PREDICTING RESPONSIVENESS TO CANCER THERAPEUTICS

Abstract
Provided herein are methods for predicting the responsiveness of a cancer to a chemotherapeutic agent using gene expression profiles. In particular, methods for predicting the responsiveness to 5-fluorouracil, adriamycin, cytotoxan, docetaxol, etoposide, taxol, topotecan, PB kinase inhibitors and Src inhibitors are provided. Methods for developing treatment plans for individuals with cancer are also provided. Kits including gene chips and instructions for predicting responsiveness and computer readable media comprising responsivity information are also provided.
Description
BACKGROUND OF THE INVENTION

The National Cancer Institute has estimated that in the United States alone, one in three people will be afflicted with cancer. Moreover, approximately 50% to 60% of people with cancer will eventually die from the disease. The inability to predict responses to specific therapies is a major impediment to improving outcome for cancer patients. Because treatment of cancer typically is approached empirically, many patients with chemo-resistant disease receive multiple cycles of often toxic therapy before the lack of efficacy becomes evident. As a consequence, many patients experience significant toxicities, compromised bone marrow reserves, and reduced quality of life while receiving chemotherapy. Further, initiation of efficacious therapy is delayed.


BRIEF SUMMARY OF THE INVENTION

In one aspect, methods for predicting responsiveness of a cancer to a chemotherapeutic agent are provided. The method includes using a comparison of a first gene expression profile of the cancer to a chemotherapy responsivity predictor set of gene expression profiles to predict the responsiveness of the cancer to the chemotherapeutic agent. The first gene expression profile and the chemotherapy responsivity predictor set each comprise at least five genes from one of Tables 1-8. Tables 1-8 comprise the chemotherapy responsivity predictor set for 5-fluorouracil, adriamycin, cytotoxan, docetaxol, etoposide, taxol, topotecan and PI3 kinase inhibitors, respectively. Also included are methods of predicting the responsiveness to PI3kinase pathway inhibitors and Src pathway inhibitors using the chemotherapy response predictor sets for docetaxol and topotecan, respectively.


In another aspect, methods of developing a treatment plan for an individual with cancer are provided. The predicted responsivity of a cancer to a chemotherapeutic agent may be used to develop a treatment plan for the individual with the cancer. The treatment plan may include administering an effective amount of a chemotherapeutic agent to the individual with the cancer which is predicted to respond to the chemotherapeutic agent.


In yet another aspect, kits including a gene chip for predicting responsivity of a cancer to a chemotherapeutic agent comprising nucleic acids capable of detecting at least five genes selected from any one of Tables 1-8 and instructions for predicting responsivity of a cancer to the chemotherapeutic agents are provided.


In a further aspect, computer readable mediums including gene expression profiles and corresponding responsivity information for chemotherapeutic agents comprising at least five genes from any of Tables 1-8 are provided.


Throughout this specification, reference numbering is sometimes used to refer to the full citation for the references, which can be found in the “Reference Bibliography” after the Examples section. The disclosure of all patents, patent applications, and publications cited herein are hereby incorporated by reference in their entirety for all purposes.





BRIEF DESCRIPTION OF THE DRAWINGS

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.



FIGS. 1A-1E show a gene expression signature that predicts sensitivity to docetaxel. (A) Strategy for generation of the chemotherapeutic response predictor. (B) Top panel—Cell lines from the NCI-60 panel used to develop the in vitro signature of docetaxel sensitivity. The figure shows a statistically significant difference (Mann Whitney U test of significance) in the IC50/GI50 and LC50 of the cell lines chosen to represent the sensitive and resistant subsets. Bottom Panel—Expression plots for genes selected for discriminating the docetaxel resistant and sensitive NCI-60 cell lines, depicted by color coding with blue representing the lowest level and red the highest. Each column in the figure represents individual samples. Each row represents an individual gene, ordered from top to bottom according to regression coefficients. (C) Top Panel—Validation of the docetaxel response prediction model in an independent set of lung and ovarian cancer cell line samples. A collection of lung and ovarian cell lines were used in a cell proliferation assay to determine the 50% inhibitory concentration (IC50) of docetaxel in the individual cell lines. A linear regression analysis demonstrates a statistically significant (p<0.01, log rank) relationship between the IC50 of docetaxel and the predicted probability of sensitivity to docetaxel. Bottom panel—Validation of the docetaxel response prediction model in another independent set of 29 lung cancer cell line samples (Gemma A, Geo accession number: GSE 4127). A linear regression analysis demonstrates a very significant (p<0.001, log rank) relationship between the IC50 of docetaxel and the predicted probability of sensitivity to docetaxel. (D) Left Panel—A strategy for assessment of the docetaxel response predictor as a function of clinical response in the breast neoadjuvant setting. Middle panel—Predicted probability of docetaxel sensitivity in a collection of samples from a breast cancer single agent neoadjuvant study. Twenty of twenty four samples (91.6%) were predicted accurately using the cell line based predictor of response to docetaxel. Right panel—A single variable scatter plot demonstrating a significance test of the predicted probabilities of sensitivity to docetaxel in the sensitive and resistant tumors (p<0.001, Mann Whitney U test of significance). (E) Left Panel—A strategy for assessment of the docetaxel response predictor as a function of clinical response in advanced ovarian cancer. Middle panel—Predicted probability of docetaxel sensitivity in a collection of samples from a prospective single agent salvage therapy study. Twelve of fourteen samples (85.7%) were predicted accurately using the cell line based predictor of response to docetaxel. Right panel—A single variable scatter plot demonstrating statistical significance (p<0.01, Mann Whitney U test of significance).



FIGS. 2A-2C show the development of a panel of gene expression signatures that predict sensitivity to chemotherapeutic drugs. (A) Gene expression patterns selected for predicting response to the indicated drugs. The genes involved the individual predictors are shown in Tables 1-8, as indicated. (B) Independent validation of the chemotherapy response predictors in an independent set of cancer cell lines37 that have dose response and Affymetrix expression data.38 A single variable scatter plot demonstrating a significance test of the predicted probabilities of sensitivity to any given drug in the sensitive and resistant cell lines (p value, Mann Whitney U test of significance). Red symbols indicate resistant cell lines, and blue symbols indicate those that are sensitive. (C) Prediction of single agent therapy response in patient samples using in vitro cell line based expression signatures of chemosensitivity. In each case, red represents non-responders (resistance) and blue represents responders (sensitivity). The top panel shows the predicted probability of sensitivity to topotecan when compared to actual clinical response data (n=48), the middle panel demonstrates the accuracy of the adriamycin predictor in a cohort of 122 samples (Evans W, GSE650 and GSE651). The bottom panel shows the predictive accuracy of the cell line based paclitaxel (taxol) predictor when used as a salvage chemotherapy in advanced ovarian cancer (n=35). The positive and negative predictive values for all the predictors are summarized in Table 16.



FIGS. 3A-3B show the prediction of response to combination therapy. (A) Top Panel—Strategy for assessment of chemotherapy response predictors in combination therapy as a function of pathologic response. Middle panel—Prediction of patient response to neoadjuvant chemotherapy involving paclitaxel, 5-fluorouracil (5-FU), adriamycin, and cyclophosphamide (TFAC) using the single agent in vitro chemosensitivity signatures developed for each of these drugs. Bottom Panel—Prediction of response (38 non-responders, 13 responders) employing a combined probability predictor assessing the probability of all four chemosensitivity signatures in 51 patients treated with TFAC chemotherapy shows statistical significance (p<0.0001, Mann Whitney) between responders (blue) and non-responders (red). Response was defined as a complete pathologic response after completion of TFAC neoadjuvant therapy. (B) Top Panel—Prediction of patient response (n=45) to adjuvant chemotherapy involving 5-FU, adriamycin, and cyclophosphamide (FAC) using the single agent in vitro chemosensitivity predictors developed for these drugs. Middle panel—Prediction of response (34 responders, 11 non-responders) employing a combined probability predictor assessing the probability of all four chemosensitivity signatures in 45 patients treated with FAC chemotherapy. Bottom panel—Kaplan Meier survival analysis for patients predicted to be sensitive (blue curve) or resistant (red curve) to FAC adjuvant chemotherapy.



FIG. 4 shows patterns of predicted sensitivity to common chemotherapeutic drugs in human cancers. Hierarchical clustering of a collection of breast (n=171), lung cancer (n=91) and ovarian cancer (n=119) samples according to patterns of predicted sensitivity to the various chemotherapeutics. These predictions were then plotted as a heatmap in which high probability of sensitivity/response is indicated by red, and low probability or resistance is indicated by blue.



FIGS. 5A-5B show the relationship between predicted chemotherapeutic sensitivity and oncogenic pathway deregulation. (A) Top Panel—Probability of oncogenic pathway deregulation as a function of predicted docetaxel sensitivity in a series of lung cancer cell lines (red=sensitive, blue=resistant). Bottom panel—Probability of oncogenic pathway deregulation as a function of predicted topotecan sensitivity in a series of ovarian cancer cell lines (red=sensitive, blue=resistant). (B) Top Left Panel—The lung cancer cell lines showing an increased probability of PI3 kinase were also more likely to respond to a PI3 kinase inhibitor (L Y −294002) (p=0.001, log-rank test)), as measured by sensitivity to the drug in assays of cell proliferation. Top Right Panel—Those cell lines predicted to be resistant to docetaxel were more likely to be sensitive to PI3 kinase inhibition (p<0.001, log-rank test). Bottom Left Panel—Ovarian cancer cell lines showing an increased probability of Src pathway deregulation were also more likely to respond to a Src inhibitor (SU6656) (p<0.007, log-rank test). Bottom Right Panel—The relationship between Src pathway deregulation and topotecan resistance can be demonstrated in a set of 13 ovarian cancer cell lines. Ovarian cell lines that are predicted to be topotecan resistant have a higher likelihood of Src pathway deregulation and there is a significant linear relationship (p=0.001, log rank) between the probability of topotecan resistance and sensitivity to a drug that inhibits the Src pathway (SU6656).



FIG. 6 shows a scheme for utilization of chemotherapeutic and oncogenic pathway predictors for identification of individualized therapeutic options.



FIGS. 7A-7C show a patient-derived docetaxel gene expression signature predicts response to docetaxel in cancer cell lines. (A) Top panel—A ROC curve analysis to show the approach used to define a cut-off, using docetaxel as an example. Middle panel—A t-test plot of significance between the probability of docetaxel sensitivity and IC 50 for docetaxel sensitive in cell lines, shown by histologic type. Bottom panel—A linear regression analysis showing the significant correlation between predicted sensitivity and actual sensitivity (IC50) for docetaxel, in lung and ovarian cancer cell lines. (B) Generation of a docetaxel response predictor based on patient data that was then validated in a leave one out cross validation and linear regression analysis (p-value obtained by log-rank), evaluated against the IC50 for docetaxel in two NCI-60 cell line drug screening experiments. (C) A comparison of predictive accuracies between a predictor for docetaxel generated from the cell line data (top panel, accuracy: 85.7%) and a predictor generated from patients treatment data (bottom panel, accuracy: 64.3%) shows the relative inferiority of the latter approach, when applied to an independent dataset of ovarian cancer patients treated with single agent docetaxel.



FIGS. 8A-8C show the development of gene expression signatures that predict sensitivity to a panel of commonly used chemotherapeutic drugs. Panel A shows the gene expression models selected for predicting response to the indicated drugs, with resistant lines on the left, sensitive on the right for each predictor. Panel B shows the leave one out cross validation accuracy of the individual predictors. Panel C demonstrates the results of an independent validation of the chemotherapy response predictors in an independent set of cancer cell lines37 shown as a plot with error bars (blue—sensitive, red—resistant).



FIG. 9 shows the specificity of chemotherapy response predictors. In each case, individual predictors of response to the various cytotoxic drugs was plotted against cell lines known to be sensitive or resistant to a given chemotherapeutic agent (e.g., adriamycin, paclitaxel).



FIG. 10A-10C shows the relationships in predicted probability of response to chemotherapies in breast (A), lung (B) and ovarian (C) cancers. In each case, a regression analysis (log rank) of predicted probability of response to two drugs is shown.



FIG. 11 shows the absolute probabilities of response to various chemotherapies in human lung and breast cancer samples.



FIG. 12 shows a gene expression based signature of PI3 kinase pathway deregulation. Image intensity display of expression levels for genes that most differentiate control cells expressing GFP from cells expressing the oncogenic activity of P13 kinase. The expression value of genes composing each signature is indicated by color, with blue representing the lowest value and red representing the highest level. The panel below shows the results of a leave one out cross validation showing a reliable differentiation between GFP controls (blue) and cells expressing P13 kinase (red).



FIGS. 13A-13C show the relationship between oncogenic pathway deregulation and chemosensitivity patterns (using docetaxel as an example). (A) Probability of oncogenic pathway deregulation as a function of predicted docetaxel sensitivity in the NCI-60 cell line panel (red=sensitive, blue=resistant). (B) Linear regression analysis (log-rank test of significance) to identify relationships between predicted docetaxel sensitivity or resistance and deregulation of PI3 kinase and E2F3 pathways. (C) A non-parametric t-test of significance demonstrating a significant difference in docetaxel sensitivity, between those cell lines predicted to be either pathway deregulated (>50% probability, red) or quiescent (<50% probability, blue), shown for both E2F and PI3 kinase pathways.



FIG. 14 shows a scatter plot demonstrating a linear regression analysis that identifies a statistically significant correlation between probability of docetaxel resistance and PI3 Kinase pathway activation in an independent cohort of 17 non-small cell lung cancer cell lines.



FIG. 15 shows a functional block diagram of general purpose computer system 1500 for performing the functions of the software provided by the invention.





BRIEF DESCRIPTION OF THE TABLES

Tables 1-8 include the chemotherapy responsivity predictor set for 5-fluorouracil, adriamycin, cytotoxan, docetaxol, etoposide, taxol, topotecan and PI3 kinase inhibitors, respectively.


Tables 9-15 list cell lines and indicate their sensitivity or resistance to 5-fluorouracil, adriamycin, cytotoxan, docetaxol, etoposide, taxol, and topotecan, respectively.


Table 16 is a summary of the chemotherapy response predictors—validations in cell line and patient data sets.


Table 17 shows an enrichment analysis shows that a genomic-guided response prediction increases the probability of a clinical response in the different data sets studied. Table 18 shows the accuracy of genomic-based chemotherapy response predictors as compared to previously reported predictors of response.


DETAILED DESCRIPTION OF THE INVENTION

The difficulty with administering one or more chemotherapeutic agents to an individual with cancer is that not all individuals with cancer will respond favorably to the chemotherapeutic agent selected by the physician. Frequently, the administration of one or more chemotherapeutic agent results in the individual becoming even more ill from the toxicity of the agent, while the cancer persists. Due to the cytotoxic nature of chemotherapeutic agents, the individual is physically weakened and immunologically compromised such that the individual cannot tolerate multiple rounds of therapy. Hence a personalized treatment plan is highly desirable.


As described in the Examples, the inventors identified gene expression patterns within primary tumors or cell lines that predict response to various chemotherapeutic agents. These predictions may be used to develop treatment plans for individual cancer patients. The invention also provides integrating gene expression profiles that predict responsiveness to combination therapies as a strategy for developing personalized treatment plans for individual patients. Treatment plans may result in individuals having a complete response, a partial response or an incomplete response to the cancer.


A “complete response” (CR) to treatment of cancer is defined as a complete disappearance of all measurable and assessable disease. In ovarian cancer a complete response includes, in the absence of measurable lesions, a normalization of the CA-125 level following adjuvant therapy. An individual who exhibits a complete response is known as a “complete responder.”


An “incomplete response” (IR) includes those who exhibited a “partial response” (PR), had “stable disease” (SD), or demonstrated “progressive disease” (PD) during primary therapy.


A “partial response” refers to a response that displays 50% or greater reduction in bi-dimensional size (area) of the lesion for at least 4 weeks or, in ovarian cancer, a drop in the CA-125 level by at least 50% for at least 4 weeks.


“Progressive disease” refers to response that is a 50% or greater increase in the product from any lesion documented within 8 weeks of initiation of therapy, the appearance of any new lesion within 8 weeks of initiation of therapy, or in the case of ovarian cancer, any increase in the CA-125 from baseline at initiation of therapy.


“Stable disease” was defined as disease not meeting any of the above criteria.


“Effective amount” refers to an amount of a chemotherapeutic agent that is sufficient to exert a prophylactic or therapeutic effect in the subject, i.e., that amount which will stop or reduce the growth of the cancer or cause the cancer to become smaller in size compared to the cancer before treatment or compared to a suitable control. In most cases, an effective amount will be known or available to those skilled in the art. The result of administering an effective amount of a chemotherapeutic agent may lead to effective treatment of the patient. It is desirable for an effective amount to be an amount sufficient to exert cytotoxic effects on cancerous cells.


“Predicting” and “prediction” as used herein includes, but is not limited to, generating a statistically based indication of whether a particular chemotherapeutic agent will be effective to treat the cancer. This does not mean that the event will happen with 100% certainty.


As used herein, “individual” and “subject” are interchangeable. A “patient” refers to an “individual” who is under the care of a treating physician.


The present invention may be practiced using any suitable technique, including techniques known to those skilled in the art. Such techniques are available in the literature or in scientific treatises, such as, Molecular Cloning: A Laboratory Manual, second edition (Sambrook et al., 1989) and Molecular Cloning: A Laboratory Manual, third edition (Sambrook and Russel, 2001), (jointly referred to herein as “Sambrook); Current Protocols in Molecular Biology (F. M. Ausubel et al., eds., 1987, including supplements); PCR: The Polymerase Chain Reaction, (Mullis et al., eds., 1994); Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York; Harlow and Lane (1999) Using Antibodies: A Laboratory Manual Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (jointly referred to herein as “Harlow and Lane”), Beaucage et al. eds., Current Protocols in Nucleic Acid Chemistry John Wiley & Sons; Inc., New York, 2000) and Casarett and Doull's Toxicology The Basic Science of Poisons, C. Klaassen, ed., 6th edition (2001).


Methods for Predicting Responsiveness to Chemotherapy

Methods of predicting responsiveness of a cancer to a chemotherapeutic agent are provided herein. Specifically, the methods rely on using a comparison of a gene expression profile of the cancer to a chemotherapy responsivity predictor set to predict the responsiveness to the chemotherapeutic agent. See Tables 1-8 for the chemotherapeutic responsivity predictor sets. The chemotherapy responsivity predictor set is expected to be distinct for each class of chemotherapeutic agents and may vary between chemotherapeutic agents within the same class. A class of chemotherapeutic agents is chemotherapeutic agents that are similar in some way. For example, the agents may be known to act through a similar mechanism, or have similar targets or structures. An example of a class of chemotherapeutic agents is agents that inhibit PI3 kinase.


The chemotherapy predictor set is, or may be derived from, a set of gene expression profiles obtained from samples (cell lines, tumor samples, etc.) with known sensitivity or resistance to the chemotherapeutic agent. The comparison of the expression of a specific set of genes in the cancer to the same set of genes in samples known to be sensitive or resistant to the chemotherapeutic agent allows prediction of the responsiveness of the cancer to the chemotherapeutic agent. The prediction may indicate that the cancer will respond completely to the chemotherapeutic agent, or it may predict that the cancer will be only partially responsive or non-responsive (i.e. resistant) to the chemotherapeutic agent. The cell lines used to generate the chemotherapy responsivity predictor sets and an indication of the cell lines' sensitivity or resistance to the chemotherapeutic agents are provided in Tables 9-15.


The methods described herein provide an indication of whether the cancer in the patient is likely to be responsive to a particular chemotherapeutic prior to beginning treatment that is more accurate than predictions using population-based approaches from clinical studies. The methods allow identification of chemotherapeutics estimated to be useful in combating a particular cancer in an individual patient, resulting in a more cost-effective, targeted therapy for the cancer patient and avoiding side effects from non-efficacious chemotherapeutic agents.


Tables 1-8 also provide the relative “weights” of each of the individual genes that make up the responsivity predictor set. The weights demonstrate that some genes are more strongly indicative of sensitivity or resistance of a cancer to a particular therapeutic agent. Predictions based on the complete set of genes are expected to provide the most accurate predictions regarding the efficacy of treating the cancer with a particular therapeutic agent. Those of skill in the art will understand based on the weights of each gene in the responsivity predictor set that some genes are more predictive of outcome than others and thus that the entire responsivity predictor set need not be used to develop a useful prediction.


Once an individual's cancer is predicted to be responsive to a particular chemotherapy, then a treatment plan can be developed incorporating the chemotherapeutic agent and an effective amount of the chemotherapeutic agent(s) may be administered to the individual with the cancer. Those of skill in the art will appreciate that the methods do not guarantee that the individuals will be responsive to the chemotherapeutic agent, but the methods will increase the probability that the selected treatment will be effective to treat the cancer. Also encompassed is the ability to predict the responsiveness of the cancer to multiple chemotherapeutic agents and then to develop a treatment plan using a combination of two or more chemotherapeutic agents. Those of skill in the art appreciate that combination therapy is often suitable.


Treatment or treating a cancer includes, but is not limited to, reduction in cancer growth or tumor burden, enhancement of an anti-cancer immune response, induction of apoptosis of cancer cells, inhibition of angiogenesis, enhancement of cancer cell apoptosis, and inhibition of metastases. Administration of an effective amount of a chemotherapeutic agent to a subject may be carried out by any means known in the art including, but not limited to intraperitoneal, intravenous, intramuscular, subcutaneous, transcutaneous, oral, nasopharyngeal or transmucosal absorption. The specific amount or dosage administered in any given case will be adjusted in accordance with the specific cancer being treated, the condition, including the age and weight, of the subject, and other relevant medical factors known to those of skill in the art.


In one embodiment, the methods involve predicting responsiveness to chemotherapeutic agents of an individual with cancer. Cancers include but are not limited to any cancer treatable with the chemotherapeutic agents described herein. Cancers include, but are not limited to, ovarian cancer, lung cancer, prostrate cancer, renal cancer, colon cancer, leukemia, skin cancer, brain or central nervous system cancer and breast cancer. In another embodiment, the individual has advanced stage cancer (e.g., Stage III/IV ovarian cancer). In other embodiments, the individual has early stage cancer. For the individuals with advanced cancer, one form of primary treatment practiced by treating physicians is to surgically remove as much of the tumor as possible, a practice sometime known as “debulking.”


The sample of the cancer used to obtain the first gene expression profile may be directly from a tumor that was surgically removed. Alternatively, the sample of the cancer could be from cells obtained in a biopsy or other tumor sample. A sample from ascites surrounding the tumor may also be used.


The sample is then analyzed to obtain a first gene expression profile. This can be achieved by any suitable means, including those available to those of skill in the art. One method that can be used is to isolate RNA (e.g., total RNA) from the cellular sample and use a publicly or commercially available micro array system to analyze the gene expression profile from the cellular sample. One microarray that may be used is Affymetrix Human U133A chip. One of skill in the art follows the standard directions that come with a commercially available microarray. Other types of microarrays may be used, for example, microarrays using RT-PCR for measurement. Other sources of microarrays include, but are not limited to, Stratagene (e.g., Universal Human Microarray), Genomic Health (e.g., Oncotype DX chip), Clontech (e.g., Atlas™ Glass Microarrays), and other types of Affymetrix microarrays. In one embodiment, the microarray may be made by a researcher or obtained from an educational institution. In other embodiments, customized microarrays, which include the particular set of genes that are particularly suitable for prediction, can be used. The gene expression profile may be obtained by any other means, including those known to those of skill in the art, e.g., Northern blots, real time rt-PCR, Western blots for the expressed proteins or protein assays.


Once a first gene expression profile has been obtained from the sample, it is compared with chemotherapy responsivity predictor set of gene expression profiles. Tables 1-8 describe the chemotherapy responsivity predictor sets for 5-FU, adriamycin, cytotoxan, docetaxol, etoposide, taxol, topotecan, and PI3 kinase inhibitors, respectively.


The use of the chemotherapy responsitivity predictor set in its entirety is contemplated; however, it is also possible to use subsets of the predictor set. For example, a subset of at least 2, 5, 10, 15, 20, 25, 30, 35 or 40 or more genes from one of Tables 1-8 can be used for predictive purposes. For example, 40, 45, 50, 55, 60, 65, 70, 75 or 80 genes from Table 7 could be used in a topotecan chemotherapy responsivity predictor set.


Thus, one of skill in art may use the chemotherapy responsitivity predictor set as detailed in the Examples to predict whether an individual or patient with cancer will be responsive to the selected chemotherapeutic agent. If the individual is a complete responder to a chemotherapeutic agent, then a treatment plan may be designed in which the therapeutic agent will be administered in an effective amount. If the complete responder stops being a complete responder, as sometimes happens, then the first gene expression profile may be further analyzed for responsivity to an alternative agent to determine which alternative agent should be administered to most effectively combat the cancer while minimizing the toxic side effects to the individual. If the individual is an incomplete responder, then the individual's gene expression profile can be further analyzed for responsivity to an alternative agent to determine which agent should be administered, or alternatively which combination of agents is predicted to be most effective to treat the cancer.


Those of skill in the art will understand that the first gene expression profile may be tested against more than one chemotherapy responsivity predictor set to allow development of a treatment plan with the best likelihood of treating the individual with the cancer. For example, an individual can be evaluated for responsiveness to one or more chemotherapeutic agents. In certain embodiments, the methods of the application are performed outside of the human body. In addition, an individual can be assessed to determine if they will be refractory to a commonly used first-line therapy such that additional alternative therapeutic intervention can be started.


For the individuals who appear to be incomplete responders to a chemotherapeutic agent or for those individuals who have ceased being complete responders, an important step in the treatment is to determine other alternative cancer therapies that may be administered to the individual to best combat the cancer while minimizing the toxicity of these additional agents.


Alternative therapeutic agents include, but are not limited to, cisplatin, denopterin, edatrexate, methotrexate, nolatrexed, pemetrexed, piritrexim, pteropterin, raltitrexed, trimetrexate, cladribine, clofarabine, fludarabine, 6-mercaptopurine, nelarabine, thiamiprine, thioguanine, tiazofurin, ancitabine, azacitidine, 6-azauridine, capecitabine, carmofur, cytarabine, decitabine, doxifluridine, enocitabine, floxuridine, fluorouracil, gemcitabine, tegafur, troxacitabine, pentostatin, hydroxyurea, cytosine arabinoside, docetaxel, paclitaxel, abraxane, topotecan, adriamycin, etoposide, fluorouracil (5-FU), and cyclophosphamide. In one embodiment, the agent may be selected from platinum-based chemotherapeutic agents (e.g., cisplatin), alkylating agents (e.g., nitrogen mustards), antimetabolites (e.g., pyrimidine analogs), radioactive isotopes (e.g., phosphorous and iodine), miscellaneous agents (e.g., substituted ureas) and natural products (e.g., vinca alkyloids and antibiotics). In another embodiment, the therapeutic agent may be selected from the group consisting of allopurinol sodium, dolasetron mesylate, pamidronate disodium, etidronate, fluconazole, epoetin alfa, levamisole HeL, amifostine, granisetron HCL, leucovorin calcium, sargramostim, dronabinol, mesna, filgrastim, pilocarpine HCl, octreotide acetate, dexrazoxane, ondansetron HCL, ondanselron, busulfan, carboplatin, cisplatin, thiotepa, melphalan HCl, melphalan, cyclophosphamide, ifosfamide, chlorambucil, mechlorethamine HCL, carmustine, lomustine, polifeprosan 20 with carmustine implant, streptozocin, doxorubicin HCL, bleomycin sulfate, daunirubicin HCL, dactinomycin, daunorucbicin citrate, idarubicin HCL, pllmycin, mitomycin, pentostatin, mitoxantrone, valrubicin, cytarabine, tludarabine phosphate, floxuridine, cladribine, methotrexate, mercaptipurine, thioguanine, capecitabine, methyltestosterone, nilutamide, testolactone, bicalutamide, flutamide, anastrozole, toremifene citrate, estramustine phosphate sodium, ethinyl estradiol, estradiol, esterified estrogens, conjugated estrogens, leuprolide acetate, goserelin acetate, medroxyprogesterone acetate, megestrol acetate, levamisole HCL, aldesleukin, irinotecan HCL, dacarbazine, asparaginase, etoposide phosphate, gemcitabine HCL, altretamine, topotecan HCL, hydroxyurea, interferon alpha-2b, mitotane, procarbazine HCL, vinorelbine tartrate, E. coli l-asparaginase, Erwinia L-asparaginase, vincristine sulfate, denileukin diftitox, aldesleukin, rituximab, interferon alpha-1a, paclitaxel, abraxane, docetaxel, BCG live (intravesical), vinblastine sulfate, etoposide, tretinoin, teniposide, porfuner sodium, tluorouracil, betamethasone sodium phosphate and betamethasone acetate, letrozole, etoposide citrororum factor, folinic acid, calcium leucouorin, 5-fluorouricil, adriamycin, c}toxan, and diamino-dichloro-platinum.


In another aspect, the first gene expression profile from the individual with cancer is analyzed and compared to gene expression profiles (or signatures) that are reflective of deregulation of various oncogenic signal transduction pathways. In one embodiment, the alternative cancer therapeutic agent is directed to a target that is implicated in oncogenic signal transduction deregulation. Such targets include, but are not limited to, Src, myc, beta-catenin and E2F3 pathways. Thus, in one aspect, the invention contemplates using an inhibitor that is directed to one of these targets as an additional therapy for cancer. One of skill in the art will be able to determine the dosages for each specific chemotherapeutic agent.


As shown in Example 1, the teachings herein provide a gene expression model that predicts response to docetaxel therapy. The other Examples provide predictors for 5-FU, adriamycin, cytotoxan, taxol, etoposide, topotecan, PI3 kinase inhibitors and Src inhibitors. The gene expression model was developed by using Bayesian binary regression analysis to identify genes highly correlated with drug sensitivity. The developed models were validated in a leave-one-out cross validation.


Chemotherapy Responsivity Predictor Set of Gene Expression Profiles

The chemotherapy responsitivity predictor sets were created by a method described in detail in the Examples and similar to that detailed in Potti et al. (Genomic signatures to guide the use of chemotherapeutics. Nature Medicine 12(11): 1294-1300, 2006, incorporated herein by reference). Unless otherwise noted in the Examples, the [−log 10(M)] GI50/IC50 and LC50 (50% cytotoxic dose) data on the NCI-60 cell line panel for each of the indicated therapeutic agents was used to populate a matrix with MATLAB software with the relevant expression data for each individual cell line. When multiple entries for a drug screen existed (by NCS number), the entry with the largest number of replicates was included. To develop in vitro gene expression based predictors for chemotherapeutic agent sensitivity from the pharmacologic data used in the NCI-60 drug screen studies, we chose cell lines within the NCI-60 panel that would represent the extremes of sensitivity (See Tables 9-15). Relevant expression data (updated data available on the Affymetrix U95A2 GeneChip) for the selected NCI-60 cell lines were then used in a supervised analysis using Bayesian regression methodologies, as described previously (Pittman J, Huang E, Nevins J, et al: Bayesian analysis of binary prediction tree models for retrospectively sampled outcomes. Biostatistics 5(4):587-601, 2004), to develop a probit model predictive of sensitivity to the indicated chemotherapeutic agent.


Method of Treating Individuals with Cancer


The methods described herein also include treating an individual afflicted with cancer. This method involves administering an effective amount of a chemotherapeutic agent to those individuals predicted to be responsive to such therapy. In the alternative, an effective amount of a combination of chemotherapeutic agents may be administered to individuals predicted to be responsive to combination therapy. In the instance where the individual is predicted to be a non-responder, a physician may decide to administer alternative therapeutic agents alone. In many instances, the treatment will comprise a combination of chemotherapeutic agents.


The methods described herein include, but are not limited to, treating individuals afflicted with NSCLC, breast cancer and ovarian cancer. In one aspect, a chemotherapeutic agent is administered in an effective amount by itself (e.g., for complete responders). In another embodiment, the therapeutic agent is administered with an alternative chemotherapeutic in an effective amount concurrently. In another embodiment, the two therapeutic agents are administered in an effective amount in a sequential manner. In yet another embodiment, the alternative therapeutic agent is administered in an effective amount by itself. In yet another embodiment, the alternative therapeutic agent is administered in an effective amount first and then followed concurrently or step-wise by a second or third chemotherapeutic agent.


Methods of Predicting/Estimating the Efficacy of a Therapeutic Agent in Treating an Individual Afflicted with Cancer


One aspect of the invention provides a method for predicting, estimating, aiding in the prediction of, or aiding in the estimation of, the efficacy of a therapeutic agent in treating a subject afflicted with cancer. In certain embodiments, the methods of the application are performed outside of the human body.


One method comprises (a) determining the expression level of multiple genes in a tumor biopsy sample from the subject; (b) defining the value of one or more metagenes from the expression levels of step (a), wherein each metagene is defined by extracting a single dominant value using singular value decomposition (SVD) from a chemotherapy responsivity predictor set; and (c) averaging the predictions of one or more statistical tree models applied to the values of the metagenes, wherein each model includes one or more nodes, each node representing a metagene, each node including a statistical predictive probability of tumor sensitivity to the therapeutic agent, thereby estimating the efficacy of a therapeutic agent in a subject afflicted with cancer. Another method comprises (a) determining the expression level of multiple genes in a tumor biopsy sample from the subject; (b) defining the value of one or more metagenes from the expression levels of step (a), wherein each metagene is defined by extracting a single dominant value using singular value decomposition (SVD) from a chemotherapy responsivity predictor set; and (c) averaging the predictions of one or more binary regression models applied to the values of the metagenes, wherein each model includes a statistical predictive probability of tumor sensitivity to the therapeutic agent, thereby estimating the efficacy of a therapeutic agent in a subject afflicted with cancer.


In one embodiment, the methods predict the efficacy of a therapeutic agent in treating a subject afflicted with cancer with at least 70% accuracy. In another embodiment, the methods predict the efficacy of a therapeutic agent in treating a subject afflicted with cancer with at least 80% accuracy. In another embodiment, the methods predict the efficacy of a therapeutic agent in treating a subject afflicted with cancer with at least 85% accuracy. In another embodiment, the methods predict the efficacy of a therapeutic agent in treating a subject afflicted with cancer with at least 90% accuracy. In another embodiment, the methods predict the efficacy of a therapeutic agent in treating a subject afflicted with cancer with at least 70%, 75%, 80%, 85%, 90% or 95% accuracy when tested against a validation sample. In another embodiment, the methods predict the efficacy of a therapeutic agent in treating a subject afflicted with cancer with at least 70%, 75%, 80%, 85%, 90% or 95% accuracy when tested against a set of training samples. In another embodiment, the methods predict the efficacy of a therapeutic agent in treating a subject afflicted with cancer with at least 70%, 75%, 80%, 85%, 90% or 95% accuracy when tested on human primary tumors ex vivo or in vivo. Accuracy is the ability of the methods to predict whether a cancer is sensitive or resistant to the chemotherapeutic agent.


The methods predict the efficacy of a therapeutic agent to treat a subject with cancer with at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or 100% sensitivity for a particular chemotherapeutic agent. In another embodiment, the methods predict the efficacy of a therapeutic agent in treating a subject afflicted with cancer with at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or 100% sensitivity when tested against a validation sample. In another embodiment, the methods predict the efficacy of a therapeutic agent in treating a subject afflicted with cancer with at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or 100% sensitivity when tested against a set of training samples. In another embodiment, the methods predict the efficacy of a therapeutic agent in treating a subject afflicted with cancer with at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or 100% sensitivity when tested on human primary tumors ex vivo or in vivo. Sensitivity measures the ability of the methods to predict all cancers that will be sensitive to the chemotherapeutic agent.


(A) Sample of the Cancer

In one embodiment, the methods comprise determining the expression level of genes in a tumor sample from the subject. In certain embodiments, the tumor is a breast tumor, an ovarian tumor, or a lung tumor. In one embodiment, the tumor is not a breast tumor. In one embodiment, the tumor is not an ovarian tumor. In one embodiment, the tumor is not a lung tumor. In one embodiment of the methods described herein, the methods comprise the step of surgically removing a tumor sample from the subject, obtaining a tumor sample from the subject, or providing a tumor sample from the subject.


Alternatively, the sample may be derived from cells from the cancer, or cancerous cells. In another embodiment, the cells may be from ascites surrounding the tumor. The sample may contain nucleic acids from the cancer. Any method may be used to remove the sample from the patient.


In one embodiment, at least 40%, 50%, 60%, 70%, 80% or 90% of the cells in the sample are cancer cells. In preferred embodiments, samples having greater than 50% cancer cell content are used. In one embodiment, the sample is a live tumor sample. In another embodiment, the sample is a frozen sample. In one embodiment, the sample is one that was frozen within less than 5, 4, 3, 2, 1, 0.75, 0.5, 0.25, 0.1, or 0.05 hours after extraction from the patient. Frozen samples include those stored in liquid nitrogen or at a temperature of about −80° C. or below.


(B) Gene Expression

The expression of the genes may be determined using any method known in the art for assaying gene expression. Gene expression may be determined by measuring mRNA or protein levels for the genes. In one embodiment, an mRNA transcript of a gene may be detected for determining the expression level of the gene. Based on the sequence information provided by the GenBank™ database entries, the genes can be detected and expression levels measured using techniques well known to one of ordinary skill in the art, including but not limited to rtPCR, Northern blot analysis and microarray analysis. For example, sequences within the sequence database entries corresponding to polynucleotides of the genes can be used to construct probes for detecting mRNAs by, e.g., Northern blot hybridization analyses. The hybridization of the probe to a gene transcript in a subject biological sample can be also carried out on a DNA array. The use of an array is suitable for detecting the expression level of a plurality of the genes. As another example, the sequences can be used to construct primers for specifically amplifying the polynucleotides in, e.g., amplification-based detection methods such as reverse-transcription based polymerase chain reaction (RT-PCR). As another example, mRNA levels can be assayed by quantitative RT-PCR. Furthermore, the expression level of the genes can be analyzed based on the biological activity or quantity of proteins encoded by the genes. Methods for determining the quantity of the protein include immunoassay methods such as Western blot analysis.


In one exemplary embodiment, about 1-50 mg of cancer tissue was added to a chilled tissue pulverizer, such as to a BioPulverizer H tube (Bio101 Systems, Carlsbad, Calif.). Lysis buffer, such as from the Qiagen RNeasy Mini kit, was added to the tissue and homogenized. A device such as a Mini-Beadbeater (Biospec Products, Bartlesville, Okla.) was used. Tubes were spun briefly as needed to pellet the mixture and reduce foam. The resulting lysate was passed through syringes, such as a 21 gauge needle, to shear DNA. Total RNA was extracted using commercially available kits, such as the Qiagen RNeasy Mini kit. The samples were prepared and arrayed using Affymetrix U133 plus 2.0 GeneChips or Affymetrix U133A GeneChips. Any suitable gene chip may be used.


In one exemplary embodiment, total RNA was extracted using the Qiashredder and Qiagen RNeasy Mini kit and the quality of RNA was checked by an Agilent 2100 Bioanalyzer. The targets for Affymetrix DNA microarray analysis were prepared according to the manufacturer's instructions. Biotin-labeled cRNA, produced by in vitro transcription, was fragmented and hybridized to the Affymetrix U133A GeneChip arrays at 45° C. for 16 hrs and then washed and stained using the GeneChip Fluidics. The arrays were scanned by a GeneArray Scanner and patterns of hybridization were detected as light emitted from the fluorescent reporter groups incorporated into the target and hybridized to oligonucleotide probes. Full details of the methods used for RNA extraction and development of gene expression data from lung and ovarian tumors have been described previously. (Bild A, Yao G, Chang J T, et al: Oncogenic pathways signatures in human cancers as guide to targeted therapies. Nature 439(7074):353-357, 200, Potti A, Dressman H K, Bild A, et al: Genomic signatures to guide the use of chemotherapeutics. Nature Medicine 12(11): 1294-1300, 2006).


In one embodiment, determining the expression level (or obtaining a first gene expression profile) of multiple genes in a tumor sample from the subject comprises extracting a nucleic acid sample from the sample from the subject. In certain embodiments, the nucleic acid sample is an mRNA sample. In one embodiment, the expression level of the nucleic acid is determined by hybridizing the nucleic acid, or amplification products thereof, to a DNA microarray. Amplification products may be generated, for example, with reverse transcription, optionally followed by PCR amplification of the products.


(C) Genes Screened

In one embodiment, the predictive methods of the invention comprise determining the expression level of all the genes in the cluster that define at least one therapeutic sensitivity/resistance determinative metagene. In one embodiment, the predictive methods of the invention comprise determining the expression level of at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% of the genes in each of the clusters that defines 1 or 2 or more therapeutic sensitivity/resistance determinative metagenes. A metagene is a cluster or set of genes which may be used to predict sensitivity or resistance to a therapeutic agent.


In one embodiment, at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% of the genes whose expression levels are used in order to predict sensitivity to the chemotherapeutic agent (or the genes in the cluster that define a metagene having said predictivity) are genes listed in one of Tables 1-8. In one embodiment, at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% of the genes whose expression levels are determined to predict sensitivity to more than one chemotherapeutic agent (or the genes in the cluster that define a metagene having said predictivity) includes genes listed in more than one of Tables 1-8.


In one embodiment, at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% of the genes listed in one of Tables 1-8 are used to predict responsiveness of a cancer to the corresponding chemotherapeutic agent. Tables 1-8 show the genes in the cluster that are used to define metagenes and indicate the therapeutic agent whose sensitivity it predicts.


In one embodiment, at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% of the genes whose expression levels are determined to predict 5-FU sensitivity (or the genes in the cluster that define a metagene having said predictivity) are genes represented by the following symbols: LOC92755 (TUBB, LOC648765), CDKN2A, TRA@, GABRA3, COL1lA2, ACTB, PDLIM4, ACTA2, FTSJ1, NBR1 (LOC727732), CFL1, ATP1A2, APOC4, KlAA1509, ZNF516, GRIK5, PDE5A, ARSF, ZC3H7B, WBP4, CSTB, TSPY1 (TSPY2, LOC653174, LOC728132, LOC728137, LOC728395, LOC728403, LOC728412), HTR2B, KBTBD11, SLC25A17, HMGN3, FIBP, IFT140, FAM63B, ZNF337, KlAA0100, FAM13C1, STK25, CPNE1, PEX19, EIF5B, EEF1A1 (APOLD1, LOC440595), SRR, THEM2, ID4, GGT1 (GGTL4), IFNα10, TUBB2A (TUBB4, TUBB2B), and TUBB3.


In one embodiment, at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% of the genes whose expression levels are determined to predict adriamycin sensitivity are genes represented by the following symbols: MLANA, CSPG4, DDR2, ETS2, EGFR, BIK, CD24, ZNF185, DSCR1, GSN, TPST1, LCN2, FAIM3, NCK2, PDZRN3, FKBP2, KRT8, NRP2, PKP2, CLDN3, CAPN1, STXBP1, LY96, WWC1, C10orf56, SPINT2, MAGED2, SYNGR2, SGCD, LAMC2, C19orf21, ZFHX1B, KRT18, CYBA, DSP, ID1, ID1, PSAP, ZNF629, ARHGAP29, ARHGAP8 (LOC553158), GPM6B, EGFR, CALU, KCNK1, RNF144, FEZ1, MEST, KLF5, CSPG4, FLNB, GYPC, SLC23A2, MITF, PITPNM1, GPNMB, PMP22, PLXNB3 (SRPK3), MIA, RAB40C, MAD2L1BP, PLOD3, VIL2, KLF9, PODXL, ATP6V1B2, SLC6A8, PLP1, KRT7, PKP3, DLG3, ZHX2, LAMAS, SASH1, GAS1, TACSTD1, GAS1, and CYP27A1.


In one embodiment, at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% of the genes whose expression levels are determined to predict cytoxan sensitivity (or the genes in the cluster that define a metagene having said predictivity) are genes represented by the following symbols: DAP3, RPS9, TTR, ACTB, MARCKS, GGT1 (GGT2), GGTL4, GGTLA4, LOC643171, LOC653590, LOC728226, LOC728441, LOC729S38, LOC73 1629), FANCA, CDC42EP3, TSPAN4, C60rf145, ARNT2, KIF22 (LOC728037), NBEAL2, CA V1, SCRN1, SCHIP1, PHLDB1, AKAP12, ST5, SNAI2, ESD, ANP32B, CD59, ACTN1, CD59, PEG10, SMARCA1, GGCX, SAMD4A, CNN3, LPP, SNRPF, SGCE, CALD1, and C220rf5.


In one embodiment, at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% of the genes whose expression levels are determined to predict docetaxel sensitivity (or the genes in the cluster that define a metagene having said predictivity) are genes represented by the following symbols: BLR1, EIF4A2, FLT1, BAD, PIP5K3, BIN1, YBX1, BCKDK, DOHH, FOXD1, TEX261, NBR1 (LOC727732), APOA4, DDX5, TBCA, USP52, SLC25A36, CHP, ANKRD28, PDXK, ATP6AP1, SETD2, CCS, BRD2, ASPHD1, B4GALT6, ASL, CAPZA2, STARD3, LIMK2 (PPPIR14BP1), BANF1, GNB2, ENSA, SH3GL1, ACVR1B, SLC6A1, PPP2R1A, PCGF1, LOC643641, INPP5A, TLE1, PLLP, ZKSCAN1, TIAL1, TK1, PPP2R1A, and PSMB6.


In one embodiment, at least 50%, 60%, 70%), 80%, 90%, 95%, 98%, 99% of the genes whose expression levels are determined to predict etoposide sensitivity are genes represented by the following symbols: LIMK1, LIG3, AXL, IFI16, MMP14, GRB7, VAV2, FLT1, JUP, FN1, FN1, PKM2, LYPLA3, RFTN1, LAD1, SPINT1, CLDN3, PTRF, SPINT2, MMP14, FAAH, CLDN4, ST14, C19orf21, KIAA0506, LLGL2 (MADD), COBL, ZFHX1B, GBP1, lER2, PPL, TMEM30B, CNKSR1, CLDN7, BTN3A2, BTN3A2, TUBB2A, MAP7, HNRNPG-T, UGCG, GAK, PKP3, DFNA5, DAB2, TACSTD1, SPARC, and PPP2R5A.


In one embodiment, at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% of the genes whose expression levels are determined to predict taxol sensitivity (or the genes in the cluster that define a metagene having said predictivity) are genes represented by the following symbols: NR2F6, TOP2B, RARG, PCNA, PTPN11, ATM, NFATC4, CACNG1, C22orf31, PIK3R2, PRSS12, MYH8, SCCPDH, PHTF2, IQSEC2, TRPC3, TRAFD1, HEPH, SOX30, GATM, LMNA, HD, YIPF3, DNPEP, PCDH9, KLHDC3, SLC10A3, LHX2, CKS2, SECTM1, SF1, RPS6KA4, DYRK2, GDI2, and IFI30.


In one embodiment, at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% of the genes whose expression levels are determined to predict topotecan sensitivity (or the genes in the cluster that define a metagene having said predictivity) are genes represented by the following symbols: DUSP1, THBS1, AXL, RAP1GAP, QSCN6, IL1R1, TGFBI, PTX3, BLM, TNFRSF1A, FGF2, VEGFC, ACO2, FARSLA, RIN2, FGF2, RRAS, FIGF, MYB, CDH2, FGFR1, FGFR1, LAMC1, HIST1H4K (HIST1H4J), COL6A2, TMC6, PEA15, MARCKS, CKAP4, GJA1, FBN1, BASP1, BASP1, BTN2A1, ITGB1, DKFZP686A01247, MYLK, LOXL2, HEG1, DEGS1, CAP2, CAP2, PTGER4, BAI2, NUAK1, DLEU1 (SPANXC), RAB11FIP5, FSTL3, MYL6, VIM, GNAl2, PRAF2, PTRF, CCL2, PLOD2, COL6A2, ATP5G3, GSR, NDUFS3, ST14, NID1, MYO1D, SDHB, CAV1, DPYSL3, PTRF, FBXL2, RIN2, PLEKHC1, CTGF, COL4A2, TPM1, TPM1, TPM1, FZD2, LOXL1, SYK, HADHA, TNFAIP1, NNMT, HPGD, MRC2, MEIS3P1, AOX1, SEMA3C, SEMA3C, SYNE1, SERPINE1, IL6, RRAS, GPD1L, AXL, WDR23, CLDN7, IL15, TNFAIP2, CYR61, LRP1, AMOTL2, PDE1B, SPOCK1, RAI14, PXDN, COL4A1, C1R, KIAA0802 (C21orf57), C50rf13, TUFM, EDIL3, BDNF, PRSS23, ATP5A1, FRAT2, C16orf51, TUSC4, NUP50, TUBA3, NFIB, TLE4, AKT3, CRIM1, RAD23A, COX5A, SMCR7L, MXRA7, STARD7, STC1, TTC28, PLK2, TGDS, CALD1, OPTN, IFITM3, DFNA5, FGFR1, HTATIP, SYK, LAMB1, FZD2, SERPINE1, THBS1, CCL2, ITGA3, ITGA3, and UBE2A.


(D) Metagene Valuation

In one embodiment, the predictive methods of the invention comprise defining the value of one or more metagenes from the expression levels of the genes. A metagene value is defined by extracting a single dominant value from a cluster of genes associated with sensitivity to an anti-cancer agent.


In one embodiment, the dominant single value is obtained using single value decomposition (SVD). In one embodiment, the cluster of genes of each metagene or at least of one metagene comprises at least 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 18, 20 or 25 genes.


In one embodiment, the predictive methods of the invention comprise defining the value of at least one metagene wherein the genes in the cluster of genes from which the metagene is defined, shares at least 50%, 60%, 70%, 80%, 90%, 95% or 98% of genes in common to the genes in one of Tables 1-8. In one embodiment, the predictive methods of the invention comprise defining the value of at least two metagenes, wherein the genes in the cluster of genes from which each metagene is defined share at least 50%, 60%, 70%, 80%, 90%, 95% or 98% of genes in common to the genes in any one of Tables 1-8. In one embodiment, the predictive methods of the invention comprise defining the value of a metagene from a cluster of genes, wherein at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 genes in the cluster are selected from the genes listed in one of Tables 1-8.


In one embodiment, the clusters of genes that define each metagene were identified using supervised classification methods of analysis as previously described. See, for example, West, M. et al. Proc Natl Acad Sci USA 98, 11462-11467 (2001). A set of genes whose expression levels are most highly correlated with the classification of tumor samples into sensitivity to an anti-cancer agent versus no sensitivity to an anti-cancer agent were selected. The dominant principal components from such a set of genes defines a relevant phenotype-related metagene, and regression models, such as binary regression models, were used to assign the relative probability of sensitivity to an anti-cancer agent.


(E) Predictions from Tree Models


In one embodiment, the methods comprise averaging the predictions of one or more statistical tree models applied to the metagene values, wherein each model includes one or more nodes, each node representing a metagene, each node including a statistical predictive probability of sensitivity to an anti-cancer agent. The statistical tree models may be generated using the methods described herein for the generation of tree models. General methods of generating tree models may also be found in the art (See for example Pitman et al, Biostatistics 2004; 5:587-601; Denison et al. Biometrika 1999; 85:363-77; Nevins et al. Hum Mol Genet 2003; 12:R153-7; Huang et al. Lancet 2003; 361: 1590-6; West et al. Proc Natl A cad Sci USA 2001; 98:11462-7; U.S. Patent Pub. Nos. 2003-0224383; 2004-0083084; 2005-0170528; 2004-0106113; and U.S. application Ser. No. 11/198,782).


In one embodiment, the methods comprise deriving a prediction from a single statistical tree model, wherein the model includes one or more nodes, each node representing a metagene, each node including a statistical predictive probability of sensitivity to an anti-cancer agent. In alternative embodiments, the tree may comprise at least 2, 3, 4, or 5 nodes.


In one embodiment, the methods comprise averaging the predictions of one or more statistical tree models applied to the metagene values, wherein each model includes one or more nodes, each node representing a metagene, each node including a statistical predictive probability of sensitivity to an anti-cancer agent. Accordingly, the invention provides methods that use mixed trees, where a tree may contain at least two nodes, where each node represents a metagene representative of the sensitivity/resistance to a particular agent.


In one embodiment, the statistical predictive probability was derived from a Bayesian analysis. In another embodiment, the Bayesian analysis included a sequence of Bayes factor based tests of association to rank and select predictors that define a node binary split, the binary split including a predictor/threshold pair. Bayesian analysis is an approach to statistical analysis that is based on the Bayes law, which states that the posterior probability of a parameter p is proportional to the prior probability of parameter p multiplied by the likelihood of p derived from the data collected. This methodology represents an alternative to the traditional (or frequentist probability) approach: whereas the latter attempts to establish confidence intervals around parameters, and/or falsify a-priori null-hypotheses, the Bayesian approach attempts to keep track of how a priori expectations about some phenomenon of interest can be refined, and how observed data can be integrated with such a priori beliefs, to arrive at updated posterior expectations about the phenomenon. Bayesian analysis has been applied to numerous statistical models to predict outcomes of events based on available data. These include standard regression models, e.g. binary regression models, as well as to more complex models that are applicable to multi-variate and essentially non-linear data.


Another such model is commonly known as the tree model which is essentially based on a decision tree. Decision trees can be used in clarification, prediction and regression. A decision tree model is built starting with a root mode, and training data partitioned to what are essentially the “children” nodes using a splitting rule. For instance, for clarification, training data contains sample vectors that have one or more measurement variables and one variable that determines that class of the sample. Various splitting rules may be used. A statistical predictive tree model to which Bayesian analysis is applied may consistently deliver accurate results with high predictive capabilities. Other statistical models known to those of skill in the art may be used.


Gene expression signatures that reflect the activity of a given pathway may be identified using supervised classification method of analysis previously described (e.g., West, M. et al. Proc Natl Acad Sci USA 98, 11462-11467, 2001). The analysis selects a set of genes whose expression levels are most highly correlated with the classification of tumor samples into sensitivity to an anti-cancer agent versus no sensitivity to an anti-cancer agent. The dominant principal components from such a set of genes then defines a relevant phenotype-related metagene, and regression models assign the relative probability of sensitivity to an anti-cancer agent.


In one embodiment, each statistical tree model generated by the methods described herein comprises 2, 3, 4, 5, 6 or more nodes. In one embodiment of the methods described herein for defining a statistical tree model predictive of sensitivity/resistance to a therapeutic, the resulting model predicts cancer sensitivity to an anti-cancer agent with at least 70%, 80%, 85%, or 90% or higher accuracy. In another embodiment, the model predicts sensitivity to an anti-cancer agent with greater accuracy than clinical variables. In one embodiment, the clinical variables are selected from age of the subject, gender of the subject, tumor size of the sample, stage of cancer disease, histological subtype of the sample and smoking history of the subject. In one embodiment, the cluster of genes that define each metagene comprise at least 3, 4, 5, 6, 7, 8, 9, 10, 12 or 15 genes. In one embodiment, the correlation-based clustering is Markov chain correlation-based clustering or K-means clustering.


Gene Chips and Kits

Arrays and microarrays which contain the gene expression profiles for determining responsivity to the chemotherapeutic agents as disclosed here are also encompassed within the scope of this invention. Methods of making arrays are well-known in the art and as such do not need to be described in detail here.


Such arrays can contain the profiles of 5, 10, 15, 20, 25, 30, 40, 50, 75, 100, 150, 200 or more genes as disclosed in the Tables. Accordingly, arrays for detection of responsivity to particular therapeutic agents can be customized for diagnosis or treatment of specific cancers, such as ovarian cancer, breast cancer, or NSCLC. The array can be packaged as part of kit comprising the customized array itself and a set of instructions for how to use the array to determine an individual's responsivity to a specific cancer therapeutic agent.


Also provided are reagents and kits for practicing one or more of the above described methods. The subject reagents and kits thereof may vary greatly. Reagents of interest include reagents specifically designed for use in production of the above described metagene values.


One type of such reagent is an array probe of nucleic acids, such as a DNA chip, in which the genes defining the metagenes in the therapeutic efficacy predictive tree models are represented. A variety of different array formats are known in the art, with a wide variety of different probe structures, substrate compositions and attachment technologies. Representative array structures of interest include those described in U.S. Pat. Nos. 5,143,854; 5,288,644; 5,324,633; 5,432,049; 5,470,710; 5,492,806; 5,503,980; 5,510,270; 5,525,464; 5,547,839; 5,580,732; 5,661,028; 5,800,992; as well as WO 95/21265; WO 96/31622; WO 97/10365; WO 97/27317; EP 373 203; and EP 785 280; the disclosures of which are herein incorporated by reference.


The DNA chip is conveniently used to compare the expression levels of a number of genes at the same time. DNA chip-based expression profiling can be carried out, for example, by the method as disclosed in “Microarray Biochip Technology” (Mark Schena, Eaton Publishing, 2000). A DNA chip comprises immobilized high-density probes to detect a number of genes. Thus, the expression levels of many genes can be estimated at the same time by a single-round analysis. Namely, the expression profile of a specimen can be determined with a DNA chip. A DNA chip may comprise probes, which have been spotted thereon, to detect the expression level of the metagene-defining genes of the present invention, i.e. the genes described in Tables 1-8. A probe may be designed for each marker gene selected, and spotted on a DNA chip. Such a probe may be, for example, an oligonucleotide comprising 5-50 nucleotide residues. Methods for synthesizing such oligonucleotides on DNA chips are known to those skilled in the art. Longer DNAs can be synthesized by PCR or chemically. Methods for spotting long DNA, which is synthesized by PCR or the like, onto a glass slide are also known to those skilled in the art. A DNA chip that is obtained by the methods described above can be used for estimating the efficacy of a therapeutic agent in treating a subject afflicted with cancer according to the present invention.


DNA microarray and methods of analyzing data from microarrays are well-described in the art, including in DNA Microarrays: A Molecular Cloning Manual. Ed. by Bowtel and Sambrook (Cold Spring Harbor Laboratory Press, 2002); Microarrays for an Integrative Genomics by Kohana (MIT Press, 2002); A Biologist's Guide to Analysis of DNA Micraarray Data, by Knudsen (Wiley, John & Sons, Incorporated, 2002); DNA Microarrays: A Practical Approach, Vol. 205 by Schema (Oxford University Press, 1999); and Methods of Microarray Data Analysis II, ed. by Lin et al. (Kluwer Academic Publishers, 2002) all of which are incorporated herein by reference.


One aspect of the invention provides a kit comprising: (a) any of the gene chips described herein; and (b) one of the computer-readable mediums described herein.


In some embodiments, the arrays include probes for at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, or 50 of the genes listed in one of Tables 1-8. In certain embodiments, the number of genes that are from one of the Tables that are represented on the array is at least 5, at least 10, at least 25, at least 50, at least 75 or more, including all of the genes listed in the table. Where the subject arrays include probes for additional genes not listed in the tables, in certain embodiments the number % of additional genes that are represented does not exceed about 50%, 40%, 30%, 20%, 15%, 10%, 8%, 6%, 5%, 4%, 3%, 2% or 1%. In some embodiments, a great majority of genes in the collection are genes that define the metagenes of the invention, whereby great majority is meant at least about 75%, usually at least about 80% and sometimes at least about 85, 90, 95% or higher, including embodiments where 100% of the genes in the collection are metagene-defining genes. In an alternative embodiment, the arrays for use in the invention may include a majority of probes that are not listed in any of Tables 1-8.


The kits of the subject invention may include the above described arrays or gene chips. The kits may further include one or more additional reagents employed in the various methods, such as primers for generating target nucleic acids, dNTPs and/or rNTPs, which may be either premixed or separate, one or more uniquely labeled dNTPs and/or rNTPs, such as biotinylated or Cy3 or Cy5 tagged dNTPs, gold or silver particles with different scattering spectra, or other post synthesis labeling reagent, such as chemically active derivatives of fluorescent dyes, enzymes, such as reverse transcriptases, DNA polymerases, RNA polymerases, and the like, various buffer mediums, e.g. hybridization and washing buffers, prefabricated probe arrays, labeled probe purification reagents and components, like spin columns, etc., signal generation and detection reagents, e.g. streptavidin-alkaline phosphatase conjugate, chemifluorescent or chemiluminescent substrate, and the like.


In addition to the above components, the subject kits further include instructions for practicing the subject methods. These instructions may be present in the subject kits in a variety of forms, one or more of which may be present in the kit. One form in which these instructions may be present is as printed information on a suitable medium or substrate, e.g., a piece or pieces of paper on which the information is printed, in the packaging of the kit, in a package insert, etc. Yet another means would be a computer readable medium, e.g., diskette, CD, etc., on which the information has been recorded. Yet another means that may be present is a website address which may be used via the internet to access the information at a remote site. Any convenient means of conveying instructions may be present in the kits.


The kits also include packaging material such as, but not limited to, ice, dry ice, styrofoam, foam, plastic, cellophane, shrink wrap, bubble wrap, paper, cardboard, starch peanuts, twist ties, metal clips, metal cans, drierite, glass, and rubber.


Diagnostic Business Methods

One aspect of the invention provides methods of conducting a diagnostic business, including a business that provides a health care practitioner with diagnostic information for the treatment of a subject afflicted with cancer. One such method comprises one, more than one, or all of the following steps: (i) obtaining an tumor sample from the subject; (ii) determining the expression level of multiple genes in the sample; (iii) defining the value of one or more metagenes from the expression levels of step (ii), wherein each metagene is defined by extracting a single dominant value using single value decomposition (SVD) from a cluster of genes associated with sensitivity to an anti-cancer agent; (iv) averaging the predictions of one or more statistical tree models applied to the values, wherein each model includes one or more nodes, each node representing a meta gene, each node including a statistical predictive probability of sensitivity to an anti-cancer agent, wherein at least one metagene is one of metagenes 1-7; and (v) providing the health care practitioner with the prediction from step (iv).


In one embodiment, obtaining a tumor sample from the subject is effected by having an agent of the business (or a subsidiary of the business) remove a tumor sample from the subject, such as by a surgical procedure. In another embodiment, obtaining a tumor sample from the subject comprises receiving a sample from a health care practitioner, such as by shipping the sample, preferably frozen. In one embodiment, the sample is a cellular sample, such as a mass of tissue. In one embodiment, the sample comprises a nucleic acid sample, such as a DNA, cDNA, mRNA sample, or combinations thereof, which was derived from a cellular tumor sample from the subject. In one embodiment, the prediction from step (iv) is provided to a health care practitioner, to the patient, or to any other business entity that has contracted with the subject.


In one embodiment, the method comprises billing the subject, the subject's insurance carrier, the health care practitioner, or an employer of the health care practitioner. A government agency, whether local, state or federal, may also be billed for the services. Multiple parties may also be billed for the service.


In some embodiments, all the steps in the method are carried out in the same general location. In certain embodiments, one or more steps of the methods for conducting a diagnostic business are performed in different locations. In one embodiment, step (ii) is performed in a first location, and step (iv) is performed in a second location, wherein the first location is remote to the second location. The other steps may be performed at either the first or second location, or in other locations. In one embodiment, the first location is remote to the second location. A remote location could be another location (e.g. office, lab, etc.) in the same city, another location in a different city, another location in a different state, another location in a different country, etc. As such, when one item is indicated as being “remote” from another, what is meant is that the two items are at least in different buildings, and may be at least one mile, ten miles, or at least one hundred miles apart In one embodiment, two locations that are remote relative to each other arc at least 1, 2, 3, 4, 5, 10, 20, 50, 100, 200, 500, 1000, 2000 or 5000 km apart. In another embodiment, the two locations are in different countries, where one of the two countries is the United States.


Some specific embodiments of the methods described herein where steps are performed in two or more locations comprise one or more steps of communicating information between the two locations. “Communicating” information means transmitting the data representing that information as electrical signals over a suitable communication channel (for example, a private or public network). “Forwarding” an item refers to any means of getting that item from one location to the next, whether by physically transporting that item or otherwise (where that is possible) and includes, at least in the case of data, physically transporting a medium carrying the data or communicating the data. The data may be transmitted to the remote location for further evaluation and/or use. Any convenient telecommunications means may be employed for transmitting the data, e.g., facsimile, modem, internet, etc.


In one specific embodiment, the method comprises one or more data transmission steps between the locations. In one embodiment, the data transmission step occurs via an electronic communication link, such as the internet. In one embodiment, the data transmission step from the first to the second location comprises experimental parameter data, such as the level of gene expression of multiple genes. In some embodiments, the data transmission step from the second location to the first location comprises data transmission to intermediate locations. In one specific embodiment, the method comprises one or more data transmission substeps from the second location to one or more intermediate locations and one or more data transmission substeps from one or more intermediate locations to the first location, wherein the intermediate locations are remote to both the first and second locations. In another embodiment, the method comprises a data transmission step in which a result from gene expression is transmitted from the second location to the first location.


In one embodiment, the methods of conducting a diagnostic business comprise the step of determining if the subject carries an allelic form of a gene whose presence correlates to sensitivity or resistance to a chemotherapeutic agent. This may be achieved by analyzing a nucleic acid sample from the patient and determining the DNA sequence of the allele. Any technique known in the art for determining the presence of mutations or polymorphisms may be used. ‘The method is not limited to any particular mutation or to any particular allele or gene. For example, mutations in the epidermal growth factor receptor (EGFR) gene are found in human lung adenocarcinomas and are associated with sensitivity to the tyrosine kinase inhibitors gefitinib and erlotinib. (See, e.g., Yi et al. Proc Natl Acad Sci USA. 2006 May 16; 103(20):7817-22; Shimato et al. Neuro-oncol. 2006 April; 8(2): 137-44). Similarly, mutations in breast cancer resistance protein (HCRP) modulate the resistance of cancer cells to BCRP-substrate anticancer agents (Yanase et al., Cancer Lett. 2006 Mar. 8; 234(1):73-80).


Computer Readable Media Comprising Gene Expression Profiles

The invention also contemplates computer readable media that comprises gene expression profiles. Such media can contain all or part of the gene expression profiles of the genes listed in the Tables that comprise the responsivity predictor set. The media can be a list of the genes or contain the raw data for running a user's own statistical calculation, such as the methods disclosed herein.


Another aspect of the invention provides a program product (i.e., software product) for use in a computer device that executes program instructions recorded in a computer-readable medium to perform one or more steps of the methods described herein, such for estimating the efficacy of a therapeutic agent in treating a subject afflicted with cancer.


One aspect of the invention provides a computer readable medium having computer readable program codes embodied therein, the computer readable medium program codes performing one or more of the following functions: defining the value of one or more metagenes from the expression levels of genes in known responsive and sensitive cells; defining a metagene value by extracting a single dominant value using singular value decomposition (SVD) from a cluster of genes associated with tumor sensitivity to a therapeutic agent; averaging the predictions of one or more statistical tree models applied to the values of the metagenes; or averaging the predictions of one or more binary regression models applied to the values of the metagenes, wherein each model includes a statistical predictive probability of tumor sensitivity to a therapeutic agent.


Another related aspect of the invention provides kits comprising the program product or the computer readable medium, optionally with a computer system. One aspect of the invention provides a system, the system comprising: a computer (See FIG. 15); a computer readable medium, operatively coupled to the computer, the computer readable medium program codes performing one or more of the following functions: defining the value of one or more metagenes from the expression levels genes; defining a metagene value by extracting a single dominant value using singular value decomposition (SVD) from a cluster of genes associated tumor sensitivity to a therapeutic agent; averaging the predictions of one or more statistical tree models applied to the values of the metagenes; or averaging the predictions of one or more binary regression models applied to the values of the metagenes, wherein each model includes a statistical predictive probability of tumor sensitivity to a therapeutic agent.


In one embodiment, the program product comprises: a recordable medium; and a plurality of computer-readable instructions executable by the computer device to analyze data from the array hybridization steps, to transmit array hybridization from one location to another, or to evaluate genome-wide location data between two or more genomes. Computer readable media include, but are not limited to, CD-ROM disks (CD-R, CD-RW), DVD-RAM disks, DVD-RW disks, floppy disks and magnetic tape.


A related aspect of the invention provides kits comprising the program products described herein. The kits may also optionally contain paper and/or computer-readable format instructions and/or information, such as, but not limited to, information on DNA microarrays, on tutorials, on experimental procedures, on reagents, on related products, on available experimental data, on using kits, on chemotherapeutic agents including their toxicity, and on other information. The kits optionally also contain in paper and/or computer-readable format information on minimum hardware requirements and instructions for running and/or installing the software. The kits optionally also include, in a paper and/or computer readable format, information on the manufacturers, warranty information, availability of additional software, technical services information, and purchasing information. The kits optionally include a video or other viewable medium or a link to a viewable format on the internet or a network that depicts the use of the software, and/or use of the kits. The kits also include packaging material such as, but not limited to, styrofoam, foam, plastic, cellophane, shrink wrap, bubble wrap, paper, cardboard, starch peanuts, twist ties, metal clips, metal cans, drierite, glass, and rubber.


The analysis of data, as well as the transmission of data steps, can be implemented by the use of one or more computer systems. Computer systems are readily available. The processing that provides the displaying and analysis of image data for example, can be performed on multiple computers or can be performed by a single, integrated computer or any variation thereof. The components contained in the computer system are those typically found in general purpose computer systems used as servers, workstations, personal computers, network terminals, and the like. See FIG. 15. In fact, these components are intended to represent a broad category of such computer components that are well known in the art.



FIG. 15 shows a functional block diagram of general purpose computer system 1500 for performing the functions of the software according to an illustrative embodiment of the invention. The exemplary computer system 1500 includes a central processing unit (CPU) 3002, a memory 1504, and an interconnect bus 1506. The CPU 1502 may include a single microprocessor or a plurality of microprocessors for configuring computer system 1500 as a multi-processor system. The memory 1504 illustratively includes a main memory and a read only memory. The computer 1500 also includes the mass storage device 1508 having, for example, various disk drives, tape drives, etc. The main memory 1504 also includes dynamic random access memory (DRAM) and high-speed cache memory. In operation, the main memory 1504 stores at least portions of instructions and data for execution by the CPU 1502.


The mass storage 1508 may include one or more magnetic disk or tape drives or optical disk drives, for storing data and instructions for use by the CPU 1502. At least one component of the mass storage system 1508, preferably in the form of a disk drive or tape drive, stores one or more databases, such as databases containing of transcriptional start sites, genomic sequence, promoter regions, or other information.


The mass storage system 1508 may also include one or more drives for various portable media, such as a floppy disk, a compact disc read only memory (CD-ROM), or an integrated circuit non-volatile memory adapter (i.e., PC-MCIA adapter) to input and output data and code to and from the computer system 1500.


The computer system 1500 may also include one or more input/output interfaces for communications, shown by way of example, as interface 1510 for data communications via a network. The data interface 1510 may be a modem, an Ethernet card or any other suitable data communications device. To provide the functions of a computer system according to FIG. 15 the data interface 1510 may provide a relatively high-speed link to a network, such as an intranet, internet, or the Internet, either directly or through an another external interface. The communication link to the network may be, for example, optical, wired, or wireless (e.g., via satellite or cellular network). Alternatively, the computer system 1500 may include a mainframe or other type of host computer system capable of Web-based communications via the network.


The computer system 1500 also includes suitable input/output ports or use the interconnect bus 1506 for interconnection with a local display 1512 and keyboard 1514 or the like serving as a local user interface for programming and/or data retrieval purposes. Alternatively, server operations personnel may interact with the system 1500 for controlling and/or programming the system from remote terminal devices via the network.


The following examples are provided to illustrate aspects of the invention but are not intended to limit the invention in any manner.


EXAMPLES
Example 1
A Gene Expression Based Predictor of Sensitivity to Docetaxel

The NCI-60 panel49 was used to develop predictors of chemotherapeutic drug response, and cell lines that were most resistant or sensitive to docetaxel were identified (FIG. 1A, B). Genes whose expression most highly correlated with drug sensitivity, using Bayesian binary regression analysis, were selected to develop a model that differentiates a pattern of docetaxel sensitivity from resistance. A gene expression signature consisting of 50 genes was identified that classified on the basis of docetaxel sensitivity (FIG. 1 B, bottom panel).


In addition to leave-one-out cross validation, we utilized an independent dataset derived from docetaxel sensitivity assays in a series of 30 lung and ovarian cancer cell lines for further validation. As shown in FIG. 1C (top panel), the correlation between the predicted probability of sensitivity to docetaxel (in both lung and ovarian cell lines) and the respective IC50 for docetaxel confirmed the capacity of the docetaxel predictor to predict sensitivity to the drug in cancer cell lines (FIG. 7). In each case, the accuracy exceeded 80%. Finally, a second independent dataset including 29 lung cancer cell lines (Gemma A, GEO accession number: GSE 4127), was used to predict and measure docetaxel sensitivity. As shown in FIG. 1C (bottom panel), the docetaxel sensitivity model developed from the NCI-60 panel again predicted sensitivity in this independent data set, again with an accuracy exceeding 80%.


Example 2
Utilization of the Expression Signature to Predict Docetaxel Response in Patients

The development of a gene expression signature capable of predicting in vitro docetaxel sensitivity provides a tool that might be useful in predicting response to the drug in patients. We made use of published studies with clinical and genomic data that linked gene expression data with clinical response to docetaxel in a breast cancer neoadjuvant study50 (FIG. 1D) to test the capacity of the in vitro docetaxel sensitivity predictor to accurately identify those patients that responded to docetaxel. Using a 0.45 predicted probability of response as the cut-off for predicting positive response, as determined by ROC curve analysis (FIG. 7A), the in vitro generated profile correctly predicted docetaxel response in 22 out of 24 patient samples, achieving an overall accuracy of 91.6% (FIG. 1D). Applying a Mann-Whitney U test for statistical significance demonstrates the capacity of the predictor to distinguish resistant from sensitive patients (FIG. 1D, right panel). We extended this further by predicting the response to docetaxel as salvage therapy for ovarian cancer. As shown in FIG. 1E, the prediction of response to docetaxel in patients with advanced ovarian cancer achieved an accuracy exceeding 85% (FIG. 1E, middle panel). Further, an analysis of statistical significance demonstrated the capacity of the predictors to distinguish patients with resistant versus sensitive disease (FIG. 1E, right panel).


We also performed a complementary analysis using the patient response data to generate a predictor and found that the in vivo generated signature of response predicted sensitivity of NCI-60 cell lines to docetaxel (FIG. 7B). This crossover is further emphasized by the fact that the genes represented in either the initial in vitro generated docetaxel predictor or the alternative in vivo predictor exhibit considerable overlap. (Table 4). We also note that the predictor of docetaxel sensitivity developed from the NCI-60 data was more accurate in predicting patient response in the ovarian samples than the predictor developed from the breast neoadjuvant patient data (85.7% vs. 64.3%) (FIG. 7C).


Example 3
Development of a Panel of Gene Expression Signatures that Predict Sensitivity to Chemotherapeutic Drugs

Given the development of a docetaxel response predictor, we examined the NCI-60 data set for other opportunities to develop predictors of chemotherapy response. Shown in FIG. 2A are a series of expression profiles developed from the NCI-60 dataset that predict response to topotecan, adriamycin, etoposide, 5-fluorouracil (5-FU), taxol (paclitaxel), and cyclophosphamide (cytotoxan). In each case, the leave-one-out cross validation analyses demonstrate a capacity of these profiles to accurately predict the samples utilized in the development of the predictor (FIG. 8B). Each profile was then further validated using in vitro response data from independent datasets; in each case, the profile developed from the NCI-60 data was capable of accurately (>85%) predicting response in the separate dataset of approximately 30 cancer cell lines for which the dose response information and relevant Affymetrix U133A gene expression data is publicly available37 (FIG. 8C) and Table 16). Once again, applying a Mann-Whitney U test for statistical significance demonstrates the capacity of the predictor to distinguish resistant from sensitive patients (FIG. 2B).


In addition to the capacity of each signature to distinguish cells that are sensitive or resistant to a particular drug, we also evaluated the extent to which a signature was also specific for an individual chemotherapeutic agent. From the example shown in FIG. 9, using the validations of chemosensitivity seen in the independent European (UC) cell line data it is clear that each of the signatures is specific for the drug that was used to develop the predictor. In each case, individual predictors of response to the various cytotoxic drugs was plotted against cell lines known to be sensitive or resistant to a given chemotherapeutic agent (e.g., adriamycin, paclitaxel).


Given the ability of the in vitro developed gene expression profiles to predict response to docetaxel in the clinical samples, we extended this approach to test the ability of additional signatures to predict response to commonly used salvage therapies for ovarian cancer and an independent data set of samples from adriamycin treated patients (Evans W, GSE650, GSE651). As shown in FIG. 2C, each of these predictors was capable of accurately predicting the response to the drugs in patient samples, achieving an accuracy in excess of 81% overall. In each case, the positive and negative predictive values confirm the validity and clinical utility of the approach (Table 16).


Example 4
Chemotherapy Response Signatures Predict Response to Multi-Drug Regimens

Many therapeutic regimens make use of combinations of chemotherapeutic drugs raising the question as to the extent to which the signatures of individual therapeutic response will also predict response to a combination of agents. To address this question, we have made use of data from a breast neoadjuvant treatment that involved the use of paclitaxel, 5-fluorouracil, adriamycin, and cyclophosphamide (TFAC)55,56 (FIG. 3A). Using available data from the 51 patients to then predict response with each of the single agent signatures (paclitaxel, 5-FU, adriamycin and cyclophosphamide) developed from the NCI-60 cell line analysis; we then compared to the clinical outcome information which was represented as complete pathologic response. As shown in FIG. 3A (middle panel), the predicted response based on each of the individual chemosensitivity signatures indicated a significant distinction between the responders (n=13) and non-responders (n=38) with the exception of 5-fluorouracil. Importantly, the combined probability of sensitivity to the four agents in this TFAC neoadjuvant regimen was calculated using the probability theorem and it is clear from this analysis that the prediction of response based on a combined probability of sensitivity, built from the individual chemosensitivity predictions yielded a statistically significant (p<0.0001, Mann Whitney U) distinction between the responders and non-responders (FIG. 3A, bottom panel).


As a further validation of the capacity to predict response to combination therapy, we made use of gene expression data generated from a collection of breast cancer (n=45) samples from patients who received 5-fluorouracil, adriamycin and cyclophosphamide (FAC) in the adjuvant chemotherapy set. As shown in FIG. 3B (top panel), the predicted response based on signatures for 5-FU, adriamycin, and cyclophosphamide indicated a significant distinction between the responders (n=34) and non-responders (n=11) for each of the single agent predictors. Furthermore, the combined probability of sensitivity to the three agents in the FAC regimen was calculated and shown in the middle panel of FIG. 3B. It is evident from this analysis that the prediction of response based on a combined probability of sensitivity to the FAC regimen yielded a clear, significant (p<0.001, Mann Whitney U) distinction between the responders and non-responders (accuracy: 82.2%, positive predictive value: 90.3%, negative predictive value: 64.3%). We note that while it is difficult to interpret the prediction of clinical response in the adjuvant setting since many of these patients were likely free of disease following surgery, the accurate identification of non-responders is a clear endpoint that does confirm the capacity of the signatures to predict clinical response.


As a further measure of the relevance of the predictions, we examined the prognostic significance of the ability to predict response to FAC. As shown in FIG. 3B (bottom panel), there was a clear distinction in the population of patients identified as sensitive or resistant to FAC, as measured by disease-free survival (sensitive=blue, resistant=red). These results, taken together with the accuracy of prediction of response in the neoadjuvant setting where clinical endpoints are uncomplicated by confounding variables such as prior surgery, and results of the single agent validations, leads us to conclude that the signatures of chemosensitivity generated from the NCI-60 panel do indeed have the capacity to predict therapeutic response in patients receiving either single agent or combination chemotherapy (Table 17).


When comparing individual genes that constitute the predictors, it was interesting to observe that the gene coding for MAP-Tau, described previously as a determinant of paclitaxel sensitivity,56 was also identified as a discriminator gene in the paclitaxel predictor generated using the NCI-60 data. Although, similar to the docetaxel example described earlier, a predictor for TFAC chemotherapy developed using the NCI-60 data was superior to the ability of the MAP-Tau based predictor described by Pusztai et al (Table 18).


Example 5
Patterns of Predicted Chemotherapy Response Across a Spectrum of Tumors

The availability of genomic-based predictors of chemotherapy response could potentially provide an opportunity for a rational approach to selection of drugs and combinations of drugs. With this in mind, we have utilized the panel of chemotherapy response predictors described in FIG. 6 to profile the potential options for use of these agents, by predicting the likelihood of sensitivity to the agents in a large collection of breast, lung, and ovarian tumor samples. We then clustered the samples according to patterns of predicted sensitivity to the various chemotherapeutics, and plotted a heatmap in which high probability of sensitivity response is indicated by red and low probability or resistance is indicated by blue (FIG. 4).


There are clearly evident patterns of predicted sensitivity to the various agents. In many cases, the predicted sensitivities to the chemotherapeutic agents are consistent with the previously documented efficacy of single agent chemotherapies in the individual tumor types57. For instance, the predicted response rate for etoposide, adriamycin, cyclophosphamide, and 5-FU approximate the observed response for these single agents in breast cancer patients (FIG. 11). Likewise, the predicted sensitivity to etoposide, docetaxel, and paclitaxel approximates the observed response for these single agents in lung cancer patients (FIG. 11). This analysis also suggests possibilities for alternate treatments. As an example, it would appear that breast cancer patients likely to respond to 5-fluorouracil are resistant to adriamycin and docetaxel (FIG. 10A). Likewise, in lung cancer, docetaxel sensitive populations are likely to be resistant to etoposide (FIG. 10B). This is a potentially useful observation considering that both etoposide and docetaxel are viable front-line options (in conjunction with cis/carboplatin) for patients with lung cancer58 A similar relationship is seen between topotecan and adriamycin, both agents used in salvage chemotherapy for ovarian cancer (FIG. 10C). Thus, by identifying patients/patient cohorts resistant to certain standard of care agents, one could avoid the side effects of that agent (e.g. topotecan) without compromising patient outcome, by choosing an alternative standard of care (e.g., adriamycin).


Example 6
Linking Predictions of Chemotherapy Sensitivity to Oncogenic Pathway Deregulation

Most patients who are resistant to chemotherapeutic agents are then recruited into a second or third line therapy or enrolled in a clinical trial.38,59 Moreover, even those patients who initially respond to a given agent are likely to eventually suffer a relapse and in either case, additional therapeutic options are needed. As one approach to identifying such options, we have taken advantage of our recent work that describes the development of gene expression signatures that reflect the activation of several oncogenic pathways.36 To illustrate the approach, we first stratified the NCI cell lines based on predicted docetaxel response and then examined the patterns of pathway deregulation associated with docetaxel sensitivity or resistance (FIG. 13A). Regression analysis revealed a significant relationship between PI3 kinase pathway deregulation and docetaxel resistance, as seen by the linear relationship (p=0.001) between the probability of PI3 kinase activation and the IC50 of docetaxel in the cell lines (FIG. 12 and Table 8).


The results linking docetaxel resistance with deregulation of the PI3 kinase pathway, suggests an opportunity to employ a PI3 kinase inhibitor in this subgroup, given our recent observations that have demonstrated a linear positive correlation between the probability of pathway deregulation and targeted drug sensitivity.36 To address this directly, we predicted docetaxel sensitivity and probability of oncogenic pathway deregulation using DNA microarray data from 17 NSCLC cell lines (FIG. 5A, top panel). Consistent with the analysis of the NCI-60 cell line panel, the cell lines predicted to be resistant to docetaxel were also predicted to exhibit PI3 kinase pathway activation (p=0.03, log-rank test, FIG. 14). In parallel, the lung cancer cell lines were subjected to assays for sensitivity to a PI3 kinase specific inhibitor (LY-294002), using a standard measure of cell proliferation.36, 38, 59 As shown by the analysis in FIG. 5B (top left panel), the cell lines showing an increased probability of PI3 kinase pathway activation were also more likely to respond to a PI3 kinase inhibitor (LY-294002) (p=0.001, log-rank test)). The same relationship held for prediction of resistance to docetaxel—these cells were more likely to be sensitive to PI3 kinase inhibition (p<0.001, log-rank test) (FIG. 5B, top right panel).


An analysis of a panel of ovarian cancer cell lines provided a second example. Ovarian cell lines that are predicted to be topotecan resistant (FIG. 5A, bottom panel) have a higher likelihood of Src pathway deregulation and there is a significant linear relationship (p=0.001, log rank) between the probability of topotecan resistance and sensitivity to a drug that inhibits the Src pathway (SU6656) (FIG. 5B, bottom right panel). The results of these assays clearly demonstrate an opportunity to potentially mitigate drug resistance (e.g., docetaxel or topotecan) using a specific pathway-targeted agent, based on a predictor developed from pathway deregulation (i.e., PI3 kinase or Src inhibition).


Taken together, these data demonstrate an approach to the identification of therapeutic options for chemotherapy resistant patients, as well as the identification of novel combinations for chemotherapy sensitive patients, and thus represents a potential strategy to a more effective treatment plan for cancer patients, after future prospective validations trials (FIG. 6).


Example 7
Methods

NCI-60 data. The (−log 10(M)) GI50/IC50, TGI (Total Growth Inhibition dose) and LC50 (50% cytotoxic dose) data was used to populate a matrix with MA TLAB software, with the relevant expression data for the individual cell lines. Where multiple entries for a drug screen existed (by NCS number), the entry with the largest number of replicates was included. Incomplete data were assigned as Nan (not a number) for statistical purposes. To develop an in vitro gene expression based predictor of sensitivity/resistance from the pharmacologic data used in the NCI-60 drug screen studies, we chose cell lines within the NCI-60 panel that would represent the extremes of sensitivity to a given chemotherapeutic agent (mean GI50+/−1 SD). Relevant expression data (updated data available on the Affymetrix U95A2 GeneChip) for the solid tumor cell lines and the respective pharmacological data for the chemotherapeutics was downloaded from the NCI website (http://dtp.nci.nih.gov/docs/cancer/cancer_data.html). The individual drug sensitivity and resistance data from the selected solid tumor NCI-60 cell lines was then used in a supervised analysis using binary regression methodologies, as described previously,60 to develop models predictive of chemotherapeutic response.


Human ovarian cancer samples. We measured expression of 22,283 genes in 13 ovarian cancer cell lines and 119 advanced (FIGO stage III/IV) serous epithelial ovarian carcinomas using Affymetrix U133A GeneChips. All ovarian cancers were obtained at initial cytoreductive surgery from patients. All tissues were collected under the auspices of respective institutional (Duke University Medical Center and H. Lee Moffitt Cancer Center) IRB approved protocols involving written informed consent.


Full details of the methods used for RNA extraction and development of gene expression signatures representing deregulation of oncogenic pathways in the tumor samples were recently described.36 Response to therapy was evaluated using standard criteria for patients with measurable disease, based upon WHO guidelines.28


Lung and ovarian cancer cell culture. Total RNA was extracted and oncogenic pathway predictions was performed similar to the methods described previously.36


Cross platform Affymetrix Gene Chip comparison. To map the probe sets across various generations of Affymetrix GeneChip arrays, we utilized an in-house program, Chip Comparer (http://tenero.duhs.duke.edu/genearray/perl/chip/chipcomparer.pl) as described previously.36


Cell proliferation assays. Growth curves for cells were produced by plating 500-10,000 cells per well in 96-well plates. The growth of cells at 12 hr time points (from t=12 hrs) was determined using the CellTiter 96 Aqueous One 23 Solution Cell Proliferation Assay Kit by Promega, which is a colorimetric method for determining the number of growing cells.36 The growth curves plot the growth rate of cells vs. each concentration of drug tested against individual cell lines. Cumulatively, these experiments determined the concentration of cells to use for each cell line, as well as the dosing range of the inhibitors. The final dose-response curves in our experiments plot the percent of cell population responding to the chemotherapy vs. the concentration of the drug for each cell line. Sensitivity to docetaxel and a phosphatidylinositol 3-kinase (PI3 kinase) inhibitor (LY-294002)36 in 17 lung cell lines, and topotecan and a Src inhibitor (SU6656) in 13 ovarian cell lines was determined by quantifying the percent reduction in growth (versus DMSO controls) at 96 hrs using a standard MTT colorimetric assay.36 Concentrations used ranged from 1-10 nM for docetaxel, 300 nM-10 μ/M (SU6656), and 300 nM-10M for LY-294002. All experiments were repeated at least three times.


Statistical analysis methods. Analysis of expression data are as previously described.36,60-62 Briefly, prior to statistical modeling, gene expression data is filtered to exclude probe sets with signals present at background noise levels, and for probe sets that do not vary significantly across samples. Each signature summarizes its constituent genes as a single expression profile, and is here derived as the top principal components of that set of genes. When predicting the chemosensitivity patterns or pathway activation of cancer cell lines or tumor samples, gene selection and identification is based on the training data, and then metagene values are computed using the principal components of the training data and additional cell line or tumor expression data. Bayesian fitting of binary probit regression models to the training data then permits an assessment of the relevance of the metagene signatures in within-sample classification,60 and estimation and uncertainty assessments for the binary regression weights mapping metagenes to probabilities. To guard against over-fitting given the disproportionate number of variables to samples, we also performed leave-one-out cross validation analysis to test the stability and predictive capability of our model. Each sample was left out of the data set one at a time, the model was refitted (both the metagene factors and the partitions used) using the remaining samples, and the phenotype of the held out case was predicted and the certainty of the classification was calculated. Given a training set of expression vectors (of values across metagenes) representing two biological states, a binary probit regression model, of predictive probabilities for each of the two states (resistant vs. sensitive) for each case is estimated using Bayesian methods. Predictions of the relative oncogenic pathway status and chemosensitivity of the validation cell lines or tumor samples are then evaluated using methods previously described36,60 producing estimated relative probabilities—and associated measures of uncertainty—of chemosensitivity/oncogenic pathway deregulation across the validation samples. In instances where a combined probability of sensitivity to a combination chemotherapeutic regimen was required based on the individual drug sensitivity patterns, we employed the theorem for combined probabilities as described by Feller: [Probability (Pr) of (A), (B), (C) . . . (N)]=ΣPr (A)+Pr (B)+Pr (C) . . . [Pr (N)−[Pr(A)×Pr(B)×Pr(C) . . . ×Pr (N)]. Hierarchical clustering of tumor predictions was performed using Gene Cluster 3.0.63 Genes and tumors were clustered using average linkage with the uncentered correlation similarity metric. Standard linear regression analyses and their significance (log rank test) were generated for the drug response data and correlation between drug response and probability of chemosensitivity/pathway deregulation using GraphPad® software.


REFERENCE BIBLIOGRAPHY



  • 1. Levin L, Simon R, Hryniuk W: Importance of multi agent chemotherapy regimens in ovarian carcinoma: dose intensity analysis. J Natl. Canc. Inst. 85: 1732-1742, 1993

  • 2. McGuire W P, Hoskins W J, Brady M F, et al: Assessment of dose-intensive therapy in suboptimally debulked ovarian cancer: a Gynecologic Oncology Group study. J Clin. Oncol. 13:1589-1599, 1995

  • 3. Jodrell Dl, Egorin M J, Canetta R M, et al: Relationships between carboplatin explosure and tumor response and toxicity in patients with ovarian cancer. J. Clin. Oncol. 10:520-528, 1992

  • 4. McGuire W P, Hoskins W J, Brady M F, et al: Cyclophosphamide and cisplatin compared with paclitaxel and cisplatin in patients with stage III and stage IV ovarian cancer. N. Engl. J. Med. 334:1-6, 1996

  • 5. McGuire W P, Brady M F, Ozols R F: The Gynecologic Oncology Group experience in ovarian cancer. Ann. Oncol. 10:29-34, 1999

  • 6. Piccart M J, Bertelsen K, Stuart G, et al: Long-term follow-up confirms a survival advantage of the paclitaxel-cisplatin regimen over the cyclophosphamide-cisplatin combination in advanced ovarian cancer. Int. J. Gyneeol. Cancer 13:144-148, 2003

  • 7. Wenham R M, Lancaster J M, Berchuck A: Molecular aspects of ovarian cancer. Best Pract. Res. Clin. Obstet. Gynaecol. 16:483-497, 2002

  • 8. Berchuck A, Kohler M F, Marks J R, et al: The p53 tumor suppressor gene frequently is altered in gynecologic cancers. Am. J. Obstet. Gynecol. 170:246-252, 1994

  • 9. Kohler M F, Marks J R, Wiseman R W, et al: Spectrum of mutation and frequency of allelic deletion of the p53 gene in ovarian cancer. J. Natl. Cane. Inst. 85:1513-1519, 1993

  • 10. Havrilesky L, Alvarez A A, Whitaker R S, et al: Loss of expression of the p16 tumor suppressor gene is more frequent in advanced ovarian cancers lacking p53 mutations. Gynecol. Oncol. 83:491-500, 2001

  • 11. Reles A, Wen W H, Schmider A, et al: Correlation of 53 mutations with resistance to platinum-based chemotherapy and shortened survival in ovarian cancer. Clinical Cancer Research 7:2984-2997, 2001

  • 12. Schmider A, Gee C, Friedmann W, et al: p21 (WAF lICIP 1) protein expression is associated with prolonged survival but not with p53 expression in epithelial ovarian carcinoma. Gynecol. On col. 77:237-242, 2000

  • 13. Wong K K, Cheng R S, Mok S C: Identification of differentially expressed genes from ovarian cancer cells by MICRO MAX cDNA microarray system. Biotechniques 30:670675, 2001

  • 14. Welsh J B, Zarrinkar P P, Sapinoso L M, et al: Analysis of gene expression profiles in normal and neoplastic ovarian tissue samples identifies candidate molecular markers of epithelial ovarian cancer. Proc. Natl. Acad. Sci. USA 98: 1176-1181, 2001

  • 15. Shridhar Y, Lee J-S, Pandita A, et al: Genetic analysis of early-versus late-state ovarian tumors. Cancer Res. 61:5895-5904, 2001

  • 16. Schummer M, Ng W W, Bumgarner R E, et al: Comparative hybridization of an array of 21,500 ovarian cDNAs for the discovery of genes overexpressed in ovarian carcinomas. Gene 238:375-385, 1999

  • 17. Ono K, Tanaka T, Tsunoda T, et al: Identification by cDNA micro array of genes involved in ovarian carcinogenesis. Cancer Res. 60:5007-5011, 2000

  • 18. Sawiris G P, Sherman-Baust C A, Becker K G, et al: Development of a highly specialized cDNA array for the study and diagnosis of epithelial ovarian cancer. Cancer Res. 62:2923-2928, 2002

  • 19. Jazaeri A A, Yee C J, Sotiriou C, et al: Gene expression profiles of BRCA1-linked, BRCA2-linked, and sporadic ovarian cancers. J Natl. Canc. Inst. 94:990-1000, 2002

  • 20. Schaner M E, Ross D T, Ciaravino G, et al: Gene expression patterns in ovarian carcinomas. Mol. Biol. Cell 14:4376-4386, 2003

  • 21. Lancaster J M, Dressman H, Whitaker R S, et al: Gene expression patterns that characterize advanced stage serous ovarian cancers. J Surgical Gynecol. Invest. 11:51-59, 2004

  • 22. Berchuck A, Iversen E S, Lancaster J M, et al: Patterns of gene expression that characterize long term survival in advanced serous ovarian cancers. Clin. Can. Res. 11:3686-3696, 2005

  • 23. Berchuck A, Iversen E, Lancaster J M, et al: Prediction of optimal versus suboptimal cytoreduction of advanced stage serous ovarian cancer using microarrays. Am. J. Obstet. Gynecol. 190:910-925, 2004

  • 24. Jazaeri A A, Awtrey Cs, Chandramouli G V, et al: Gene expression profiles associated with response to chemotherapy in epithelial ovarian cancers. Clin. Cancer Res. 11:6300-6310, 2005

  • 25. Helleman J, Jansen M P, Span P N, et al: Molecular profiling of platinum resistant ovarian cancer. Int. J. Cancer 118: 1963-1971, 2005

  • 26. Spentzos D, Levine D A, Kolia s, et al: Unique gene expression profile based on pathologic response in epithelial ovarian cancer. J. Clin. Oncol. 23:7911-7918, 2005

  • 27. Spentzos D, Levine D A, Ramoni M F, et al: Gene expression signature with independent prognostic significance in epithelial ovarian cancer. J. Clin. Oneal. 22:4700-4710, 2004

  • 28. Miller A B, Hoogstraten B, Staquet M, et al: Reporting results of cancer treatment. Cancer 47:207-214, 1981

  • 29. Rustin G J, Nelstrop A E, Bentzen S M, et al: Use of tumor markers in monitoring the course of ovarian cancer. Ann. Oncol. 10:21-27, 1999

  • 30. Rustin G J, Nelstrop A E, McClean P, et al: Defining response of ovarian carcinoma to initial chemotherapy according to serum CA 125. J. Clin. Oncol. 14:1545-1551, 1996

  • 31. Irizarry R A, Hobbs B, Collin F, et al: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4:249-263, 2003

  • 32. Bolstad B M, Irizarry R A, Astrand M, et al: A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19: 185-193, 2003

  • 33. Lucus J, Carvalho C, Wang Q, et al: Sparse statistical modeling in gene expression genomics. Cambridge, Cambridge University Press, 2006

  • 34. Rich J, Jones B, Hans C, et al: Gene expression profiling and genetic markers in glioblastoma survival. Cancer Res. 65:4051-4058, 2005

  • 35. Hans C, Dobra A, West M: Shotgun stochastic search for regression with many candidate predictors. JASA in press. 2006

  • 36. Bild A, Yao G, Chang J T, et al: Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature 439:353-357, 2006.

  • 37. Gyorrfy B, Surowiak P, Kiesslich O, Denkert C, Schafer R, Dietel M, Lage H: Gene expression profiling of 30 cancer cell lines predicts resistance towards 11 anticancer drugs at clinically achieved concentrations. Int. J Cancer 118(7): 1699-712, 2006

  • 38. Minna, J D, Gazdar, A F, Sprang, S R & Herz, J: Cancer. A bull's eye for targeted lung cancer therapy. Science 304: 1458-1461, 2004

  • 39. Jemal et al., CA Cancer J Clin., 53, 5-26, 2003

  • 40. Cancer Facts and Figures: American Cancer Society, Atlanta, p. 11, 2002

  • 41. Travis et al., Lung Cancer Principles and Practice, Lippincott-Raven, New York, pps. 361-395, 1996

  • 42. Gazdar et al., Anticancer Res. 14:261-267

  • 43. Niklinska et al., Folia Histochem. Cytobiol. 39:147-148, 2001

  • 44. Parker et al, CA Cancer J Clin. 47:5-27, 1997

  • 45. Chu et al, J Nat. Cancer Inst. 88:1571-1579, 1996

  • 46. Baker, V V: Salvage therapy for recurrent epithelial ovarian cancer. Hematol. 011COI Clin. N. Am, 17: 977-988, 2003

  • 47. Hansen, H H, Eisenhauer, E A, Hasen M, Neijt J P, Piccart M J, Sessa C, Thigpen J T: New cytostatis drugs in ovarian cancer. Ann. Oncol. 4:S63-S70, 1993

  • 48. Herrin, V E, Thigpen J T: Chemotherapy for ovarian cancer: current concepts. Semin. Surg. On col. 17: 181-188, 1999

  • 49. Staunton, J. E. et al. Chemosensitivity prediction by transcriptional profiling. Proc Natl Acad Sci USA 98: 10787-19792, 2001

  • 50. Chang, J. c. et al. Gene expression profiling for the prediction of therapeutic response to docetaxel in patients with breast cancer. Lancet 362:362-369, 2003

  • 51. Emi, M., Kim, R., Tanabe, K., Uchida, Y. & toge, T. Targeted therapy against Bcl-2-related proteins in breast cancer cells. Breast Cancer Res 7: R940-R952, 2005

  • 52. Takahashi, T. et al. Cyclin A-associated kinase activity is needed for paclitaxel sensitivity. Mol Cancer Ther 4: 1039-1046, 2005

  • 53. Modi, S. et al. Phosphorylated/activated HER2 as a marker of clinical resistance to single agent taxane chemotherapy for metastatic breast cancer. Cancer Invest 23: 483-487, 2005

  • 54. Langer, R. et al. Association of pre therapeutic expression of chemotherapy-related genes with response to neoadjuvant chemotherapy in Barrett carcinoma. Clin Cancer Res. 11: 7462-7469, 2005

  • 55. Rouzier, R. et al. Breast cancer molecular subtypes respond differently to preoperative chemotherapy. Clin Cancer Res. 11: 5678-5685, 2005

  • 56. Rouzier, R. et al. Microbubule-associated protein tau: a marker of paclitaxel sensitivity on breast cancer. Proc Natl Acad Sci USA 102: 8315-8320, 2005

  • 57. DeVita, V. T., Hellman, S. & Rosenberg, S. A. Cancer: Principles and Practice of Oncology, Lippincott-Raven, Philadelphia, 2005

  • 58. Herbst, R. S. et at Clinical Cancer Advances 2005; Major research advances in cancer treatment, prevention, and screening—a report from the American Society of Clinical Oncology. J. Clin. Oncol. 24: 190-205, 2006

  • 59. Broxterman, H. J. & Georgopapadakou, N. H. Anticancer therapeutics: Addictive targets, multi-targeted drugs, new drug combinations. Drug Resist Update 8: 183-197, 2005

  • 60. Pittman, J. Huang, E., Wang, Q., Nevins, J R. & West, M. Bayesian analysis of binary prediction tree models for retrospectively sampled outcomes. Biostatistics 5: 587-601, 2004

  • 61. West, M. et at Predicting the clinical status of human breast cancer by using gene expression profiles. Proc Natl Acad Sci USA 98: 11462-11467, 2001

  • 62. Ihaka, R. & Gentleman, R. A language for data analysis and graphics. J. Comput. Graph. Stat. 5: 299-314, 1996

  • 63. Eisen, M. B., Spellman, P. T., Brown, P. O. & Botstein, D. Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci USA 95:14863-14868, 1998










TABLE 1







5-Flourouracil responsivity predictor set











5FUProbe



Entrez


Set ID web
Weight
Gene Symbol
Go biological process term
Gene ID














151_s_at
−3.83685
LOC92755 ///
fructose metabolic process ///
203068 ///




TUBB
glycolysis /// cell motility ///
92755





microtubule-based process ///





microtubule-based movement ///





metabolic process /// natural killer





cell mediated cytotoxicity /// protein





polymerization


1713_s_at
4.712802
CDKN2A
cell cycle checkpoint /// G1/S
1029





transition of mitotic cell cycle ///





negative regulation of cell-matrix





adhesion /// DNA fragmentation





during apoptosis /// transcription ///





regulation of transcription, DNA-





dependent /// rRNA processing ///





negative regulation of protein kinase





activity /// apoptosis /// induction of





apoptosis /// induction of apoptosis





/// caspase activation /// cell cycle ///





cell cycle arrest /// negative





regulation of cell proliferation ///





apoptotic mitochondrial changes ///





senescence /// regulation of G2/M





transition of mitotic cell cycle ///





negative regulation of cell growth ///





negative regulation of B cell





proliferation /// regulation of protein





stability /// negative regulation of NF-





kappaB transcription factor activity





/// negative regulation of immature T





cell proliferation in the thymus ///





negative regulation of





phosphorylation /// negative





regulation of cyclin-dependent





protein kinase activity /// negative





regulation of cell cycle /// somatic





stem cell division /// negative





regulation of ubiquitin-protein ligase





activity


1882_g_at
0.861954





31322_at
1.401
TRA@
immune response /// cellular
6955





defense response


31726_at

GABRA3
transport /// transport /// ion transport
2556





/// chloride transport /// gamma-





aminobutyric acid signaling pathway





/// gamma-aminobutyric acid





signaling pathway


32308_r_at
−1.10479
COL1A2
skeletal development /// phosphate
1278





transport /// transmembrane





receptor protein tyrosine kinase





signaling pathway /// sensory





perception of sound


32318_s_at
−1.23171
ACTB
transport /// amino acid transport ///
60





cell motility /// sensory perception of





sound /// arginine transport /// lysine





transport /// response to calcium ion


32610_at
2.301947
PDLIM4

8572


32755_at
0.912152
ACTA2

59


33437_at
−0.66656
FTSJ1
rRNA processing
24140


33444_at

NBR1 ///

100133166




LOC100133166

/// 4077


33659_at
0.622566
CFL1
anti-apoptosis /// Rho protein signal
1072





transduction /// actin cytoskeleton





organization and biogenesis


34377_at
1.171995
ATP1A2
transport /// ion transport /// cation
477





transport /// potassium ion transport





/// potassium ion transport /// sodium





ion transport /// sodium ion transport





/// regulation of striated muscle





contraction /// metabolic process ///





monovalent inorganic cation





transport /// proton transport ///





sperm motility /// regulation of





cellular pH


34454_r_at
−0.8324
APOC2 ///
lipid metabolic process /// lipid
344 /// 346




APOC4
metabolic process /// triacylglycerol





metabolic process /// phospholipid





metabolic process /// transport ///





lipid transport /// lipid catabolic





process /// cholesterol efflux ///





phospholipid efflux /// positive





regulation of lipoprotein lipase





activity


34545_at

CCDC88C
regulation of protein amino acid
440193





phosphorylation /// Wnt receptor





signaling pathway /// protein





destabilization /// protein





homooligomerization


34843_at
−2.25281





34905_at
1.045288
GRIK5
transport /// ion transport /// synaptic
2901





transmission


34954_r_at
1.084054
PDE5A
signal transduction /// signal
8654





transduction /// cyclic nucleotide





metabolic process


35056_at
−2.52505
ARSF
metabolic process
416


35144_at
0.689025
ZC3H7B

23264


35213_at
0.693573
WBP4
mRNA processing /// RNA splicing
11193


35816_at
−1.29531
CSTB
adult locomotory behavior
1476


35929_s_at
1.027644
TSPY1 ///
nucleosome assembly ///
64591 ///




TSPY2 ///
multicellular organismal
7258 ///




LOC728137 ///
development /// spermatogenesis ///
728137 ///




LOC728395 ///
spermatogenesis /// gonadal
728395 ///




LOC728412
mesoderm development /// sex
728412





differentiation /// cell proliferation ///





cell differentiation


36245_at
−1.6361
HTR2B
signal transduction /// G-protein
3357





coupled receptor protein signaling





pathway /// G-protein signaling,





coupled to IP3 second messenger





(phospholipase C activating) ///





heart development /// blood





circulation /// positive regulation of I-





kappaB kinase/NF-kappaB cascade


36453_at
1.22492
KBTBD11

9920


36549_at
−0.92503
SLC25A17
transport /// transport ///
10478





mitochondrial transport


37349_r_at
1.984623
HMGN3

9324


37361_at
−1.79231
FIBP
fibroblast growth factor receptor
9158





signaling pathway


37437_at
−0.9449
IFT140
cell communication
9742


37802_r_at
2.334769
FAM63B

54629


37860_at
1.40471
ZNF337
transcription /// regulation of
26152





transcription, DNA-dependent


39783_at
0.802025
KIAA0100
tricarboxylic acid cycle
9703


39898_at
−0.9176
FAM13C1

220965


40104_at
−0.70116
STK25
protein amino acid phosphorylation
10494





/// response to oxidative stress ///





signal transduction


40452_at
−1.26932
CPNE1
lipid metabolic process /// vesicle-
8904





mediated transport


40471_at

PEX19
protein targeting to peroxisome ///
5824





peroxisome organization and





biogenesis /// peroxisome





organization and biogenesis


40536_f_at

EIF5B
translation /// regulation of
9669





translational initiation /// regulation of





translational initiation


40886_at
−1.81682
EEF1A1 ///
angiogenesis /// translation ///
100132804




EEF1AL3 ///
translational elongation ///
/// 158078




LOC100132804
translational elongation ///
/// 1915





translational elongation /// lipid





transport /// multicellular organismal





development /// cell differentiation ///





lipoprotein metabolic process


40983_s_at
1.828091
SRR
amino acid metabolic process ///
63826





amino acid metabolic process /// L-





serine metabolic process ///





metabolic process /// serine family





amino acid metabolic process


41058_g_at
0.545125
THEM2

55856


41536_at
0.453047
ID4
regulation of transcription from RNA
3400





polymerase II promoter ///





neuroblast proliferation /// positive





regulation of cell proliferation ///





negative regulation of transcription





/// regulation of transcription ///





negative regulation of neuron





differentiation /// negative regulation





of astrocyte differentiation


41868_at
−1.46067
GGT1 ///
amino acid metabolic process ///
2678 ///




GGTLC2
glutathione biosynthetic process ///
91227





glutathione biosynthetic process


427_f_at
−0.72026
IFNA10
defense response /// cell-cell
3446





signaling /// response to virus ///





response to virus


429_f_at
0.512718
TUBB4
microtubule-based process ///
10382





microtubule-based movement ///





mitosis /// neuron differentiation ///





protein polymerization


471_f_at
−0.74815
TUBB3
microtubule-based process ///
10381





microtubule-based movement ///





mitosis /// signal transduction /// G-





protein coupled receptor protein





signaling pathway /// G-protein





signaling, coupled to cyclic





nucleotide second messenger ///





multicellular organismal





development /// UV protection ///





neuron differentiation /// protein





polymerization
















TABLE 2







Adriamycin responsivity predictor set











Web site






Adria


Probe Set

Gene

Entrez


ID
Weight
Symbol
Go biological process term
Gene ID














1051_g_at
−0.94348
MLANA

2315


110_at
1.234027
CSPG4
angiogenesis /// cell motility /// signal
1464





transduction /// multicellular organismal





development /// cell differentiation ///





tissue remodeling


1319_at
0.677949
DDR2
protein amino acid phosphorylation /// cell
4921





adhesion /// cell adhesion /// signal





transduction /// transmembrane receptor





protein tyrosine kinase signaling pathway





/// positive regulation of cell proliferation


1519_at
−1.85295
ETS2
skeletal development /// regulation of
2114





transcription, DNA-dependent


1537_at
2.591759
EGFR
ossification /// protein amino acid
1956





phosphorylation /// response to stress ///





cell cycle /// signal transduction /// cell





surface receptor linked signal





transduction /// transmembrane receptor





protein tyrosine kinase signaling pathway





/// epidermal growth factor receptor





signaling pathway /// epidermal growth





factor receptor signaling pathway ///





activation of phospholipase C activity ///





cell proliferation /// positive regulation of





cell proliferation /// cell-cell adhesion ///





positive regulation of cell migration ///





positive regulation of phosphorylation ///





calcium-dependent phospholipase A2





activation /// positive regulation of MAP





kinase activity /// positive regulation of





nitric oxide biosynthetic process ///





negative regulation of cell cycle /// positive





regulation of epithelial cell proliferation ///





regulation of peptidyl-tyrosine





phosphorylation /// regulation of nitric-





oxide synthase activity /// protein insertion





into membrane


2011_s_at
−2.46428
BIK
apoptosis /// induction of apoptosis ///
638





induction of apoptosis /// apoptotic





program /// regulation of apoptosis


266_s_at
1.920993
CD24
response to hypoxia /// cell activation ///
934





regulation of cytokine and chemokine





mediated signaling pathway /// regulation





of cytokine and chemokine mediated





signaling pathway /// response to





molecule of bacterial origin /// response to





molecule of bacterial origin /// immune





response-regulating cell surface receptor





signaling pathway /// elevation of cytosolic





calcium ion concentration ///





neuromuscular synaptic transmission ///





induction of apoptosis by intracellular





signals /// Wnt receptor signaling pathway





/// cell-cell adhesion /// cell migration ///





cell migration /// regulation of epithelial





cell differentiation /// T cell costimulation





/// B cell receptor transport into membrane





raft /// chemokine receptor transport out of





membrane raft /// negative regulation of





transforming growth factor-beta3





production /// positive regulation of





activated T cell proliferation /// regulation





of phosphorylation /// cholesterol





homeostasis /// cholesterol homeostasis





/// positive regulation of MAP kinase





activity /// regulation of MAPKKK cascade





/// response to estrogen stimulus ///





respiratory burst /// synaptic vesicle





endocytosis


32139_at
2.120382
ZNF185

7739


32168_s_at
−0.56607
RCAN1
signal transduction /// central nervous
1827





system development /// blood circulation





/// calcium-mediated signaling


32612_at
−1.93636
GSN
actin filament polymerization /// actin
2934





filament polymerization /// actin filament





severing /// actin filament severing ///





barbed-end actin filament capping ///





barbed-end actin filament capping


32718_at
−1.07596
TPST1
peptidyl-tyrosine sulfation /// inflammatory
8460





response


32821_at
1.172906
LCN2
transport
3934


32967_at
2.725153
FAIM3
anti-apoptosis /// immune response ///
9214





cellular defense response


33004_g_at
1.165497
NCK2
regulation of translation /// signal
8440





transduction /// signal complex assembly





/// epidermal growth factor receptor





signaling pathway /// regulation of





epidermal growth factor receptor activity





/// negative regulation of cell proliferation





/// positive regulation of actin filament





polymerization /// positive regulation of T





cell proliferation /// T cell activation


33240_at
−2.07044
PDZRN3

23024


33409_at
−0.84774
FKBP2
protein folding
2286


33824_at
0.8914
KRT8
cytoskeleton organization and biogenesis
3856





/// response to other organism


33853_s_at
2.187597
NRP2
angiogenesis /// cell adhesion /// cell
8828





adhesion /// multicellular organismal





development /// nervous system





development /// axon guidance /// cell





differentiation /// cell redox homeostasis


33892_at
−1.14625
PKP2
cell adhesion /// cell-cell adhesion
5318


33904_at
−1.29549
CLDN3
response to hypoxia /// calcium-
1365





independent cell-cell adhesion /// calcium-





independent cell-cell adhesion


33908_at
−1.35671
CAPN1
proteolysis /// positive regulation of cell
823





proliferation


33942_s_at
−1.20596
STXBP1
transport /// vesicle docking during
6812





exocytosis /// protein transport /// vesicle-





mediated transport


33956_at
1.164645
LY96
inflammatory response /// immune
23643





response /// cellular defense response ///





cell surface receptor linked signal





transduction


34213_at
−1.32674
WWC1

23286


34303_at
1.15829
C10orf56

219654


34348_at
−0.97728
SPINT2 ///
cell motility
100130414




LOC100130414

///






10653


34859_at
1.376672
MAGED2
translation
10916


34885_at
1.474456
SYNGR2

9144


34993_at
3.241691
SGCD
cytoskeleton organization and biogenesis
6444





/// muscle development


35280_at
−0.95845
LAMC2
cell adhesion /// epidermis development
3918


35444_at
−1.24187
C19orf21

126353


35681_r_at
2.082145
ZEB2
transcription /// regulation of transcription,
9839





DNA-dependent /// nervous system





development /// negative regulation of





transcription /// regulation of transcription


35766_at
1.257264
KRT18
cell cycle /// anatomical structure
3875





morphogenesis /// Golgi to plasma





membrane CFTR protein transport ///





negative regulation of apoptosis


35807_at
−1.93358
CYBA
superoxide metabolic process /// transport
1535





/// oxidation reduction


36133_at
−0.92039
DSP
epidermis development /// peptide cross-
1832





linking /// keratinocyte differentiation


36618_g_at
0.683166
ID1
regulation of transcription from RNA
3397





polymerase II promoter /// multicellular





organismal development /// negative





regulation of transcription /// negative





regulation of transcription factor activity ///





regulation of transcription


36619_r_at
2.621706
ID1
regulation of transcription from RNA
3397





polymerase II promoter /// multicellular





organismal development /// negative





regulation of transcription /// negative





regulation of transcription factor activity ///





regulation of transcription


36795_at
−0.62444
PSAP
lipid metabolic process /// sphingolipid
5660





metabolic process /// glycosphingolipid





metabolic process /// lipid transport ///





lysosome organization and biogenesis


36828_at
−0.58068
ZNF629
transcription /// regulation of transcription,
23361





DNA-dependent


36849_at
0.612692
ARHGAP29
signal transduction /// intracellular
9411





signaling cascade /// Rho protein signal





transduction


37117_at
−0.53853
ARHGAP8 ///
cell cycle /// signal transduction /// actin
23779 ///




PRR5 ///
cytoskeleton organization and biogenesis
553158




LOC553158
/// positive regulation of cell migration ///
/// 55615





negative regulation of cell cycle


37251_s_at

GPM6B
multicellular organismal development ///
2824





nervous system development /// nervous





system development /// cell differentiation


37327_at
1.236668
EGFR
ossification /// protein amino acid
1956





phosphorylation /// response to stress ///





cell cycle /// signal transduction /// cell





surface receptor linked signal





transduction /// transmembrane receptor





protein tyrosine kinase signaling pathway





/// epidermal growth factor receptor





signaling pathway /// epidermal growth





factor receptor signaling pathway ///





activation of phospholipase C activity ///





cell proliferation /// positive regulation of





cell proliferation /// cell-cell adhesion ///





positive regulation of cell migration ///





positive regulation of phosphorylation ///





calcium-dependent phospholipase A2





activation /// positive regulation of MAP





kinase activity /// positive regulation of





nitric oxide biosynthetic process ///





negative regulation of cell cycle /// positive





regulation of epithelial cell proliferation ///





regulation of peptidyl-tyrosine





phosphorylation /// regulation of nitric-





oxide synthase activity /// protein insertion





into membrane


37345_at
−1.49834
CALU

813


37552_at
1.600714
KCNK1
transport /// ion transport /// potassium ion
3775





transport /// potassium ion transport


37695_at
1.263311
RNF144A
ubiquitin cycle
9781


37743_at
−2.36633
FEZ1
cell adhesion /// nervous system
9638





development /// axon guidance


37749_at
0.830285
MEST
mesoderm development
4232


37926_at
1.591841
KLF5
angiogenesis /// transcription /// regulation
688





of transcription, DNA-dependent ///





transcription from RNA polymerase II





promoter /// positive regulation of cell





proliferation /// microvillus biogenesis ///





positive regulation of transcription


38004_at
−0.6707
CSPG4
angiogenesis /// cell motility /// signal
1464





transduction /// multicellular organismal





development /// cell differentiation ///





tissue remodeling


38078_at
−1.44495
FLNB
cytoskeletal anchoring /// signal
2317





transduction /// multicellular organismal





development /// skeletal muscle





development /// actin cytoskeleton





organization and biogenesis /// cell





differentiation


38119_at
−3.15147
GYPC
protein amino acid N-linked glycosylation
2995





/// protein amino acid O-linked





glycosylation /// organ morphogenesis


38122_at
1.601555
SLC23A2
nucleobase, nucleoside, nucleotide and
9962





nucleic acid metabolic process ///





transport /// ion transport /// sodium ion





transport /// nucleobase transport ///





molecular hydrogen transport /// L-





ascorbic acid metabolic process


38227_at

MITF
transcription /// regulation of transcription,
4286





DNA-dependent /// regulation of





transcription, DNA-dependent ///





multicellular organismal development ///





sensory perception of sound ///





melanocyte differentiation /// regulation of





transcription


38297_at
0.897307
PITPNM1
lipid metabolic process /// transport ///
9600





brain development /// phototransduction ///





protein transport


38379_at
−1.22485
GPNMB
negative regulation of cell proliferation
10457


38653_at
−1.26954
PMP22
synaptic transmission /// peripheral
5376





nervous system development /// sensory





perception of sound /// mechanosensory





behavior /// negative regulation of cell





proliferation


39214_at
−2.83871
PLXNB3
protein amino acid phosphorylation ///
5365





signal transduction /// multicellular





organismal development


39271_at
−2.40695
MIA
cell-matrix adhesion /// cell proliferation ///
8190





extracellular matrix organization and





biogenesis


39316_at
1.090483
RAB40C
ubiquitin cycle /// intracellular signaling
57799





cascade /// small GTPase mediated signal





transduction /// protein transport


39386_at
−2.27665
MAD2L1BP
regulation of exit from mitosis
9587


39801_at
−1.2452
PLOD3
protein modification process /// protein
8985





metabolic process


40103_at
1.069164
EZR
cytoskeletal anchoring /// regulation of cell
7430





shape /// actin filament bundle formation


40202_at
1.070163
KLF9
transcription /// regulation of transcription,
687





DNA-dependent /// regulation of





transcription from RNA polymerase II





promoter /// embryo implantation ///





regulation of transcription /// progesterone





receptor signaling pathway


40434_at
1.126169
PODXL
negative regulation of cell adhesion ///
5420





leukocyte migration


40568_at
−0.48906
ATP6V1B2
ATP biosynthetic process /// transport ///
526





ion transport /// ATP synthesis coupled





proton transport /// energy coupled proton





transport, against electrochemical





gradient /// proton transport /// proton





transport


40926_at
−1.2209
SLC6A8
neurotransmitter uptake /// transport ///
6535





transport /// ion transport /// sodium ion





transport /// neurotransmitter transport ///





muscle contraction


41158_at
1.088079
PLP1
synaptic transmission /// axon
5354





ensheathment


41294_at
−1.29363
KRT7
DNA replication /// regulation of
3855





translation /// cytoskeleton organization





and biogenesis /// interphase


41359_at
1.004171
PKP3
cell adhesion
11187


41378_at
−2.53685





41453_at
1.276055
DLG3
negative regulation of cell proliferation
1741


41503_at
0.635937
ZHX2
transcription /// regulation of transcription,
22882





DNA-dependent /// mRNA catabolic





process /// regulation of transcription ///





negative regulation of transcription, DNA-





dependent


41610_at
−2.14951
LAMA5
angiogenesis /// cytoskeleton organization
3911





and biogenesis /// cell adhesion ///





integrin-mediated signaling pathway ///





cell recognition /// cell proliferation ///





embryonic development /// cell migration





/// cell differentiation /// regulation of cell





adhesion /// regulation of cell migration ///





endothelial cell differentiation /// regulation





of embryonic development /// focal





adhesion formation


41644_at
−1.27145
SASH1
cell cycle /// negative regulation of cell
23328





cycle


41839_at
−1.30948
GAS1
cell cycle /// cell cycle arrest /// cell cycle
2619





arrest /// negative regulation of cell





proliferation /// programmed cell death ///





cell fate commitment /// negative





regulation of S phase of mitotic cell cycle





/// eye morphogenesis /// negative





regulation of epithelial cell proliferation


575_s_at
0.929663
TACSTD1

4072


661_at
2.176958
GAS1
cell cycle /// cell cycle arrest /// cell cycle
2619





arrest /// negative regulation of cell





proliferation /// programmed cell death ///





cell fate commitment /// negative





regulation of S phase of mitotic cell cycle





/// eye morphogenesis /// negative





regulation of epithelial cell proliferation


953_g_at
−0.89283





999_at
1.075154
CYP27A1
oxidation reduction
1593
















TABLE 3







Cytotaxan responsivity predictor set











Cytoxan






Probe Set



Entrez


ID
Weight
Gene Symbol
Go biological process term
Gene ID














1356_at
4.874357
DAP3
apoptosis /// induction of apoptosis by
7818





extracellular signals


31511_at
3.788663
RPS9
translation /// translation
6203


32252_at
5.394493
TTR
thyroid hormone generation /// transport ///
7276





transport


32318_s_at
3.101326
ACTB
transport /// amino acid transport /// cell
60





motility /// sensory perception of sound ///





arginine transport /// lysine transport ///





response to calcium ion


32434_at
−2.569863
MARCKS
cell motility
4082


32893_s_at
1.254223
GGT1 ///
amino acid metabolic process ///
2678 ///




GGT3P ///
glutathione biosynthetic process ///
2679 ///




GGTLC2 ///
glutathione biosynthetic process
650860 ///




GGTLC1 ///

728226 ///




LOC650860 ///

728441 ///




GGTLC3 ///

91227 ///




GGT2

92086


33145_at
−1.175404
FANCA
DNA repair /// DNA repair /// protein
2175





complex assembly /// response to DNA





damage stimulus


33362_at
2.071209
CDC42EP3
signal transduction /// regulation of cell
10602





shape


33919_at
2.705049
TSPAN4
protein complex assembly
7106


34246_at
−2.812789
C6orf145
cell communication
221749


35352_at
4.019153
ARNT2
response to hypoxia /// in utero embryonic
9915





development /// in utero embryonic





development /// transcription /// regulation





of transcription, DNA-dependent /// signal





transduction /// central nervous system





development /// central nervous system





development /// regulation of transcription





/// regulation of transcription /// positive





regulation of transcription /// positive





regulation of transcription /// positive





regulation of transcription from RNA





polymerase II promoter


356_at
1.879373
KIF22
microtubule-based movement /// mitosis
3835


35763_at
−0.870241
NBEAL2

23218


36119_at
2.689896
CAV1
inactivation of MAPK activity ///
857





vasculogenesis /// response to hypoxia ///





negative regulation of endothelial cell





proliferation /// triacylglycerol metabolic





process /// calcium ion transport /// cellular





calcium ion homeostasis /// endocytosis ///





regulation of smooth muscle contraction ///





skeletal muscle development /// protein





localization /// vesicle organization and





biogenesis /// regulation of fatty acid





metabolic process /// sequestering of lipid





/// regulation of blood coagulation ///





cholesterol transport /// negative regulation





of epithelial cell differentiation ///





mammary gland development /// nitric





oxide homeostasis /// cholesterol





homeostasis /// cholesterol homeostasis ///





negative regulation of MAPKKK cascade





/// negative regulation of nitric oxide





biosynthetic process /// positive regulation





of vasoconstriction /// negative regulation





of vasodilation /// negative regulation of





JAK-STAT cascade /// positive regulation





of metalloenzyme activity /// protein





homooligomerization /// membrane





depolarization /// regulation of peptidase





activity /// calcium ion homeostasis ///





mammary gland involution


36192_at
1.514496
SCRN1
proteolysis /// exocytosis /// exocytosis
9805


36536_at
4.000312
SCHIP1

29970


37375_at
3.575144
PHLDB1

23187


37680_at
−1.228293
AKAP12
protein targeting /// signal transduction ///
9590





G-protein coupled receptor protein





signaling pathway


37745_s_at
3.741699
ST5

6764


38288_at
4.706992
SNAI2
negative regulation of transcription from
6591





RNA polymerase II promoter ///





transcription /// regulation of transcription,





DNA-dependent /// multicellular





organismal development /// ectoderm and





mesoderm interaction /// sensory





perception of sound /// response to





radiation


38375_at
0.969629
ESD
release of cytochrome c from mitochondria
2098





/// apoptosis /// induction of apoptosis via





death domain receptors /// apoptotic





mitochondrial changes /// regulation of





apoptosis /// positive regulation of





apoptosis /// neuron apoptosis


38479_at
1.720282
ANP32B

10541


39170_at
3.438103
CD59
defense response /// immune response ///
966





cell surface receptor linked signal





transduction /// blood coagulation


39329_at
2.366823
ACTN1
regulation of apoptosis /// focal adhesion
87





formation /// actin filament bundle





formation /// negative regulation of cell





motility


39351_at
−1.350962
CD59
defense response /// immune response ///
966





cell surface receptor linked signal





transduction /// blood coagulation


39696_at
5.888778
PEG10
proteolysis /// apoptosis /// cell
23089





differentiation /// transposition


39750_at
−2.622822





40213_at
3.382652
SMARCA1
chromatin remodeling /// transcription ///
6594





regulation of transcription, DNA-





dependent /// brain development ///





chromatin modification /// neuron





differentiation /// ATP-dependent





chromatin remodeling /// positive





regulation of gene-specific transcription


40394_at
3.379691
GGCX
protein modification process /// blood
2677





coagulation /// peptidyl-glutamic acid





carboxylation


40855_at
−0.689293
SAMD4A
positive regulation of translation
23034


40953_at
2.35874
CNN3
smooth muscle contraction /// muscle
1266





development /// actomyosin structure





organization and biogenesis


41195_at
−1.931524
LPP
cell adhesion
4026


41403_at
1.485089
SNRPF
mRNA processing /// RNA splicing /// RNA
6636





splicing /// mRNA metabolic process


41449_at
2.035738
SGCE
cell-matrix adhesion /// muscle
8910





development


41739_s_at
1.395914
CALD1
cell motility /// muscle contraction
800


41758_at
2.920572
TMEM184B

25829
















TABLE 4







Docetaxol responsivity predictor set











Doce






Probe Set



Entrez


ID
Weight
Gene Symbol
Go biological process term
Gene ID














1003_s_at
−1.586523
CXCR5
cell motility /// signal transduction ///
643





G-protein coupled receptor protein





signaling pathway /// G-protein





coupled receptor protein signaling





pathway /// B cell activation /// lymph





node development


1420_s_at
−1.306835
EIF4A2
translation /// regulation of
1974





translational initiation


1567_at
−1.007947
FLT1
angiogenesis /// patterning of blood
2321





vessels /// protein amino acid





phosphorylation /// transmembrane





receptor protein tyrosine kinase





signaling pathway ///





transmembrane receptor protein





tyrosine kinase signaling pathway ///





multicellular organismal





development /// female pregnancy ///





positive regulation of cell





proliferation /// cell migration /// cell





differentiation /// vascular endothelial





growth factor receptor signaling





pathway


1861_at
−1.210512
BAD
apoptosis /// induction of apoptosis
572





/// apoptotic program


32085_at
1.984601
PIP5K3
intracellular signaling cascade ///
200576





intracellular signaling cascade ///





calcium-mediated signaling ///





cellular protein metabolic process ///





phosphatidylinositol metabolic





process


32218_at
−1.090595





32238_at
1.432857
BIN1
ATP biosynthetic process ///
274





transport /// ion transport ///





endocytosis /// cell cycle ///





multicellular organismal





development /// cell proliferation ///





ATP synthesis coupled proton





transport /// proton transport ///





regulation of endocytosis /// cell





differentiation /// negative regulation





of cell cycle


32340_s_at
−1.66312
YBX1
transcription /// regulation of
4904





transcription, DNA-dependent ///





regulation of transcription, DNA-





dependent /// transcription from RNA





polymerase II promoter /// mRNA





processing /// RNA splicing


32828_at
−2.216035
BCKDK
signal transduction /// amino acid
10295





catabolic process /// branched chain





family amino acid catabolic process





/// branched chain family amino acid





catabolic process /// phosphorylation





/// phosphorylation ///





phosphorylation /// peptidyl-histidine





phosphorylation


33176_at
−1.302196
DOHH
peptidyl-lysine modification to
83475





hypusine /// peptidyl-lysine





modification to hypusine


33204_at
−0.607135
FOXD1
transcription /// regulation of
2297





transcription, DNA-dependent


33388_at
2.300796
TEX261

113419


33444_at
−2.738947
NBR1 ///

100133166




LOC100133166

/// 4077


34523_at
−2.692606
APOA4
innate immune response in mucosa
337





/// transport /// lipid transport /// lipid





transport /// response to lipid





hydroperoxide /// leukocyte adhesion





/// cholesterol metabolic process ///





removal of superoxide radicals ///





regulation of cholesterol transport ///





cholesterol efflux /// phospholipid





efflux /// lipoprotein metabolic





process /// lipoprotein modification ///





cholesterol homeostasis /// hydrogen





peroxide catabolic process ///





reverse cholesterol transport ///





multicellular organismal lipid





catabolic process ///





phosphatidylcholine metabolic





process /// lipid homeostasis ///





protein-lipid complex assembly


34647_at
3.145899
DDX5
mRNA processing /// RNA splicing ///
1655





cell growth


34773_at
1.963235
TBCA
tubulin folding /// post-chaperonin
6902





tubulin folding pathway /// beta-





tubulin folding


34801_at
−1.144803
PAN2
nuclear-transcribed mRNA catabolic
9924





process, nonsense-mediated decay





/// ubiquitin-dependent protein





catabolic process


34804_at
−0.852211
SLC25A36
transport /// mitochondrial transport
55186


35018_at
−1.802839
CHP
potassium ion transport /// small
11261





GTPase mediated signal





transduction


35655_at
−1.746975
ANKRD28

23243


35714_at
−2.095963
PDXK
pyridoxine biosynthetic process
8566


35770_at
3.159298
ATP6AP1
protein import into nucleus,
537





translocation /// ATP biosynthetic





process /// transport /// ion transport





/// response to stress /// fibroblast





growth factor receptor signaling





pathway /// ATP synthesis coupled





proton transport /// proton transport





/// proton transport /// regulation of





transcription /// positive regulation of





receptor-mediated endocytosis ///





positive regulation of urothelial cell





proliferation


35815_at
−2.192203
SETD2
transcription /// regulation of
29072





transcription, DNA-dependent ///





chromatin modification


36068_at
−1.112913
CCS
superoxide metabolic process ///
9973





superoxide metabolic process ///





intracellular copper ion transport ///





metal ion transport /// positive





regulation of oxidoreductase activity


36209_at
−2.145239
BRD2
spermatogenesis
6046


36250_at
−1.191006
ASPHD1
peptidyl-amino acid modification
253982


36366_at
1.997465
B4GALT6
carbohydrate metabolic process
9331


36395_at
2.355726





36528_at
0.743151
ASL
urea cycle /// argininosuccinate
435





metabolic process /// response to





hypoxia /// kidney development ///





liver development /// arginine





biosynthetic process /// arginine





catabolic process /// response to





nutrient /// amino acid biosynthetic





process /// arginine biosynthetic





process via ornithine /// response to





peptide hormone stimulus ///





response to glucocorticoid stimulus





/// response to cAMP


36641_at
−0.973701
CAPZA2
protein complex assembly /// cell
830





motility /// actin cytoskeleton





organization and biogenesis ///





barbed-end actin filament capping


37355_at
−3.431367
STARD3
lipid metabolic process /// steroid
10948





biosynthetic process /// C21-steroid





hormone biosynthetic process ///





transport /// mitochondrial transport





/// lipid transport /// steroid metabolic





process /// cholesterol metabolic





process


38618_at
0.761043
LIMK2 ///
protein amino acid phosphorylation
3985 ///




PPP1R14BP1
/// phosphorylation /// regulation of
50516





phosphorylation


38663_at
−0.841092
BANF1
response to virus
8815


38831_f_at
−1.128399
GNB2
signal transduction /// G-protein
2783





coupled receptor protein signaling





pathway


39012_g_at
−1.366345
ENSA
transport /// response to nutrient
2029


39159_at
−0.871773
SH3GL1
endocytosis /// signal transduction ///
6455





central nervous system development


39199_at
−1.58861
ACVR1B
G1/S transition of mitotic cell cycle
91





/// in utero embryonic development





/// hair follicle development /// protein





amino acid phosphorylation ///





protein amino acid phosphorylation





/// induction of apoptosis /// signal





transduction /// transmembrane





receptor protein serine/threonine





kinase signaling pathway ///





embryonic development /// negative





regulation of cell growth /// positive





regulation of activin receptor





signaling pathway /// regulation of





transcription /// positive regulation of





erythrocyte differentiation


39599_at
1.466566
SLC6A1
transport /// transport ///
6529





neurotransmitter transport ///





synaptic transmission


40867_at
2.484647
PPP2R1A
inactivation of MAPK activity ///
5518





regulation of DNA replication ///





protein complex assembly /// protein





amino acid dephosphorylation ///





ceramide metabolic process ///





induction of apoptosis /// RNA





splicing /// response to organic





substance /// second-messenger-





mediated signaling /// regulation of





Wnt receptor signaling pathway ///





regulation of cell adhesion ///





negative regulation of cell growth ///





regulation of growth /// negative





regulation of tyrosine





phosphorylation of Stat3 protein ///





regulation of transcription ///





regulation of cell differentiation


41063_g_at
1.696948
PCGF1
transcription /// regulation of
84759





transcription, DNA-dependent


41077_at
2.21384
LOC643641
transcription /// regulation of
643641





transcription, DNA-dependent


41285_at
0.839605
INPP5A
cell communication
3632


41489_at
−1.356162
TLE1
transcription /// regulation of
7088





transcription, DNA-dependent ///





signal transduction /// multicellular





organismal development /// organ





morphogenesis /// Wnt receptor





signaling pathway /// negative





regulation of transcription ///





negative regulation of Wnt receptor





signaling pathway /// regulation of





transcription


41689_at
1.121172
PLLP
transport /// ion transport
51090


41713_at
−1.680428
ZKSCAN1
transcription /// regulation of
7586





transcription, DNA-dependent ///





regulation of transcription, DNA-





dependent


41762_at
2.014074
TIAL1
regulation of transcription from RNA
7073





polymerase II promoter /// apoptosis





/// induction of apoptosis /// defense





response


910_at
−1.438133
TK1
nucleobase, nucleoside, nucleotide
7083





and nucleic acid metabolic process





/// DNA replication


922_at
0.851269
PPP2R1A
inactivation of MAPK activity ///
5518





regulation of DNA replication ///





protein complex assembly /// protein





amino acid dephosphorylation ///





ceramide metabolic process ///





induction of apoptosis /// RNA





splicing /// response to organic





substance /// second-messenger-





mediated signaling /// regulation of





Wnt receptor signaling pathway ///





regulation of cell adhesion ///





negative regulation of cell growth ///





regulation of growth /// negative





regulation of tyrosine





phosphorylation of Stat3 protein ///





regulation of transcription ///





regulation of cell differentiation


941_at
2.247213
PSMB6
ubiquitin-dependent protein
5694





catabolic process /// ubiquitin-





dependent protein catabolic process


954_s_at
−1.514611



















TABLE 5







Etoposide responsivity predictor set











Etopo






Probe Set



Entrez


ID
Weight
Gene Symbol
Go biological process term
Gene ID














1015_s_at
1.784401
LIMK1
protein amino acid phosphorylation ///
3984





protein amino acid phosphorylation ///





cell motility /// signal transduction ///





Rho protein signal transduction ///





nervous system development /// actin





cytoskeleton organization and





biogenesis /// positive regulation of





axon extension


1188_g_at
−1.618287
LIG3
DNA replication /// DNA repair /// DNA
3980





repair /// DNA recombination ///





response to DNA damage stimulus ///





cell cycle /// meiotic recombination ///





spermatogenesis /// V(D)J





recombination /// cell division


1233_s_at
−1.351889
AXL
protein amino acid phosphorylation ///
558





signal transduction


1456_s_at
−1.981805
IFI16
transcription /// regulation of
3428





transcription, DNA-dependent ///





regulation of transcription, DNA-





dependent /// cell proliferation ///





response to virus /// hemopoiesis ///





myeloid cell differentiation /// monocyte





differentiation /// DNA damage





response, signal transduction by p53





class mediator resulting in induction of





apoptosis


160020_at
1.795838
MMP14
ossification /// angiogenesis /// ovarian
4323





follicle development /// response to





hypoxia /// endothelial cell proliferation





/// proteolysis /// proteolysis ///





response to oxidative stress ///





metabolic process /// response to





mechanical stimulus /// response to





hormone stimulus /// cell migration ///





lung development /// zymogen





activation /// astrocyte cell migration ///





response to estrogen stimulus ///





branching morphogenesis of a tube ///





tissue remodeling /// negative





regulation of focal adhesion formation


1680_at
−4.528865
GRB7
signal transduction /// epidermal
2886





growth factor receptor signaling





pathway


1704_at
−0.820263
VAV2
signal transduction /// intracellular
7410





signaling cascade /// small GTPase





mediated signal transduction /// cell





migration /// lamellipodium biogenesis





/// regulation of Rho protein signal





transduction /// positive regulation of





phosphoinositide 3-kinase activity


1963_at
−1.84567
FLT1
angiogenesis /// patterning of blood
2321





vessels /// protein amino acid





phosphorylation /// transmembrane





receptor protein tyrosine kinase





signaling pathway /// transmembrane





receptor protein tyrosine kinase





signaling pathway /// multicellular





organismal development /// female





pregnancy /// positive regulation of cell





proliferation /// cell migration /// cell





differentiation /// vascular endothelial





growth factor receptor signaling





pathway


2047_s_at
3.095477
JUP
cell adhesion /// cell adhesion /// cell-
3728





cell adhesion


296_at
−2.126599





297_g_at
−1.354399





311_s_at
0.902404





31719_at
−0.768085
FN1
acute-phase response /// cell adhesion
2335





/// cell adhesion /// transmembrane





receptor protein tyrosine kinase





signaling pathway /// metabolic





process /// response to wounding ///





cell migration


31720_s_at
−0.997247
FN1
acute-phase response /// cell adhesion
2335





/// cell adhesion /// transmembrane





receptor protein tyrosine kinase





signaling pathway /// metabolic





process /// response to wounding ///





cell migration


32378_at
−1.419169
PKM2
glycolysis /// glycolysis
5315


32387_at
−2.0891
LYPLA3
lipid metabolic process /// fatty acid
23659





metabolic process /// ceramide





metabolic process /// fatty acid





catabolic process


32593_at
1.245739
RFTN1

23180


33282_at
−1.860632
LAD1

3898


33448_at
2.351885
SPINT1
morphogenesis of a branching
6692





structure /// embryonic placenta





development


33904_at
−5.280012
CLDN3
response to hypoxia /// calcium-
1365





independent cell-cell adhesion ///





calcium-independent cell-cell adhesion


34320_at
0.752612
PTRF
transcription /// transcription
284119





termination /// regulation of





transcription, DNA-dependent ///





transcription initiation from RNA





polymerase I promoter


34348_at
−4.77987
SPINT2 ///
cell motility
100130414




LOC100130414

/// 10653


34747_at
−1.301025
MMP14
ossification /// angiogenesis /// ovarian
4323





follicle development /// response to





hypoxia /// endothelial cell proliferation





/// proteolysis /// proteolysis ///





response to oxidative stress ///





metabolic process /// response to





mechanical stimulus /// response to





hormone stimulus /// cell migration ///





lung development /// zymogen





activation /// astrocyte cell migration ///





response to estrogen stimulus ///





branching morphogenesis of a tube ///





tissue remodeling /// negative





regulation of focal adhesion formation


34769_at
−0.957897
FAAH
fatty acid metabolic process
2166


35276_at
−0.982402
CLDN4
pathogenesis /// calcium-independent
1364





cell-cell adhesion


35309_at
2.289795
ST14
proteolysis /// proteolysis
6768


35444_at
1.213463
C19orf21

126353


35541_r_at
0.994896
KIAA0506

57239


35630_at
1.157887
LLGL2
cell cycle /// cell division
3993


35669_at
−5.408485
COBL

23242


35681_r_at
0.991531
ZEB2
transcription /// regulation of
9839





transcription, DNA-dependent ///





nervous system development ///





negative regulation of transcription ///





regulation of transcription


35735_at
−0.843365
GBP1
immune response
2633


36097_at
2.89579
IER2

9592


36890_at
−1.433033
PPL
keratinization
5493


37934_at
1.072557
TMEM30B

161291


38221_at
0.839583
CNKSR1
transmembrane receptor protein
10256





tyrosine kinase signaling pathway ///





Ras protein signal transduction /// Rho





protein signal transduction


38482_at
3.716125
CLDN7
calcium-independent cell-cell adhesion
1366


38759_at
0.944703
BTN3A2

11118


38760_f_at
−1.596566
BTN3A2

11118


39331_at
2.411297
TUBB2A
microtubule-based process ///
7280





microtubule-based movement ///





mitosis /// neuron differentiation ///





protein polymerization


39732_at
2.325338
MAP7
microtubule cytoskeleton organization
9053





and biogenesis /// establishment





and/or maintenance of cell polarity


39870_at
−1.522243
RBMXL2

27288


40215_at
1.420872
UGCG
glucosylceramide biosynthetic process
7357





/// glycosphingolipid biosynthetic





process /// epidermis development


40225_at
2.377423
GAK
protein amino acid phosphorylation ///
2580





cell cycle


41359_at
0.513065
PKP3
cell adhesion
11187


41872_at
0.58884
DFNA5
sensory perception of sound ///
1687





sensory perception of sound /// inner





ear receptor cell differentiation


479_at
−1.886719
DAB2
cell proliferation
1601


575_s_at
3.082061
TACSTD1

4072


671_at
1.491038
SPARC
ossification /// transmembrane receptor
6678





protein tyrosine kinase signaling





pathway


903_at
−2.447505
PPP2R5A
signal transduction
5525
















TABLE 6







Taxol responsivity predictor set











Taxol






Probe Set



Entrez


ID
Weight
Gene Symbol
Go biological process term
Gene ID














1218_at
2.174617
NR2F6
transcription /// regulation of
2063





transcription, DNA-dependent ///





signal transduction /// entrainment of





circadian clock by photoperiod ///





neuron development /// detection of





temperature stimulus involved in





sensory perception of pain


1581_s_at
−1.252126
TOP2B
neuron migration /// DNA metabolic
7155





process /// DNA topological change





/// axonogenesis /// forebrain





development


1587_at
1.902324
RARG
transcription /// regulation of
5916





transcription, DNA-dependent ///





multicellular organismal





development


1824_s_at
1.645376
PCNA
DNA replication /// DNA replication
5111





/// regulation of DNA replication ///





DNA repair /// base-excision repair,





gap-filling /// intracellular protein





transport /// cell proliferation ///





phosphoinositide-mediated signaling


1871_g_at
1.490669
PTPN11
protein amino acid
5781





dephosphorylation /// signal





transduction /// sensory perception





of sound /// dephosphorylation


1882_g_at
1.181144





1903_at
2.373819





2001_g_at
−1.02787
ATM
DNA repair /// DNA repair ///
472





response to DNA damage stimulus





/// cell cycle /// mitotic cell cycle





spindle assembly checkpoint ///





meiotic recombination /// signal





transduction /// response to ionizing





radiation /// negative regulation of





cell cycle


249_at
2.686971
NFATC4
transcription /// regulation of
4776





transcription, DNA-dependent ///





transcription from RNA polymerase





II promoter /// inflammatory response





/// heart development /// cellular





respiration /// regulation of





transcription


32386_at
0.851159
LOC100130134

100130134


33064_at
−2.893535
CACNG1
transport /// transport /// ion transport
786





/// calcium ion transport /// muscle





contraction


33557_at
2.052513
C22orf31

25770


335_r_at
−2.423531





34197_at
1.523938
PIK3R2
signal transduction /// negative
5296





regulation of anti-apoptosis ///





negative regulation of anti-apoptosis


34247_at
2.406482





34471_at
2.325227
MYH8
muscle contraction /// striated
4626





muscle contraction


34862_at
0.634438
SCCPDH
metabolic process
51097


34909_at
−1.087802
PHTF2
transcription /// regulation of
57157





transcription, DNA-dependent


34923_at
2.607029
IQSEC2
regulation of ARF protein signal
23096





transduction


34984_at
3.099121
TRPC3
transport /// ion transport /// calcium
7222





ion transport /// calcium ion transport





/// phototransduction


35254_at
0.711369
TRAFD1

10906


35644_at
4.69153
HEPH
transport /// ion transport /// copper
9843





ion transport /// iron ion transport


35908_at
−1.593513
SOX30
transcription /// regulation of
11063





transcription, DNA-dependent


36595_s_at
1.499879
GATM
creatine biosynthetic process
2628


37378_r_at
−1.0972
LMNA

4000


37767_at
2.505571
HTT
apoptosis /// apoptosis /// induction
3064





of apoptosis /// behavior ///





pathogenesis /// organ





morphogenesis


38680_at
1.486391
SNRPE
spliceosome assembly /// mRNA
6635





processing /// RNA splicing /// mRNA





metabolic process


38697_at
−2.522602
YIPF3
cell differentiation
25844


38703_at
−0.908363
DNPEP
proteolysis /// peptide metabolic
23549





process


39488_at
1.644714
PCDH9
cell adhesion /// homophilic cell
5101





adhesion


39537_at
−3.282921
KLHDC3
ossification /// transport /// Golgi to
116138





endosome transport /// endocytosis





/// endocytosis /// meiosis /// meiotic





recombination /// G-protein coupled





receptor protein signaling pathway ///





neuropeptide signaling pathway ///





multicellular organismal





development /// endosome to





lysosome transport /// induction of





apoptosis by extracellular signals ///





regulation of gene expression ///





myotube differentiation /// vesicle





organization and biogenesis /// cell





differentiation /// endosome transport





via multivesicular body sorting





pathway /// response to insulin





stimulus /// negative regulation of





apoptosis /// glucose import /// nerve





growth factor receptor signaling





pathway /// plasma membrane to





endosome transport /// negative





regulation of lipoprotein lipase





activity


40360_at
1.718898
SLC10A3
transport /// sodium ion transport ///
8273





sodium ion transport /// organic





anion transport


40529_at
−1.361106
LHX2
transcription /// regulation of
9355





transcription, DNA-dependent ///





nervous system development ///





brain development /// mesoderm





development /// dorsal/ventral





pattern formation /// regulation of





transcription


40690_at
0.503167
CKS2
regulation of cyclin-dependent
1164





protein kinase activity /// cell cycle ///





cell cycle /// spindle organization and





biogenesis /// meiosis I /// cell





proliferation /// phosphoinositide-





mediated signaling /// cell division


41045_at
0.75712
SECTM1
immune response /// mesoderm
6398





development /// positive regulation of





I-kappaB kinase/NF-kappaB





cascade


41204_s_at
−1.781384
SF1
spliceosome assembly /// nuclear
7536





mRNA 3′-splice site recognition ///





nuclear mRNA splicing, via





spliceosome /// transcription ///





regulation of transcription, DNA-





dependent /// mRNA processing ///





RNA splicing /// negative regulation





of smooth muscle cell proliferation


41404_at
−1.779211
RPS6KA4
regulation of transcription, DNA-
8986





dependent /// protein amino acid





phosphorylation /// protein amino





acid phosphorylation /// protein





amino acid phosphorylation ///





protein kinase cascade /// protein





kinase cascade


761_g_at
−2.000734
DYRK2
protein amino acid phosphorylation
8445





/// protein amino acid





phosphorylation /// protein amino





acid phosphorylation /// apoptosis ///





DNA damage response, signal





transduction by p53 class mediator





resulting in induction of apoptosis ///





positive regulation of glycogen





biosynthetic process


777_at
0.746673
GDI2
signal transduction /// protein
2665





transport /// regulation of GTPase





activity


925_at
−1.943148
PIK3R2 ///
signal transduction /// negative
10437 ///




IFI30
regulation of anti-apoptosis ///
5296





negative regulation of anti-apoptosis
















TABLE 7







Topotecan responsivity predictor set











Topo Probe



Entrez


Set ID
Weight
Gene Symbol
Go biological process term
Gene ID














1005_at
−1.583466
DUSP1
protein amino acid
1843





dephosphorylation /// response to





stress /// response to oxidative





stress /// cell cycle /// intracellular





signaling cascade ///





dephosphorylation


115_at
−0.533912
THBS1
cell motility /// cell adhesion ///
7057





multicellular organismal





development /// nervous system





development /// blood coagulation


1233_s_at
0.416455
AXL
protein amino acid phosphorylation
558





/// signal transduction


1251_g_at
−2.051381
RAP1GAP
signal transduction /// signal
5909





transduction /// signal transduction





/// regulation of small GTPase





mediated signal transduction


1257_s_at
−0.915209
QSOX1
protein thiol-disulfide exchange ///
5768





negative regulation of cell





proliferation /// cell redox





homeostasis


1278_at
−1.053777





1368_at
0.95653
IL1R1
inflammatory response /// immune
3554





response /// signal transduction ///





cell surface receptor linked signal





transduction /// cytokine and





chemokine mediated signaling





pathway /// innate immune





response


1385_at
−0.254775
TGFBI
cell adhesion /// negative
7045





regulation of cell adhesion /// visual





perception /// visual perception ///





cell proliferation /// response to





stimulus


1491_at
−1.32518
PTX3
response to yeast /// inflammatory
5806





response /// opsonization ///





positive regulation of nitric oxide





biosynthetic process /// positive





regulation of phagocytosis


1544_at
−2.317916
BLM
regulation of cyclin-dependent
641





protein kinase activity /// G2 phase





of mitotic cell cycle /// telomere





maintenance /// double-strand





break repair via homologous





recombination /// DNA replication





/// DNA repair /// DNA repair ///





DNA recombination /// DNA





recombination /// response to DNA





damage stimulus /// response to X-





ray /// G2/M transition DNA





damage checkpoint /// cellular





metabolic process /// negative





regulation of DNA recombination ///





positive regulation of transcription





/// negative regulation of mitotic





recombination /// alpha-beta T cell





differentiation /// positive regulation





of alpha-beta T cell proliferation ///





replication fork protection ///





regulation of binding /// protein





oligomerization /// chromosome





organization and biogenesis ///





negative regulation of cell division


1563_s_at
−1.13338
TNFRSF1A
prostaglandin metabolic process ///
7132





apoptosis /// inflammatory





response /// inflammatory





response /// signal transduction ///





cytokine and chemokine mediated





signaling pathway /// cytokine and





chemokine mediated signaling





pathway /// positive regulation of I-





kappaB kinase/NF-kappaB





cascade /// positive regulation of





transcription from RNA polymerase





II promoter /// positive regulation of





transcription from RNA polymerase





II promoter /// positive regulation of





inflammatory response /// positive





regulation of inflammatory





response


1593_at
−0.931912
FGF2
activation of MAPKK activity ///
2247





activation of MAPK activity ///





angiogenesis /// induction of an





organ /// positive regulation of





protein amino acid phosphorylation





/// chemotaxis /// signal





transduction /// Ras protein signal





transduction /// cell-cell signaling ///





multicellular organismal





development /// nervous system





development /// muscle





development /// cell proliferation ///





positive regulation of cell





proliferation /// negative regulation





of cell proliferation /// organ





morphogenesis /// glial cell





differentiation /// positive regulation





of granule cell precursor





proliferation /// cell differentiation ///





lung development /// positive





regulation of cell differentiation ///





positive regulation of angiogenesis





/// regulation of retinal cell





programmed cell death /// positive





regulation of epithelial cell





proliferation


159_at
−1.131544
VEGFC
angiogenesis /// positive regulation
7424





of neuroblast proliferation ///





substrate-bound cell migration ///





signal transduction /// multicellular





organismal development /// cell





proliferation /// positive regulation





of cell proliferation /// positive





regulation of cell proliferation ///





organ morphogenesis ///





morphogenesis of embryonic





epithelium /// cell differentiation ///





vascular endothelial growth factor





receptor signaling pathway


160044_g_at
−1.330621
ACO2
generation of precursor
50





metabolites and energy ///





tricarboxylic acid cycle ///





tricarboxylic acid cycle /// citrate





metabolic process /// citrate





metabolic process /// metabolic





process


1751_g_at
−1.870035
FARSA
translation /// tRNA aminoacylation
2193





for protein translation ///





phenylalanyl-tRNA aminoacylation


1783_at
0.658517
RIN2
endocytosis /// signal transduction
54453





/// small GTPase mediated signal





transduction


1828_s_at
−1.142258
FGF2
activation of MAPKK activity ///
2247





activation of MAPK activity ///





angiogenesis /// induction of an





organ /// positive regulation of





protein amino acid phosphorylation





/// chemotaxis /// signal





transduction /// Ras protein signal





transduction /// cell-cell signaling ///





multicellular organismal





development /// nervous system





development /// muscle





development /// cell proliferation ///





positive regulation of cell





proliferation /// negative regulation





of cell proliferation /// organ





morphogenesis /// glial cell





differentiation /// positive regulation





of granule cell precursor





proliferation /// cell differentiation ///





lung development /// positive





regulation of cell differentiation ///





positive regulation of angiogenesis





/// regulation of retinal cell





programmed cell death /// positive





regulation of epithelial cell





proliferation


1879_at
−1.345807
RRAS
small GTPase mediated signal
6237





transduction /// Ras protein signal





transduction /// negative regulation





of cell migration


1958_at
−0.849681
FIGF
angiogenesis /// multicellular
2277





organismal development /// cell





proliferation /// positive regulation





of cell proliferation /// positive





regulation of cell proliferation ///





cell differentiation /// vascular





endothelial growth factor receptor





signaling pathway


2042_s_at
−2.085535
MYB
transcription /// regulation of
4602





transcription, DNA-dependent ///





regulation of transcription, DNA-





dependent /// regulation of





transcription


2053_at
−1.376639
CDH2
cell adhesion /// cell adhesion ///
1000





homophilic cell adhesion


2056_at
−1.331695
FGFR1
MAPKKK cascade /// skeletal
2260





development /// protein amino acid





phosphorylation /// protein amino





acid phosphorylation /// fibroblast





growth factor receptor signaling





pathway /// fibroblast growth factor





receptor signaling pathway /// cell





growth


2057_g_at
−1.931123
FGFR1
MAPKKK cascade /// skeletal
2260





development /// protein amino acid





phosphorylation /// protein amino





acid phosphorylation /// fibroblast





growth factor receptor signaling





pathway /// fibroblast growth factor





receptor signaling pathway /// cell





growth


232_at
−1.506829
LAMC1
protein complex assembly /// cell
3915





adhesion /// cell adhesion ///





endoderm development /// cell





migration /// extracellular matrix





disassembly /// hemidesmosome





assembly /// positive regulation of





epithelial cell proliferation


31521_f_at
−2.857943
HIST1H4I ///
establishment and/or maintenance
121504 ///




HIST1H4A ///
of chromatin architecture ///
554313 ///




HIST1H4D ///
nucleosome assembly ///
8294 ///




HIST1H4F ///
phosphoinositide-mediated
8359 ///




HIST1H4K ///
signaling
8360 ///




HIST1H4J ///

8361 ///




HIST1H4C ///

8362 ///




HIST1H4H ///

8363 ///




HIST1H4B ///

8364 ///




HIST1H4E ///

8365 ///




HIST1H4L ///

8366 ///




HIST2H4A ///

8367 ///




HIST4H4 ///

8368 ///




HIST2H4B

8370


32098_at
0.474113
COL6A2
phosphate transport /// cell
1292





adhesion /// cell-cell adhesion ///





extracellular matrix organization





and biogenesis


32116_at
−1.798732
TMC6

11322


32260_at
0.772105
PEA15
transport /// transport /// apoptosis
8682





/// anti-apoptosis /// carbohydrate





transport /// regulation of apoptosis





/// regulation of apoptosis ///





negative regulation of glucose





import


32434_at
−0.868285
MARCKS
cell motility
4082


32529_at
−1.490314
CKAP4

10970


32531_at
0.748264
GJA1
in utero embryonic development ///
2697





neuron migration /// heart looping





/// epithelial cell maturation ///





transport /// apoptosis /// muscle





contraction /// cell communication





/// cell-cell signaling /// cell-cell





signaling /// heart development ///





adult heart development ///





sensory perception of sound ///





regulation of heart contraction ///





negative regulation of cell





proliferation /// response to pH ///





vascular transport /// ATP transport





/// gap junction assembly ///





embryonic heart tube development





/// positive regulation of I-kappaB





kinase/NF-kappaB cascade ///





skeletal muscle regeneration ///





positive regulation of protein





catabolic process /// positive





regulation of striated muscle





development /// blood vessel





morphogenesis /// neurite





morphogenesis /// protein





oligomerization /// regulation of





calcium ion transport


32535_at
−0.37409
FBN1
skeletal development /// heart
2200





development /// blood coagulation


32606_at
0.49158





32607_at
−0.910611
BASP1

10409


32673_at
−0.94355
BTN2A1
lipid metabolic process
11120


32808_at
−1.041547
ITGB1
cellular defense response /// cell
3688





adhesion /// homophilic cell





adhesion /// cell-matrix adhesion ///





integrin-mediated signaling





pathway /// multicellular organismal





development /// cell migration


32812_at
−0.762285
LIMCH1
actomyosin structure organization
22998





and biogenesis


32847_at
−1.519068
MYLK
protein amino acid phosphorylation
4638





/// protein amino acid





phosphorylation


33127_at
−0.928141
LOXL2 ///
UDP catabolic process /// protein
4017 ///




ENTPD4
modification process /// cell
9583





adhesion /// aging


33328_at
−0.492804
HEG1

57493


33337_at
−1.951393
DEGS1
lipid metabolic process /// fatty acid
8560





biosynthetic process ///





unsaturated fatty acid biosynthetic





process /// lipid biosynthetic





process


33404_at
−1.525294
CAP2
cytoskeleton organization and
10486





biogenesis /// establishment and/or





maintenance of cell polarity ///





signal transduction /// activation of





adenylate cyclase activity


33405_at
−1.580917
CAP2
cytoskeleton organization and
10486





biogenesis /// establishment and/or





maintenance of cell polarity ///





signal transduction /// activation of





adenylate cyclase activity


33440_at
0.52335
ZEB1
negative regulation of transcription
6935





from RNA polymerase II promoter





/// transcription /// regulation of





transcription, DNA-dependent ///





regulation of transcription from





RNA polymerase II promoter ///





immune response /// cell





proliferation /// regulation of





transcription


33772_at
0.366823
PTGER4
immune response /// signal
5734





transduction /// G-protein coupled





receptor protein signaling pathway





/// G-protein signaling, coupled to





cAMP nucleotide second





messenger /// regulation of





ossification


33785_at
−0.600057
BAI2
signal transduction /// G-protein
576





coupled receptor protein signaling





pathway /// G-protein coupled





receptor protein signaling pathway





/// neuropeptide signaling pathway


33787_at
0.350378
NUAK1
protein amino acid phosphorylation
9891


33791_at
−1.38754
DLEU1
cell cycle /// negative regulation of
10301





cell cycle


33882_at
−1.204573
RAB11FIP5
transport /// metabolic process ///
26056





protein transport


33900_at
−1.640013
FSTL3
negative regulation of BMP
10272





signaling pathway


33994_g_at
−1.586516
MYL6 ///
muscle contraction /// skeletal
140465 ///




MYL6B
muscle development /// muscle
4637





filament sliding


34091_s_at
−0.849163
VIM
cell motility /// intermediate
7431





filament-based process


34106_at
−0.605788
GNA12
signal transduction /// G-protein
2768





coupled receptor protein signaling





pathway /// G-protein coupled





receptor protein signaling pathway





/// blood coagulation


34318_at
−1.594771
PRAF2
transport /// protein transport /// L-
11230





glutamate transport


34320_at
−0.729318
PTRF
transcription /// transcription
284119





termination /// regulation of





transcription, DNA-dependent ///





transcription initiation from RNA





polymerase I promoter


34375_at
−1.016405
CCL2
protein amino acid phosphorylation
6347





/// cellular calcium ion homeostasis





/// anti-apoptosis /// chemotaxis ///





chemotaxis /// inflammatory





response /// immune response ///





humoral immune response /// cell





adhesion /// signal transduction ///





cell surface receptor linked signal





transduction /// G-protein coupled





receptor protein signaling pathway





/// G-protein signaling, coupled to





cyclic nucleotide second





messenger /// JAK-STAT cascade





/// cell-cell signaling /// organ





morphogenesis /// viral genome





replication


34795_at
−0.96244
PLOD2
response to hypoxia /// protein
5352





modification process /// protein





metabolic process


34802_at
−1.080254
COL6A2
phosphate transport /// cell
1292





adhesion /// cell-cell adhesion ///





extracellular matrix organization





and biogenesis


34811_at
0.554813
ATP5G3
generation of precursor
518





metabolites and energy ///





transport /// ion transport /// ATP





synthesis coupled proton transport





/// proton transport /// ATP





metabolic process


35130_at
−1.014434
GSR
glutathione metabolic process ///
2936





cell redox homeostasis


35264_at
−1.524088
NDUFS3
mitochondrial electron transport,
4722





NADH to ubiquinone ///





mitochondrial electron transport,





NADH to ubiquinone /// protein





amino acid dephosphorylation ///





oxygen and reactive oxygen





species metabolic process ///





transport /// induction of apoptosis





/// dephosphorylation /// negative





regulation of cell growth ///





oxidation reduction


35309_at
−0.925755
ST14
proteolysis /// proteolysis
6768


35366_at
−0.947779
NID1
cell adhesion /// cell-matrix
4811





adhesion /// bioluminescence ///





protein-chromophore linkage


35729_at
−0.923216
MYO1D

4642


35751_at
−1.131622
SDHB
tricarboxylic acid cycle ///
6390





tricarboxylic acid cycle /// transport





/// aerobic respiration /// oxidation





reduction


36119_at
−0.40934
CAV1
inactivation of MAPK activity ///
857





vasculogenesis /// response to





hypoxia /// negative regulation of





endothelial cell proliferation ///





triacylglycerol metabolic process ///





calcium ion transport /// cellular





calcium ion homeostasis ///





endocytosis /// regulation of





smooth muscle contraction ///





skeletal muscle development ///





protein localization /// vesicle





organization and biogenesis ///





regulation of fatty acid metabolic





process /// sequestering of lipid ///





regulation of blood coagulation ///





cholesterol transport /// negative





regulation of epithelial cell





differentiation /// mammary gland





development /// nitric oxide





homeostasis /// cholesterol





homeostasis /// cholesterol





homeostasis /// negative regulation





of MAPKKK cascade /// negative





regulation of nitric oxide





biosynthetic process /// positive





regulation of vasoconstriction ///





negative regulation of vasodilation





/// negative regulation of JAK-





STAT cascade /// positive





regulation of metalloenzyme





activity /// protein





homooligomerization /// membrane





depolarization /// regulation of





peptidase activity /// calcium ion





homeostasis /// mammary gland





involution


36149_at
−1.468146
DPYSL3
nucleobase, nucleoside, nucleotide
1809





and nucleic acid metabolic process





/// signal transduction /// nervous





system development /// nervous





system development


36369_at
0.937326
PTRF
transcription /// transcription
284119





termination /// regulation of





transcription, DNA-dependent ///





transcription initiation from RNA





polymerase I promoter


36525_at
−0.978609
FBXL2
protein modification process ///
25827





proteolysis /// ubiquitin cycle


36550_at
−1.009504
RIN2
endocytosis /// signal transduction
54453





/// small GTPase mediated signal





transduction


36577_at
−1.446371
FERMT2
cell adhesion /// cell adhesion ///
10979





regulation of cell shape /// actin





cytoskeleton organization and





biogenesis


36638_at
0.911469
CTGF
cartilage condensation ///
1490





ossification /// angiogenesis ///





regulation of cell growth /// DNA





replication /// cell motility /// cell





adhesion /// cell-matrix adhesion ///





integrin-mediated signaling





pathway /// intracellular signaling





cascade /// fibroblast growth factor





receptor signaling pathway ///





epidermis development ///





response to wounding /// cell





migration /// cell differentiation


36659_at
−2.116964
COL4A2
phosphate transport /// negative
1284





regulation of angiogenesis ///





extracellular matrix organization





and biogenesis


36790_at
−1.46788
TPM1
cell motility /// regulation of muscle
7168





contraction /// regulation of heart





contraction


36791_g_at
0.634657
TPM1
cell motility /// regulation of muscle
7168





contraction /// regulation of heart





contraction


36792_at
−0.771539
TPM1
cell motility /// regulation of muscle
7168





contraction /// regulation of heart





contraction


36799_at
−0.81719
FZD2
establishment of tissue polarity ///
2535





signal transduction /// signal





transduction /// cell surface





receptor linked signal transduction





/// G-protein coupled receptor





protein signaling pathway /// cell-





cell signaling /// multicellular





organismal development /// Wnt





receptor signaling pathway ///





epithelial cell differentiation


36811_at
−0.394702
LOXL1
protein amino acid deamination ///
4016





oxidation reduction


36885_at
−2.44668
SYK
serotonin secretion /// protein
6850





complex assembly /// protein





amino acid phosphorylation ///





protein amino acid phosphorylation





/// leukocyte adhesion /// signal





transduction /// enzyme linked





receptor protein signaling pathway





/// integrin-mediated signaling





pathway /// intracellular signaling





cascade /// activation of JNK





activity /// cell proliferation /// organ





morphogenesis /// peptidyl-tyrosine





phosphorylation /// leukotriene





biosynthetic process /// neutrophil





chemotaxis /// positive regulation





of mast cell degranulation /// beta





selection /// positive regulation of





interleukin-3 biosynthetic process





/// positive regulation of





granulocyte macrophage colony-





stimulating factor biosynthetic





process /// positive regulation of B





cell differentiation /// positive





regulation of gamma-delta T cell





differentiation /// positive regulation





of alpha-beta T cell differentiation





/// positive regulation of alpha-beta





T cell proliferation /// protein amino





acid autophosphorylation ///





positive regulation of peptidyl-





tyrosine phosphorylation ///





positive regulation of calcium-





mediated signaling /// B cell





receptor signaling pathway


36952_at
−1.151528
HADHA
lipid metabolic process /// fatty acid
3030





metabolic process /// fatty acid





beta-oxidation /// metabolic





process /// response to drug


36988_at
−1.204986
TNFAIP1
DNA replication /// DNA replication
7126





/// regulation of transcription, DNA-





dependent /// translation ///





translation /// potassium ion





transport /// immune response ///





immune response /// embryonic





development /// embryonic





development


37032_at
−1.245426
NNMT

4837


37322_s_at
−2.270441
HPGD
lipid metabolic process /// fatty acid
3248





metabolic process /// prostaglandin





metabolic process /// prostaglandin





metabolic process /// transforming





growth factor beta receptor





signaling pathway /// female





pregnancy /// parturition ///





metabolic process /// lipoxygenase





pathway /// negative regulation of





cell cycle


37408_at
−1.250313
MRC2
endocytosis
9902


37486_f_at
−0.773001
MEIS3P1
regulation of transcription, DNA-
4213





dependent /// regulation of





transcription


37599_at
−0.440975
AOX1
oxygen and reactive oxygen
316





species metabolic process ///





inflammatory response /// oxidation





reduction


376_at
0.358759
SEMA3C
immune response ///
10512





transmembrane receptor protein





tyrosine kinase signaling pathway





/// multicellular organismal





development /// response to drug


377_g_at
1.565062
SEMA3C
immune response ///
10512





transmembrane receptor protein





tyrosine kinase signaling pathway





/// multicellular organismal





development /// response to drug


38113_at
−0.971934
SYNE1
nuclear organization and
23345





biogenesis /// Golgi organization





and biogenesis /// keratinization ///





muscle cell differentiation


38125_at
−1.45776
SERPINE1
blood coagulation /// fibrinolysis ///
5054





regulation of angiogenesis


38299_at
−0.839698
IL6
neutrophil apoptosis /// neutrophil
3569





apoptosis /// acute-phase response





/// inflammatory response ///





immune response /// humoral





immune response /// cell surface





receptor linked signal transduction





/// cell-cell signaling /// cell-cell





signaling /// positive regulation of





cell proliferation /// negative





regulation of cell proliferation ///





positive regulation of peptidyl-





serine phosphorylation /// defense





response to protozoan ///





regulation of apoptosis /// negative





regulation of apoptosis /// positive





regulation of MAPKKK cascade ///





negative regulation of chemokine





biosynthetic process /// negative





regulation of chemokine





biosynthetic process /// positive





regulation of T-helper 2 cell





differentiation /// positive regulation





of translation /// positive regulation





of transcription, DNA-dependent ///





positive regulation of transcription





from RNA polymerase II promoter





/// negative regulation of hormone





secretion /// positive regulation of





peptidyl-tyrosine phosphorylation





/// response to glucocorticoid





stimulus


38338_at
−0.844895
RRAS
small GTPase mediated signal
6237





transduction /// Ras protein signal





transduction /// negative regulation





of cell migration


38394_at
−1.836007
GPD1L
carbohydrate metabolic process ///
23171





glycerol-3-phosphate metabolic





process /// metabolic process ///





glycerol-3-phosphate catabolic





process


38396_at
−0.915548
MAP1B
microtubule bundle formation ///
4131





negative regulation of microtubule





depolymerization /// dendrite





development


38433_at
0.24027
AXL
protein amino acid phosphorylation
558





/// signal transduction


38449_at
−2.322063
WDR23

80344


38482_at
−2.584731
CLDN7
calcium-independent cell-cell
1366





adhesion


38488_s_at
−1.103501
IL15
immune response /// immune
3600





response /// signal transduction ///





cell-cell signaling /// positive





regulation of cell proliferation


38631_at
0.472464
TNFAIP2
angiogenesis /// multicellular
7127





organismal development /// cell





differentiation


38772_at
−1.647628
CYR61
regulation of cell growth ///
3491





chemotaxis /// cell adhesion /// cell





proliferation /// anatomical





structure morphogenesis


38775_at
−0.628315
LRP1 ///
lipid metabolic process ///
100134190




LOC100134190
endocytosis /// multicellular
/// 4035





organismal development /// cell





proliferation


38842_at
0.382258
AMOTL2

51421


38921_at
−0.553897
PDE1B
apoptosis /// signal transduction
5153


39100_at
0.440558
SPOCK1
cell motility /// cell adhesion ///
6695





multicellular organismal





development /// nervous system





development /// cell proliferation


39254_at
−1.452007
RAI14

26064


39277_at
0.600055
OSMR
cell surface receptor linked signal
9180





transduction /// cell proliferation


39327_at
−0.423697
PXDN
immune response /// response to
7837





oxidative stress /// hydrogen





peroxide catabolic process ///





oxidation reduction


39333_at
−3.12657
COL4A1
phosphate transport
1282


39409_at
0.638946
C1R
proteolysis /// immune response ///
715





immune response /// complement





activation, classical pathway ///





innate immune response


39614_at
−0.683932
KIAA0802

23255


39710_at
−0.703314
C5orf13
regulation of transforming growth
9315





factor beta receptor signaling





pathway


39867_at
−1.328942
TUFM
translation /// translational
7284





elongation /// translational





elongation


39901_at
0.370034
EDIL3
cell adhesion /// multicellular
10085





organismal development


40023_at
0.527535
BDNF
nervous system development
627


40078_at
0.311987
PRSS23
proteolysis
11098


40096_at
−2.35259
ATP5A1
negative regulation of endothelial
498





cell proliferation /// ATP





biosynthetic process /// transport ///





ion transport /// ATP synthesis





coupled proton transport /// proton





transport


40171_at
−2.001626
FRAT2
multicellular organismal
23401





development /// cell proliferation ///





Wnt receptor signaling pathway


40341_at
−0.929303
TMEM186

25880


40497_at
−1.09089
TUSC4
cell cycle /// negative regulation of
10641





cell cycle


40564_at
−1.429238
NUP50
transport /// protein transport ///
10762





intracellular transport /// mRNA





transport /// intracellular protein





transport across a membrane


40567_at
−0.263158
TUBA1A
microtubule-based process ///
7846





microtubule-based movement ///





protein polymerization


40642_at
−2.210099
NFIB
DNA replication /// transcription ///
4781





regulation of transcription, DNA-





dependent


40692_at
−0.519972
TLE4
transcription /// regulation of
7091





transcription, DNA-dependent ///





Wnt receptor signaling pathway ///





regulation of transcription


40781_at
0.252463
AKT3
protein amino acid phosphorylation
10000





/// protein amino acid





phosphorylation /// signal





transduction


40936_at
−1.518856
CRIM1
regulation of cell growth ///
51232





proteolysis /// nervous system





development


41197_at
1.636782
RAD23A
DNA repair /// nucleotide-excision
5886





repair /// nucleotide-excision repair





/// protein modification process ///





response to DNA damage stimulus





/// proteasomal ubiquitin-





dependent protein catabolic





process


41223_at
0.425264
COX5A
oxidation reduction
9377


41236_at
0.532242
SMCR7L

54471


41273_at
−0.865492
MXRA7

439921


41295_at
−1.468337
STARD7

56910


41354_at
0.22724
STC1
cellular calcium ion homeostasis ///
6781





cell surface receptor linked signal





transduction /// cell-cell signaling ///





response to nutrient


41478_at
0.785668
TTC28

23331


41544_at
0.959715
PLK2
mitotic cell cycle /// protein amino
10769





acid phosphorylation /// positive





regulation of I-kappaB kinase/NF-





kappaB cascade


41667_s_at
−1.111511
TGDS
metabolic process /// cellular
23483





metabolic process


41738_at
−1.668471
CALD1
cell motility /// muscle contraction
800


41744_at
−1.398598
OPTN
protein targeting to Golgi /// Golgi
10133





organization and biogenesis ///





signal transduction /// cell death ///





Golgi to plasma membrane protein





transport


41745_at
−0.837378
IFITM3
immune response /// response to
10410





biotic stimulus


41872_at
−1.128956
DFNA5
sensory perception of sound ///
1687





sensory perception of sound ///





inner ear receptor cell





differentiation


424_s_at
−1.471912
FGFR1
MAPKKK cascade /// skeletal
2260





development /// protein amino acid





phosphorylation /// protein amino





acid phosphorylation /// fibroblast





growth factor receptor signaling





pathway /// fibroblast growth factor





receptor signaling pathway /// cell





growth


465_at
−1.303867
HTATIP
regulation of cell growth /// double-
10524





strand break repair /// chromatin





assembly or disassembly ///





transcription /// regulation of





transcription, DNA-dependent ///





transcription from RNA polymerase





II promoter /// chromatin





modification /// histone acetylation





/// androgen receptor signaling





pathway /// positive regulation of





transcription, DNA-dependent


548_s_at
−2.690624
SYK
serotonin secretion /// protein
6850





complex assembly /// protein





amino acid phosphorylation ///





protein amino acid phosphorylation





/// leukocyte adhesion /// signal





transduction /// enzyme linked





receptor protein signaling pathway





/// integrin-mediated signaling





pathway /// intracellular signaling





cascade /// activation of JNK





activity /// cell proliferation /// organ





morphogenesis /// peptidyl-tyrosine





phosphorylation /// leukotriene





biosynthetic process /// neutrophil





chemotaxis /// positive regulation





of mast cell degranulation /// beta





selection /// positive regulation of





interleukin-3 biosynthetic process





/// positive regulation of





granulocyte macrophage colony-





stimulating factor biosynthetic





process /// positive regulation of B





cell differentiation /// positive





regulation of gamma-delta T cell





differentiation /// positive regulation





of alpha-beta T cell differentiation





/// positive regulation of alpha-beta





T cell proliferation /// protein amino





acid autophosphorylation ///





positive regulation of peptidyl-





tyrosine phosphorylation ///





positive regulation of calcium-





mediated signaling /// B cell





receptor signaling pathway


581_at
−1.565877
LAMB1
cell adhesion /// cell adhesion ///
3912





positive regulation of cell migration





/// neurite development ///





odontogenesis /// positive





regulation of epithelial cell





proliferation


628_at
−0.861772
FZD2
establishment of tissue polarity ///
2535





signal transduction /// signal





transduction /// cell surface





receptor linked signal transduction





/// G-protein coupled receptor





protein signaling pathway /// cell-





cell signaling /// multicellular





organismal development /// Wnt





receptor signaling pathway ///





epithelial cell differentiation


672_at
−1.059245
SERPINE1
blood coagulation /// fibrinolysis ///
5054





regulation of angiogenesis


867_s_at
0.423802
THBS1
cell motility /// cell adhesion ///
7057





multicellular organismal





development /// nervous system





development /// blood coagulation


875_g_at
−0.889978
CCL2
protein amino acid phosphorylation
6347





/// cellular calcium ion homeostasis





/// anti-apoptosis /// chemotaxis ///





chemotaxis /// inflammatory





response /// immune response ///





humoral immune response /// cell





adhesion /// signal transduction ///





cell surface receptor linked signal





transduction /// G-protein coupled





receptor protein signaling pathway





/// G-protein signaling, coupled to





cyclic nucleotide second





messenger /// JAK-STAT cascade





/// cell-cell signaling /// organ





morphogenesis /// viral genome





replication


884_at
−1.840205
ITGA3
cell adhesion /// cell-matrix
3675





adhesion /// integrin-mediated





signaling pathway


885_g_at
−2.1017
ITGA3
cell adhesion /// cell-matrix
3675





adhesion /// integrin-mediated





signaling pathway


890_at
−1.809728
UBE2A
DNA repair /// postreplication
7319





repair /// ubiquitin-dependent





protein catabolic process ///





ubiquitin cycle /// response to DNA





damage stimulus /// post-





translational protein modification ///





regulation of protein metabolic





process


919_at
−0.953292



















TABLE 8







PI3 Kinase inhibitor responsivity predictor set









Gene Symbol
Affymetrix Probe ID
Gene Title





RFC2
1053_at
replication factor C (Activator 1) 2, 40 kDa


KIAA0153
1552257_a_at
KIAA0153 protein


EXOSC6
1553947_at
exosome component 6


RHOB
1553962_s_at
ras homolog gene family, member B


MAD2L1
1554768_a_at
MAD2 mitotic arrest deficient-like 1 (yeast)


RBM15
1555762_s_at
RNA binding motif protein 15


SPEN
1556059_s_at
spen homolog, transcriptional regulator




(Drosophilia)


C6orf150
1559051_s_at
chromosome 6 reading frame 150


HSPA1A
200799_at
heat shock 70 kDa protein 1A


HSPA1A///HSPA1B
200800_s_at
heat shock 70 kDa protein 1A///heat shock




70 kDa protein 1B


NOL5A
200875_s_at
nucleolar protein 5A (56 kDa with KKE/D




repeat)


CSE1L
201112_s_at
CSE1 chromosome segregation 1-like (yeast)


PCNA
201202_at
proliferating cell nuclear antigen


JUN
201464_x_at
v-jun sarcoma virus 17 oncogene homolog




(avian)


JUN
201465_s_at
v-jun sarcoma virus 17 oncogene homolog




(avian)


JUN
201466_s_at
v-jun sarcoma virus 17 oncogene homolog




(avian)


JUNB
201473_at
jun B proto-oncogene


MCM3
201555_at
MCM3 minichromosome maintenance deficient




3 (S. cerevisiae)


EGR1
201693_s_at
early growth response 1


DNMT1
201697_s_at
DNA (cytosine-5-)-methyltransferase 1


MCM5
201755_at
MCM5 minichromosome maintenance deficient




5, cell division cycle 46 (S. cerevisiae)


RRM2
201890_at
ribonucleotide reductase M2 polypeptide


MCM6
201930_at
MCM6 minichromosome maintenance deficient




6, (MISS homolog, S. pombe) (S. cerevisiae)


NASP
201970_s_at
nuclear autoantigenic sperm protein (histone-




binding)


SPEN
201997_s_at
spen homolog, transcriptional regulator




(Drosophilia)


IER2
202081_at
immediate early response 2


MCM2
202107_s_at
MCM2 minichromosome maintenance deficient




2, mitotin (S. cerevisiae)


MTHFD1
202309_at
methylenetetrahydrofolate dehydrogenase




(NADP+dependant) 1,




methylenetetrahydrofolate cyclohydrolase,




formyltetrahydrofolate synthetase


UNG
202330_s_at
uracil-DNA glycosylase


HSPA1B
202581_at
heat shock 70 kDa protein 1B


MSH6
202911_at
mutS homolog 6 (E. coli)


SSX2IP
203017_s_at
synovial sarcoma, X breakpoint 2 interacting




protein


RNASEH2A
203022_at
ribonuclease H2, large subunit


PEX5
203244_at
peroxisomal biogenesis factor 5


LMNB1
203276_at
lamin B1


POLD1
203422_at
polymerase (DNA directed), delta 1, catalytic




subunit 125 kDa


CDC6
203968_s_at
CDC6 cell division cycle 6 homolog (S. cerevisiae)


ZWINT
204026_s_at
ZW10 interactor


CDC45L
204126_s_at
CDC45 cell division cycle 45-like (S. cerevisiae)


RFC3
204128_s_at
replication factor C (activator 1) 3, 38 kDa


POLA2
204441_s_at
polymerase (DNA directed), alpha 2 (70 kD




subunit


CDC7
204510_at
CDC7 cell division cycle 7 (S. cerevisiae)


DIPA
204610_s_at
hepatitis delta antigen-interacting protein A


ACD
204617_s_at
adrenocortical dysplasia homolog (mouse)


CDC25A
204695_at
cell division cycle 25A


FEN1
204767_s_at
flap structure-specific endonuclease 1


FEN1
204768_s_at
flap structure-specific endonuclease 1


MYB
204798_at
v-myb myeloblastosis viral oncogene homolog




(avian)


TOP3A
204946_s_at
topoisomerase (DNA) III alpha


DDX10
204977_at
DEAD (Asp-Glu-Ala-Asp) box polypeptide 10


RAD51
205024_s_at
RAD51 homolog (RecA homolog, E. coli) S. cerevisiae)


CCNE2
205034_at
cyclin E2


PRIM1
205053_at
primase, polypeptide 1, 49 kDa


BARD1
205345_at
BRCA1 associated RING domain 1


CHEK1
205393_s_at
CHK1 checkpoint homolog (S. pombe)


H2AFX
205436_s_at
H2A histone family, member X


FLJI2973
205519_at
hypothetical protein FLJI2973


GEMIN4
205527_s_at
gem (nuclear organelle) associated protein 4


SLBP
206052_s_at
stem-loop (histone) binding protein


KIAA0186
206102_at
KIAA0186 gene product


AKR7A3
206469_x_at
aldo-keto reductase family 7, member A3




(aflatoxin aldehyde reductase)


TLE3
206472_s_at
transducin-like enhancer of split 3 (E(spl)




homolog, Drosophilia)


GADD45B
207574_s_at
growth arrest and DNA-damage-inducible, beta


PRPS1
207447_s_at
phosphoribosyl pyrophosphate synthetase 1


BRD2
208685_x_at
bromodomain containing 2


MCM7
208795_s_at
MCM7 minichromosome maintenance deficient




7 (S. cerevisiae)


ID1
208937_s_at
inhibitor of DNA binding 1, dominant negative




helix-loop-helix protein


GADD45B
209304_x_at
growth arrest and DNA-damage-inducible, beta


GADD45B
209305_s_at
growth arrest and DNA-damage-inducible, beta


POLR1C
209317_at
polymerase (RNA) I polypeptide C, 30 kDa


PRKRIR
209323_at
protein-kinase, interferon-inducible double




stranded RNA dependent inhibitor, repressor of




(P58 repressor)


MSH2
209421_at
mutS homolog 2, colon cancer, nonpolyposis




type 1 (E. coli)


PPAT
209433_s_at
phosphoribosyl pyrophosphate




amidotransferase


PPAT
209434_s_at
phosphoribosyl pyrophosphate




amidotransferase


PRPS1
209440_at
phosphoribosyl pyrophosphate synthetase 1


RPA3
209507_at
replication protein A3, 14 kDa


EED
209572_s_at
embryonic ectoderm development


GAS2L1
209729_at
growth arrest-specific 2 like 1


RPM2
209773_s_at
ribonucleotide reductase M2 polypeptide


SLC19A1
209777_s_at
solute carrier family 19 (folate transporter),




member 1


CDT1
209832_s_at
DNA replication factor


SHMT1
209980_s_at
serine hydroxymethyltranse 1 (soluable)


TAF5
210053_at
TAF5 RNA polymerase II, TATA box binding




protein (TBP)-associated factor, 100 kDa


MCM7
210983_s_at
MCM7 minichromosome maintenance deficient




7 (S. cerevisiae)


MSH6
211450_s_at
mutS homolog 6 (E. coli)


CCNE2
211814_s_at
cyclin E2


RHOB
212099_at
ras homolog gene family, member B


MCM4
212141_at
MCM4 minichromosome maintenance deficient




4 (S. cerevisiae)


MCM4
212142_at
MCM4 minichromosome maintenance deficient




4 (S. cerevisiae)


KCTD12
212188_at
potassium channel tetramerisation domain




containing 12/// potassium channel




tetramerisation domain containing 12


KCTD12
212192_at
potassium channel tetramerisation domain




containing 12


MAC30
212281_s_at
hypothetical protein MAC30


POLD3
212836_at
polymerase (DNA-directed), delta 3, accessory




subunit


KIAA0406
212898_at
KIAA0406 gene product


FLJ10719
213007_at
hypothetical protein FLJ10719


ITPKC
213076_at
inositol 1,4,5-triphosphate 3-kinase C


ZNF473
213124_at
zinc finger protein



213281_at



CCNE1
213523_at
cyclin E1


GADD45B
213560_at
Growth arrest and DNA-damage-inductible,




beta


GAL
214240_at
galanin


BRD2
214911_s_at
bromodomain containing 2


UMPS
215165_x_at
uridine monophosphate synthetase (orotate




phosphoribosyl transferase and orotidine-5′-




decarboxylase)


MCM5
216237_s_at
MCM5 minichromosome maintenance deficient




5, cell division cycle 46 (S. cerevisiae)


LAMNB2
216952_s_at
lamin B2


GEMIN4
217099_s_at
gem (nuclear organelle) associated protein 4


SUPT16H
217815_at
suppressor of Ty 16 homolog (S. cerevisae)


GMNN
218350_s_at
geminin, DNA replication inhibitor


RAMP
218585_s_at
RA-regulated nuclear matrix-associated protein


SLC25A15
218653_at
solute carrier family 25 (mitochondrial carrier;




ornithine transporter) member 15


FLJ13912
218719_s_at
hypothetical protein FLJ13912


ATAD2
218782_s_at
ATpase family, AAA domain containing 2


C10orf117
21889_at
chromosome 10 open reading frame 117


MGC10993
218897_at
hypothetical protein MGC10993


C21orf45
219004_s_at
chromosome 21 open reading frame 45


RPP25
219143_s_at
ribonuclease P 25 kDa subunit


FJL20516
219258_at
timeless-interacting protein


MGC4504
219270_at
hypothetical protein MGC4504


RBM15
219286_s_at
RNA binding motif protein 15


FLJ11078
219254_at
hypothetical protein FLJ11078


DCLRE1B
219490_s_at
DNA cross-link repair 1B (PSO2 homolog, S. cerevisiae)


FLJ34077
219731_at
weakly similar to zinc finger protein


FLJ20257
219798_s_at
hypothetical protein FLJ20257


MCM10
220651_s_at
MCM10 minichromosome maintenance




deficient 10 (S. cerevisiae)


TBRG4
220789_s_at
transforming growth factor beta regulator 4


Pfs2
221521_s_at
DNA replication complex GINS protein PSF2


LEF1
221558_s_at
lymphoid enhancer-binding factor 1


ZNF45
222028_at
zinc finger protein 45


MCM4
222036_s_at
MCM4 minichromosome maintenance deficient




4 (S. cerevisiae)


MCM4
222037_at
MCM4 minichromosome maintenance deficient




4 (S. cerevisiae)


CASP8AP2
22201_s_at
CASP8 associated protein 2


MGC4692
222622_at
Hypothetical protein MGC4692


RAMP
222680_s_at
RA-regulated nuclear matrix-associated protein


FIGNL1
222843_at
fidgetin-like 1


SLC25A19
223222_at
solute carrier family 25 (mitochondrial




deoxynucleotide carrier) member 19


UBE2T
223229_at
ubiquitin-conjugating enzyme E2T (putative)


TCF19
223274_at
transcription factor 19 (SC1)


PDXP
223290_at
pyridoxal (pyridoxine, vitamin B6) phosphatase


POLR1B
223403_s_at
polymerase (RNA) I polypeptide B, 128 kDa


ANKRD32
223542_at
ankyrin repeat domain 32


IL17RB
224361_s_at
interleukin 17 receptor B/// interleukin 17




receptor B


CDCA7
224428_s_at
cell division cycle associated 7 /// cell division




cycle associated 7


MGC13096
224467_s_at
hypothetical protein MGC13096 /// hypothetical




protein MGC13096


CDCA5
224752_at
cell division cycle associated 5


TMEM18
225489_at
transmembrane protein 18


MGC20419
225641_at
hypothetical protein BC012173


UHRF1
225655_at
ubiquitin-like, containing PHD and RING finger




domains, 1



225716_at
Full-length cDNA clone CS0DK008Y109 of




HeLa cells Cot 25-normalized of Homo sapiens




(human)


MGC23280
226121_at
hypothetical protein MGC23280


C13orf8
226194_at
chromosome 13 open reading frame 8



226832_at
Hypothetical LOC389188


EGR1
227404_s_at
Early growth response 1


ZMYND19
227477_at
zinc finger, MYND domain containing 19


BARD1
227545_at
BRCA1 associated RING domain 1


KIAA1393
227653_at
KIAA1393


GPR27
227769_at
G protein-coupled receptor 27


RP13-15M17.2
228671_at
Novel protein


IL17D
228977_at
Interleukin 17D


JPH1
229139_at
junctophilin 1


ZNF367
229551_x_at
zinc finger protein 367


MGC35521
235431_s_at
pellino 3 alpha



239312_at
Transcribed locus


CSPG5
39966_at
chondroitin sulfate proteoglycan 5 (neuroglycan




C)
















TABLE 9







5-Flourouracil cell lines













Resistant or Sensitive



5-FU
Tissue of Origin
(Res or Sen)







MCF7
Breast
Sen



COLO 205
Colon
Sen



HCT-116
Colon
Sen



NCI-H460
Non-Small Cell Lung
Sen



LOX IMVI
Melanoma
Sen



SK-MEL-5
Melanoma
Sen



A498
Renal
Sen



UO-31
Renal
Sen



NCI/ADR-RES
Ovarian
Res



MDA-MB-435
Melanoma
Res



SW-620
Colon
Res



EKVX
Non-Small Cell Lung
Res



M14
Melanoma
Res



SN12C
Renal
Res



OVCAR-8
Ovarian
Res

















TABLE 10







Adriamycin cell lines













Resistant or Sensitive



Adriamycin
Tissue of Origin
(Res or Sen)







SF-539
CNS
Sen



SNB-75
CNS
Sen



MDA-MB-435
Melanoma
Sen



NCI-H23
Non-Small Cell Lung
Sen



M14
Melanoma
Sen



MALME-3M
Melanoma
Sen



SK-MEL-2
Melanoma
Sen



SK-MEL-28
Melanoma
Sen



SK-MEL-5
Melanoma
Sen



UACC-62
Melanoma
Sen



NCI/ADR-RES
Ovarian
Res



HCT-15
Colon
Res



HT29
Colon
Res



EKVX
Non-Small Cell Lung
Res



NCI-H322M
Non-Small Cell Lung
Res



IGROV1
Ovarian
Res



OVCAR-3
Ovarian
Res



OVCAR-4
Ovarian
Res



OVCAR-5
Ovarian
Res



OVCAR-8
Ovarian
Res



SK-OV-3
Ovarian
Res



CAKI-1
Renal
Res

















TABLE 11







Cytotoxan cell lines











Resistant or Sensitive


Cytotoxan
Tissue of Origin
(Res or Sen)





K-562
Leukemia
Sen


MOLT-4
Leukemia
Sen


HL-60(TB)
Leukemia
Sen


MCF7
Breast
Sen


HCC-2998
Colon
Sen


HCT-116
Colon
Sen


NCI-H460
Non-Small Cell Lung
Sen


TK-10
Renal
Sen


SNB-19
CNS
Res


HS 578T
Breast
Res


MDA-MB-231/A
Breast
Res


MDA-MB-435
Melanoma
Res


NCI-H226
Non-Small Cell Lung
Res


M14
Melanoma
Res


MALME-3M
Melanoma
Res


SK-MEL-2
Melanoma
Res
















TABLE 12







Taxotere (docetaxel) cell lines













Resistant or Sensitive



Taxotere
Tissue of Origin
(Res or Sen)







EKVX
Non-Small Cell Lung
Res



IGROV1
Ovarian
Res



OVCAR-4
Ovarian
Res



786-0
Renal
Res



CAKI-1
Renal
Res



SN12C
Renal
Res



TK-10
Renal
Res



HL-60(TB)
Leukemia
Sen



SF-539
CNS
Sen



HT29
Colon
Sen



HOP-62
Non-Small Cell Lung
Sen



SK-MEL-2
Melanoma
Sen



SK-MEL-5
Melanoma
Sen



NCI-H522
Non-Small Cell Lung
Sen

















TABLE 13







Etoposide cell lines











Resistant or Sensitive (Res


Etoposide
Tissue of Origin
or Sen)





SF-539
CNS
Sen


BT-549
Breast
Sen


MDA-MB-231
Breast
Sen


HCC-2998
Colon
Sen


HOP-62
Non-Small Cell Lung
Sen


NCI-H226
Non-Small Cell Lung
Sen


M14
Melanoma
Sen


PC-3
Prostate
Sen


786-0
Renal
Sen


MCF7
Breast
Res


NCI/ADR-RES
Ovarian
Res


HCT-15
Colon
Res


SW-620
Colon
Res


NCI-H322M
Non-Small Cell Lung
Res


UACC-257
Melanoma
Res


OVCAR-4
Ovarian
Res


OVCAR-5
Ovarian
Res
















TABLE 14







Taxol cell lines













Resistant or Sensitive



Taxol
Tissue of Origin
(Res or Sen)







SF-295
CNS
Sen



SF-539
CNS
Sen



HS 578T
Breast
Sen



MDA-MB-435
Melanoma
Sen



COLO 205
Colon
Sen



HCC-2998
Colon
Sen



HT29
Colon
Sen



OVCAR-3
Ovarian
Sen



NCI-H522
Non-Small Cell Lung
Sen



CCRF-CEM
Leukemia
Res



SW-620
Colon
Res



A549/ATCC
Non-Small Cell Lung
Res



EKVX
Non-Small Cell Lung
Res



MALME-3M
Melanoma
Res



SK-MEL-28
Melanoma
Res



OVCAR-8
Ovarian
Res



786-0
Renal
Res

















TABLE 15







Topotecan cell lines













Resistant or Sensitive



Topotecan
Tissue of Origin
(Res or Sen)







SF-539
CNS
Sen



SNB-75
CNS
Sen



U251
CNS
Sen



HS 578T
Breast
Sen



HOP-62
Non-Small Cell Lung
Sen



NCI-H226
Non-Small Cell Lung
Sen



NCI-H23
Non-Small Cell Lung
Sen



LOXIMVI
Melanoma
Sen



OVCAR-8
Ovarian
Sen



A498
Renal
Sen



ACHN
Renal
Sen



CAKI-1
Renal
Sen



UO-31
Renal
Sen



K-562
Leukemia
Res



RPMI-8226
Leukemia
Res



MDA-MB-435
Melanoma
Res



MDA-MB-231
Breast
Res



HCC-2998
Colon
Res



HCT-116
Colon
Res



HCT-15
Colon
Res



NCI-H322M
Non-Small Cell Lung
Res



SK-MEL-28
Melanoma
Res



COLO 205
Colon
Res

















TABLE 16







Validation of predictor sets in cell line and patient data sets











Genomic-based



Actual Overall
Prediction of Response


Tumor Data set/Response
Response
(i.e. PPV for Response)





Breast Tumor Data




MDACC
13/51 (25.4%)
11/13 (85.7%)


Adjuvant
33/45 (66.6%)
28/31 (90.3%)


Neoadjuvant Docetaxel
13/24 (54.1%)
11/13 (85.7%)


Ovarian


Topotecan
20/48 (41.6%)
17/22 (77.3%)


Paclitaxel
20/35 (57.1%)
20/28 (71.5%)


Docetaxel
7/14 (50%) 
 6/7 (85.7%)


Adriamycin (Evans et al.)
24/122 (19.6%) 
19/33 (57.5%)
















TABLE 17







Accuracy of predictions in cell lines and patients









Drugs














Validations
Topotecan
Adriamycin
Etoposide
5-Flourouracil
Paclitaxed
Cytoxan
Doceaxel





In Vitro Data









Accuracy
18/20 (90%)
18/25 (86%)
21/24
21/24
26/28 (92.8%)
25/29
P < 0.001**





(87%)
(87%)

(86.2%)



PPV
12/14 (86%)
 13/13 (100%)
6/8
14/14
21/21 (100%)
13/15






(75%)
(100%) 

(86.6%)



NPV
 6/6 (100%)
   5/8 (62.5%)
15/16
 7/10
 5/7 (71.5%)
12/14






(94%)
(70%)

  (86%)



In Vivo (Patient)









Data






Breast


Accuracy
40/48 (83.2%)
99/122 (81%) 


28/35 (80%)

22/24









(91.6%)


PPV
17/22 (77.34%)
  19/33 (57.6%)


20/28 (71.4%)

11/13









(85.7%)


NPV
23/26 (88.5%)
  80/89 (89.8%)


 7/7 (100%)

11/11









 (100%)









Ovarian









12/14









(85.7%)









6/7









(85.7%)









6/7









(85.7%)





PPV—positive predictive value,


NPV—negative predictive value.


**Determining accuracy for the docetaxel predictor in the IJC cellline data set was not possible since docetaxel was not one of the drugs studied. Instead, the docetaxel predictor was validated in two independent cell line experiments, correlating predicted probability of response to docetaxel in vitro with actual IC50 of docetaxel by cell line (FIG. 1C).













TABLE 18







Comparison of different predictors















Predictor of





Genomic predictor of
response to



Docetaxel
Docetaxel
response to TFAC
TFAC


Validations/
predictor
predictor (Chang
chemotherapy (Potti
chemotherapy


Predictors
(Potti et al.)
et al.)
et al.)
(Pusztai et al.)





Breast






neoadjuvant data


(Chang et al.)


Accuracy
22/24
87.5%  



(91.6%)


PPV
13/13
92%



(85.7%)


NPV of
11/11
83%


ROC
 (100%)



0.97
0.96


MDACC data


(Pusztai et al.)


Accuracy


42/51 (82.3%)
74%


PPV


11/18 (61.1%)
44%


NPV


31/33 (94%)  
93%





PPV—positive predictive value,


NPV—negative predictive value.


**For both the Chang and Pusztai data, the actual numbers of predicted responders was not available, just the predictive accuracies. Also, the predictive accuracy reported for the Chang data is not in an independent validation, instead it is for leave-one cross out validation.





Claims
  • 1. A method for predicting responsiveness of a cancer to a chemotherapeutic agent comprising: a) comparing a first gene expression profile of the cancer to a chemotherapy responsivity predictor set of gene expression profiles, the first gene expression profile and the chemotherapy responsivity predictor set each comprising at least five genes from one of Tables 1-8, wherein Tables 1-8 comprise the chemotherapy responsivity predictor set for 5-fluorouracil, adriamycin, cytotoxan, docetaxol, etoposide, taxol, topotecan and PI3 kinase inhibitors, respectively; andb) using the comparison of step (a) to predict the responsiveness of the cancer to the chemotherapeutic agent.
  • 2. The method of claim 1, wherein the chemotherapeutic agent is an inhibitor of the PI3kinase pathway and the first gene expression profile and the chemotherapy responsivity predictor set each comprise at least five genes from Table 4.
  • 3. The method of claim 1, wherein the chemotherapeutic agent is an inhibitor of the Src pathway and the first gene expression profile and the chemotherapy responsivity predictor set each comprise at least five genes from Table 7.
  • 4. The method of claim 1, wherein the first gene expression profile is obtained by analyzing a nucleic acid sample from the cancer.
  • 5. The method of claim 1, wherein the first gene expression profile is obtained by analyzing a sample from a tumor or ascites.
  • 6. The method of claim 1, wherein the first gene expression profile is determined using a nucleic acid microarray.
  • 7. The method of claim 1, wherein the first gene expression profile and the chemotherapy responsivity predictor set each comprises at least 10 genes.
  • 8. The method of claim 1, wherein the first gene expression profile and the chemotherapy responsivity predictor set each comprises at least 20 genes.
  • 9. The method of claim 1, wherein the cancer is from an individual and wherein step (b) identifies the individual as a complete responder or as an incomplete responder to the chemotherapeutic agent.
  • 10. The method of claim 1, wherein the first gene expression profile is compared to at least two chemotherapy responsivity predictor sets each comprising at least five genes from the corresponding Tables 1-8.
  • 11. The method of claim 1, wherein the cancer is selected from the group consisting of lung, breast, ovarian, prostrate, renal, colon, leukemia, skin, and brain cancer.
  • 12. The method of claim 1, wherein the chemotherapy responsivity predictor set is defined by extracting a single dominant value using singular value decomposition (SVD) and determining the value of the chemotherapy responsivity predictor set in the cancer.
  • 13. The method of claim 1, wherein step (b) comprises applying one or more statistical models to the comparison of step (a), each model producing a statistical probability of the sensitivity of the cancer to the chemotherapeutic agent.
  • 14. The method of claim 13, wherein the statistical model is a binary regression model.
  • 15. The method of claim 13, wherein the statistical model is a tree model, the tree model including one or more nodes, each node representing a metagene, each node including a statistical probability of sensitivity of the cancer to the chemotherapeutic agent.
  • 16. The method of claim 1, wherein the method predicts responsiveness to the chemotherapeutic agent with at least 80% accuracy.
  • 17. The method of claim 1, wherein the chemotherapy responsivity predictor set is developed using at least one resistant cell line and at least one sensitive cell line of one of Tables 9-15, Tables 9-15 listing cell lines sensitive or resistant to 5-fluorouracil, adriamycin, cytotoxan, docetaxol, etoposide, taxol, and topotecan, respectively.
  • 18. A method of developing a treatment plan for an individual with cancer comprising using the predicted responsivity of a cancer to a chemotherapeutic agent obtained by the method of claim 1 to develop a treatment plan.
  • 19. The method of claim 18, wherein the treatment plan includes administering an effective amount of a chemotherapeutic agent to the individual with the cancer if the cancer is predicted to respond to the chemotherapeutic agent.
  • 20. The method of claim 18, further comprising comparing the first gene expression profile to an alternative chemotherapy responsivity predictor set of gene expression profiles predictive of responsivity to alternative chemotherapeutic agents; predicting responsiveness of the cancer to the alternative chemotherapeutic agents and administering an alternative chemotherapeutic agent to the individual with the cancer.
  • 21. The method of claim 20, wherein the alternative chemotherapeutic agent is selected from the group comprising docetaxel, paclitaxel, abraxane, topotecan, adriamycin, etoposide, fluorouracil (5-FU), cyclophosphamide, denopterin, edatrexate, methotrexate, nolatrexed, pemetrexed, piritrexim, pteropterin, raltitrexed, trimetrexate, cladribine, ctofarabine, fludarabine, 6-mercaptopurine, nelarabine, thiamiprine, thioguanine, tiazofurin, ancitabine, azacibdine, 6-azauridine, capecitabine, carmofur, cytarabine, decitabine, doxifluridine, enocitabine, floxuridine, fluorouracil, gemcitabine, tegafur, troxacitabine, pentostatin, hydroxyurea, cytosine arabinoside.
  • 22. The method of claim 18, wherein the plan includes administering the chemotherapeutic agent before, after or concurrently with the administration of one or more alternative chemotherapeutic agents.
  • 23. The method of claim 18, wherein the alternative chemotherapeutic agent targets a signal transduction pathway.
  • 24. The method of claim 23, wherein the first gene expression profile of the cancer comprises at least one gene expression profile indicative of deregulation of the signal transduction pathway.
  • 25. The method of claim 23, wherein the alternative chemotherapeutic agent is selected from inhibitors of a signal transduction pathway selected from the group consisting of Src, E2F3, Myc, PI3kinase and β-catenin.
  • 26. The method of claim 18, wherein the cancer is predicted to be responsive to more than one chemotherapeutic agent.
  • 27. The method of claim 26, wherein the treatment plan administering an effective amount of at least two chemotherapeutic agents to the individual with the cancer.
  • 28. The method of claim 27, wherein the plan includes administering at least two chemotherapeutic agents before, after or concurrently with each other.
  • 29. The method of claim 18, wherein the treatment plan has an estimated efficacy of at least 50%.
  • 30. A kit comprising a gene chip for predicting responsivity of a cancer to a chemotherapeutic agent comprising nucleic acids capable of detecting at least five genes selected from Tables 1-8 and instructions for predicting responsivity of a cancer to the chemotherapeutic agent.
  • 31. A computer readable medium comprising gene expression profiles and corresponding responsivity information for chemotherapeutic agents comprising at least five genes from any of Tables 1-8.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority to U.S. Utility application Ser. No. 11/975,722, filed Oct. 19, 2007, which is incorporated herein by reference in its entirety.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT

This invention was made with government support under NCI-U54 CA112952-02 and ROI-CA106520 awarded by the National Cancer Institute. The government has certain rights in the invention.

PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US08/80481 10/20/2008 WO 00 7/16/2010
Continuations (1)
Number Date Country
Parent 11975722 Oct 2007 US
Child 12738470 US