein-Barr virus induced









gene 3 precursor









S0616
zinc finger protein
58495
ZNF339; zinc finger protein 339
RRSLGVSV
N/A
N/A
1:2500



339


RSWDELPD









EKR (376)








S0617
DAB2 interacting
153090
DAB2IP; DAB2 interacting protein
DEGLGPDP
N/A
N/A
1:600



protein


PHRDRLRS









K (377)








S0618
protein tyrosine
8500
MGC26800; LIP1; PPFIA1; LIP.1;
SGKRSSDG
N/A
N/A
1:150



phosphatase, receptor

LAR-interacting protein 1; PTPRF
SLSHEEDL






type, f polypeptide

interacting protein alpha 1 isoform a;
AK (378)






(PTPRF), interacting

PTPRF interacting protein alpha 1







protein (liprin), alpha

isoform b; protein tyrosine







1

phosphatase, receptor type, f









polypeptide (PTPRF), interacting









protein (liprin), alpha 1









S0631
RGM domain family,
56963
RGMA; REPULSIVE GUIDANCE
SQERSDSPE
N/A
N/A
1:600



member A

MOLECULE; RGM domain family,
ICHYEKSFH








member A
K (379)








S0633
hypothetical protein
144347
LOC144347; hypothetical protein
KVNPEPTH
N/A
N/A
1:100-



LOC144347

LOC144347
EIRCNSEVK


1:200






(380)








S0639
tetratricopeptide
57217
TTC7; tetratricopeptide repeat domain 7
RELREVLR
N/A
N/A
1:2000-



repeat domain 7


TVETKATQ


1:3000






N (381)








S0640
protein C (inactivator
5624
PROC; 3.4.21.69; PROC
RDTEDQED
N/A
N/A
1:1000-



of coagulation factors

DEFICIENCY PROTEIN C;
QVDPRLID


1:1800



Va and VIIIa)

THROMBOPHILIA, HEREDITARY,
GK (382)








DUE TO PC DEFICIENCY;









PROTEIN C DEFICIENCY,









CONGENITAL THROMBOTIC









DISEASE DUE TO; protein C









(inactivator of coagulation factors Va









and VIIIa)









S0643
transducin-like
7090
HsT18976; KIAA1547; ESG3; TLE3;
KNHHELDH
N/A
N/A
1:200-



enhancer of split 3

transducin-like enhancer protein 3;
RERESSAN


1:1440



(E(sp1) homolog,

enhancer of split groucho 3;
(383)






Drosophila)

transducin-like enhancer of split 3









(E(sp1) homolog, Drosophila)









S0645
frizzled homolog 7
8324
FzE3; FZD7; frizzled 7; frizzled
SDGRGRPA
N/A
N/A
1:900



(Drosophila)

homolog 7 (Drosophila); Frizzled,
FPFSCPRQ








drosophila, homolog of, 7
(384)








S0646
solute carrier family 3
6520
MDU1; 4T2HC; SLC3A2; NACAE;
GSKEDFDS
N/A
N/A
1:3600-



(activators of dibasic

4F2HC; 4F2 HEAVY CHAIN; CD98
LLQSAKK


1:5400



and neutral amino

HEAVY CHAIN; CD98
(385)






acid transport),

MONOCLONAL ANTIBODY 44D7;







member 2

ANTIGEN DEFINED BY









MONOCLONAL ANTIBODY 4F2,









HEAVY CHAIN; antigen identified by









monoclonal antibodies 4F2, TRA1.10,









TROP4, and T43; SOLUTE









CARRIER FAMILY 3









S0648
KIAA0738 gene
9747
KIAA0738; KIAA0738 gene product
EYRNQTNL
N/A
N/A
1:200



product


PTENVDK









(386)








S0651
phospholipase A2
22925
PLA2IR; PLA2-R; PLA2R1;
QKEEKTW
N/A
N/A
1:3600



receptor 1, 180 kDa

PLA2G1R; PHOSPHOLIPASE A2
HEALRSCQ








RECEPTOR, 180-KD; phospholipase
ADN (387)








A2 receptor 1, 180 kDa









S0654
KIAA0182 protein
23199
KIAA0182; KIAA0182 protein
EKAEEGPR
N/A
N/A
1:400






KREPAPLD









K (388)








S0659
thymidine kinase 2,
7084
TK2; THYMIDINE KINASE,
EQNRDRIL
N/A
N/A
1:300



mitochondrial

MITOCHONDRIAL; thymidine
TPENRK








kinase 2, mitochondrial
(389)








S0663
chromosome 14 open
64430
C14orf135; chromosome 14 open
RDWYIGLV
N/A
N/A
1:900



reading frame 135

reading frame 135
SDEKWK









(390)








S0665
KIAA1007 protein
23019
KIAA1007; KIAA1007 protein;
DSYLKTRS
N/A
N/A
1:1500-





adrenal gland protein AD-005;
PVTFLSDLR


1:3000





KIAA1007 protein isoform a;
(391)








KIAA1007 protein isoform b









S0670
DKFZP56601646
25936
DC8; DKFZP56601646 protein
KCRGETVA
N/A
N/A
1:900



protein


KEISEAMK









S (392)








S0672
B-cell
605
BCL7A; B-cell CLL/lymphoma-7; B-
QRGSQIGR
N/A
N/A
1:800



CLL/lymphoma 7A

cell CLL/lymphoma 7A
EPIGLSGD









(393)








S0673
likely ortholog of
28987
ART-4; NOB1P; adenocarcinoma
KPPQETEK
N/A
N/A
1:50



mouse nin one

antigen recognized by T lymphocytes
GHSACEPE






binding protein

4; likely ortholog of mouse nin one
N (394)








binding protein









S0676
guanine nucleotide
2768
RMP; NNX3; GNA12; GUANINE
ERRAGSGA
N/A
N/A
1:1200-



binding protein (G

NUCLEOTIDE-BINDING PROTEIN,
RDAERE


1:2400



protein) alpha 12

ALPHA-12; guanine nucleotide
(395)








binding protein (G protein) alpha 12









S0677
GrpE-like 1,
80273
HMGE; GRPEL1; HUMAN
SEQKADPP
N/A
N/A
1:500-



mitochondrial (E.

MITOCHONDRIAL GrpE PROTEIN;
ATEKTLLE


1:1000




coli)


GrpE-like 1, mitochondrial (E. coli);
(396)








GrpE, E. COLI, HOMOLOG OF, 1









S0684
hypothetical protein
91607
FLJ34922; hypothetical protein
EAEWSQGV
N/A
N/A
1:8100



FLJ34922

FLJ34922
QGTLRIKK









YLT (397)








S0687
hypothetical protein
54942
FLJ20457; hypothetical protein
EESKSITEG
N/A
N/A
1:600-



FLJ20457

FLJ20457
LLTQKQYE


1:1260






(398)








S0691
solute carrier family
23657
CCBR1; SLC7A11; xCT;
QNFKDAFS
N/A
N/A
1:1000-



7, (cationic amino

cystine/glutamate transporter;
GRDSSITR


1:1575



acid transporter, y+

SYSTEM Xc(-) TRANSPORTER-
(399)






system) member 11

RELATED PROTEIN; SOLUTE









CARRIER FAMILY 7, MEMBER 11;









solute carrier family 7, (cationic amino









acid transporter, y+ system) member









11









S0692
glutamate-cysteine
2729
GLCLC; GCLC; 6.3.2.2; GCS;
EKIHLDDA
N/A
N/A
1:100-



ligase, catalytic

GAMMA-GLUTAMYLCYSTEINE
NESDHFEN


1:400



subunit

SYNTHETASE, CATALYTIC
(400)








SUBUNIT; glutamate-cysteine ligase,









catalytic subunit









S0695
integrin, beta 4
3691
ITGB4; INTEGRIN, BETA-4;
TEDVDEFR
N/A
N/A
1:2700-





integrin, beta 4
NKLQGER


1:4050






(401)








S0702
solute carrier family 7
8140
SLC7A5; MPE16; D16S469E; CD98;
KGDVSNLD
N/A
N/A
1:21160-



(cationic amino acid

LAT1; 4F2 light chain; Membrane
PNFSFEGTK


1:178200



transporter, y+

protein E16; L-TYPE AMINO ACID
LDV (402)






system), member 5

TRANSPORTER 1; Solute carrier









family 7, member 5; solute carrier









family 7 (cationic amino acid









transporter, y+ system), member 5









S0705
breast cancer
25855
DKFZp564A063; BRMS1; breast
KARAAVSP
N/A
N/A
1:1000-



metastasis suppressor

cancer metastasis-suppressor 1; breast
QKRKSDGP


1:2000



1

cancer metastasis suppressor 1
(403)








S0706
KiSS-1 metastasis-
3814
MGC39258; KISS1; KiSS-1
RQIPAPQG
N/A
N/A
1:180



suppressor

metastasis-suppressor; KISS1
AVLVQREK








METASTIN; malignant melanoma
D (404)








metastasis-suppressor; KISS1









METASTASIS SUPPRESSOR









S0708
cofactor required for
9439
DKFZp434H0117; CRSP133; SUR2;
SVKEQVEK
N/A
N/A
1:2430



Sp1 transcriptional

DRIP130; CRSP3; mediator;
IICNLKPAL






activation, subunit 3,

transcriptional co-activator CRSP130;
K (138)






130 kDa

CRSP, 130-KD SUBUNIT; CRSP









130-kD subunit; 133 kDa









transcriptional co-activator; 130 kDa









transcriptional co-activator; vitamin









D3 receptor interacting protein; c



















S5002
keratin 14
3861
CK; KRT14; K14; EBS4; EBS3;
Antibody obtained from Chemicon
1:50



(epidermolysis bullosa

cytokeratin 14; CK 14; KERATIN,





simplex, Dowling-

TYPE I CYTOSKELETAL 14; keratin





Meara, Koebner)

14 (epidermolysis bullosa simplex,







Dowling-Meara, Koebner)







S5003
keratin 17
3872
PCHC1; PC; PC2; 39.1; KRT17; K17;
Antibody obtained from Dako
1:10-





CYTOKERATIN 17; VERSION 1;

1:25





CK 17; KERATIN, TYPE I







CYTOSKELETAL 17







S5004
keratin 18
3875
K18; CYK18; KRT18;
Antibody obtained from Dako
1:200-





CYTOKERATIN 18; CK 18;

1:400





KERATIN, TYPE I







CYTOSKELETAL 18







S5005
keratin 18
3875
K18; CYK18; KRT18;
Antibody obtained from Becton Dickinson
1:50-





CYTOKERATIN 18; CK 18;

1:100





KERATIN, TYPE I







CYTOSKELETAL 18







S5012
tumor-associated
4072
TROP1; LY74; Ep-CAM; GA733-2;
Antibody obtained from Oncogene
1:40



calcium signal

EGP40; MK-1; CO17-1A; EPCAM;
Research Products (Calbiochem)




transducer 1

M4S1; KSA; TACSTD1; EGP; MK-1







antigen; EPITHELIAL CELLULAR







ADHESION MOLECULE;







GASTROINTESTINAL TUMOR-







ASSOCIATED ANTIGEN 2, 35-KD







GLYCOPROTEIN; tumor-associated







calcium signal transducer 1 precurso







S5014
estrogen receptor 2
2100
ER-BETA; ESR-BETA; ESR2; Erb;
Antibody obtained from Oncogene
1:2500



(ER beta)

ESRB; NR3A2; ESTROGEN
Research Products (Calbiochem)






RECEPTOR, BETA; estrogen receptor







2 (ER beta)







S5038
mucin 1,
4582
PEMT; MUC1; episialin; EMA; PUM;
Antibody obtained from Imperial Cancer
1:1



transmembrane

H23AG; CD227; PEM;
Research Technology (ICRT)






CARCINOMA-ASSOCIATED







MUCIN; H23 antigen; TUMOR-







ASSOCIATED MUCIN; DF3 antigen;







peanut-reactive urinary mucin; mucin







1, transmembrane; polymorphic







epithelial mucin; MUCIN 1,







URINARY; MUCIN, TUMOR-







ASSOCIATE







S5044
transferrin receptor
7037
P90; TR; TFRC; TFR; CD71; T9;
Antibody obtained from NeoMarkers
1:20



(p90, CD71)

TRFR; ANTIGEN CD71;







TRANSFERRIN RECEPTOR







PROTEIN; transferrin receptor (p90,







CD71)







S5045
v-erb-b2
2064
HER-2; ERBB2; NGL; P185ERBB2;
Antibody obtained from NeoMarkers
1:600



erythroblastic

HER2; C-ERBB-2; NEU; MLN 19;





leukemia viral

EC 2.7.1.112; TKR1 HERSTATIN;





oncogene homolog 2,

NEU PROTO-ONCOGENE;





neuro/glioblastoma

ONCOGENE ERBB2; RECEPTOR





derived oncogene

PROTEIN-TYROSINE KINASE





homolog (avian)

ERBB-2 PRECURSOR; ONCOGENE







NGL, NEUROBLASTOMA- OR







GLIOBLASTOMA-DERIVED;







TYROSINE KINASE-TYPE CELL







S5047
major vault protein
9961
MVP; LRP; VAULT1; LUNG
Antibody obtained from NeoMarkers
1:300





RESISTANCE-RELATED PROTEIN;







MAJOR VAULT PROTEIN, RAT,







HOMOLOG OF







S5064
tumor protein p73-like
8626
LMS; TP73L; KET; SHFM4; p73H;
Antibody obtained from Dako
1:50





EEC3; TP63; p51; TUMOR PROTEIN







p63; TUMOR PROTEIN p73-LIKE;







p53-RELATED PROTEIN p63; tumor







protein 63 kDa with strong homology







to p53







S5065
estrogen receptor 1
2099
ER; NR3A1; ESR1; Era; ESR; ER-
Antibody obtained from Dako
1:20





ALPHA; ESRA; ESTRADIOL







RECEPTOR; ESTROGEN







RECEPTOR, ALPHA; estrogen







receptor 1 (alpha)







S5066
v-erb-b2
2064
HER-2; ERBB2; NGL; P185ERBB2;
Antibody obtained from Dako
1:300



erythroblastic

HER2; C-ERBB-2; NEU; MLN 19;





leukemia viral

EC 2.7.1.112; TKR1 HERSTATIN;





oncogene homolog 2,

NEU PROTO-ONCOGENE;





neuro/glioblastoma

ONCOGENE ERBB2; RECEPTOR





derived oncogene

PROTEIN-TYROSINE KINASE





homolog (avian)

ERBB-2 PRECURSOR; ONCOGENE







NGL, NEUROBLASTOMA- OR







GLIOBLASTOMA-DERIVED;







TYROSINE KINASE-TYPE CELL







S5067
cathepsin D
1509
CTSD; MGC2311; CPSD; EC
Antibody obtained from Dako
1:20-



(lysosomal aspartyl

3.4.23.5; cathepsin D preproprotein;

1:50



protease)

Cathepsin D precursor; cathepsin D







(lysosomal aspartyl protease);







S5069
CA 125
n/a

Antibody obtained from Dako
1:20





S5070
CA 15-3
n/a

Antibody obtained from Dako
1:50





S5071
CA 19-9
n/a

Antibody obtained from Dako
1:50





S5072
v-myc
4609
c-Myc; MYC; ONCOGENE MYC;
Antibody obtained from Dako
1:50



myelocytomatosis

Myc proto-oncogene protein;





viral oncogene

PROTOONCOGENE





homolog (avian)

HOMOLOGOUS TO







MYELOCYTOMATOSIS VIRUS; v-







myc myelocytomatosis viral oncogene







homolog (avian); v-myc avian







myelocytomatosis viral oncogene







homolog; Avian myelocytomatosis







viral (v-myc) onco







S5073
cadherin 1, type 1, E-
999
CDH1; Cadherin-1; Arc-1; ECAD;
Antibody obtained from Dako
1:100-



cadherin (epithelial)

CDHE; Uvomorulin; LCAM;

1:150





Epithelial-cadherin precursor; cell-







CAM 120/80; CADHERIN,







EPITHELIAL; calcium-dependent







adhesion protein, epithelial; cadherin







1, E-cadherin (epithelial); cadherin 1,







type 1 preproprotein; cadherin 1,







S5074
glutathione S-
2950
GSTP1; DFN7; GSTP1-1; GST3;
Antibody obtained from Dako
1:50



transferase pi

GSTPP; GST class-pi; glutathione







transferase; EC 2.5.1.18; glutathione







S-transferase pi; GST, CLASS PI;







deafness, X-linked 7;







GLUTATHIONE S-TRANSFERASE







3; GLUTATHIONE S-







TRANSFERASE, PI; FAEES3







GLUTATHIONE S-TRANSFERASE







PI PSEUD







S5075
tumor protein p53 (Li-
7157
p53; TP53; TRP53;
Antibody obtained from Dako
1:50



Fraumeni syndrome)

PHOSPHOPROTEIN P53;







TRANSFORMATION-RELATED







PROTEIN 53; TUMOR







SUPPRESSOR P53; CELLULAR







TUMOR ANTIGEN P53; tumor







protein p53 (Li-Fraumeni syndrome)







S5076
progesterone receptor
5241
NR3C3; PR; PGR; PROGESTERONE
Antibody obtained from Dako
1:50





RESISTANCE; PSEUDOCORPUS







LUTEUM INSUFFICIENCY







PROGESTERONE RECEPTOR







S5077
trefoil factor 1 (breast
7031

Antibody obtained from Dako
1:50-



cancer, estrogen-



1:100



inducible sequence







expressed in)









S5079
enolase 2, (gamma,
2026
NSE; ENO2; 2-phospho-D-glycerate
Antibody obtained from Dako
1:400



neuronal)

hydro-lyase; ENOLASE, GAMMA;







neurone-specific enolase; ENOLASE,







NEURON-SPECIFIC; 2-phospho-D-







glycerate hydrolyase; EC 4.2.1.11;







Neural enolase; enolase-2, gamma,







neuronal; neuron specific gamma







enolase; enolase 2, (gamma,







S5080
B-cell
596
BCL2; FOLLICULAR LYMPHOMA;
Antibody obtained from Dako
1:50



CLL/lymphoma 2

APOPTOSIS REGULATOR BCL-2;







B-cell CLL/lymphoma 2; B-cell







lymphoma protein 2 alpha; B-cell







lymphoma protein 2 beta;







ONCOGENE B-CELL LEUKEMIA 2







LEUKEMIA, CHRONIC







LYMPHATIC, TYPE 2







S5081
retinoblastoma 1
5925
p105-Rb; PP110; Retinoblastoma-1;
Antibody obtained from Dako
1:20



(including

RB; RB1; RETINOBLASTOMA-





osteosarcoma)

ASSOCIATED PROTEIN; RB







OSTEOSARCOMA,







RETINOBLASTOMA-RELATED;







retinoblastoma 1 (including







osteosarcoma)







S5082
synaptophysin
6855
SYP; Synaptophysin; Major synaptic
Antibody obtained from Dako
1:50





vesicle protein P38







S5083
BCL2-associated X
581
BAX; BCL2-associated X protein;
Antibody obtained from Dako
1:500



protein

APOPTOSIS REGULATOR BAX,







MEMBRANE ISOFORM ALPHA







S5086
estrogen receptor 2
2100
ER-BETA; ESR-BETA; ESR2; Erb;
Antibody obtained from Abcam
1:200



(ER beta)

ESRB; NR3A2; ESTROGEN







RECEPTOR, BETA; estrogen receptor







2 (ER beta)







S5087
mucin 1,
4582
PEMT; MUC1; episialin; EMA; PUM;
Antibody obtained from Zymed
1:200-



transmembrane

H23AG; CD227; PEM;

1:1600





CARCINOMA-ASSOCIATED







MUCIN; H23 antigen; TUMOR-







ASSOCIATED MUCIN; DF3 antigen;







peanut-reactive urinary mucin; mucin







1, transmembrane; polymorphic







epithelial mucin; MUCIN 1,







URINARY; MUCIN, TUMOR-







ASSOCIATE







S6001
estrogen receptor 1
2099
ER; NR3A1; ESR1; Era; ESR; ER-
Antibody obtained from US Labs
1:1





ALPHA; ESRA; ESTRADIOL







RECEPTOR; ESTROGEN







RECEPTOR, ALPHA; estrogen







receptor 1 (alpha)







S6002
progesterone receptor
5241
NR3C3; PR; PGR; PROGESTERONE
Antibody obtained from US Labs
1:1





RESISTANCE; PSEUDOCORPUS







LUTEUM INSUFFICIENCY







PROGESTERONE RECEPTOR







S6003
v-erb-b2
2064
HER-2; ERBB2; NGL; P185ERBB2;
Antibody obtained from US Labs
1:1



erythroblastic

HER2; C-ERBB-2; NEU; MLN 19;





leukemia viral

EC 2.7.1.112; TKR1 HERSTATIN;





oncogene homolog 2,

NEU PROTO-ONCOGENE;





neuro/glioblastoma

ONCOGENE ERBB2; RECEPTOR





derived oncogene

PROTEIN-TYROSINE KINASE





homolog (avian)

ERBB-2 PRECURSOR; ONCOGENE







NGL, NEUROBLASTOMA- OR







GLIOBLASTOMA-DERIVED;







TYROSINE KINASE-TYPE CELL







S6004
B-cell
596
BCL2; FOLLICULAR LYMPHOMA;
Antibody obtained from US Labs
1:1



CLL/lymphoma 2

APOPTOSIS REGULATOR BCL-2;







B-cell CLL/lymphoma 2; B-cell







lymphoma protein 2 alpha; B-cell







lymphoma protein 2 beta;







ONCOGENE B-CELL LEUKEMIA 2







LEUKEMIA, CHRONIC







LYMPHATIC, TYPE 2







S6005
keratin 5
3852
KRT5; EBS2; Keratin-5; K5;
Antibody obtained from US Labs
1:1



(epidermolysis bullosa

CYTOKERATIN 5; CK 5; 58 KDA





simplex, Dowling-

CYTOKERATIN; KERATIN, TYPE





Meara/Kobner/Weber-

II CYTOSKELETAL 5; keratin 5





Cockayne types)

(epidermolysis bullosa simplex,







Dowling-Meara/Kobner/Weber-







Cockayne types)







S6006
tumor protein p53 (Li-
7157
p53; TP53; TRP53;
Antibody obtained from US Labs
1:1



Fraumeni syndrome)

PHOSPHOPROTEIN P53;







TRANSFORMATION-RELATED







PROTEIN 53; TUMOR







SUPPRESSOR P53; CELLULAR







TUMOR ANTIGEN P53; tumor







protein p53 (Li-Fraumeni syndrome)







S6007
KI67
n/a

Antibody obtained from US Labs
1:1





S6008
epidermal growth
1956
S7; EGFR; 2.7.1.112; ERBB;
Antibody obtained from US Labs
1:1



factor receptor

ONCOGENE ERBB; ERBB1





(erythroblastic

SPECIES ANTIGEN 7; V-ERB-B





leukemia viral (v-erb-

AVIAN ERYTHROBLASTIC





b) oncogene homolog,

LEUKEMIA VIRAL ONCOGENE





avian)

HOMOLOG; epidermal growth factor







receptor (avian erythroblastic leukemia







viral (v-erb-b) oncogene homolog)







S6011
enolase 2, (gamma,
2026
NSE; ENO2; 2-phospho-D-glycerate
Antibody obtained from US Labs
1:1



neuronal)

hydro-lyase; ENOLASE, GAMMA;







neurone-specific enolase; ENOLASE,







NEURON-SPECIFIC; 2-phospho-D-







glycerate hydrolyase; EC 4.2.1.11;







Neural enolase; enolase-2, gamma,







neuronal; neuron specific gamma







enolase; enolase 2, (gamma,







S6012
thyroid transcription
7080
benign chorea; chorea, hereditary
Antibody obtained from US Labs
1:1



factor 1

benign; NK-2 (Drosophila) homolog A







(thyroid nuclear factor); Thyroid







transcription factor 1 (NK-2,







Drosophila, homolog of, A); BCH;







BHC; TEBP; TTF1; NKX2A; TTF-1;







NKX2.1







S6013
v-erb-b2
2064
HER-2; ERBB2; NGL; P185ERBB2;
Antibody obtained from US Labs
1:1



erythroblastic

HER2; C-ERBB-2; NEU; MLN 19;





leukemia viral

EC 2.7.1.112; TKR1 HERSTATIN;





oncogene homolog 2,

NEU PROTO-ONCOGENE;





neuro/glioblastoma

ONCOGENE ERBB2; RECEPTOR





derived oncogene

PROTEIN-TYROSINE KINASE





homolog (avian)

ERBB-2 PRECURSOR; ONCOGENE







NGL, NEUROBLASTOMA- OR







GLIOBLASTOMA-DERIVED;







TYROSINE KINASE-TYPE CELL









OTHER EMBODIMENTS

Other embodiments of the invention will be apparent to those skilled in the art from a consideration of the specification or practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with the true scope of the invention being indicated by the following claims.

Claims
  • 1. (canceled)
  • 2. A method of identifying a breast cancer that will not respond favorably to a chemotherapy treatment that includes a taxane or taxane derivative, the method comprising: (a) performing immunohistochemical staining for TLE3 polypeptide on a cancer sample obtained from a breast cancer patient, wherein the breast cancer patient is triple negative for estrogen receptor, progesterone receptor, and HER-2 markers;(b) detecting a percentage and/or intensity of immunohistochemical staining for TLE3 polypeptide in the cancer sample, wherein the detected percentage and/or intensity is lower than a reference; and(c) identifying the patient's breast cancer as one that will not respond favorably to a chemotherapy treatment that includes a taxane or taxane derivative,wherein the step of performing immunohistochemical staining comprises contacting the cancer sample with an antibody that binds TLE3 polypeptide thereby forming a complex comprising the antibody and TLE3 polypeptide, and the step of detecting comprises detecting the complex in the cancer sample.
  • 3. The method of claim 2, wherein the step of detecting comprises quantifying a percentage of cells in which immunohistochemical staining for TLE3 polypeptide is detected.
  • 4. The method of claim 2, wherein the step of detecting comprises quantifying an intensity of immunohistochemical staining for TLE3 polypeptide in the cancer sample.
  • 5. The method of claim 2, wherein the step of identifying comprises identifying the patient's breast cancer as one that will not respond favorably to a chemotherapy treatment that includes a taxane.
  • 6. The method of claim 5, wherein the taxane is paclitaxel or docetaxel.
  • 7. The method of claim 2, further comprising administering a chemotherapy treatment that does not include a taxane or taxane derivative to the breast cancer patient.
  • 8. A method of identifying a breast cancer that will not respond favorably to a chemotherapy treatment that includes a taxane or taxane derivative, the method comprising: (a) performing immunohistochemical staining for TLE3 polypeptide on a cancer sample obtained from a breast cancer patient, wherein the breast cancer patient is triple negative for estrogen receptor, progesterone receptor, and HER-2 markers;(b) detecting a percentage and/or intensity of immunohistochemical staining for TLE3 polypeptide in the cancer sample, wherein the detected percentage and/or intensity is lower than a reference; and(c) identifying the patient's breast cancer as one that will not respond favorably to a chemotherapy treatment that includes a taxane or taxane derivative,wherein the step of performing immunohistochemical staining comprises contacting the cancer sample with a primary antibody that binds TLE3 polypeptide and a labeled secondary antibody that binds the primary antibody thereby forming a complex comprising the primary antibody, secondary antibody and TLE3 polypeptide, and the step of detecting comprises detecting the complex in the cancer sample.
  • 9. The method of claim 8, wherein the step of detecting comprises quantifying a percentage of cells in which immunohistochemical staining for TLE3 polypeptide is detected.
  • 10. The method of claim 8, wherein the step of detecting comprises quantifying an intensity of immunohistochemical staining for TLE3 polypeptide in the cancer sample.
  • 11. The method of claim 8, wherein the step of identifying comprises identifying the patient's breast cancer as one that will not respond favorably to a chemotherapy treatment that includes a taxane.
  • 12. The method of claim 11, wherein the taxane is paclitaxel or docetaxel.
  • 13. The method of claim 8, further comprising administering a chemotherapy treatment that does not include a taxane or taxane derivative to the breast cancer patient.
  • 14. A method of predicting the likelihood that a breast cancer will not respond to a chemotherapy treatment that includes a taxane or taxane derivative, the method comprising: (a) performing immunohistochemical staining for TLE3 polypeptide on a cancer sample obtained from a breast cancer patient, wherein the breast cancer patient is triple negative for estrogen receptor, progesterone receptor, and HER-2 markers;(b) detecting a percentage and/or intensity of immunohistochemical staining for TLE3 polypeptide in the cancer sample, wherein the detected percentage and/or intensity is lower than a reference; and(c) predicting that the patient's breast cancer will likely not respond favorably to a chemotherapy treatment that includes a taxane or taxane derivative,wherein the step of performing immunohistochemical staining comprises contacting the cancer sample with an antibody that binds TLE3 polypeptide thereby forming a complex comprising the antibody and TLE3 polypeptide, and the step of detecting comprises detecting the complex in the cancer sample.
  • 15. The method of claim 14, wherein the step of detecting comprises quantifying a percentage of cells in which immunohistochemical staining for TLE3 polypeptide is detected.
  • 16. The method of claim 14, wherein the step of detecting comprises quantifying an intensity of immunohistochemical staining for TLE3 polypeptide in the cancer sample.
  • 17. The method of claim 14, wherein the step of predicting comprises predicting that the patient's breast cancer will likely not respond to a chemotherapy treatment that includes a taxane.
  • 18. The method of claim 17, wherein the taxane is paclitaxel.
  • 19. The method of claim 17, wherein the taxane is docetaxel.
  • 20. The method of claim 14, further comprising administering a chemotherapy treatment that does not include a taxane or taxane derivative to the breast cancer patient.
CROSS REFERENCE TO RELATED APPLICATION

This application is a continuation of U.S. patent application Ser. No. 12/889,630, filed Sep. 24, 2010, which is a continuation of U.S. patent application Ser. No. 12/277,920, filed Nov. 25, 2008, (issued as U.S. Pat. No. 7,816,084), which claims priority to U.S. Provisional Patent Application Ser. No. 60/991,487, filed Nov. 30, 2007, each of which is hereby incorporated herein by reference in its entirety.

Provisional Applications (1)
Number Date Country
60991487 Nov 2007 US
Continuations (2)
Number Date Country
Parent 12889630 Sep 2010 US
Child 14671015 US
Parent 12277920 Nov 2008 US
Child 12889630 US