TLE3 as a marker for chemotherapy

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 Oct. 13, 2009, 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 (RPI) 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, 11th 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 (G0). 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, sulfhydryl 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. Nos. 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 600sl 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






GENE

(SEQ ID
(SEQ ID
(SEQ ID



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







S0011
vav 3 oncogene
10451
VAV3; VAV3 ONCOGENE; ONCOGENE
TEESINDEDIY
EKRTNGLR
DYISKSKED
1:90-






VAV3; vav 3 oncogene
KGLPDLIDE
RTPKQVD
VKLK (3)
1:300






(1)
(2)





S0017
WAP four-disul-
10406
WFDC2; WAP5; dJ461P17.6; major
EKTGVCPELQ
PNDKEGSC
RDQCQVDT
1:25-



fide core domain

epididymis-specific protein
ADQNCTQE
PQVNIN (5)
QCPGQMK
1:500



2

E4; epididymal secretory pro-
(4)

(6)





tein E4; WAP 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,
4250
UGB2; MGB1; SCGB2A2; mamma-
SKTINPQVSK
DDNATTNAI
NQTDETLSN
1:300-



family 2A,

globin 1; secretoglobin,
TEYKELLQE
DELKEC (8)
VEVFMQ
1:1000



member 2

family 2A, member 2
(7)

(9)





S0020
PPAR binding
5469
RB18A; TRIP2; PPARGBP; PBP;
SSDDGIRPLP
DGKSKDKP
NKTKKKKSS
1:100



protein

CRSP1; PPARBP; CRSP200;
EYSTEKHKK
PKRKKADTE
RLPPEK





DRIP230; PPAR-BINDING PROTEIN;
(10)
(11)
(12)





PPARG binding protein; PPAR





binding protein; CRSP, 200-KD





SUBUNIT; PEROXISOME PROLIFERA-





TOR-ACTIVATED RECEPTOR-BINDING





PROTEIN; THYROID HORMONE RE-





CEPTOR INTERACTOR 2; RECOGN





S0021
hypothetical
222256
FLJ23834; hypothetical protein
KNKEPLTKKG
KLTCTDLDS
EVDYENPS
1:200-



protein FLJ23834

FLJ23834
ETKTAERD
SPRSFRYS
NLAAGNKYT
1:2500






(13)
(14)
(15)





S0022
cytochrome P450
199974
CYP4Z1; cytochrome P450 4Z1;
KTLQVFNPLR
QHFAIIECKV
RKFLAPDHS
1:50-



4Z1

cytochrome P450, family 4,
FSRENSEKIH
AVALT (17)
RPPQPVRQ
1:500





sub-family Z, polypeptide 1
(16)

(18)





S0024
RAS-like,
85004
RERG; RAS-like, estrogen-
MAKSAEVKLA
VLPLKNILDE
YELCREVRR
1:900-



estrogen-regu-

regulated, growth-inhibitor
IFGRAGVGK
IKKPKN (20)
RRMVQGKT
1:2700



lated, growth-


(19)

(21)



inhibitor





S0032
fatty acid bind-
2170
MDGI; O-FABP; FABP3; FABP11;
TKPTTIIEKNG
KNTEISFKL
HLQKWDGQ
1:225



ing protein 3,

H-FABP; FATTY ACID-BINDING
DILTLKTH
GVEFDE
ETTLVRE



muscle and heart

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



(mammary-derived

Fatty acid-binding protein



growth

3, muscle; fatty acid



inhibitor)

binding protein 11; FATTY





ACID-BINDING PROTEIN, MUSCLE





AND HEART; fatty acid binding





protein 3, muscle and heart





(mammary-de





S0036
gamma-aminobuty-
2568
GABRP; GAMMA-AMINOBUTYRIC ACID
DGNDVEFTW
LQQMAAKD
KRKISFASIE
1:250-



ric acid (GABA)

RECEPTOR, PI; GABA-A RECEPTOR,
LRGNDSVRG
RGTTKEVEE
ISSDNVDYS
1:500



A receptor, pi

PI POLYPEPTIDE; gamma-aminobu-
LEH (25)
VS (26)
D (27)





tyric acid (GABA) A receptor,





pi





S0037
annexin A8
244
ANX8; ANXA8; annexin VIII;
QRQQIAKSFK
REIMKAYEE
EEYEKIANK
1:30-





annexin A8
AQFGKDLTE
DYGSSLEED
SIEDSIKSE
1:40






(28)
IQ (29)
(30)





S0039
CDNA FLJ25076
134111
similar to 3110006E14Rik pro-
EGGSLVPAA
RKAGKSKK
KTHEKYGW
1:50-



fis, clone

tein; CDNA FLJ25076 fis, clone
RQQHCTQVR
SFSRKEAE
VTPPVSDG
1:30000



CBL06117

CBL06117
SRR (31)
(32)
(33)





S0040
ATP-binding cas-
5243
P-gp; PGY1; CLCS; ABCB1;
MDLEGDRNG
NLEDLMSNI
RGSQAQDR
1:200-



sette, sub-

ABC20; CD243; GP170; MDR1;
GAKKKN (34)
TNRSDINDT
KLSTKEA
1:400



family B

doxorubicin resistance; col-

G (35)
(36)



(MDR/TAP),

chicin sensitivity; P-GLYCO-



member 1

PROTEIN 1; multidrug resis-





tance 1; P glycoprotein 1;





ATP-binding cassette sub-





family B member 1; ATP-





BINDING CASSETTE, SUBFAMILY B,





MEMBER 1; ATP-bin





S0041
ATP-binding cas-
5244
MDR3; PGY3; PFIC-3; ABCB4;
MDLEAAKNG
NFSFPVNFS
KNSQMCQK
1:60-



sette, sub-

ABC21; MDR2/3; P-GLYCOPROTEIN
TAWRPTSAE
LSLLNPGK
SLDVETDG
1:300



family B

3; MULTIDRUG RESISTANCE 3; P-
(37)
(38)
(39)



(MDR/TAP),

glycoprotein-3/multiple drug



member 4

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 cas-
4363
ABCC1; MRP1; GS-X; ABC29;
MALRGFCSA
KNWKKECA
DSIERRPVK
1:40-



sette, sub-

multidrug resistance protein;
DGSD (40)
KTRKQPVK
DGGGTNS
1:500



family C

MULTIDRUG RESISTANCE-ASSOCI-

(41)
(42)



(CFTR/MRP),

ATED PROTEIN 1; multiple drug



member 1

resistance-associated protein;





multiple drug resistance pro-





tein 1; ATP-BINDING CASSETTE,





SUBFAMILY C, MEMBER 1; ATP-





binding cassette, sub-fami





S0043
ATP-binding cas-
1244
MRP2; cMRP; CMOAT; ABCC2;
MLEKFCNSTF
SILCGTFQF
ENNESSNN
1:50-



sette, sub-

ABC30; DJS; MULTIDRUG RESIS-
WNSSFLDSP
QTLIRT (44)
PSSIAS
1:333



family C

TANCE-ASSOCIATED PROTEIN 2;
E (43)

(45)



(CFTR/MRP),

canalicular multispecific or-



member 2

ganic anion transporter; MUL-





TISPECIFIC ORGANIC ANION





TRANSPORTER, CANALICULAR; ATP-





BINDING CASSETTE, SUBFAMILY C,





MEMBER 2; ATP-binding





cassette,





S0044
ATP-binding cas-
10257
MOAT-B; MRP4; MOATB; ABCC4;
QEVKPNPLQ
DEISQRNRQ
VQDFTAFW
1:20-



sette, sub-

EST170205; MULTIDRUG RESIS-
DANICSR
LPSDGKK
DKASETPTL
1:100



family C

TANCE-ASSOCIATED PROTEIN 4;
(46)
(47)
Q (48)



(CFTR/MRP),

MULTISPECIFIC ORGANIC ANION



member 4

TRANSPORTER B; ATP-binding





cassette, sub-family C, member





4; ATP-BINDING CASSETTE, SUB-





FAMILY C, MEMBER 4; ATP-bind-





ing cassette, sub-family C





(CFT





S0045
ATP-binding cas-
8714
MOAT-D; ABC31; MLP2; ABCC3;
MDALCGSGE
RKQEKQTA
DPQSVERK
1:2000



sette, sub-

E5T90757; cMOAT2; MULTIDRUG
LGSKFWDSN
RHKASAA
TISPG



family C

RESISTANCE-ASSOCIATED PROTEIN
(49)
(50)
(51)



(CFTR/MRP),

3; canicular multispecific or-



member 3

ganic anion transporter; CAN-





ALICULAR MULTISPECIFIC ORGANIC





ANION TRANSPORTER 2; ATP-





BINDING CASSETTE, SUBFAMILY C,





MEMBER 3; ATP-binding cas





S0046
ATP-binding cas-
10057
MOAT-C; ABCC5; MRP5;
MKDIDIGKEYI
RDREDSKF
SKHESSDV
1:100-



sette, sub-

EST277145; ABC33; SMRP;
IPSPGYRS
RRTRPLEC
NCRRLER
1:450



family C

pABC11; MOATC; MULTIDRUG RE-
(52)
QD (53)
(54)



(CFTR/MRP),

SISTANCE-ASSOCIATED PROTEIN 5;



member 5

canalicular multispecific or-





ganic anion transporter C;





ATP-binding cassette, sub-





family C, member 5; ATP-





BINDING CASSETTE, SUBFAMILY C,





MEMBER 5; ATP-bi





S0047
ATP-binding cas-
368
MRP6; ARA; E5T349056; ABCC6;
MAAPAEPCA
DPGVVDSS
HTLVAENAM
1:50



sette, sub-

MOATE; PXE; MLP1; ABC34;
GQGVWNQTE
SSGSAAGK
NAEK (57)



family C

ANTHRACYCLINE RESISTANCE-
PE (55)
D (56)



(CFTR/MRP),

ASSOCIATED PROTEIN; MULTIDRUG



member 6

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 cas-
8647
BSEP; ABCB11; PFIC-2; SPGP;
MSDSVILRSIK
TNSSLNQN
QEVLSKIQH
1:600



sette, sub-

PGY4; PFIC2; ABC16; SISTER OF
KFGEEND
MTNGTR
GHTIIS



family B

P-GLYCOPROTEIN; bile salt ex-
(58)
(59)
(60)



(MDR/TAP),

port pump; progressive famil-



member 11

ial intrahepatic cholestasis





2; ABC member 16, MDR/TAP sub-





family; ATP-BINDING CASSETTE,





SUBFAMILY B, MEMBER 11; ATP-





binding cassette, sub-fam





S0049
ATP-binding cas-
23456
MTABC2; E5T20237; MABC2;
GADDPSSVT
NAVASPEFP
KPNGIYRKL
1:10-



sette, sub-

M-ABC2; ABCB10; MITOCHONDRIAL
AEEIQR (61)
PRFNT (62)
MNKQSFISA
1:25



family B

ABC PROTEIN 2; ATP-BINDING


(63)



(MDR/TAP),

CASSETTE, SUBFAMILY B, MEMBER



member 10

10; ATP-binding cassette, sub-





family B, member 10; ATP-





binding cassette, sub-family B





(MDR/TAP), member 10





S0050
transporter 1,
6890
RING4; ABC17; D6S114E; ABCB2;
MASSRCPAP
QGGSGNPV
EFVGDGIYN
1:80



ATP-binding cas-

TAP1; APT1; PEPTIDE TRANS-
RGCR (64)
RR (65)
NTMGHVHS



sette, sub-

PORTER PSF1; TRANSPORTER, ABC,


(66)



family B

MHC, 1; ABC transporter, MHC



(MDR/TAP)

1; antigen peptide transporter





1; peptide supply factor 1;





ABC TRANSPORTER, MHC, 1; ATP-





BINDING CASSETTE, SUBFAMILY B,





MEMBER 2; TRANSPORTER





S0052
ATP-binding cas-
6833
SUR1; MRP8; PHHI; ABC36;
MPLAFCGSE
DHLGKEND
EIREEQCAP
1:25-



sette, sub-

ABCC8; HRINS; sulfonylurea
NHSAAYR
VFQPKTQFL
HEPTPQG
1:150



family C

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



(CFTR/MRP),

SULFONYLUREA RECEPTOR, BETA-



member 8

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 cas-
10060
ABCC9; ABC37; sulfonylurea re-
MSLSFCGNNI
QRVNETQN
DEIGDDSW
1:25-



sette, sub-

ceptor 2A; ATP-BINDING CAS-
SS (70)
GTNNTTGIS
RTGESSLPF
1:50



family C

SETTE, SUBFAMILY C, MEMBER 9;

E (71)
E (72)



(CFTR/MRP),

ATP-binding cassette, sub-



member 9

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 mem-
9445
E25B; ABRI; E3-16; FBD; BRI2;
MVKVTFNSAL
QTIEENIKIF
HDKETYKLQ
1:450 -



brane protein 2B

BRICD2B; ITM2B; BRI GENE;
AQKEAKKDE
EEEEVE (74)
RRETIKGIQ
1:500





BRICHOS domain containing 2B;
PK (73)

KRE (75)





integral membrane protein 2B





S0057
ankyrin 3, node
288
ankyrin-G; ANK3; ankyrin-3,
MAHAASQLK
HKKETESD
EGFKVKTKK
1:750



of Ranvier

node of Ranvier; ankyrin 3
KNRDLEINAE
QDDEIEKTD
EIRHVEKKS



(ankyrin G)

isoform 1; ankyrin 3 isoform
E (76)
RRQ (77)
HS (78)





2; ankyrin 3, node of Ranvier





(ankyrin G)





S0058
hypothetical
80004
FLJ21918; hypothetical protein
ERALAAAQR
TAGMKDLLS
DPPRTVLQA
1:20



protein FLJ21918

FLJ21918
CHKKVMKER
VFQAYQ
PKEWVCL






(79)
(80)
(81)





S0059
tripartite
23650
ATDC; TRIM29; tripartite mo-
MEAADASRS
ELHLKPHLE
EGEGLGQS
1:50-



motif-containing

tif-containing 29; ataxia-
NGSSPEARD
GAAFRDHQ
LGNFKDDLL
1:3000



29

telangiectasia group D-
AR (82)
(83)
N (84)





associated protein; tripartite





motif protein TRIM29 isoform





alpha; tripartite motif pro-





tein TRIM29 isoform beta





S0059P
tripartite
23650
ATDC; TRIM29; tripartite mo-
ELHLKPHLEG
N/A
N/A
1:30-


2
motif-containing

tif-containing 29; ataxia-
AAFRDHQ


1:90



29

telangiectasia group D-
(85)





associated protein; tripar-





tite motif protein TRIM29 iso-





form alpha; tripartite motif





protein TRIM29 isoform beta





S0063
iroquois homeo-
79191
IRX3; iroquois homeobox
GSEERGAGR
KIWSLAETA
KKLLKTAFQ
1:200-



box protein 3

protein 3
GSSGGREE
TSPDNPRR
PVPRRPQN
1:1200






(86)
S (87)
HLD (88)





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



estrogen-regu-

regulated, growth-inhibitor
QAINKMLTKIS


1:40000



lated, growth-


S (89)



inhibitor





S0070
G protein-
26996
GPCR150; GPR160; putative G
MRRKNTCQN
NETILYFPFS
KVQIPAYIE
1:10-



coupled receptor

protein-coupled receptor; G
FMEYFCISLA
SHSSYTVRS
MNIPLVILCQ
1:100



160

protein-coupled receptor 160
F (90)
KK (91)
(92)





S0072
S100 calcium
6279
CP-10; L1Ag; CALPROTECTIN;
MLTELEKALN
RDDLKKLLE
KMGVAAHK
1:6500-



binding protein

60B8AG; S100A8; MIF; CAGA;
SIIDVYHK
TECPQYIRK
KSHEESHKE
1:10000



A8 (calgranulin

NIF; MRP8; MA387; CFAG; CGLA
(93)
KGAD (94)
(95)



A)

S100A8/S100A9 COMPLEX; cystic





fibrosis antigen; S100





calcium-binding protein A8;





S100 calcium binding protein





A8 (calgranulin A)





S0073
forkhead box A1
3169
HNF3A; MGC33105; TCF3A; FOXA1;
PESRKDPSG
HGLAPHES
EQQHKLDF
1:100-





forkhead box A1; HEPATOCYTE
ASNPSADS
QLHLKGD
KAYEQALQY
1:2700





NUCLEAR FACTOR 3-ALPHA;
(96)
(97)
S (98)





hepatocyte nuclear factor 3,





alpha





S0073P
forkhead box A1
3169
HNF3A; MGC33105; TCF3A; FOXA1;
HGLAPHESQ
N/A
N/A
1:50-


2


forkhead box A1; HEPATOCYTE
LHLKGD (99)


1:450





NUCLEAR FACTOR 3-ALPHA;





hepatocyte nuclear factor 3,





alpha





S0074
trefoil factor 3
7033
TFF3; trefoil factor 3 (intes-
EEYVGLSAN
RVDCGYPH
VPWCFKPL
1:2500-



(intestinal)

tinal); trefoil factor 3,
QCAVPAKDR
VTPKECN
QEAECTF
1:30000





HITF, human intestinal trefoil
VD (100)
(101)
(102)





factor





S0074P
trefoil factor 3
7033
TFF3; trefoil factor 3 (intes-
VPWCFKPLQ
N/A
N/A
1:400-


3
(intestinal)

tinal); trefoil factor 3,
EAECTF


1:810





HITF, human intestinal trefoil
(103)





factor





S0076x
keratin 17
3872
PC2; PCHC1; KRT17; K17;
KKEPVTTRQV
QDGKVISSR
SSSIKGSSG
1:200


1


CYTOKERATIN 17; keratin 17
RTIVEE
EQVHQTTR
LGGGSS






(104)
(105)
(106)





S0078
kynureninase (L-
8942
3.7.1.3; XANTHURENICACIDURIA;
DEEDKLRHFR
KPREGEETL
EERGCQLTI
1:180-



kynurenine

KYNU; HYDROXYKYNURENINURIA;
ECFYIPKIQD
RIEDILEVIE
TFSVPNKDV
1:200



hydrolase)

KYNURENINASE DEFICIENCY;
(107)
KE (108)
FQE (109)





XANTHURENIC ACIDURIA;





kynureninase (L-kynurenine





hydrolase)





S0079
solute carrier
25800
SLC39A6; LIV-1 protein, estro-
DHNHAASGK
EEPAMEMK
QRYSREELK
1:200-



family 39 (zinc

gen regulated; solute carrier
NKRKALCPD
RGPLFSHLS
DAGVATL
1:800



transporter),

family 39 (zinc transporter),
HD (110)
SQNI (111)
(112)



member 6

member 6; solute carrier fami-





ly 39 (metal ion transporter),





member 6





S0081
N-acetyltrans-
9
AAC1; 2.3.1.5; NAT1; arylamine
MDIEAYLERI
QMWQPLELI
FNISLQRKL
1:10-



ferase 1 (aryla-

N-acetyltransferase-1; ACETYL-
GYKKSRNKL
SGKDQPQV
VPKHGDRF
1:240



mine N-acetyl-

CoA: ARYLAMINE N-
DLE (113)
PCVFR
FTI (115)



transferase)

ACETYLTRANSFERASE; ARYLAMINE

(114)





N-ACETYLTRANSFERASE 1; N-





acetyltransferase 1 (arylamine





N-acetyltransferase); aryla-





mide acetylase 1 (N-acetyl-





transferase 1)





S0086
X-box binding
7494
XBP2; TREB5; XBP1; X-box-
RQRLTHLSPE
EKTHGLVVE
QPPFLCQW
1:180-



protein 1

binding protein-1; X BOX-
EKALRRKLKN
NQELRQRL
GRHQPSWK
1:400





BINDING PROTEIN 1; X BOX-
R (116)
GMD (117)
PLMN (118)





BINDING PROTEIN 2; X-box





binding protein 1





S0088
claudin 10
9071
CPETRL3; OSP-L; CLDN10; clau-
NKITTEFFDPL
FSISDNNKT
EDFKTTNPS
1:333-





din 10; claudin 10 isoform a;
FVEQK (119)
PRYTYNGAT
KQFDKNAY
1:1000





claudin 10 isoform b

(120)
V (121)





S0090
sparc/osteonec-
9806
KIAA0275; testican-2; SPOCK2;
EGDAKGLKE
EWCFCFWR
EEEGEAGE
1:100-



tin, cwcv and

TESTICAN 2; SPARC/OSTEONECTIN,
GETPGNFME
EKPPCLAEL
ADDGGYIW
1:800



kazal-like do-

CWCV, AND KAZAL-LIKE DOMAINS
DE (122)
ER (123)
(124)



mains proteo-

PROTEOGLYCAN 2; sparc/osteo-



glycan

nectin, cwcv and kazal-like



(testican) 2

domains proteoglycan





(testican) 2





S0091
lipocalin 2
3934
UTEROCALIN; NGAL; LCN2;
DKDPQKMYA
KKCDYWIRT
ENFIRESKY
1:100



(oncogene 24p3)

NEUTROPHIL GELATINASE-
TIYE (125)
FVPGCQ
LGLPEN





ASSOCIATED LIPOCALIN; ONCOGEN-

(126)
(127)





IC LIPOCALIN 24P3; lipocalin 2





(oncogene 24p3)





S0092
paired box gene
7849
PAX8; paired box gene 8;
DDSDQDSCR
RQHYPEAY
NTPLGRNLS
1:30-



8

paired box gene 8 isoform
LSIDSQ
ASPSHTK
THQTYPVVA
1:100





PAX8C; paired box gene 8 iso-
(128)
(129)
D (130)





form PAX8D; paired box gene 8





isoform PAX8E; paired box 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;
RLVSCPGPLD
KMSPEDIRK
SPEELSSVP
1:500





MEGAKARYOCYTE-POTENTIATING
QDQQE
WNVTSLETL
PSSIWAVRP





FACTOR; SOLUBLE MPF/MESO-
(131)
K (132)
QD (133)





THELIN-RELATED PROTEIN; meso-





thelin isoform 2 precursor;





mesothelin isoform 1 precur-





sor; megakaryocyte potentiat-





ing factor precursor; ANTIGEN





RECOGNIZED BY MONOCLONAL





ANTIBODY





S0094
kallikrein 6
5653
Bssp; PRSS18; KLK6; Klk7;
EEQNKLVHG
ELIQPLPLER
GKTADGDF
1:150-



(neurosin, zyme)

SP59; PRSS9; MGC9355; protease
GPCDKTSH
DCSANT
PDTIQC
1:300





M; kallikrein 6 preproprotein;
(134)
(135)
(136)





protease, serine, 18; pro-





tease, serine, 9; kallikrein 6





(neurosin, zyme)





S0095
Rap guanine nu-
10411
bcm910; MGC21410;
REQWPERRR
KVNSAGDAI
QQLKVIDNQ
1:250-



cleotide ex-

9330170P05Rik; EPAC; RAPGEF3;
CHRLENGCG
GLQPDAR
RELSRLSRE
1:1000



change factor

cAMP-GEFI; RAP guanine-
NA (137)
(139)
LE (140)



(GEF) 3

nucleotide-exchange factor 3;





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;
REHMQAVTR
KKSKAVLDY
DEFYSREG
1:100-



transporting,

3.6.3.14; VATB; ATP6B1; V-
NYITHPR
HDDN (142)
RLQDLAPDT
1:800



lysosomal 56/58

ATPase B1 subunit; H+-ATPase
(141)

AL (143)



kDa, V1 subunit

beta 1 subunit; H(+)-trans-



B, isoform 1

porting two-sector ATPase, 58



(Renal tubular

kD subunit; vacuolar proton



acidosis with

pump, subunit 3; endomembrane



deafness)

proton pump 58 kDa subunit;





ATPase, H+ transporting, lysos





S0097
frizzled homolog
8325
FZ-8; hFZ8; FZD8; frizzled 8;
KQQDGPTKT
ELRVLSKAN
RRGGEGGE
1:100-



8 (Drosophila)

frizzled homolog 8
HKLEKLMIR
AIVPGLSGG
ENPSAAKG
1:500





(Drosophila); FRIZZLED,
(144)
E (145)
HLMG (146)






DROSOPHILA, HOMOLOG OF, 8






S0099
histone 1, H2ba
255626
HIST1H2BA; histone 1, H2ba
MPEVSSKGA
GFKKAVVKT
KEGKKRKR
1:333-






TISKK (147)
QK (148)
TRKE (149)
1:500





S0110
hypothetical
84259
MGC2714; hypothetical protein
RYAFDFARDK
SVFYQYLEQ
EDGAWPVL
1:500-



protein MGC2714

MGC2714
DQRSLDID
SKYRVMNK
LDEFVEWQ
1:2500






(150)
DQ (151)
KVRQTS








(152)





S0117
reproduction 8
7993
D8S2298E; REP8; reproduction
SFKSPQVYLK
RKKQQEAQ
EDIGITVDTV
1:200-





8; Reproduction/chromosome 8
EEEEKNEKR
GEKASRYIE
LILEEKEQT
1:375






(153)
(154)
N (155)





S0119
slit homolog 1
6585
SLIT3; MEGF4; SLIL1; Slit-1;
KAFRGATDLK
DERCEEGQ
DGTSFAEEV
1:900



(Drosophila)

SLIT1; slit homolog 1
NLRLDKNQ
EEGGCLPR
EKPTKCGC





(Drosophila); SLIT,
(156)
PQ (157)
ALCA (158)






DROSOPHILA, HOMOLOG OF, 1;






MULTIPLE EPIDERMAL GROWTH





FACTOR-LIKE DOMAINS 4





S0122
leucyl-tRNA
23395
6.1.1.4; MGC26121; KIAA0028;
QRIKEQASKI
HAKTKEKLE
KSPQPQLLS
1:150



synthetase 2,

LEURS; LARS2; leucine trans-
SEADKSKPKF
VTWEKMSK
NKEKAEARK



mitochondrial

lase; leucine-tRNA ligase;
(159)
SKHN (160)
(161)





LEUCYL-tRNA SYNTHETASE, MITO-





CHONDRIAL; leucyl-tRNA synthe-





tase 2, mitochondrial; leucyl-





tRNA synthetase 2, mitochon-





drial precursor





S0123
homeo box D4
3233
HOX4B; HOXD4; HHO.C13; HOX-
MLFEQGQQA
KDQKAKGIL
HSSQGRLP
1:100-





5.1; HOMEOBOX D4; HOMEOBOX 4B;
LELPECT
HSPASQSP
EAPKLTHL
1:500





HOMEOBOX X; homeo box D4;
(162)
ERS (163)
(164)





homeobox protein Hox-D4;





Hox-4.2, mouse, homolog of





homeo box X





S0124
sphingosine-1-
8879
KIAA1252; SPL; SGPL1;
KRGARRGGW
KIVRVPLTK
QFLKDIRES
1:990-



phosphate lyase

sphingosine-1-phosphate lyase
KRKMPSTDL
MMEVDVR
VTQIMKNPK
1:1500



1

1
(165)
(166)
A (167)





S0126
HBxAg transacti-
55789
XTP1; HBxAg transactivated
SKQGVVILDD
VQTFSRCIL
LKKPFQPFQ
1:450-



vated protein 1

protein 1
KSKELPHW
CSKDEVDLD
RTRSFRM
1:1600






(168)
EL (169)
(170)





S0132
SRY (sex deter-
6662
SRA1; CMD1; CMPD1; SOX9; SRY-
MNLLDPFMK
NTFPKGEPD
KNGQAEAE
1:100 -



mining region

BOX 9; transcription factor
MTDEQEKGL
LKKESEEDK
EATEQTHIS
1:500



Y)-box 9 (campo-

SOX9; SRY-RELATED HMG-BOX GENE
S (171)
(172)
PN (173)



melic dysplasia

9; SEX REVERSAL, AUTOSOMAL, 1;



autosomal sex-

SRY (sex-determining region



reversal)

Y)-box 9 protein; SRY (sex-





determining region Y)-box 9





(campomelic dysplasia, auto-





somal sex-reversal); SRY (





S0137
cadherin, EGF
1952
Flamingo1; CELSR2; EGFL2;
QASSLRLEPG
ELKGFAERL
RSGKSQPS
1:1800-



LAG seven-pass

KIAA0279; MEGF3; CDHF10; EGF-
RANDGDWH
QRNESGLD
YIPFLLREE
1:5000



G-type receptor

like-domain, multiple 2; epi-
(174)
SGR (175)
(176)



2 (flamingo

dermal growth factor-like 2;



homolog,

multiple epidermal growth




Drosophila)


factor-like domains 3; cad-





herin 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
RRSDYAKVA
KNFTMNEKL
EFFVNEARK
1:2500-



hydrolase

hydrolase precursor; gamma-
KIFYNLSIQSF
KKFFNVLTT
NNHHFKSE
1:30000



(conjugase,

glutamyl hydrolase (conjugase,
DD (177)
N (178)
SEE (179)



folylpolygamma-

folylpolygammaglutamyl



glutamyl

hydrolase)



hydrolase)





S0140
bullous pemphi-
667
BP240; FLJ13425; FLJ32235;
KNTQAAEALV
QENQPENS
KQMEKDLA
1:250-



goid antigen 1,

FLJ21489; FLJ30627; CATX-15;
KLYETKLCE
KTLATQLNQ
FQKQVAEK
1:20000



230/240 kDa

KIAA0728; BPAG1; dystonin;
(180)
(181)
QLK (182)





hemidesmosomal plaque protein;





bullous pemphigoid antigen 1,





230/240 kDa; bullous pemphi-





goid antigen 1 (230/240 kD);





bullous pemphigoid antigen 1





isoform 1eA precursor; bullo





S0143
fatty acid
2194
2.3.1.85; OA-519; FASN;
EFVEQLRKEG
DRHPQALE
REVRQLTLR
1:5000-



synthase

MGC14367; MGC15706; fatty acid
VFAKEVR
AAQAELQQ
KLQELSSKA
1:30000





synthase
(183)
HD (184)
DE (185)





S0143P
fatty acid
2194
2.3.1.85; OA-519; FASN;
REVRQLTLRK
N/A
N/A
1:200-


3
synthase

MGC14367; MGC15706; fatty acid
LQELSSKADE


1:630





synthase
(186)





S0144
matrix metallo-
4323
MMP-X1; 3.4.24.-; MMP14;
AYIREGHEKQ
DEASLEPGY
RGSFMGSD
1:500-



proteinase 14

MTMMP1; MT1-MMP; membrane-
ADIMIFFAE
PKHIKELGR
EVFTYFYK
1:20000



(membrane-

type-1 matrix metalloprotein-
(187)
(188)
(189)



inserted)

ase; matrix metalloproteinase





14 preproprotein; MATRIX





METALLOPROTEINASE 14, MEM-





BRANE-TYPE; matrix metal-





loproteinase 14 (membrane-





inserted); membrane-type





matrix metalloprotein





S0147
cystatin A
1475
STF1; CSTA; STFA; cystatin AS;
MIPGGLSEAK
NETYGKLEA
DLVLTGYQV
1:100-



(stefin A)

cystatin A (stefin A)
PATPEIQEIV
VQYKTQ
DKNKDDELT
1:5000






(190)
(191)
GF (192)





S0149
transient recep-
55503
TRPV6; ECAC2; CAT1; CATL;
RQEHCMSEH
QGHKWGES
RACGKRVS
1:400-



tor potential

CALCIUM TRANSPORTER 1; CALCIUM
FKNRPACLGA
PSQGTQAG
EGDRNGSG
1:20000



cation channel,

TRANSPORTER-LIKE PROTEIN;
R (193)
AGK (194)
GGKWG



subfamily V,

EPITHELIAL CALCIUM CHANNEL 2


(195)



member 6

transient receptor potential





cation channel, subfamily V,





member 6





S0156
fatty acid
2173
B-FABP; FABP7; FABPB; MRG;
MVEAFCATW
QVGNVTKP
KVVIRTLSTF
1:100-



binding protein

mammary-derived growth inhibi-
KLTNSQN
TVIISQE
KNTE (198)
1:20000



7, brain

tor-related; FATTY ACID-
(196)
(197)





BINDING PROTEIN 7; FATTY ACID-





BINDING PROTEIN, BRAIN; fatty





acid binding protein 7, brain





S0158
cadherin 3, type
1001
CDHP; HJMD; PCAD; CDH3; pla-
RAVFREAEVT
QEPALFSTD
QKYEAHVP
1:150-



1, P-cadherin

cental cadherin; CADHERIN,
LEAGGAEQE
NDDFTVRN
ENAVGHE
1:2000



(placental)

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





cadherin (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;
KKIIEKMLNSD
N/A
N/A
1:100-



motif) ligand 1

GROa; GRO1, FORMERLY; GRO PRO-
KSN (202)


1:500



(melanoma growth

TEIN, ALPHA; GRO1 ONCOGENE,



stimulating ac-

FORMERLY; MELANOMA GROWTH



tivity, alpha)

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-
GKPGNQNSK
QAEAPLVPL
NCFLTERKA
1:22500-



repeat-contain-

containing 5 (survivin)
NEPPKKRER
SRQNK
QPDE (205)
1:30000



ing 5 (survivin)


ER (203)
(204)





S0193
procollagen-
5352
PLOD2; LYSYL HYDROXYLASE 2;
EFDTVDLSAV
NKEVYHEK
KQVDLENV
1:20000



lysine, 2-oxo-

LYSINE HYDROXYLASE 2
DVHPN (206)
DIKVFFDKA
WLDFIRE



glutarate 5-

PROCOLLAGEN-LYSINE, 2-

K (207)
(208)



dioxygenase

OXOGLUTARATE 5-DIOXYGENASE 2;



(lysine hydroxy-

procollagen-lysine, 2-oxoglu-



lase) 2

tarate 5-dioxygenase (lysine





hydroxylase) 2; procollagen-





lysine, 2-oxoglutarate 5-





dioxygenase (lysine hydroxy-





lase) 2 isoform





S0202
PTK7 protein
5754
PTK7; CCK4; protein-tyrosine
LKKPQDSQLE
KAKRLQKQ
KDRPSFSEI
1:500-



tyrosine kinase

kinase PTK7; colon carcinoma
EGKPGYLD
PEGEEPEM
ASALGDSTV
1:800



7

kinase-4; PTK7 protein tyro-
(209)
E (210)
DSKP (211)





sine kinase 7; PTK7 protein





tyrosine kinase 7 isoform e





precursor; PTK7 protein tyro-





sine kinase 7 isoform a pre-





cursor; PTK7 protein tyrosine





kinase 7 isoform d precursor;





S0211
cytochrome P450,
1549
CYPIIA7; P450-IIA4; 1.14.14.1;
KRGIEERIQE
DRVIGKNRQ
NPQHFLDD
1:500-



family 2, sub-

CPA7; CYP2A7; CPAD; CYTOCHROME
ESGFLIE
PKFEDRTK
KGQFKKSD
1:2500



family A, poly-

P450, SUBFAMILY IIA, POLYPEP-
(212)
(213)
(214)



peptide 7

TIDE 7; cytochrome P450, sub-





family IIA (phenobarbital-





inducible), polypeptide 7;





cytochrome P450, family 2,





subfamily A, polypeptide 7;





cytochrome P450, family 2, su





S0218
solute carrier
222962
SLC29A4; solute carrier family
RHCILGEWLP
KQRELAGN
RNAHGSCL
1:20-



family 29

29 (nucleoside transporters),
ILIMAVFN
TMTVSYMS
HASTANGSI
1:50



(nucleoside

member 4
(215)
(216)
LAGL (217)



transporters),



member 4





S0221
solute carrier
9153
HCNT2; SLC28A2; HsT17153;
ELMEKEVEPE
KARSFCKTH
KNKRLSGM
1:500-



family 28

SPNT1; CONCENTRATIVE NUCLEO-
GSKRTD
ARLFKK
EEWIEGEK
1:1200



(sodium-coupled

SIDE TRANSPORTER 2; SODIUM-
(218)
(219)
(220)



nucleoside

DEPENDENT PURINE NUCLEOSIDE



transporter),

TRANSPORTER 1; solute carrier



member 2

family 28 (sodium-coupled





nucleoside transporter),





member 2





S0223
angiopoietin-
51129
HFARP; FIAF; ANGPTL4; PGAR;
EGSTDLPLAP
KVAQQQRH
DHKHLDHE
1:30-



like 4

angiopoietin-like 4; FASTING-
ESRVDPE
LEKQHLR
VAKPARRK
1:10000





INDUCED ADIPOSE FACTOR; PPARG
(221)
(222)
RLPE (223)





ANGIOPOIETIN-RELATED PROTEIN;





HEPATIC FIBRINOGENIANGIOPOIE-





TIN-RELATED PROTEIN





S0235
carcinoembryonic
1048
CEACAM5; CD66e; carcinoembryo-
KLTIESTPFNV
KSDLVNEEA
KPVEDKDAV
1:500-



antigen-related

nic antigen-related cell adhe-
AEGKEC
TGQFRVYP
AFTCEPEAQ
1:4500



cell adhesion

sion molecule 5
(224)
ELPK (225)
(226)



molecule 5





S0237
podocalyxin-like
5420
podocalyxin-like; Gp200; PCLP;
DEKLISLICRA
KDKWDELK
DSWIVPLDN
1:1000-





PODXL; PODOCALYXIN-LIKE
VKATFNPAQD
EAGVSDMK
LTKDDLDEE
1:2000





PROTEIN; podocalyxin-like
K (227)
LGD (228)
EDTHL (229)





precursor





S0238
xenotropic and
9213
X3; XPR1; X RECEPTOR; SYG1,
EAVVTNELED
RRYRDTKR
KARDTKVLI
1:100-



polytropic re-

YEAST, HOMOLOG OF; xenotropic
GDRQKAMKR
AFPHLVNAG
EDTDDEANT
1:500



tro virus

and polytropic retrovirus
LR (230)
K (231)
(232)



receptor

receptor





S0241
glycyl-tRNA
2617
GlyRS; GARS; CMT2D; 6.1.1.14;
RKRVLEAKEL
RHGVSHKV
EARYPLFEG
1:500-



synthetase

SMAD1; GLYCYL-tRNA SYNTHETASE;
ALQPKDDIVD
DDSSGSIGR
QETGKKETI
1:7500





glycine tRNA ligase; Charcot-
(233)
RYAR (234)
EE (235)





Marie-Tooth neuropathy,





neuronal type, D





S0244
dachshund homo-
1602
DACH1; FLJ10138; dachshund
DLAGHDMGH
EKQVQLEKT
EADRSGGR
1:100-



log 1

homolog (Drosophila);
ESKRMHIEKD
ELKMDFLRE
TDAERTIQD
1:3000



(Drosophila)

DACHSHUND, DROSOPHILA,
E (236)
RE (237)
GR (238)





HOMOLOG OF; dachshund homolog





1 (Drosophila); dachshund





homolog 1 isoform a; dachshund





homolog 1 isoform b; dachshund





homolog 1 isoform c





S0251
transcription
29841
TFCP2L2; LBP-32; MGR; GRHL1;
EALYPQRRS
DYYKVPRE
DKYDVPHD
1:5400



factor CP2-like

mammalian grainyhead; LBP pro-
YTSEDEAWK
RRSSTAKPE
KIGKIFKKCK



2

tein 32; transcription CP2-
(239)
VE (240)
K (241)





like 2; leader-binding protein





32 isoform 2; leader-binding





protein 32 isoform 1





S0253
lysosomal as-
55353
LAPTM4B; lysosomal associated
DPDQYNFSS
EYIRQLPPN
DTTVLLPPY
1:500-



sociated protein

protein transmembrane 4 beta
SELGGDFEF
FPYRDD
DDATVNGA
1:2000



transmembrane


MDD (242)
(243)
AKE (244)



4 beta





S0255
cyclin E2
9134
CYCE2; CCNE2; cyclin E2; G1/S-
RREEVTKKH
KESRYVHD
DFFDRFMLT
1:1000-





specific cyclin E2; cyclin E2
QYEIR (245)
KHFEVLHSD
QKDINK
1:2000





isoform 2; cyclin E2 isoform

LE (246)
(247)





3; cyclin E2 isoform 1





S0260
nicastrin
23385
KIAA0253; nicastrin; NCSTN;
ESKHFTRDLM
ETDRLPRCV
ESRWKDIRA
1:2400-





APH2; ANTERIOR PHARYNX DEFEC-
EKLKGRTSR
RSTARLAR
RIFLIASKEL
1:5400





TIVE 2, C. ELEGANS, HOMOLOG OF
(248)
(249)
E (250)





S0265
FXYD domain
5349
MAT-8; MAT8; PLML; FXYD3;
KVTLGLLVFL
SEWRSSGE
KCKCKFGQ
1:400-



containing ion

phospholemman-like protein;
AGFPVLDAND
QAGR (252)
KSGHHPGE
1:1200



transport regu-

MAMMARY TUMOR, 8-KD; FXYD do-
LED (251)

(253)



lator 3

main-containing ion transport





regulator 3; FXYD domain con-





taining ion transport regula-





tor 3; FXYD domain containing





ion transport regulator 3 iso-





form 2 precursor; FXYD domai





S0267
immunoglobulin
3321
EWI-3; V8; IGSF3; immunoglobin
KVAKESDSVF
EREKTVTGE
KRAEDTAG
1:200-



superfamily,

superfamily, member 3; immuno-
VLKIYHLRQE
FIDKESKRP
QTALTVMRP
1:250



member 3

globulin superfamily, member 3
D (254)
K (255)
D (256)





S0270
signal trans-
10254
STAM2B; STAM2; DKFZp564C047;
KVARKVRALY
ETEVAAVDK
EIKKSEPEP
1:1000-



ducing adaptor

Hbp; STAM2A; SIGNAL-TRANSDUC-
DFEAVEDNE
LNVIDDDVE
VYIDEDKMD
1:9000



molecule (SH3

ING ADAPTOR MOLECULE 2; signal
(257)
(258)
R (259)



domain and ITAM

transducing adaptor molecule



motif) 2

2; STAM-like protein contain-





ing SH3 and ITAM domains 2;





signal transducing adaptor





molecule (SH3 domain and ITAM





motif) 2





S0273
dickkopf homolog
22943
DKK1; DKK-1; SK; dickkopf-1
DEECGTDEY
RGEIEETITE
N/A
1:400-



1 (Xenopus

like; dickkopf (Xenopus
CASPTRGGD
SFGNDHSTL

1:500




laevis)



laevis) homolog 1; dickkopf

(260)
D (261)





homolog 1 (Xenopus laevis);





DICKKOPF, XENOPUS, HOMOLOG OF,





1





S0280
solute carrier
65010
SLC26A6; solute carrier family
MDLRRRDYH
DTDIYRDVA
EFYSDALKQ
1:1800-



family 26,

26, member 6
MERPLLNQE
EYSEAKE
RCGVDVDF
1:2400



member 6


HLEE (262)
(263)
LISQKKK








(264)





S0286
WNT inhibitory
11197
WIF1; WIF-1; WNT inhibitory
DAHQARVLIG
ERRICECPD
KRYEASLIH
1:90



factor 1

factor 1; Wnt inhibitory
FEEDILIVSE
GFHGPHCE
ALRPAGAQL





factor-1 precursor
(265)
K (266)
R (267)





S0288
preferentially
23532
MAPE; PRAME; OPA-INTERACTING
KRKVDGLSTE
KEGACDELF
DIKMILKMV
1:1200



expressed anti-

PROTEIN 4; Opa-interacting
AEQPFIPVE
SYLIEKVKR
QLDSIEDLE



gen in melanoma

protein OIP4; preferentially
(268)
KK (269)
(270)





expressed antigen in melanoma;





melanoma antigen preferential-





ly expressed in tumors





S0295
prostaglandin E
9536
PGES; TP53112; MGST1L1;
RLRKKAFANP
RSDPDVER
RVAHTVAYL
1:100-



synthase

PP1294; PP102; PTGES;
EDALR (271)
CLRAHRND
GKLRAPIR
1:2400





MGC10317; PIG12; MGST1-L1;

(272)
(273)





MGST-IV; MGST1-like 1; p53-





INDUCED GENE 12; prostaglandin





E synthase; p53-induced apop-





tosis protein 12; prostaglan-





din E synthase isoform 2;





prostaglandin E synthase





isoform 1; micros





S0296
solute carrier
8140
SLC7A5; MPE16; D16S469E; CD98;
KRRALAAPAA
EAREKMLAA
MIWLRHRKP
1:300-



family 7

LAT1; 4F2 light chain; Mem-
EEKEEAR
KSADGSAP
ELERPIK
1:5000



(cationic amino

brane protein E16; L-TYPE
(274)
AGE (275)
(276)



acid transpor-

AMINO ACID TRANSPORTER 1;



ter, y+ system),

Solute carrier family 7, mem-



member 5

ber 5; solute carrier family 7





(cationic amino acid trans-





porter, y+ system), member 5





S0296P
solute carrier
8140
SLC7A5; MPE16; D165469E; CD98;
KRRALAAPAA
N/A
N/A
1:225-


1
family 7

LAT1; 4F2 light chain; Mem-
EEKEEAR


1:3150



(cationic amino

brane protein E16; L-TYPE
(277)



acid transpor-

AMINO ACID TRANSPORTER 1;



ter, y+ system),

Solute carrier family 7,



member 5

member 5; solute carrier





family 7 (cationic amino acid





transporter, y+ system),





member 5





S0297
v-maf
7975
FLJ32205; NFE2U; MAFK; NFE2,
KPNKALKVKK
KRVTQKEEL
RLELDALRS
1:333-



musculoaponeuro-

18-KD SUBUNIT; nuclear factor
EAGE (278)
ERQRVELQ
KYE (280)
1:800



tic fibrosarcoma

erythroid-2, ubiquitous (p18);

QEVEK (279)



oncogene homolog

NUCLEAR FACTOR ERYTHROID 2,



K (avian)

UBIQUITOUS SUBUNIT; v-maf mus-





culoaponeurotic fibrosarcoma





oncogene homolog K (avian);





v-maf avian musculoaponeurotic





fibrosarcoma oncogen





S0301
signal peptide,
57758
SCUBE2; signal peptide, CUB
KMHTDGRSC
KKGFKLLTD
KRTEKRLRK
1:3500-



CUB domain,

domain, EGF-like 2
LEREDTVLEV
EKSCQDVD
AIRTLRKAV
1:5400



EGF-like 2


TE (281)
E (282)
HRE (283)





S0303
gamma-amino-
2564
GABRE; GABA-A RECEPTOR, EPSI-
RVEGPQTES
EETKSTETE
KWENFKLEI
1:300-



butyric acid

LON POLYPEPTIDE; GAMMA-
KNEASSRD
TGSRVGKLP
NEKNSWKL
1:500



(GABA) A recep-

AMINOBUTYRIC ACID RECEPTOR,
(284)
E (285)
FQFD (286)



tor, epsilon

EPSILON; gamma-aminobutyric





acid (GABA) A receptor, epsi-





lon; gamma-aminobutyric acid





(GABA) A receptor, epsilon





isoform 2; gamma-aminobutyric





acid (GABA) A receptor,





epsilon is





S0305
S100 calcium
6281
CAL1L; GP11; p10; 42C;
DKGYLTKEDL
KDPLAVDKI
N/A
1:8332-



binding protein

S100A10; ANX2LG; CLP11; Ca[1];
RVLMEKE
MKDLDQCR

1:24996



A10 (annexin II

CALPACTIN I, p11 SUBUNIT; AN-
(287)
DGK (288)



ligand, calpac-

NEXIN II, LIGHT CHAIN; CAL-



tin I, light

PACTIN I, LIGHT CHAIN; S100



polypeptide

calcium-binding protein A10



(p11))

(annexin II ligand, calpactin





I, light polypeptide (p11));





S100 calcium binding protein





A10





S0311
v-myb myeloblas-
4605
MYBL2; MGC15600; MYB-RELATED
MSRRTRCED
EEDLKEVLR
RRSPIKKVR
1:750-



tosis viral on-

GENE BMYB; MYB-related protein
LDELHYQDTD
SEAGIELIIE
KSLALDIVD
1:5000



cogene homolog

B; v-myb myeloblastosis viral
SD (289)
DDIR (290)
ED (291)



(avian)-like 2

oncogene homolog (avian)-like





2; V-MYB AVIAN MYELOBLASTOSIS





VIRAL ONCOGENE HOMOLOG-LIKE 2





S0312
nucleoside
4860
NP; 2.4.2.1; nucleoside phos-
EDYKNTAEW
DERFGDRF
KVIMDYESL
1:1000-



phosphorylase

phorylase; PURINE-
LLSHTKHR
PAMSDAYD
EKANHEE
1:3600





NUCLEOSIDE: ORTHOPHOSPHATE
(292)
RTMRQR
(294)





RIBOSYLTRANSFERASE; purine

(293)





nucleoside phosphorylase; PNP





NUCLEOSIDE PHOSPHORYLASE





DEFICIENCY; ATAXIA WITH DE-





FICIENT CELLULAR IMMUNITY





S0314
chaperonin con-
22948
KIAA0098; CCT5; chaperonin
DQDRKSRLM
KGVIVDKDF
RMILKIDDIR
1:6000-



taining TCP1,

containing TCP1, subunit 5
GLEALKSHIM
SHPQMPKK
KPGESEE
1:30000



subunit 5

(epsilon)
AAK (295)
VED (296)
(297)



(epsilon)





S0315
non-metastatic
4830
GAAD; NME1; NDPKA; 2.7.4.6;
RLQPEFKPKQ
KFMQASED
DSVESAEKE
1:9000-



cells 1, protein

NM23-H1; AWD NM23H1B; GZMA-
LEGTMANCE
LLKEHYVDL
IGLWFHPEE
1:18000



(NM23A) ex-

ACTIVATED DNase; NUCLEOSIDE
R (298)
KDR (299)
LVD (300)



pressed in

DIPHOSPHATE KINASE-A; AWD,






DROSOPHILA, HOMOLOG OF;






METASTASIS INHIBITION FACTOR





NM23; nucleoside-diphosphate





kinase 1 isoform b; NONMETA-





STATIC PROTEIN 23, HOMOLOG 1;





nucleo





S0316
squalene
6713
SQLE; 1.14.99.7; squalene
KSPPESENKE
RDGRKVTVI
DHLKEPFLE
1:1000-



epoxidase

epoxidase; squalene
QLEARRRR
ERDLKEPDR
ATDNSHLR
1:10000





monooxygenase
(301)
(302)
(303)





S0319
pregnancy-
29948
OKL38; pregnancy-induced
DLEVKDWMQ
EYHKVHQM
RHQLLCFKE
1:900



induced growth

growth inhibitor; PREGNANCY-
KKRRGLRNS
MREQSILSP
DCQAVFQD



inhibitor

INDUCED GROWTH INHIBITOR OKL38
R (304)
SPYEGYR
LEGVEK







(305)
(306)





S0326
mal, T-cell
114569
MAL2; mal, T-cell differentia-
GPDILRTYSG
CSLGLALRR
N/A
1:120-



differentiation

tion protein 2
AFVCLE
WRP (308)

1:1200



protein 2


(307)





S0330
aldo-keto re-
1645
1.1.1.213; 2-ALPHA-HSD;
RYLTLDIFAG
N/A
N/A
1:2500-



ductase family

1.3.1.20; 20-ALPHA-HSD;
PPNYPFSDEY


1:75000



1, member C 1/2

MGC8954; H-37; HAKRC; MBAB;
(309)



(dihydrodiol

C9; DDH1; AKR1C1; trans-1,2-



dehydrogenase 1;

dihydrobenzene-1,2-diol dehy-



20-alpha (3-

drogenase; chlordecone reduc-



alpha)-hydroxy-

tase homolog; aldo-keto reduc-



steroid dehydro-

tase C; 20 alpha-hydroxyster-



genase)

oid dehydrogenase; hepatic





dihydrodiol





S0330-
aldo-keto re-
1645
1.1.1.213; 2-ALPHA-HSD;
RYLTLDIFAG
N/A
N/A
1:600


x1
ductase family

1.3.1.20; 20-ALPHA-HSD;
PPNYPFSDEY



1, member C 1/2

MGC8954; H-37; HAKRC; MBAB;
(310)



(dihydrodiol

C9; DDH1; AKR1C1; trans-1,2-



dehydrogenase 1;

dihydrobenzene-1,2-diol de-



20-alpha (3-

hydrogenase; chlordecone re-



alpha)-hydroxy-

ductase homolog; aldo-keto



steroid dehydro-

reductase C; 20 alpha-hydroxy-



genase)

steroid dehydrogenase; hepatic





dihydrodiol





S0331
aldo-keto re-
8644
HA1753; 1.1.1.188; DD3;
HYFNSDSFAS
N/A
N/A
1:300-



ductase family

hIuPGFS; HSD17B5; 1.3.1.20;
HPNYPYSDE


1:999



1, member C3 (3-

1.1.1.213; AKR1C3; KIAA0119;
Y (311)



alpha hydroxy-

HAKRB; HAKRe; trans-1,2-dihy-



steroid dehydro-

drobenzene-1,2-diol dehydro-



genase, type II)

genase; chlordecone reductase





homolog; dihydrodiol dehydro-





genase 3; prostaglandin F syn-





thase; ALDO-KETO REDUCTASE B;





3-





S0331-
aldo-keto re-
8644
HA1753; 1.1.1.188; DD3;
HYFNSDSFAS
N/A
N/A
1:150-


x1
ductase family

hIuPGFS; HSD17B5; 1.3.1.20;
HPNYPYSDE


1:300



1, member C3 (3-

1.1.1.213; AKR1C3; KIAA0119;
Y (312)



alpha hydroxy-

HAKRB; HAKRe; trans-1,2-dihy-



steroid dehydro-

drobenzene-1,2-diol dehydro-



genase, type II)

genase; chlordecone reductase





homolog; dihydrodiol dehydro-





genase 3; prostaglandin F syn-





thase; ALDO-KETO REDUCTASE B;





3-





S0332
aldo-keto re-
1645
1.1.1.213; 2-ALPHA-HSD;
RYVVMDFLM
N/A
N/A
1:300-



ductase family

1.3.1.20; 20-ALPHA-HSD;
DHPDYPFSD


1:400



1, member C4

MGC8954; H-37; HAKRC; MBAB;
EY (313)



(dihydrodiol

C9; DDH1; AKR1C1; trans-1,2-



dehydrogenase 1;

dihydrobenzene-1,2-diol dehy-



20-alpha (3-

drogenase; chlordecone reduc-



alpha)-hydroxy-

tase homolog; aldo-keto reduc-



steroid

tase C; 20 alpha-hydroxyster-



dehydrogenase)

oid dehydrogenase; hepatic





dihydrodiol





S0332-
aldo-keto re-
1645
1.1.1.213; 2-ALPHA-HSD;
RYVVMDFLM
N/A
N/A
1:75-


x1
ductase family

1.3.1.20; 20-ALPHA-HSD;
DHPDYPFSD


1:150



1, member C4

MGC8954; H-37; HAKRC; MBAB; C9;
EY (314)



(dihydrodiol

DDH1; AKR1C1; trans-1,2-di-



dehydrogenase 1;

hydrobenzene-1,2-diol dehydro-



20-alpha (3-

genase; chlordecone reductase



alpha)-hydroxy-

homolog; aldo-keto reductase



steroid

C; 20 alpha-hydroxysteroid de-



dehydrogenase)

hydrogenase; hepatic





dihydrodiol





S0336
chromosome 20
140809
C20orf139; chromosome 20 open
DPAKVQSLV
RETIPAKLV
N/A
1:1600-



open reading

reading frame 139
DTIREDPD
QSTLSDLR

1:2400



frame 139


(315)
(316)





S0342
solute carrier
154091
SLC2A12; solute carrier family
SDTTEELTVIK
N/A
N/A
1:400-



family 2 (facil-

2 (facilitated glucose trans-
SSLKDE


1:1250



itated glucose

porter), member 12
(317)



transporter),



member 12





S0343
solute carrier
154091
SLC2A12; solute carrier family
HSRSSLMPLR
N/A
N/A
1:50-



family 2 (facil-

2 (facilitated glucose trans-
NDVDKR


1:125



itated glucose

porter), member 12
(318)



transporter),



member 12





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






YFPTKPMFVI


1:300






A (319)





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






LEATRRQRM


1:300






D (320)





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



acyl-CoA

thioesterase 2B
SSIP (321)


1:600



thioesterase 2B





S0374
chloride intra-
53405
CLIC5; chloride intracellular
DANTCGEDK
N/A
N/A
1:5000-



cellular channel

channel 5
GSRRKFLDG


1:9000



5


DE (322)





S0380
keratinocyte
200634
KRTCAP3; keratinocyte associ-
QLEEMTELES
N/A
N/A
1:2000-



associated

ated protein 3
PKCKRQENE


1:9000



protein 3


Q (323)





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



(ARHGEF) and

chondrocyte-derived ezrin-like
TEEEEEVVKD



pleckstrin do-

protein; FERM, RhoGEF, and
R (324)



main protein 1

pleckstrin domain protein 1;



(chondrocyte-

FERM, ARHGEF, AND PLECKSTRIN



derived)

DOMAIN-CONTAINING PROTEIN 1;





FERM, RhoGEF (ARHGEF) and





pleckstrin domain protein 1





(chondrocyte-derived)





S0388
trichorhino-
7227
GC79; TRPS1; TRPS1 GENE;
SGDSLETKED
N/A
N/A
1:600



phalangeal

trichorhinophalangeal syndrome
QKMSPKATE



syndrome I

I; zinc finger transcription
E (325)





factor TRPS1





S0396
cytochrome,
1576
1.14.14.1; HLP; CYP3A3;
RKSVKRMKE
N/A
N/A
1:15



P450, family 3,

CYP3A4; P450C3; NF-25; CP33;
SRLEDTQKH



subfamily A,

CP34; P450-III, STEROID-INDUC-
RV (326)



polypeptide 4

IBLE; nifedipine oxidase; glu-





cocorticoid-inducible P450;





CYTOCHROME P450PCN1; P450,





FAMILY III; P450-III, steroid





inducible; cytochrome P450,





subfamily IIIA, polypeptide 4;





S0398
FAT tumor sup-
2195
CDHF7; FAT; cadherin ME5; FAT
KIRLPEREKP
N/A
N/A
1:45-



pressor homolog

tumor suppressor precursor;
DRERNARRE


1:200



1 (Drosophila)

cadherin-related tumor sup-
P (327)





pressor homolog precursor;





cadherin family member 7 pre-





cursor; homolog of Drosophila





Fat protein precursor; FAT





tumor suppressor homolog 1





(Drosophila); FAT TUMOR





SUPPRESS





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





PROGRANULIN; PEPI; PCDGF;
APRWDAPLR


1:3000





granulin; GRN; EPITHELIN
DP (328)





PRECURSOR





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



regulated gene 1

PROXY1; NDRG1; GC4; NMSL;
EGTRSRSHT


1:900





TDD5; RIT42; NDR1; differenti-
SE (329)





ation-related gene 1 protein;





nickel-specific induction





protein Cap43; protein regu-





lated by oxygen-1; NMYC DOWN-





STREAM-REGULATED GENE 1; re-





ducing agents and tunicamycin-





respon





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



ing protein 5

PA-FABP; FATTY ACID-BINDING
QTVCNFTD



(psoriasis-

PROTEIN, EPIDERMAL; FATTY
(330)



associated)

ACID-BINDING PROTEIN 5; FATTY





ACID-BINDING PROTEIN, PSORIA-





SIS-ASSOCIATED; fatty acid





binding protein 5 (psoriasis-





associated)





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



kinase inhibitor

BWS; Beckwith-Wiedemann syn-
KSSGDVP



1C (p57, Kip2)

drome; cyclin-dependent kinase
(331)





inhibitor 1C (p57, Kip2)





S0414
alpha-methyl-
23600
AMACR; 5.1.99.4; ALPHA-METHYL-
RVDRPGSRY
N/A
N/A
1:100



acyl-CoA

ACYL-CoA RACEMASE; AMACR DE-
DVSRLGRGK



racemase

FICIENCY; AMACR ALPHA-METHYL-
RS (332)





ACYL-CoA RACEMASE DEFICIENCY;





alpha-methylacyl-CoA racemase





isoform 1; alpha-methylacyl-





CoA racemase isoform 2





S0415
gamma-amino-
2562
MGC9051; GABRB3; GABA-A
ETVDKLLKGY
N/A
N/A
1:600-



butyric acid

RECEPTOR, BETA-3 POLYPEPTIDE;
DIRLRPD


1:1800



(GABA) A recep-

GAMMA-AMINOBUTYRIC ACID
(333)



tor, beta 3

RECEPTOR, BETA-3; gamma-





aminobutyric acid (GABA) A





receptor, beta 3; gamma-amino-





butyric acid (GABA) A recep-





tor, beta 3 isoform 2 precur-





sor; gamma-aminobutyric acid





(GABA) A rece





S0417
HSV-1 stimula-
22879
HSRG1; KIAA0872; HSV-1 stim-
APGGAEDLE
N/A
N/A
1:9000



tion-related

ulation-related 1; HSV-1 stim-
DTQFPSEEAR



gene 1

ulation-related gene 1
E (334)





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



factor receptor

related death receptor 6;
PRQDPSAIVE



superfamily,

tumor necrosis factor receptor
(335)



member 21

superfamily, member 21; tumor





necrosis factor receptor





superfamily, member 21





precursor





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



containing 1C

containing 1C; THYROID HORMONE
SESKSEQRTK





RECEPTOR INTERACTOR 8
R (336)





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



open reading

reading frame 140
LPRARGERR


1:300



frame 140


RH (337)





S0440
cell division
994
3.1.3.48; CDC25B; cell divi-
RLERPQDRD
N/A
N/A
1:350-



cycle 25B

sion cycle 25B; cell division
TPVQNKRRR


1:3600





cycle 25B isoform 4; cell di-
S (338)





vision cycle 25B isoform 5;





cell division cycle 25B iso-





form 1; cell division cycle





25B isoform 2; cell division





cycle 25B isoform 3





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





LAXA-MARFANOID SYNDROME;
RESQFKEKQ


1:1800





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





1 precursor; LAMB1 NEONATAL





CUTIS LAXA WITH MARFANOID





PHENOTYPE





S0447
papillary renal
5546
TPRC; MGC17178; MGC4723; PRCC;
DEAFKRLQG
N/A
N/A
1:4000-



cell carcinoma

proline-rich protein PRCC;
KRNRGREE


1:6000



(translocation-

RCCP1 PRCC/TFE3 FUSION GENE;
(340)



associated)

papillary renal cell carcinoma





(translocation-associated);





RENAL CELL CARCINOMA, PAPIL-





LARY, 1 GENE; papillary renal





cell carcinoma translocation-





associated gene product





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



factor (ligand)

Apo-2 ligand; APO2 LIGAND;
KNDKQ (341)



superfamily,

TNF-RELATED APOPTOSIS-INDUCING



member 10

LIGAND; TNF-related apoptosis





inducing ligand TRAIL; tumor





necrosis factor (ligand)





superfamily, member 10; TUMOR





NECROSIS FACTOR LIGAND SUPER-





FAMILY, MEMBER 10





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





CMPD4; TTN; FLJ32040; CMH9,
TKCNKKTLTD


1:8100





included; titin isoform N2-A;
(342)





titin isoform N2-B; titin iso-





form novex-1; titin isoform





novex-2; titin isoform novex-





3; cardiomyopathy, dilated 1G





(autosomal dominant); TTN





CARDIOMYOPATHY, FAMILIAL





S0469
DNA fragmenta-
1676
DFF45; DFF1; DFFA; ICAD; DFF-
KEGSLLSKQE
N/A
N/A
1:600



tion factor, 45

45; INHIBITOR OF CASPASE-
ESKAAFGEE



kDa, alpha poly-

ACTIVATED DNase; DNA FRAGMEN-
(343)



peptide

TATION FACTOR, 45-KD, ALPHA





SUBUNIT; DNA fragmentation





factor, 45 kDa, alpha poly-





peptide; DNA fragmentation





factor, 45 kD, alpha subunit;





DNA fragmentation factor, 45





kD, alp





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



apoptosis-

2; ICH-1 protease; caspase 2
KMRLPTRSD



related cysteine

isoform 3; caspase 2 isoform
(344)



protease (neural

4; NEDD2 apoptosis regulatory



precursor cell

gene; caspase 2 isoform 2 pre-



expressed, de-

cursor; caspase 2 isoform 1



velopmentally

preproprotein; NEURAL PRECUR-



down-regulated

SOR CELL EXPRESSED, DEVELOP-



2)

MENTALLY DOWNREGULATED 2;





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



transition 1

YEAST, HOMOLOG OF; PEPTIDE
VGRAYFETEK





CHAIN RELEASE FACTOR 3A; G1-
(345)





TO S-PHASE TRANSITION 1; G1 to





S phase transition 1





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



control of

control of amino-acid synthe-
VPEKR



amino-acid

sis, yeast, homolog)-like 2;
(346)



synthesis 5-

GCN5 general control of amino-



like 2 (yeast)

acid synthesis 5-like 2





(yeast); General control of





amino acid synthesis, yeast,





homolog-like 2





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



replication

inhibitor
YEALKENEK



inhibitor


(347)





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



factor-like 6

factor-like 6 interacting
NSDEVMKKP


1:9000



interacting

protein 2
(348)



protein 2





S0511
DNA replication
51659
Pfs2; DNA replication complex
PKADEIRTLV
N/A
N/A
1:2000



complex GINS

GINS protein PSF2
KDMWDTR



protein PSF2


(349)





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



domain 10

10
DFGVKKARTE






(350)





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



channel tetra-

tetramerisation domain
CQEDPELDS


1:1500



merisation do-

containing 2
(351)



main containing



2





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






SDDVDYRGS


1:1200






(352)





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



rich) nuclear

nuclear phosphoprotein 32
DTEKELEF



phosphoprotein

family, member E
(353)



32 family,



member E





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



open reading

r eading frame 100
RACTARE


1:240



frame 100


(354)





S0545
Hpall tiny
27037
D22S1733E; HTF9C; Hpall tiny
ERKQLECEQ
N/A
N/A
1:900-



fragments locus

fragments locus 9C; Hpall tiny
VLQKLAKE


1:5400



9C

fragments locus 9C isoform2;
(355)





Hpall tiny fragments locus 9C





isoform 1





S0546
cell division
157313
CDCA2; cell division cycle
RNSETKVRR
N/A
N/A
1:1200



cycle associated

associated 2
STRLQKDLEN



2


(356)





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



phoprotein 44

NUCLEOPORIN, 35-KD; mitotic
TPKKDE


1:5400





phosphoprotein 44
(357)





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



maintenance of

some-associated polypeptide C;
QELKE (358)



chromosomes 4-

SMC4 (structural maintenance



like 1 (yeast)

of chromosomes 4, yeast)-like





1; SMC4 structural maintenance





of chromosomes 4-like 1





(yeast); structural mainten-





ance of chromosomes (SMC)





family member, chromosome-ass





S0564
phosphatidyl-
9791
KIAA0024; PSSA; PTDSS1;
DDVNYKMHF
N/A
N/A
1:1000-



serine synthase

phosphatidylserine synthase 1
RMINEQQVE


1:8000



1


D (359)





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



1 (Drosophila)

like kinase (Drosophila); polo
KEEPVVR


1:100





(Drosophia)-like kinase;
(360)





SERINE/THREONINE PROTEIN





KINASE 13; polo-like kinase 1





(Drosophila)





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






RVRRSIGRPE






(361)





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



cassette, sub-

EST111653; ABC transporter 3;
EDSDPEKALR



family A(ABC1),

ATP-binding cassette 3; ATP-
(362)



member 3

BINDING CASSETTE TRANSPORTER





3; ATP-BINDING CASSETTE, SUB-





FAMILY A, MEMBER 3; ATP-bind-





ing cassette, sub-family A





member 3; ATP-binding cas-





sette, subfamily A (ABC1),





memb





S0579
ATP-binding
10347
ABCX; ABCA7; ABCA-SSN; autoan-
EKADTDMEG
N/A
N/A
1:300-



cassette, sub-

tigen SS-N; macrophage ABC
SVDTRQEK


1:400



family A (ABC1),

transporter; SJOGREN SYNDROME
(363)



member 7

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
22
ABCB7; Atm1p; ASAT; ABC7;
RVQNHDNPK
N/A
N/A
1:4000-



cassette, sub-

E5T140535; ABC TRANSPORTER 7;
WEAKKENISK


1:10000



family B

ATP-binding cassette 7; ATP-
(364)



(MDR/TAP),

BINDING CASSETTE TRANSPORTER



member 7

7; Anemia, sideroblastic, with





spinocerebellar ataxia; ATP-





BINDING CASSETTE, SUBFAMILY





B, MEMBER 7; ATP-binding cas-





sette, sub-family B, member





S0585
ATP-binding
94160
MRP9; ABCC12; MULTIDRUG
RSPPAKGAT
N/A
N/A
1:500



cassette, sub-

RESISTANCE-ASSOCIATED PROTEIN
GPEEQSDSL



family C

9; ATP-BINDING CASSETTE,
K (365)



(CFTR/MRP),

SUBFAMILY C, MEMBER 12; ATP-



member 12

binding cassette, sub-family C





(CFTR/MRP), member 12





S0586
ATP-binding
9429
ABC15; MXR1; ABCP; EST157481;
REEDFKATEII
N/A
NA
1:333-



cassette, sub-

MRX; ABCG2; BCRP1; BMDP;
EPSKQDKP
1:400



family G

MITOXANTRONE-RESISTANCE
(366)



(WHITE), member

PROTEIN; mitoxantrone resis-



2

tance protein; placenta speci-





fic MDR protein; ATP-BINDING





CASSETTE TRANSPORTER,





PLACENTA-SPECIFIC; breast can-





cer resistance protein; ATP-





BINDING CASS





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



organic anion

SLCO1B3; ORGANIC ANION TRANS-
NSCGAQ


1:2400



transporter

PORTER 8; solute carrier or-
(367)



family, member

ganic anion transporter fam-



1B3

ily, member 1B3; SOLUTE CAR-





RIER FAMILY 21, MEMBER 8 (OR-





GANIC ANION TRANSPORTER);





solute carrier family 21





(organic anion transporter),





member 8





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



family 22 (or-

SLC22A6; PAH TRANSPORTER;
LEVWL (368)



ganic anion

para-aminohippurate transpor-



transporter),

ter; renal organic anion



member 6

transporter 1; solute carrier





family 22 member 6 isoform b;





solute carrier family 22 mem-





ber 6 isoform c; solute car-





rier family 22 member 6





isoform





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



family 35 (UDP-

UGT1; UDP-galactose translo-
(369)



galactose

cator; UDP-GALACTOSE TRANS-



transporter),

PORTER, ISOFORM 2; UGALT UDP-



member A2

GALACTOSE TRANSPORTER, ISOFORM





1; solute carrier family 35





(UDP-galactose transporter),





member A2; solute carrier





family 35 (UDP-galactose





transpo





S0607
cell division
994
3.1.3.48; CDC25B; cell divi-
RKSEAGSGA
N/A
N/A
1:1800



cycle 25B

sion cycle 25B; cell division
ASSSGEDKE





cycle 25B isoform 4; cell di-
N (370)





vision cycle 25B isoform 5;





cell division cycle 25B iso-





form 1; cell division cycle





25B isoform 2; cell division





cycle 25B isoform 3





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



desaturase

stearoyl-CoA desaturase
KEGPSPKVE


1:5000



(delta-9-

(delta-9-desaturase); fatty
(371)



desaturase)

acid desaturase





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



activated pro-

GAMMA; PRKM12; MAPK12; ERK3;
KGLPELEKKD



tein kinase 12

ERK6; EXTRACELLULAR SIGNAL-
(372)





REGULATED KINASE 6; mitogen-





activated protein kinase 3;





stress-activated protein





kinase 3; mitogen-activated





protein kinase 12





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



of kappa light

LYT 10; TRANSCRIPTION FACTOR
QRELEQEAK



polypeptide gene

NFKB2; NFKB, p52/p100 SUBUNIT;
(373)



enhancer in B-

LYMPHOCYTE TRANSLOCATION



cells 2

CHROMOSOME 10; NUCLEAR FACTOR



(p49/p100)

KAPPA-B, SUBUNIT 2; Nuclear





factor of kappa light chain





gene enhancer in B-cells 2;





nuclear factor of kappa I





S0613
tumor necrosis
958
Bp50; TNFRSF5; MGC9013; CDW40;
RVQQKGTSE
N/A
N/A
1:250-



factor receptor

CD40 antigen; CD40L receptor;
TDTIC (374)


1:270



superfamily,

B CELL-ASSOCIATED MOLECULE



member 5

CD40; CD40 type II isoform; B





cell surface antigen CD40;





nerve growth factor receptor-





related B-lymphocyte activa-





tion molecule; tumor necrosis





factor receptor superfam





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



virus induced

EPSTEIN-BARR VIRUS-INDUCED
LQVQWE


1:3000



gene 3

GENE 3; INTERLEUKIN 27, EBI3
(375)





SUBUNIT; Epstein-Barr virus





induced gene 3; Epstein-Barr





virus induced gene 3 precursor





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



protein 339

339
SWDELPDEK






R (376)





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



protein

protein
HRDRLRSK






(377)





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



phosphatase,

LAR-interacting protein 1;
LSHEEDLAK



receptor type,

PTPRF interacting protein al-
(378)



f polypeptide

pha 1 isoform a; PTPRF inter-



(PTPRF), inter-

acting protein alpha 1 isoform



acting protein

b; protein tyrosine phospha-



(liprin), alpha

tase, receptor type, f poly-



1

peptide (PTPRF), interacting





protein (liprin), alpha 1





S0631
RGM domain fam-
56963
RGMA; REPULSIVE GUIDANCE
SQERSDSPEI
N/A
N/A
1:600



ily, member A

MOLECULE; RGM domain family,
CHYEKSFHK





member A
(379)





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



protein

protein LOC144347
RCNSEVK


1:200



LOC144347


(380)





S0639
tetratricopep-
57217
TTC7; tetratricopeptide repeat
RELREVLRTV
N/A
N/A
1:2000-



tide repeat

domain 7
ETKATQN


1:3000



domain 7


(381)





S0640
protein C (in-
5624
PROC; 3.4.21.69; PROC DEFICI-
RDTEDQEDQ
N/A
N/A
1:1000-



activator of

ENCY PROTEIN C; THROMBOPHILIA,
VDPRLIDGK


1:1800



coagulation fac-

HEREDITARY, DUE TO PC
(382)



tors Va and

DEFICIENCY; PROTEIN C DEFICI-



VIIIa)

ENCY, CONGENITAL THROMBOTIC





DISEASE DUE TO; protein C (in-





activator of coagulation fac-





tors Va and VIIIa)





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



enhancer of

TLE3; transducin-like enhancer
ERESSAN


1:1440



split 3 (E(sp1)

protein 3; enhancer of split
(383)



homolog,

groucho 3; transducin-like




Drosophila)


enhancer of split 3 (E(sp1)





homolog, Drosophila)





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



7 (Drosophila)

frizzled homolog 7
PFSCPRQ





(Drosophila); Frizzled,
(384)






drosophila, homolog of, 7






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



family 3 (acti-

4F2HC; 4F2 HEAVY CHAIN; CD98
QSAKK (385)


1:5400



vators of di-

HEAVY CHAIN; CD98 MONOCLONAL



basic and neu-

ANTIBODY 44D7; ANTIGEN DEFINED



tral amino acid

BY MONOCLONAL ANTIBODY 4F2,



transport), mem-

HEAVY CHAIN; antigen identi-



ber 2

fied by monoclonal antibodies





4F2, TRA1.10, TROP4, and T43;





SOLUTE CARRIER FAMILY 3





S0648
KIAA0738 gene
9747
KIAA0738; KIAA0738 gene
EYRNQTNLPT
N/A
N/A
1:200



product

product
ENVDK (386)





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



receptor 1,

PLA2G1R; PHOSPHOLIPASE A2 RE-
ALRSCQADN



180 kDa

CEPTOR, 180-KD; phospholipase
(387)





A2 receptor 1, 180 kDa





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






REPAPLDK






(388)





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



2, mitochondrial

MITOCHONDRIAL; thymidine
ENRK (389)





kinase 2, mitochondrial





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



open reading

reading frame 135
EKWK (390)



frame 135





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





adrenal gland protein AD-005;
TFLSDLR


1:3000





KIAA1007 protein isoform a;
(391)





KIAA1007 protein isoform b





S0670
DKFZP566O1646
25936
DC8; DKFZP566O1646 protein
KCRGETVAK
N/A
N/A
1:900



protein


EISEAMKS






(392)





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



CLL/lymphoma 7A

B-cell CLL/lymphoma 7A
PIGLSGD






(393)





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



of mouse nin

antigen recognized by T
HSACEPEN



one binding

lymphocytes 4; likely ortholog
(394)



protein

of mouse nin one binding





protein





S0676
guanine nucleo-
2768
RMP; NNX3; GNA12; GUANINE
ERRAGSGAR
N/A
N/A
1:1200-



tide binding

NUCLEOTIDE-BINDING PROTEIN,
DAERE (395)


1:2400



protein (G pro-

ALPHA-12; guanine nucleotide



tein) alpha 12

binding protein (G protein)





alpha 12





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



mitochondrial

MITOCHONDRIAL GrpE PROTEIN;
EKTLLE


1:1000



(E. coli)

GrpE-like 1, mitochondrial
(396)





(E. coli); GrpE, E. COLI,





HOMOLOG OF, 1





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



protein FLJ34922

FLJ34922
GTLRIKKYLT






(397)





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



protein FLJ20457

FLJ20457
LTQKQYE


1:1260






(398)





S0691
solute carrier
23657
CCBR1; SLC7A11; xCT; cystine/
QNFKDAFSG
N/A
N/A
1:1000-



family 7, (ca-

glutamate transporter; SYSTEM
RDSSITR


1:1575



tionic amino

Xc(−) TRANSPORTER-RELATED
(399)



acid transpor-

PROTEIN; SOLUTE CARRIER FAMILY



ter, y+ system)

7, MEMBER 11; solute carrier



member 11

family 7, (cationic amino acid





transporter, y+ system) member





11





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



cysteine ligase,

GAMMA-GLUTAMYLCYSTEINE SYNTHE-
SDHFEN


1:400



catalytic

TASE, CATALYTIC SUBUNIT; glu-
(400)



subunit

tamate-cysteine ligase, cata-





lytic subunit





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





integrin, beta 4
LQGER (401)


1:4050





S0702
solute carrier
8140
SLC7A5; MPE16; D165469E; CD98;
KGDVSNLDP
N/A
N/A
1:21160-



family 7 (ca-

LAT1; 4F2 light chain; Mem-
NFSFEGTKLD


1:178200



tionic amino

brane protein E16; L-TYPE
V (402)



acid transpor-

AMINO ACID TRANSPORTER 1;



ter, y+ system),

Solute carrier family 7, mem-



member 5

ber 5; solute carrier family 7





(cationic amino acid transpor-





ter, y+ system), member 5





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



metastasis sup-

cancer metastasis-suppressor
KRKSDGP


1:2000



pressor 1

1; breast cancer metastasis
(403)





suppressor 1





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



metastasis-

metastasis-suppressor; KISS 1
LVQREKD



suppressor

METASTIN; malignant melanoma
(404)





metastasis-suppressor; KISS1





METASTASIS SUPPRESSOR





S0708
cofactor re-
9439
DKFZp434H0117; CRSP133; SUR2;
SVKEQVEKIIC
N/A
N/A
1:2430



quired for Sp1

DRIP130; CRSP3; mediator;
NLKPALK



transcriptional

transcriptional co-activator
(138)



activation, sub-

CRSP130; CRSP, 130-KD SUBUNIT;



unit 3, 130 kDa

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

cytokeratin 14; CK 14; KERA-



bullosa simplex,

TIN, TYPE I CYTOSKELETAL 14;



Dowling-Meara,

keratin 14 (epidermolysis



Koebner)

bullosa simplex, Dowling-





Meara, Koebner)





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





K17; CYTOKERATIN 17; VERSION

1:25





1; CK 17; KERATIN, TYPE I





CYTOSKELETAL 17





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





18; CK 18; KERATIN, TYPE I

1:400





CYTOSKELETAL 18





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





18; CK 18; KERATIN, TYPE I
Dickinson
1:100





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; GASTRO-





INTESTINAL TUMOR-ASSOCIATED





ANTIGEN 2, 35-KD GLYCOPROTEIN;





tumor-associated calcium





signal transducer 1 precurso





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



tor 2 (ER beta)

ESRB; NR3A2; ESTROGEN RECEP-
Research Products (Calbiochem)





TOR, BETA; estrogen receptor 2





(ER beta)





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



transmembrane

PUM; H23AG; CD227; PEM;
Cancer Research Technology (ICRT)





CARCINOMA-ASSOCIATED MUCIN;





H23 antigen; TUMOR-ASSOCIATED





MUCIN; DF3 antigen; peanut-





reactive urinary mucin;





mucin 1, transmembrane; poly-





morphic epithelial mucin;





MUCIN 1, URINARY; MUCIN,





TUMOR-ASSOCIATE





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



ceptor

TRFR; ANTIGEN CD71; TRANSFER-



(p90, CD71)

RIN RECEPTOR PROTEIN; trans-





ferrin 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

NEU PROTO-ONCOGENE; ONCOGENE



2, neuro/

ERBB2; RECEPTOR PROTEIN-



glioblastoma

TYROSINE KINASE ERBB-2



derived oncogene

PRECURSOR; ONCOGENE NGL,



homolog (avian)

NEUROBLASTOMA- OR





GLIOBLASTOMA-DERIVED; TYROSINE





KINASE-TYPE CELL





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



protein

RESISTANCE-RELATED PROTEIN





MAJOR VAULT PROTEIN, RAT,





HOMOLOG OF





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



p73-like

EEC3; TP63; p51; TUMOR PRO-





TEIN p63; TUMOR PROTEIN p73-





LIKE; p53-RELATED PROTEIN p63;





tumor protein 63 kDa with





strong homology to p53





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



ceptor 1

ALPHA; ESRA; ESTRADIOL RECEP-





TOR; 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

NEU PROTO-ONCOGENE; ONCOGENE



2, neuro/

ERBB2; RECEPTOR PROTEIN-



glioblastoma

TYROSINE KINASE ERBB-2



derived oncogene

PRECURSOR; ONCOGENE NGL,



homolog (avian)

NEUROBLASTOMA- OR





GLIOBLASTOMA-DERIVED; TYROSINE





KINASE-TYPE CELL





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



(lysosomal

3.4.23.5; cathepsin D prepro-

1:50



aspartyl

protein; Cathepsin D precur-



protease)

sor; 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 myelo-
4609
c-Myc; MYC; ONCOGENE MYC; Myc
Antibody obtained from Dako
1:50



cytomatosis

proto-oncogene protein;



viral oncogene

PROTOONCOGENE HOMOLOGOUS TO



homolog (avian)

MYELOCYTOMATOSIS VIRUS; v-myc





myelocytomatosis viral onco-





gene homolog (avian); v-myc





avian myelocytomatosis viral





oncogene homolog; Avian myelo-





cytomatosis viral (v-myc) onco





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



1, E-cadherin

CDHE; Uvomorulin; LCAM;

1:150



(epithelial)

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-trans-





ferase pi; GST, CLASS PI;





deafness, X-linked 7; GLUTA-





THIONE S-TRANSFERASE 3;





GLUTATHIONE S-TRANSFERASE, PI;





FAEES3 GLUTATHIONE S-





TRANSFERASE PI PSEUD





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



p53 (Li-Fraumeni

TEIN P53; TRANSFORMATION-



syndrome)

RELATED PROTEIN 53; TUMOR





SUPPRESSOR P53; CELLULAR





TUMOR ANTIGEN P53; tumor





protein p53 (Li-Fraumeni





syndrome)





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



receptor

RESISTANCE; PSEUDOCORPUS





LUTEUM INSUFFICIENCY





PROGESTERONE RECEPTOR





S5077
trefoil factor 1
7031

Antibody obtained from Dako
1:50-



(breast cancer,



1:100



estrogen-



inducible se-



quence



expressed in)





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



(gamma,

glycerate hydro-lyase; ENO-



neuronal)

LASE, 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 CLL/
596
BCL2; FOLLICULAR LYMPHOMA;
Antibody obtained from Dako
1:50



lymphoma 2

APOPTOSIS REGULATOR BCL-2;





B-cell CLL/lymphoma 2; B-cell





ymphoma 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; Retinoblas-
Antibody obtained from Dako
1:20



(including

toma-1; RB; RB1; RETINOBLAS-



osteosarcoma)

TOMA-ASSOCIATED PROTEIN; RB





OSTEOSARCOMA, RETINOBLASTOMA-





RELATED; retinoblastoma 1





(including osteosarcoma)





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





synaptic vesicle protein P38





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



X protein

tein; APOPTOSIS REGULATOR BAX,





MEMBRANE ISOFORM ALPHA





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



tor 2 (ER beta)

ESRB; NR3A2; ESTROGEN RECEP-





TOR, BETA; estrogen receptor 2





(ER beta)





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



transmembrane

PUM; 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 recep-
2099
ER; NR3A1; ESR1; Era; ESR; ER-
Antibody obtained from US Labs
1:1



tor 1

ALPHA; ESRA; ESTRADIOL RECEP-





TOR; ESTROGEN RECEPTOR, ALPHA;





estrogen receptor 1 (alpha)





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



receptor

RESISTANCE; PSEUDOCORPUS





LUTEUM INSUFFICIENCY





PROGESTERONE RECEPTOR





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



erythroblastic

C-ERBB-2; NEU; MLN 19;



leukemia viral

EC 2.7.1.112; TKR1 HERSTATIN;



oncogene homolog

NEU PROTO-ONCOGENE; ONCOGENE



2, neuro/

ERBB2; RECEPTOR PROTEIN-



glioblastoma

TYROSINE KINASE ERBB-2



derived oncogene

PRECURSOR; ONCOGENE NGL,



homolog (avian)

NEUROBLASTOMA- OR





GLIOBLASTOMA-DERIVED; TYROSINE





KINASE-TYPE CELL





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



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

CYTOKERATIN 5; CK 5; 58 KDA



bullosa simplex,

CYTOKERATIN; KERATIN, TYPE II



Dowling-Meara/

CYTOSKELETAL 5; keratin 5



Kobner/Weber-

(epidermolysis bullosa sim-



Cockayne types)

plex, Dowling-Meara/Kobner/





Weber-Cockayne types)





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



p53 (Li-Fraumeni

TEIN P53; TRANSFORMATION-



syndrome)

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; ON-
Antibody obtained from US Labs
1:1



factor receptor

COGENE ERBB; ERBB1 SPECIES



(erythroblastic

ANTIGEN 7; V-ERB-B AVIAN



leukemia viral

ERYTHROBLASTIC LEUKEMIA VIRAL



(v-erb-b) onco-

ONCOGENE HOMOLOG; epidermal



gene homolog,

growth factor receptor (avian



avian)

erythroblastic leukemia viral





(v-erb-b) oncogene homolog)





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



(gamma,

glycerate hydro-lyase; ENO-



neuronal)

LASE, GAMMA; neurone-specific





enolase; ENOLASE, NEURN-





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
7080
benign chorea; chorea, heredi-
Antibody obtained from US Labs
1:1



transcription

tary benign; NK-2 (Drosophila)



factor 1

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

NEU PROTO-ONCOGENE; ONCOGENE



2, neuro/

ERBB2; RECEPTOR PROTEIN-



glioblastoma

TYROSINE KINASE ERBB-2



derived oncogene

PRECURSOR; ONCOGENE NGL,



homolog (avian)

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. A method for selecting a chemotherapy for a breast cancer patient comprising steps of: providing a cancer sample from a breast cancer patient;detecting the presence of expression of TLE3 in the cancer sample; andselecting a taxane or taxane derivative for chemotherapy of the cancer patient when the TLE3 expression is present.
  • 2. The method of claim 1, wherein the step of detecting comprises steps of: providing a negative control sample;detecting a level of TLE3 expression in the negative control sample;detecting a level of TLE3 expression in the cancer sample; andcomparing the level of TLE3 expression in the cancer sample with the level of TLE3 expression in the negative control sample.
  • 3. The method of claim 1, wherein the step of detecting comprises steps of: providing a positive control sample;detecting a level of TLE3 expression in the positive control sample;detecting a level of TLE3 expression in the cancer sample; andcomparing the level of TLE3 expression in the cancer sample with the level of TLE3 expression in the positive control sample.
  • 4. The method of claim 1, wherein the step of detecting comprises contacting the cancer sample with an interaction partner that binds a TLE3 polypeptide.
  • 5. The method of claim 4, wherein the interaction partner is an antibody.
  • 6. The method of claim 1, wherein the step of detecting comprises contacting the cancer sample with one or more primers that hybridize with a TLE3 polynucleotide.
  • 7. The method of claim 6, wherein the taxane is paclitaxel.
  • 8. The method of claim 6, wherein the taxane is docetaxel.
  • 9. The method of claim 4, wherein the step of selecting comprises selecting a taxane for chemotherapy.
  • 10. The method of claim 9, wherein the taxane is paclitaxel.
  • 11. The method of claim 9, wherein the taxane is docetaxel.
  • 12. The method of claim 5, wherein the step of selecting comprises selecting a taxane for chemotherapy.
  • 13. The method of claim 12, wherein the taxane is paclitaxel.
  • 14. The method of claim 12, wherein the taxane is docetaxel.
  • 15. The method of claim 6, wherein the step of selecting comprises selecting a taxane for chemotherapy.
  • 16. The method of claim 15, wherein the taxane is paclitaxel.
  • 17. The method of claim 15, wherein the taxane is docetaxel.
CROSS REFERENCE TO RELATED APPLICATION

This application is a continuation of U.S. patent application Ser. No. 12/277,920, filed Nov. 25, 2008, which claims priority to U.S. Provisional Patent Application Ser. No. 60/991,487, filed Nov. 30, 2007, both of which are hereby incorporated herein by reference in their entireties.

US Referenced Citations (20)
Number Name Date Kind
5637471 Artavanis-Tsakonas et al. Jun 1997 A
20040029114 Mack et al. Feb 2004 A1
20040038207 Orntoft Feb 2004 A1
20040072154 Morris et al. Apr 2004 A1
20040156854 Mulligan et al. Aug 2004 A1
20040253606 Aziz et al. Dec 2004 A1
20050181375 Aziz et al. Aug 2005 A1
20060121511 Lee et al. Jun 2006 A1
20060234287 Erlander et al. Oct 2006 A1
20060286586 Drexhage et al. Dec 2006 A1
20070037145 Morris et al. Feb 2007 A1
20070059706 Yu et al. Mar 2007 A1
20070065888 Ring et al. Mar 2007 A1
20070172844 Lancaster et al. Jul 2007 A1
20070178090 Sukumar et al. Aug 2007 A1
20070237770 Lai et al. Oct 2007 A1
20080318228 Lee et al. Dec 2008 A1
20090004173 Evans et al. Jan 2009 A1
20090092973 Erlander et al. Apr 2009 A1
20090105167 Potti et al. Apr 2009 A1
Foreign Referenced Citations (7)
Number Date Country
1538218 Jun 2005 EP
WO-9407522 Apr 1994 WO
WO-9408037 Apr 1994 WO
WO 2004053066 Jun 2004 WO
WO-2005054513 Jun 2005 WO
WO 2006089233 Aug 2006 WO
WO-2006125195 Nov 2006 WO
Related Publications (1)
Number Date Country
20100062444 A1 Mar 2010 US
Provisional Applications (1)
Number Date Country
60991487 Nov 2007 US
Continuations (1)
Number Date Country
Parent 12277920 Nov 2008 US
Child 12578255 US