Methods and Systems for Diagnosis, Prognosis and Selection of Treatment of Leukemia

Information

  • Patent Application
  • 20080280774
  • Publication Number
    20080280774
  • Date Filed
    February 16, 2006
    18 years ago
  • Date Published
    November 13, 2008
    15 years ago
Abstract
The present invention provides methods, systems and equipment for the prognosis, diagnosis and selection of treatment of AML or other types of leukemia. Genes prognostic of clinical outcome of leukemia patients can be identified according to the present invention. Leukemia disease genes can also be identified according to the present invention. These genes are differentially expressed in PBMCs of AML patients relative to disease-free humans. These genes can be used for the diagnosis or monitoring the development, progression or treatment of AML.
Description
TECHNICAL FIELD

The present invention relates to leukemia diagnostic and prognostic genes and methods of using the same for the diagnosis, prognosis, and selection of treatment of AML or other types of leukemia.


BACKGROUND

Acute myeloid leukemia (AML) is a heterogeneous clonal disorder typified by hyperproliferation of immature leukemic blast cells in the bone marrow. Approximately 90% of all AML cases exhibit proliferation of CD33+ blast cells, and CD33 is a cell surface antigen that appears to be specifically expressed in myeloblasts and myeloid progenitors but is absent from normal hematopoetic stem cells. Gemtuzumab ozogamicin (Mylotarg® or GO) is an anti-CD33 antibody conjugated to calicheamicin specifically designed to target CD33+ blast cells of AML patients for destruction. For reviews, see Matthews, LEUKEMIA, 12(Suppl 1):S33-S36 (1998); and Bernstein, LEUKEMIA, 14:474-475 (2000).


While gemtuzumab ozogamicin has demonstrated efficacy in patients with advanced AML, it is sometimes not completely effective as a single line agent. Both in vitro and in vivo studies have demonstrated that p-glycoprotein expression and the multi-drug resistance (MDR) phenotype are associated with reduced responsiveness to gemtuzumab ozogamicin therapy, suggesting that extrusion of gemtuzumab ozogamicin by this mechanism may be one of several important molecular pathways of gemtuzumab ozogamicin resistance (Naito, et al., LEUKEMIA, 14:1436-1443 (2000); and Linenberger, et al., BLOOD, 98:988-994 (2001)). However, the MDR phenotype fails to account for all cases found to be gemtuzumab ozogamicin resistant. While gemtuzumab ozogamicin exhibits a favorable safety profile in the majority of patients receiving Mylotarg® therapy (Sievers, et al., J CLIN. ONCOL., 19(13):3244-3254 (2001)), a small but significant number of cases of hepatic veno-occlusive disease have been reported following exposure to this therapy (Neumeister, et al., ANN. HEMATOL., 80:119-120 (2001)). Recently, GO has also been evaluated in combination with an anthracycline and cytarabine in an attempt to increase the effectiveness of GO administered as a single agent therapy (Alvarado, et al., CANCER CHEMOTHER PHARMACOL., 51:87-90 (2003)).


SUMMARY OF THE INVENTION

It is therefore an object of the present invention to provide effective pharmacogenomic analysis to assess any relationship between gene expression and response to therapy.


It is an object of the present invention to identify leukemia prognostic genes whose expression levels are predictive of clinical outcome of leukemia patients who undergo an anti-cancer therapy.


It is a further object of the present invention to provide a method for predicting a clinical outcome of a leukemia patient as well as a method for selecting a treatment for a leukemia patient based on pharmacogenomic analysis.


It is another object of the present invention to identify leukemia diagnostic genes and to provide a method for diagnosis, or monitoring the occurrence, development, progression or treatment, of a leukemia based on the analysis of the expression levels of the diagnostic genes.


Thus, in one aspect, the present invention provides a method for predicting a clinical outcome in response to a treatment of a leukemia. The method includes the following steps: (1) measuring expression levels of one or more prognostic genes of the leukemia in a peripheral blood mononuclear cell sample derived from a patient prior to the treatment; and (2) comparing each of the expression levels to a corresponding control level, wherein the result of the comparison is predictive of a clinical outcome. “Prognostic genes” referred to in the application include, but are not limited to, any genes that are differentially expressed in peripheral blood mononuclear cells (PBMCs) or other tissues of leukemia patients with different clinical outcomes. In particular, prognostic genes include genes whose expression levels in PBMCs or other tissues of leukemia patients are correlated with clinical outcomes of the patients. Exemplary prognostic genes are shown in Table 1, Table 2, Table 3, Table 4, Table 5 and Table 6. A “clinical outcome” referred to in the application includes, but is not limited to, any response to any leukemia treatment.


The present invention is suitable for prognosis of any leukemias, including acute leukemia, chronic leukemia, lymphocytic leukemia or nonlymphocytic leukemia. In particular, the present invention is suitable for prognosis of acute myeloid leukemia (AML). Typically, the clinical outcome is measured by a response to an anti-cancer therapy. For example, the anti-cancer therapy includes administering one or more compounds selected from the group consisting of an anti-CD33 antibody, a daunorubicin, a cytarabine, a gemtuzumab ozogamicin, an anthracycline, and a pyrimidine or purine nucleotide analog. In one particular example, the present invention may be used to predict a response to a gemtuzumab ozogamicin (GO) combination therapy.


In one embodiment, the one or more prognostic genes suitable for the invention include at least a first gene selected from a first class and a second gene selected from a second class. The first class includes genes having higher expression levels in peripheral blood mononuclear cells in patients predicted to have a less desirable clinical outcome in response to the treatment. Exemplary first class genes are shown in Table 1 and Table 3. The second class includes genes having higher expression levels in peripheral blood mononuclear cells in patients predicted to have a more desirable clinical outcome in response to the treatment. Exemplary second class genes are shown in Table 2 and 4. In one embodiment, the first gene is selected from Table 3 and the second gene is selected from Table 4.


In one particular embodiment, the first gene is selected from the group consisting of zinc finger protein 217, peptide transporter 3, forkhead box O3A, T cell receptor alpha locus and putative chemokine receptor/GTP-binding protein, and the second gene is selected from the group consisting of metallothionein, fatty acid desaturase 1, an uncharacterized gene corresponding to Affymetrix ID 216336, deformed epidermal autoregulatory factor 1 and growth arrest and DNA-damage-inducible alpha. In another embodiment, the first gene is serum glucocorticoid regulated kinase and the second gene is metallothionein 1X/1L.


In some embodiments, each of the expression levels of the prognostic genes is compared to the corresponding control level which is a numerical threshold.


In some embodiments, the method of the present invention may be used to predict development of an adverse event in a leukemia patient in response to a treatment. For example, the method may be used to assess the possibility of development of veno-occlusive disease (VOD). Exemplary prognostic genes predictive of VOD are shown in Table 5 and Table 6. In one particular embodiment, the expression level of p-selectin ligand is measured to predict the risk for VOD.


In another aspect, the present invention provides a method for predicting a clinical outcome of a leukemia by taking the following steps: (1) generating a gene expression profile from a peripheral blood sample of a patient having the leukemia; and (2) comparing the gene expression profile to one or more reference expression profiles, wherein the gene expression profile and the one or more reference expression profiles contain expression patterns of one or more prognostic genes of the leukemia in peripheral blood mononuclear cells, and wherein the difference or similarity between the gene expression profile and the one or more reference expression profiles is indicative of the clinical outcome for the patient.


In one embodiment, the gene expression profile of the one or more prognostic genes may be compared to the one or more reference expression profiles by, for example, a k-nearest neighbor analysis or a weighted voting algorithm. Typically, the one or more reference expression profiles represent known or determinable clinical outcomes. In some embodiments, the gene expression profile from the patient may be compared to at least two reference expression profiles, each of which represents a different clinical outcome. For example, each reference expression profile may represent a different clinical outcome selected from the group consisting of remission to less than 5% blasts in response to the anti-cancer therapy; remission to no less than 5% blasts in response to the anti-cancer therapy; and non-remission in response to the anti-cancer therapy. In some embodiments, the one or more reference expression profiles may include a reference expression profile representing a leukemia-free human.


In some embodiments, the gene expression profile may be generated by using a nucleic acid array. Typically, the gene expression profile is generated from the peripheral blood sample of the patient prior to the anti-cancer therapy.


In one embodiment, the one or more prognostic genes include one or more genes selected from Table 3 or Table 4. In another embodiment, the one or more prognostic genes include ten or more genes selected from Table 3 or Table 4. In yet another embodiment, the one or more prognostic genes include twenty or more genes selected from Table 3 or Table 4.


In yet another aspect, the present invention provides a method for selecting a treatment for a leukemia patient. The method includes the following steps: (1) generating a gene expression profile from a peripheral blood sample derived from the leukemia patient; (2) comparing the gene expression profile to a plurality of reference expression profiles, each representing a clinical outcome in response to one of a plurality of treatments; and (3) selecting from the plurality of treatments a treatment which has a favorable clinical outcome for the leukemia patient based on the comparison in step (2), wherein the gene expression profile and the one or more reference expression profiles comprise expression patterns of one or more prognostic genes of the leukemia in peripheral blood mononuclear cells. In one embodiment, the gene expression profile may be compared to the plurality of reference expression profiles by, for example, a k-nearest neighbor analysis or a weighted voting algorithm.


In one embodiment, the one or more prognostic genes include one or more genes selected from Table 3 or Table 4. In another embodiment, the one or more prognostic genes include ten or more genes selected from Table 3 or Table 4. In yet another embodiment, the one or more prognostic genes include twenty or more genes selected from Table 3 or Table 4.


In another aspect, the present invention provides a method for diagnosis, or monitoring the occurrence, development, progression or treatment, of a leukemia. The method includes the following steps: (1) generating a gene expression profile from a peripheral blood sample of a patient having the leukemia; and (2) comparing the gene expression profile to one or more reference expression profiles, wherein the gene expression profile and the one or more reference expression profiles contain the expression patterns of one or more diagnostic genes of the leukemia in peripheral blood mononuclear cells, and wherein the difference or similarity between the gene expression profile and the one or more reference expression profiles is indicative of the presence, absence, occurrence, development, progression, or effectiveness of treatment of the leukemia in the patient. In one embodiment, the leukemia is AML. “Diagnostic genes” referred to in the application include, but are not limited to, any genes that are differentially expressed in peripheral blood mononuclear cells (PBMCs) or other tissues of leukemia patients with different disease status. In particular, diagnostic genes include genes that are differentially expressed in PBMCs or other tissues of leukemia patients relative to PBMCs of leukemia-fee patients. Exemplary diagnostic genes are shown in Table 7, Table 8 and Table 9. Diagonistic genes are also referred to as disease genes in this application.


Typically, the one or more reference expression profiles include a reference expression profile representing a disease-free human. Typically, the one or more diagnostic genes include one or more genes selected from Table 7. Preferably, the one or more diagnostic genes comprise one or more genes selected from Table 8 or Table 9. In some embodiments, the one or more diagnostic genes include ten or more genes selected from Table 7. Preferably, the one or more diagnostic genes include ten or more genes selected from Table 8 or Table 9.


In another aspect, the present invention provides an array for use in a method for predicting a clinical outcome for an AML patient. The array of the invention includes a substrate having a plurality of addresses, each of which has a distinct probe disposed thereon. In some embodiments, at least 15% of the plurality of addresses have disposed thereon probes that can specifically detect prognostic genes of AML in peripheral blood mononuclear cells. In some embodiments, at least 30% of the plurality of addresses have disposed thereon probes that can specifically detect prognostic genes of AML in peripheral blood mononuclear cells. In some embodiments, at least 50% of the plurality of addresses have disposed thereon probes that can specifically detect prognostic genes of AML in peripheral blood mononuclear cells. In some embodiments, the prognostic genes are selected from Table 1, Table 2, Table 3, Table 4, Table 5 or Table 6. The probe suitable for the present invention may be a nucleic acid probe. Alternatively, the probe suitable for the invention may be an antibody probe.


In a further aspect, the present invention provides an array for use in a method for diagnosis of AML including a substrate having a plurality of addresses, each of which has a distinct probe disposed thereon. In some embodiments, at least 15% of the plurality of addresses have disposed thereon probes that can specifically detect diagnostic genes of AML in peripheral blood mononuclear cells. In some embodiments, at least 30% of the plurality of addresses have disposed thereon probes that can specifically detect diagnostic genes of AML in peripheral blood mononuclear cells. In some embodiments, at least 50% of the plurality of addresses have disposed thereon probes that can specifically detect diagnostic genes of AML in peripheral blood mononuclear cells. In some embodiments, the diagnostic genes are selected from Table 7, Table 8 or Table 9. The probe suitable for the present invention may be a nucleic acid probe. Alternatively, the probe suitable for the present invention may be an antibody probe.


In yet another aspect, the present invention provides a computer-readable medium containing a digitally-encoded expression profile having a plurality of digitally-encoded expression signals, each of which includes a value representing the expression of a prognostic gene of AML in a peripheral blood mononuclear cell. In some embodiments, each of the plurality of digitally-encoded expression signals has a value representing a prognostic gene selected from Table 1, Table 2, Table 3, Table 4, Table 5 or Table 6. In some embodiments, each of the plurality of digitally-encoded expression signals has a value representing the expression of the prognostic gene of AML in a peripheral blood mononuclear cell of a patient with a known or determinable clinical outcome. In some embodiments, the computer-readable medium of the present invention contains a digitally-encoded expression profile including at least ten digitally-encoded expression signals.


In another aspect, the present invention provides a computer-readable medium containing a digitally-encoded expression profile having a plurality of digitally-encoded expression signals, each of which has a value representing the expression of a diagnostic gene of AML in a peripheral blood mononuclear cell. In some embodiments, each of the plurality of digitally-encoded expression signals has a value representing a diagnostic gene selected from Table 7, Table 8 or Table 9. In some embodiments, each of the plurality of digitally-encoded expression signals has a value representing the expression of the diagnostic gene of AML in a peripheral blood mononuclear cell of an AML-free human. In some embodiments, the computer-readable medium of the present invention contains a digitally-encoded expression profile including at least ten digitally-encoded expression signals.


In yet another aspect, the present invention provides a kit for prognosis of a leukemia, e.g., AML. The kit includes a) one or more probes that can specifically detect prognostic genes of AML in peripheral blood mononuclear cells; and b) one or more controls, each representing a reference expression level of a prognostic gene detectable by the one or more probes. In some embodiments, the kit of the present invention includes one or more probes that can specifically detect prognostic genes selected from Table 1, Table 2, Table 3, Table 4, Table 5 or Table 6.


In another aspect, the present invention provides a kit for diagnosis of a leukemia, e.g., AML. The kit includes a) one or more probes that can specifically detect diagnostic genes of AML in peripheral blood mononuclear cells; and b) one or more controls, each representing a reference expression level of a prognostic gene detectable by the one or more probes. In some embodiments, the kit of the present invention includes one or more probes that can specifically detect diagnostic genes selected from Table 7, Table 8 or Table 9.


Other features, objects, and advantages of the present invention are apparent in the detailed description that follows. It should be understood, however, that the detailed description, while indicating embodiments of the present invention, is given by way of illustration only, not limitation. Various changes and modifications within the scope of the invention will become apparent to those skilled in the art from the detailed description.





BRIEF DESCRIPTION OF THE DRAWINGS

The drawings are provided for illustration, not limitation.



FIG. 1A demonstrates relative PBMC expression levels of 98 class correlated genes selected from Tables 1 and 2. Among the 98 genes, 49 genes had elevated expression levels in PBMCs of patients who responded to Mylotarg combination therapy (R) relative to patients who did not respond to the therapy (NR), and the other 49 genes had elevated expression levels in PBMCs of the non-responding patients (NR) compared to the responding patients (R).



FIG. 1B shows cross validation results for each sample using a 154-gene class predictor consisting of the genes in Tables 1 and 2, where a leave-one out cross validation was performed and the prediction strengths were calculated for each sample. Samples are ordered in the same order as in FIG. 1A.



FIG. 2 illustrates an unsupervised hierarchical clustering of PBMC gene expression profiles from normal patients, patients with AML, or patients with MDS using the 7879 transcripts detected in one or more profiles with a maximal frequency greater than or equal to 10 ppm. Data were log transformed and gene expression values were median centered, and profiles were clustered using an average linkage clustering approach with an uncentered correlation similarity metric. The two main clusters of normal and non-normal are denoted as clusters 1 and 2. The subgroup in cluster 2 possessing a preponderance of AML is indicated as “AML-like” while the subgroup in cluster 2 possessing a preponderance of MDS is indicated as “MDS-like.”



FIG. 3 illustrates a gene ontology based annotation of transcripts altered during GO combination therapy of AML patients. The 52 transcripts exhibiting 3-fold or greater repression over treatment were annotated into each of the twelve categories listed. Transcripts in the immune response category were most significantly overrepresented in the group of transcripts elevated over therapy, while uncategorized transcripts were most significantly overrepresented in the group of transcripts repressed during therapy.



FIG. 4 illustrates levels of p-selectin ligand transcript in the pretreatment PBMCs of 4 AML patients who eventually experienced veno-occlusive disease (VOD) (left panel) and in pretreatment PBMCs of 32 patients who did not experience VOD (right panel). Frequency (in ppm) based on microarray analysis is plotted on the y-axis and the level of p-selectin ligand in each individual sample in each group is plotted as a discrete symbol.



FIG. 5 illustrates levels of MDR1 transcript in pretreatment PBMCs of 8 AML patients who failed to respond (NR) and in pretreatment PBMCs of 28 patients who responded (R). Frequency (in ppm) based on microarray analysis is plotted on the y-axis and the level of MDR1 transcript in each individual of the 36 pretreatment PBMC samples is indicated by each column. The p-value is based on an unpaired Student's t-test assuming unequal variances.



FIG. 6 illustrates the transcript levels of various ABC cassette transporters in PBMC samples of AML patients prior to therapy. Frequency (in ppm) based on microarray analysis is plotted on the y-axis and the average level plus standard deviation of each transporter in the NR and R groups is indicated. No significant differences in expression between NR and R were detected for any of the sequences encoding ABC transporters evaluated on U133A.



FIG. 7 illustrates levels of CD33 cell surface antigen transcript in pretreatment PBMCs of 8 patients who failed to respond (NR) and in pretreatment PBMCs of 28 patients who responded (R). Frequency (in ppm) based on microarray analysis is plotted on the y-axis and the level of CD33 transcript in each individual of the 36 pretreatment PBMC samples is indicated by each column. The p-value is based on an unpaired Student's t-test assuming unequal variances.



FIG. 8 illustrates the accuracy of a 10-gene classifier for distinguishing pretreatment PBMCs from eventual responders and eventual nonresponders to therapy. Data from baseline PBMC profiles from AML patients were scale-frequency normalized together using a total of 11382 sequences possessing at least one present call and one value of greater than or equal to 10 ppm across baseline profiles from each of two independent clinical studies involving GO-based therapy. Analyses were conducted following a z-score normalization step in Genecluster. Panel A depicts overall accuracy in a 36 member training set for models containing increasing numbers of features (transcript sequences) built using a binary classification approach with a S2N similarity metric that used median values for the class estimate. The smallest classifier (10-gene) yielding the highest overall accuracy is indicated (arrow). Panel B depicts ten-fold cross validation accuracy of the 10-gene classifier. A weighted voting algorithm was used to assign class membership using the 10-gene classifier. Confidence scores for each prediction call are indicated by columns where a downward deflection indicates a call of “NR” and an upward deflection indicates a call of “R.” True non-responders are indicated by light columns and true responders are indicated by dark columns. In this cross-validation 4/8 non-responders were correctly identified and 24/28 responders were correctly identified.



FIG. 9 illustrates the use of the 10-gene classifier to evaluate baseline PBMCs from AML patients from an independent clinical trial. The weighted voting algorithm was used to assign class membership using the 10-gene classifier. Confidence scores for each prediction call are indicated by columns where a downward deflection indicates a call of “NR” and an upward deflection indicates a call of “R.” True non-responders are indicated by light columns and true responders are indicated by dark columns. In this independent test set, 4/7 non-responders were correctly identified and 7/7 responders were correctly identified.



FIG. 10 illustrates expression levels of two genes in AML PBMCs inversely correlated with response to GO-based therapies. Panel A represents a two-dimensional plot of Affymetrix-based expression levels (in ppm) of serum/glucocorticoid regulated kinase (Y-axes) and metallothionein 1X, 1L (X-axes) in PMBC samples from AML patients. Levels of each transcript in each patient are plotted where non-responders are indicated by squares and responders are indicated by circles. The shadow indicates the area of the X-Y plot encompassing the largest number of non-responders and the smallest number of responders, defining the boundaries for this pairwise classifier. Implementing requirements for expression levels of less than 30 ppm for serum glucocorticoid regulated kinase and expression levels of greater than 30 ppm for metallothionein 1X, 1L, would have successfully identified 6/8 non-responders and only falsely identified 2 of 28 responders as non-responders in the original dataset of 36 samples. Panel B illustrates an evaluation of the 2-gene classifier in 14 AML samples from an independent clinical trial. Implementation of the same requirements correctly identified 4/7 non-responders and all responders (7/7) were also correctly identified.





DETAILED DESCRIPTION

The present invention provides methods, reagents and systems useful for prognosis or selection of treatment of AML or other types of leukemia. These methods, reagents and systems employ leukemia prognostic genes which are differentially expressed in peripheral blood samples of leukemia patients who have different clinical outcomes. The present invention also provides methods, reagents and systems for diagnosis, or monitoring the occurrence, development, progression or treatment, of AML or other types of leukemia. These methods, reagents and systems employ diagnostic genes which are differentially expressed in peripheral blood samples of leukemia patients with different disease status. Thus, the present invention represents a significant advance in clinical pharmacogenomics and leukemia treatment.


Various aspects of the invention are described in further detail in the following subsections. The use of subsections is not meant to limit the invention. Each subsection may apply to any aspect of the invention. In this application, the use of “or” means “and/or” unless stated otherwise.


Leukemia and Leukemia Treatment

The types of leukemia that are amenable to the present invention include, but are not limited to, acute leukemia, chronic leukemia, lymphocytic leukemia, or nonlymphocytic leukemia (e.g., myelogenous, monocytic, or erythroid). Acute leukemia includes, for example, AML or ALL (acute lymphoblastic leukemia). Chronic leukemia includes, for example, CML (chronic myelogenous leukemia), CLL (chronic lymphocytic leukemia), or hairy cell leukemia. The present invention also contemplates genes that are prognostic of clinical outcome of patients having myelodysplastic syndromes (MDS).


Any leukemia treatment regime can be analyzed according to the present invention. Examples of these leukemia treatments include, but are not limited to, chemotherapy, drug therapy, gene therapy, immunotherapy, biological therapy, radiation therapy, bone marrow transplantation, surgery, or a combination thereof. Other conventional, non-conventional, novel or experimental therapies, including treatments under clinical trials, can also be evaluated according to the present invention.


A variety of anti-cancer agents can be used to treat leukemia. Examples of these agents include, but are not limited to, alkylators, anthracyclines, antibiotics, biphosphonates, folate antagonists, inorganic arsenates, microtubule inhibitors, nitrosoureas, nucleoside analogs, retinoids, or topoisomerase inhibitors.


Examples of alkylators include, but are not limited to, busulfan (Myleran, Busulfex), chlorambucil (Leukeran), cyclophosphamide (Cytoxan, Neosar), melphalan, L-PAM (Alkeran), dacarbazine (DTIC-Dome), and temozolamide (Temodar). Examples of anthracyclines include, but are not limited to, doxorubicin (Adriamycin, Doxil, Rubex), mitoxantrone (Novantrone), idarubicin (Idamycin), valrubicin (Valstar), and epirubicin (Ellence). Examples of antibiotics include, but are not limited to, dactinomycin, actinomycin D (Cosmegen), bleomycin (Blenoxane), and daunorubicin, daunomycin (Cerubidine, DanuoXome). Examples of biphosphonate inhibitors include, but are not limited to, zoledronate (Zometa). Examples of folate antagonists include, but are not limited to, methotrexate and tremetrexate. Examples of inorganic arsenates include, but are not limited to, arsenic trioxide (Trisenox). Examples of microtubule inhibitors, which may inhibit either microtubule assembly or disassembly, include, but are not limited to, vincristine (Oncovin), vinblastine (Velban), paclitaxel (Taxol, Paxene), vinorelbine (Navelbine), docetaxel (Taxotere), epothilone B or D or a derivative of either, and discodermolide or its derivatives. Examples of nitrosoureas include, but are not limited to, procarbazine (Matulane), lomustine, CCNU (CeeBU), carmustine (BCNU, BiCNU, Gliadel Wafer), and estramustine (Emcyt). Examples of nucleoside analogs include, but are not limited to, mercaptopurine, 6-MP (Purinethol), fluorouracil, 5-FU (Adrucil), thioguanine, 6-TG (Thioguanine), hydroxyurea (Hydrea), cytarabine (Cytosar-U, DepoCyt), floxuridine (FUDR), fludarabine (Fludara), pentostatin (Nipent), cladribine (Leustatin, 2-CdA), gemcitabine (Gemzar), and capecitabine (Xeloda). Examples of retinoids include, but are not limited to, tretinoin, ATRA (Vesanoid), alitretinoin (Panretin), and bexarotene (Targretin). Examples of topoisomerase inhibitors include, but are not limited to, etoposide, VP-16 (Vepesid), teniposide, VM-26 (Vumon), etoposide phosphate (Etopophos), topotecan (Hycamtin), and irinotecan (Camptostar). Therapies including the use of any of these anti-cancer agents can be evaluated according to the present invention.


Leukemia can also be treated by antibodies that specifically recognize diseased or otherwise unwanted cells. Antibodies suitable for this purpose include, but are not limited to, polyclonal, monoclonal, mono-specific, poly-specific, humanized, human, single-chain, chimeric, synthetic, recombinant, hybrid, mutated, grafted, or in vitro generated antibodies. Suitable antibodies can also be Fab, F(ab′)2, Fv, scFv, Fd, dAb, or other antibody fragments that retain the antigen-binding function. In many cases, an antibody employed in the present invention can bind to a specific antigen on the diseased or unwanted cells (e.g., the CD33 antigen on myeloblasts or myeloid progenitor cells) with a binding affinity of at least 10−6 M−1, 10−7 M−1, 10−8 M−1, 10−9 M−1, or stronger.


Many antibodies employed in the present invention are conjugated with a cytotoxic or otherwise anticellular agent which can kill or suppress the growth or division of cells. Examples of cytotoxic or anticellular agents include, but are not limited to, the anti-neoplastic agents described above, and other chemotherapeutic agents, radioisotopes or cytotoxins. Two or more different cytotoxic moieties can be coupled to one antibody, thereby accommodating variable or even enhanced anti-cancer activities.


Linking or coupling one or more cytotoxic moieties to an antibody may be achieved by a variety of mechanisms, for example, covalent binding, affinity binding, intercalation, coordinate binding and complexation. Preferred binding methods are those involving covalent binding, such as using chemical cross-linkers, natural peptides or disulfide bonds.


Covalent binding can be achieved, for example, by direct condensation of existing side chains or by the incorporation of external bridging molecules. Many bivalent or polyvalent agents are useful in coupling protein molecules to other proteins, peptides or amine functions. Examples of coupling agents are, without limitation, carbodiimides, diisocyanates, glutaraldehyde, diazobenzenes, and hexamethylene diamines.


In one embodiment, an antibody employed in the present invention is first derivatized before being attaching with a cytotoxic moiety. “Derivatize” means chemical modification(s) of the antibody substrate with a suitable cross-linking agent. Examples of cross-linking agents for use in this manner include the disulfide-bond containing linkers SPDP (N-succinimidyl-3-(2-pyridyldithio)propionate) and SMPT (4-succinimidyl-oxycarbonyl-α-methyl-α(2-pyridyldithio)toluene). Biologically releasable bonds can also be used to construct a clinically active antibody, such that a cytotoxic moiety can be released from the antibody once it binds to or enters the target cell. Numerous types of linking constructs are known for this purpose (e.g., disulfide linkages).


Anti-neoplastic agent(s) employed in a leukemia treatment regime can be administered via any common route so long as the target tissue or cell is available via that route. This includes, but is not limited to, intravenous, catheterization, orthotopic, intradermal, subcutaneous, intramuscular, intraperitoneal intrtumoral, oral, nasal, buccal, rectal, vaginal, or topical administration. Selection of anti-neoplastic agents and dosage regimes may depend on various factors, such as the drug combination employed, the particular disease being treated, and the condition and prior history of the patient. Specific dose regimens for known and approved anti-neoplastic agents can be found in the current version of Physician's Desk Reference, Medical Economics Company, Inc., Oradell, N.J.


In addition, a leukemia treatment regime can include a combination of different types of therapies, such as chemotherapy plus antibody therapy. The present invention contemplates identification of prognostic genes for all types of leukemia treatment regime.


In one aspect, the present invention features identification of genes that are prognostic of clinical outcome of AML patients who undergo an anti-cancer treatment. An AML treatment can include a remission induction therapy, a postremission therapy, or a combination thereof. The purpose of the remission induction therapy is to attain remission by killing the leukemia cells in the blood or bone marrow. The purpose of the postremission therapy is to maintain remission by killing any remaining leukemia cells that may not be active but could begin to regrow and cause a relapse.


Standard remission induction therapies for AML patients include, but are not limited to, combination chemotherapy, stem cell transplantation, high-dose combination chemotherapy, all-trans retinoic acid (ATRA) plus chemotherapy, or intrathecal chemotherapy. Standard postremission therapies include, but are not limited to, combination chemotherapy, high-dose chemotherapy and stem cell transplantation using donor stem cells, or high-dose chemotherapy and stem cell transplantation using the patient's stem cells with or without radiation therapy. For recurrent AML patients, standard treatments include, but are not limited to, combination chemotherapy, biologic therapy with monoclonal antibodies, stem cell transplantation, low dose radiation therapy as palliative therapy to relieve symptoms and improve quality of life, or arsenic trioxide therapy. Nonstandard therapies, including treatments under clinical trials, are also contemplated by the present invention.


In many embodiments, the treatment regimes described in U.S. Patent Application Publication No. 20040152632 are employed to treat AML or MDS. Genes prognostic of patient outcome under these treatment regimes can be identified according to the present invention. In one example, the treatment regime includes administration of at least one chemotherapy drug and an anti-CD33 antibody conjugated with a cytotoxic agent. The chemotherapy drug can be selected, without limitation, from the group consisting of an anthracycline and a pyrimidine or purine nucleoside analog. The cytotoxic agent can be, for example, a calicheamicin or an esperamicin.


Anthracyclines suitable for treating AML or MDS include, but are not limited to, doxorubicin, daunorubicin, idarubicin, aclarubicin, zorubicin, mitoxantrone, epirubicin, carubicin, nogalamycin, menogaril, pitarubicin, and valrubicin. Pyrimidine or purine nucleoside analogs useful for treating AML or MDS include, but are not limited to, cytarabine, gemcitabine, trifluridine, ancitabine, enocitabine, azacitidine, doxifluridine, pentostatin, broxuridine, capecitabine, cladribine, decitabine, floxuridine, fludarabine, gougerotin, puromycin, tegafur, tiazofurin, or tubercidin. Other anthracyclines and pyrimidine/purine nucleoside analogs can also be used in the present invention.


In a further example, the AML/MDS treatment regime includes administration of gemtuzumab ozogamicin (GO), daunorubicin and cytarabine to a patient in need of the treatment. Gemtuzumab ozogamicin can be administered, without limitation, in an amount of about 3 mg/m2 to about 9 mg/m2 per day, such as about 3, 4, 5, 6, 7, 8 or 9 mg/m2 per day. Daunorubicin can be administered, for example, in an amount of about 45 mg/m2 to about 60 mg/m2 per day, such as about 45, 50, 55 or 60 mg/m2 per day. Cytarabine can be administered, without limitation, in an amount of about 100 mg/m2 to about 200 mg/m2 per day, such as about 100, 125, 150, 175 or 200 mg/m2 per day. In one example, the daunorubicin employed in the treatment regime is daunorubicin hydrochloride.


Clinical Outcome

Clinical outcome of leukemia patients can be assessed by a number of criteria. Examples of clinical outcome measures include, but are not limited to, complete remission, partial remission, non-remission, survival, development of adverse events, or any combination thereof. Patients with complete remission show less than 5% blast cells in the bone marrow after the treatment. Patients with partial remission exhibit a decrease in the blast percentage to certain degree but do not achieve normal hematopoiesis with less than 5% blast cells. The blast percentage in the bone marrow of non-remission patients does not decrease in a significant way in response to the treatment.


In many cases, the peripheral blood samples used for the identification of the prognostic genes are “baseline” or “pretreatment” samples. These samples are isolated from respective leukemia patients prior to a therapeutic treatment and can be used to identify genes whose baseline peripheral blood expression profiles are correlated with clinical outcome of these leukemia patients in response to the treatment. Peripheral blood samples isolated at other treatment or disease stages can also be used to identify leukemia prognostic genes.


A variety of types of peripheral blood samples can be used in the present invention. In one embodiment, the peripheral blood samples are whole blood samples. In another embodiment, the peripheral blood samples comprise enriched PBMCs. By “enriched,” it means that the percentage of PBMCs in the sample is higher than that in whole blood. In some cases, the PBMC percentage in an enriched sample is at least 1, 2, 3, 4, 5 or more times higher than that in whole blood. In some other cases, the PBMC percentage in an enriched sample is at least 90%, 95%, 98%, 99%, 99.5%, or more. Blood samples containing enriched PBMCs can be prepared using any method known in the art, such as Ficoll gradients centrifugation or CPTs (cell purification tubes).


Gene Expression Analysis

The relationship between peripheral blood gene expression profiles and patient outcome can be evaluated by using global gene expression analyses. Methods suitable for this purpose include, but are not limited to, nucleic acid arrays (such as cDNA or oligonucleotide arrays), 2-dimensional SDS-polyacrylamide gel electrophoresis/mass spectrometry, and other high throughput nucleotide or polypeptide detection techniques.


Nucleic acid arrays allow for quantitative detection of the expression levels of a large number of genes at one time. Examples of nucleic acid arrays include, but are not limited to, Genechip® microarrays from Affymetrix (Santa Clara, Calif.), cDNA microarrays from Agilent Technologies (Palo Alto, Calif.), and bead arrays described in U.S. Pat. Nos. 6,288,220 and 6,391,562.


The polynucleotides to be hybridized to a nucleic acid array can be labeled with one or more labeling moieties to allow for detection of hybridized polynucleotide complexes. The labeling moieties can include compositions that are detectable by spectroscopic, photochemical, biochemical, bioelectronic, immunochemical, electrical, optical or chemical means. Exemplary labeling moieties include radioisotopes, chemiluminescent compounds, labeled binding proteins, heavy metal atoms, spectroscopic markers such as fluorescent markers and dyes, magnetic labels, linked enzymes, mass spectrometry tags, spin labels, electron transfer donors and acceptors, and the like. Unlabeled polynucleotides can also be employed. The polynucleotides can be DNA, RNA, or a modified form thereof.


Hybridization reactions can be performed in absolute or differential hybridization formats. In the absolute hybridization format, polynucleotides derived from one sample, such as PBMCs from a patient in a selected outcome class, are hybridized to the probes on a nucleic acid array. Signals detected after the formation of hybridization complexes correlate to the polynucleotide levels in the sample. In the differential hybridization format, polynucleotides derived from two biological samples, such as one from a patient in a first outcome class and the other from a patient in a second outcome class, are labeled with different labeling moieties. A mixture of these differently labeled polynucleotides is added to a nucleic acid array. The nucleic acid array is then examined under conditions in which the emissions from the two different labels are individually detectable. In one embodiment, the fluorophores Cy3 and Cy5 (Amersham Pharmacia Biotech, Piscataway N.J.) are used as the labeling moieties for the differential hybridization format.


Signals gathered from a nucleic acid array can be analyzed using commercially available software, such as those provided by Affymetrix or Agilent Technologies. Controls, such as for scan sensitivity, probe labeling and cDNA/cRNA quantitation, can be included in the hybridization experiments. In many embodiments, the nucleic acid array expression signals are scaled or normalized before being subject to further analysis. For instance, the expression signals for each gene can be normalized to take into account variations in hybridization intensities when more than one array is used under similar test conditions. Signals for individual polynucleotide complex hybridization can also be normalized using the intensities derived from internal normalization controls contained on each array. In addition, genes with relatively consistent expression levels across the samples can be used to normalize the expression levels of other genes. In one embodiment, the expression levels of the genes are normalized across the samples such that the mean is zero and the standard deviation is one. In another embodiment, the expression data detected by nucleic acid arrays are subject to a variation filter which excludes genes showing minimal or insignificant variation across all samples.


Correlation Analysis

The gene expression data collected from nucleic acid arrays can be correlated with clinical outcome using a variety of methods. Methods suitable for this purpose include, but are not limited to, statistical methods (such as Spearman's rank correlation, Cox proportional hazard regression model, ANOVA/t test, or other rank tests or survival models) and class-based correlation metrics (such as nearest-neighbor analysis).


In one embodiment, patients with a specified leukemia (e.g., AML) are divided into at least two classes based on their responses to a therapeutic treatment. The correlation between peripheral blood gene expression (e.g., PBMC gene expression) and the patient outcome classes is then analyzed by a supervised cluster or learning algorithm. Supervised algorithms suitable for this purpose include, but are not limited to, nearest-neighbor analysis, support vector machines, the SAM method, artificial neural networks, and SPLASH. Under a supervised analysis, clinical outcome of each patient is either known or determinable. Genes that are differentially expressed in peripheral blood cells (e.g., PBMCs) of one class of patients relative to another class of patients can be identified. These genes can be used as surrogate markers for predicting clinical outcome of a leukemia patient of interest. Many of the genes thus identified are correlated with a class distinction that represents an idealized expression pattern of these genes in patients of different outcome classes.


In another embodiment, patients with a specified leukemia (e.g., AML) can be divided into at least two classes based on their peripheral blood gene expression profiles. Methods suitable for this purpose include unsupervised clustering algorithms, such as self-organized maps (SOMs), k-means, principal component analysis, and hierarchical clustering. A substantial number (e.g., at least 50%, 60%, 70%, 80%, 90%, or more) of patients in one class may have a first clinical outcome, and a substantial number of patients in another class may have a second clinical outcome. Genes that are differentially expressed in the peripheral blood cells of one class of patients relative to another class of patients can be identified. These genes can also be used as prognostic markers for predicting clinical outcome of a leukemia patient of interest.


In yet another embodiment, patients with a specified leukemia (e.g., AML) can be divided into three or more classes based on their clinical outcomes or peripheral blood gene expression profiles. Multi-class correlation metrics can be employed to identify genes that are differentially expressed in one class of patients relative to another class. Exemplary multi-class correlation metrics include, but are not limited to, those employed by GeneCluster 2 software provided by MIT Center for Genome Research at Whitehead Institute (Cambridge, Mass.).


In a further embodiment, nearest-neighbor analysis (also known as neighborhood analysis) is used to correlate peripheral blood gene expression profiles with clinical outcome of leukemia patients. The algorithm for neighborhood analysis is described in Golub, et al., SCIENCE, 286: 531-537 (1999); Slonim, et al., PROCS. OF THE FOURTH ANNUAL INTERNATIONAL CONFERENCE ON COMPUTATIONAL MOLECULAR BIOLOGY, Tokyo, Japan, April 8-11, p 263-272 (2000); and U.S. Pat. No. 6,647,341. Under one version of the neighborhood analysis, the expression profile of each gene can be represented by an expression vector g=(e1, e2, e3, . . . , en), where ei corresponds to the expression level of gene “g” in the ith sample. A class distinction can be represented by an idealized expression pattern c=(c1, c2, c3, . . . , cn), where ci=1 or −1, depending on whether the ith sample is isolated from class 0 or class 1. Class 0 may include patients having a first clinical outcome, and class 1 includes patients having a second clinical outcome. Other forms of class distinction can also be employed. Typically, a class distinction represents an idealized expression pattern, where the expression level of a gene is uniformly high for samples in one class and uniformly low for samples in the other class.


The correlation between gene “g” and the class distinction can be measured by a signal-to-noise score:






P(g,c)=[μ1(g)−μ2(g)]/[σ1(g)+σ2(g)]


where μ1(g) and μ2(g) represent the means of the log-transformed expression levels of gene “g” in class 0 and class 1, respectively, and σ1(g) and σ2(g) represent the standard deviation of the log-transformed expression levels of gene “g” in class 0 and class 1, respectively. A higher absolute value of a signal-to-noise score indicates that the gene is more highly expressed in one class than in the other. In one example, the samples used to derive the signal-to-noise scores comprise enriched or purified PBMCs and, therefore, the signal-to-noise score P(g,c) represents a correlation between the class distinction and the expression level of gene “g” in PBMCs.


The correlation between gene “g” and the class distinction can also be measured by other methods, such as by the Pearson correlation coefficient or the Euclidean distance, as appreciated by those skilled in the art.


The significance of the correlation between peripheral blood gene expression profiles and the class distinction can be evaluated using a random permutation test. An unusually high density of genes within the neighborhoods of the class distinction, as compared to random patterns, suggests that many genes have expression patterns that are significantly correlated with the class distinction. The correlation between genes and the class distinction can be diagrammatically viewed through a neighborhood analysis plot, in which the y-axis represents the number of genes within various neighborhoods around the class distinction and the x-axis indicates the size of the neighborhood (i.e., P(g,c)). Curves showing different significance levels for the number of genes within corresponding neighborhoods of randomly permuted class distinctions can also be included in the plot.


In many embodiments, the prognostic genes employed in the present invention are above the median significance level in the neighborhood analysis plot. This means that the correlation measure P(g,c) for each prognostic gene is such that the number of genes within the neighborhood of the class distinction having the size of P(g,c) is greater than the number of genes within the corresponding neighborhoods of randomly permuted class distinctions at the median significance level. In many other embodiments, the prognostic genes employed in the present invention are above the 40%, 30%, 20%, 10%, 5%, 2%, or 1% significance level. As used herein, x % significance level means that x % of random neighborhoods contain as many genes as the real neighborhood around the class distinction.


Class predictors can be constructed using the prognostic genes of the present invention. These class predictors can be used to assign a leukemia patient of interest to an outcome class. In one embodiment, the prognostic genes employed in a class predictor are limited to those shown to be significantly correlated with a class distinction by the permutation test, such as those at above the 1%, 2%, 5%, 10%, 20%, 30%, 40%, or 50% significance level. In another embodiment, the PBMC expression level of each prognostic gene in a class predictor is substantially higher or substantially lower in one class of patients than in another class of patients. In still another embodiment, the prognostic genes in a class predictor have top absolute values of P(g,c). In yet another embodiment, the p-value under a Student's t-test (e.g., two-tailed distribution, two sample unequal variance) for each prognostic gene in a class predictor is no more than 0.05, 0.01, 0.005, 0.001, 0.0005, 0.0001, or less. For each prognostic gene, the p-value suggests the statistical significance of the difference observed between the average PBMC expression profiles of the gene in one class of patients versus another class of patients. Lesser p-values indicate more statistical significance for the differences observed between different classes of leukemia patients.


The SAM method can also be used to correlate peripheral blood gene expression profiles with different outcome classes. The prediction analysis of microarrays (PAM) method can then be used to identify class predictors that can best characterize a predefined outcome class and predict the class membership of new samples. See Tibshirani, et al., PROC. NATL. ACAD. SCI. U.S.A., 99:6567-6572 (2002).


In many embodiments, a class predictor of the present invention has high prediction accuracy under leave-one-out cross validation, 10-fold cross validation, or 4-fold cross validation. For instance, a class predictor of the present invention can have at least 50%, 60%, 70%, 80%, 90%, 95%, or 99% accuracy under leave-one-out cross validation, 10-fold cross validation, or 4-fold cross validation. In a typical k-fold cross validation, the data is divided into k subsets of approximately equal size. The model is trained k times, each time leaving out one of the subsets from training and using the omitted subset as the test samples to calculate the prediction error. If k equals the sample size, it becomes the leave-one-out cross validation.


Other class-based correlation metrics or statistical methods can also be used to identify prognostic genes whose expression profiles in peripheral blood samples are correlated with clinical outcome of leukemia patients. Many of these methods can be performed by using commercial or publicly accessible softwares.


Other methods capable of identifying leukemia prognostic genes include, but are not limited, RT-PCR, Northern Blot, in situ hybridization, and immunoassays such as ELISA, RIA or Western Blot. These genes are differentially expressed in peripheral blood cells (e.g., PBMCs) of one class of patients relative to another class of patients. In many cases, the average peripheral blood expression level of each of these genes in one class of patients is statistically different from that in another class of patients. For instance, the p-value under an appropriate statistical significance test (e.g., Student's t-test) for the observed difference can be no more than 0.05, 0.01, 0.005, 0.001, 0.0005, 0.0001, or less. In many other cases, each prognostic gene thus identified has at least 2-, 3-, 4-, 5-, 10-, or 20-fold difference in the average PBMC expression level between one class of patients and another class of patients.


Identification of AML Prognostic Genes Using HG-U133A Microarrays

As an example, the present invention characterized signatures in peripheral blood of AML patients that are indicative of remission in response to a chemotherapy regimen consisting of daunorubicin and cytarabine induction therapy with concomitant administration of GO. In particular, the present invention employed a pharmacogenomic approach to identify transcriptional patterns in peripheral blood samples taken from AML patients prior to treatment that were correlated with positive response to the therapy regimen.


Of the 36 AML patients who consented for pharmacogenomic analysis, 28 achieved a positive response and 8 failed to respond to the treatment regimen following 36 days of induction therapy. Genecluster's default correlation metric (Golub, et al., SCIENCE, 286: 531-537 (1999)) was used to identify genes with expression levels highly correlated with responder and non-responder profiles in the entire set of samples. The low number of non-responders in the pharmacogenomic consented patients precluded division of the pretreatment blood samples into a training and test set. Therefore all samples were used to identify gene classifiers that displayed high accuracies for classification of responder samples versus non-responder samples.


Table 1 lists genes which had higher pretreatment PBMC expression levels in AML patients who eventually failed to respond to the GO combination chemotherapy (non-remission or partial remission), compared to AML patients who responded to the therapy (remission to less than 5% blasts). Genes showing greatest fold elevation in non-responding patients at baseline PBMCs are listed in Table 3. Table 2 describes transcripts that had higher pretreatment expression levels in PBMCs of AML patients who eventually respond to the GO combination chemotherapy, compared to AML patients who did not respond to the therapy. Genes showing greatest fold elevation in responding patients at baseline PBMCs are listed in Table 4. “Fold Change (NR/R)” denotes the ratio of the mean expression level of a gene in PBMCs of non-responding AML patients over that in responding AML patients. “Fold Change (R/NR)” represents the ratio of the mean expression level of a gene in PBMCs of responding AML patients over that in non-responding AML patients. In each table, the transcripts are presented in order of the signal to noise metric score calculated by the supervised algorithm described in Examples. Each gene depicted in Tables 1-4 and the corresponding unigene(s) were identified according to Affymetrix annotations.


Classifiers consisting of genes selected from Tables 1 and 2 were built and evaluated for class prediction accuracy. Each classifier included the top n gene(s) in Table 1 and the top n gene(s) in Table 2, where n represents an integer no less than 1. For example, a first classifier being evaluated included Gene Nos. 1 and 78, a second classifier included Gene Nos. 1-2 and 78-79, a third classifier included Gene Nos. 1-3 and 78-80, a fourth classifier included Gene Nos. 1-4 and 78-81, and so on. Each classifier thus constructed produced significant prediction accuracy. For instance, a classifier consisting of all of the 154 genes in Tables 1 and 2 yielded 81% overall prediction accuracy by 4-fold cross validation on the peripheral blood profiles used in the present study.


Correlation analysis between the pretreatment transcriptional patterns and the clinical outcomes, including occurrence of adverse events, are further discussed in Examples. Additional classifiers are also disclosed in Examples.









TABLE 1







Genes Having Higher Baseline Peripheral Blood Expression


Levels in Non-Responding Patients















SEQ

Fold




Gene

ID
Unigene
Change
Gene


No.
Qualifier
NO:
No.
(NR/R)
Symbol
Gene Name
















1
208581_x_at
1
Hs.278462
2.04
MT1L,
metallothionein 1L, metallothionein







MT1X
1X


2
208963_x_at
2
Hs.132898
1.34
FADS1
fatty acid desaturase 1


3
216336_x_at
3

1.73

unknown


4
209407_s_at
4
Hs.6574
1.88
DEAF1
deformed epidermal autoregulatory








factor 1 (Drosophila)


5
203725_at
5
Hs.80409
1.84
GADD45A
growth arrest and DNA-damage-








inducible, alpha


6
205366_s_at
6
Hs.98428
1.69
HOXB6
homeo box B6


7
209480_at
7
Hs.73931
1.61
HLA-DQB1
major histocompatibility complex,








class II, DQ beta 1


8
204430_s_at
8
Hs.33084
1.61
SLC2A5
solute carrier family 2 (facilitated








glucose/fructose transporter),








member 5


9
204468_s_at
9
Hs.78824
3.62
TIE
tyrosine kinase with immunoglobulin








and epidermal growth factor








homology domains


10
212747_at
10
Hs.20060
1.10
KIAA0229
KIAA0229 protein


11
205227_at
11
Hs.173880
1.88
IL1RAP
interleukin 1 receptor accessory








protein


12
201539_s_at
12
Hs.239069
1.09
FHL1
four and a half LIM domains 1


13
203373_at
13
Hs.110776
2.94
STATI2
STAT induced STAT inhibitor-2


14
210093_s_at
14
Hs.57904
1.52
MAGOH
mago-nashi homolog, proliferation-








associated (Drosophila)


15
209392_at
15
Hs.174185
2.64
ENPP2
ectonucleotide








pyrophosphatase/phosphodiesterase








2 (autotaxin)


16
203372_s_at
16
Hs.110776
2.44
STATI2
STAT induced STAT inhibitor-2


17
212813_at
17
Hs.334703
1.48
FLJ14529
hypothetical protein FLJ14529


18
204326_x_at
18
Hs.199263
1.78
MT1L,
metallothionein 1L, metallothionein







MT1X,
1X, serine threonine kinase 39







STK39
(STE20/SPS1 homolog, yeast)


19
203177_x_at
19
Hs.75133
1.39
TFAM
transcription factor A, mitochondrial


20
212173_at
20
Hs.171811
1.61
AK2
adenylate kinase 2


21
204438_at
21
Hs.75182
2.26
MRC1
mannose receptor, C type 1


22
212185_x_at
22
Hs.118786
1.89
MT2A
metallothionein 2A


23
214281_s_at
23
Hs.48297
1.56
ZNF363
zinc finger protein 363


24
217975_at
24
Hs.15984
1.65
LOC51186
pp21 homolog


25
220974_x_at
25
Hs.283844
2.10
BA108L7.2
similar to rat tricarboxylate carrier-








like protein


26
218807_at
26
Hs.267659
1.52
VAV3
vav 3 oncogene


27
201263_at
27
Hs.84131
1.43
TARS
threonyl-tRNA synthetase


28
217165_x_at
28
n/a
2.02

unknown


29
201013_s_at
29
Hs.117950
1.54
PAICS
phosphoribosylaminoimidazole








carboxylase,








phosphoribosylaminoimidazole








succinocarboxamide synthetase


30
208835_s_at
30
Hs.3688
1.46
LUC7A
cisplatin resistance-associated








overexpressed protein


31
218049_s_at
31
Hs.333823
1.48
MRPL13
mitochondrial ribosomal protein L13


32
217824_at
32
Hs.184325
1.25
NCUBE1
non-canonical ubquitin conjugating








enzyme 1


33
220059_at
33
Hs.121128
1.56
BRDG1
BCR downstream signaling 1


34
202942_at
34
Hs.74047
1.78
ETFB
electron-transfer-flavoprotein, beta








polypeptide


35
200986_at
35
Hs.151242
1.38
SERPING1
serine (or cysteine) proteinase








inhibitor, clade G (C1 inhibitor),








member 1, (angioedema, hereditary)


36
221652_s_at
36
Hs.22595
1.33
FLJ10637
hypothetical protein FLJ10637


37
211456_x_at
37
Hs.367850
1.75

unknown


38
201487_at
38
Hs.10029
1.74
CTSC
cathepsin C


39
220668_s_at
39
Hs.251673
2.00
DNMT3B
DNA (cytosine-5-)-methyltransferase








3 beta


40
215088_s_at
40
Hs.355964
1.43
SDHC
succinate dehydrogenase complex,








subunit C, integral membrane








protein, 15 kD


41
205394_at
41
Hs.20295
1.07
CHEK1
CHK1 checkpoint homolog (S. pombe)


42
218364_at
42
Hs.57672
1.38
LRRFIP2
leucine rich repeat (in FLII)








interacting protein 2


43
222010_at
43
Hs.4112
1.27
TCP1
t-complex 1


44
218286_s_at
44
Hs.14084
1.47
RNF7
ring finger protein 7


45
208955_at
45
Hs.367676
1.21
DUT
dUTP pyrophosphatase


46
210715_s_at
46
Hs.31439
2.04
SPINT2
serine protease inhibitor, Kunitz








type, 2


47
218055_s_at
47
Hs.16470
1.21
FLJ10904
hypothetical protein FLJ10904


48
202946_s_at
48
Hs.7935
2.65
BTBD3
BTB (POZ) domain containing 3


49
201397_at
49
Hs.3343
1.14
PHGDH
phosphoglycerate dehydrogenase


50
204050_s_at
50
Hs.104143
1.54
CLTA
clathrin, light polypeptide (Lca)


51
201425_at
51
Hs.195432
2.29
ALDH2
aldehyde dehydrogenase 2 family








(mitochondrial)


52
204484_at
52
Hs.132463
1.58
PIK3C2B
phosphoinositide-3-kinase, class 2,








beta polypeptide


53
212072_s_at
53
n/a
1.40

unknown


54
215905_s_at
54
Hs.10290
1.34
HPRP8BP
U5 snRNP-specific 40 kDa protein








(hPrp8-binding)


55
201827_at
55
Hs.250581
1.47
SMARCD2
SWI/SNF related, matrix associated,








actin dependent regulator of








chromatin, subfamily d, member 2


56
211031_s_at
56
Hs.104717
1.21
CYLN2
cytoplasmic linker 2


57
217963_s_at
57
Hs.169248
2.49
HCS,
cytochrome c, nerve growth factor







NGFRAP1
receptor (TNFRSF16) associated








protein 1


58
208029_s_at
58
Hs.296398
6.87
LC27
putative integral membrane








transporter


59
202184_s_at
59
Hs.12457
1.37
NUP133
nucleoporin 133 kD


60
214228_x_at
60
Hs.129780
2.36
TNFRSF4
tumor necrosis factor receptor








superfamily, member 4


61
214113_s_at
61
Hs.10283
1.42
RBM8A
RNA binding motif protein 8A


62
217957_at
62
Hs.279818
1.26
AF093680
similar to mouse Glt3 or D. malanogaster








transcription factor IIB


63
218622_at
63
Hs.5152
1.30
MGC5585
hypothetical protein MGC5585


64
208937_s_at
64
Hs.75424
1.20
ID1
inhibitor of DNA binding 1, dominant








negative helix-loop-helix protein


65
213258_at
65
Hs.288582
1.94

unknown


66
206480_at
66
Hs.456
2.05
LTC4S
leukotriene C4 synthase


67
203405_at
67
Hs.5198
1.47
DSCR2
Down syndrome critical region gene 2


68
202430_s_at
68
Hs.198282
1.50
PLSCR1
phospholipid scramblase 1


69
218289_s_at
69
Hs.170737
1.23
FLJ23251
hypothetical protein FLJ23251


70
209757_s_at
70
Hs.25960
1.36
MYCN
v-myc myelocytomatosis viral related








oncogene, neuroblastoma derived








(avian)


71
210298_x_at
71
Hs.239069
1.14
FHL1
four and a half LIM domains 1


72
217814_at
72
Hs.8207
1.50
GK001
GK001 protein


73
201690_s_at
73
Hs.2384
1.63
TPD52
tumor protein D52


74
201923_at
74
Hs.83383
1.18
PRDX4
peroxiredoxin 4


75
210665_at
75
Hs.170279
1.81
TFPI
tissue factor pathway inhibitor








(lipoprotein-associated coagulation








inhibitor)


76
212859_x_at
76
Hs.74170
1.47

unknown


77
221504_s_at
77
Hs.19575
1.60
ATP6V1H
ATPase, H+ transporting, lysosomal








50/57 kD V1 subunit H
















TABLE 2







Genes Having Higher Baseline Peripheral Blood Expression


Levels in Responding Patients

















Fold




Gene

SEQ

Change


No.
Qualifier
ID NO:
Unigene No.
(R/NR)
Gene Symbol
Gene Name
















78
203739_at
78
Hs.155040
1.50
ZNF217
zinc finger protein 217


79
219593_at
79
Hs.237856
3.57
PHT2
peptide transporter 3


80
204132_s_at
80
Hs.14845
1.93
FOXO3A
forkhead box O3A


81
210972_x_at
81
Hs.74647
3.89
TRA@
T cell receptor alpha locus


82
205220_at
82
Hs.137555
3.11
HM74
putative chemokine receptor;








GTP-binding protein


83
201235_s_at
83
Hs.75462
2.35
BTG2
BTG family, member 2


84
209535_s_at
84
Hs.301946
1.69
LBC
lymphoid blast crisis








oncogene


85
209671_x_at
85
Hs.74647
3.95
TRA@
T cell receptor alpha locus


86
203945_at
86
Hs.172851
1.62
ARG2
arginase, type II


87
219434_at
87
Hs.283022
2.61
TREM1
triggering receptor expressed








on myeloid cells 1


88
221558_s_at
88
Hs.44865
2.63
LEF1
lymphoid enhancer-binding








factor 1


89
214056_at
89
Hs.86386
1.91
MCL1
myeloid cell leukemia








sequence 1 (BCL2-related)


90
203907_s_at
90
Hs.4764
2.63
KIAA0763
KIAA0763 gene product


91
217022_s_at
91
Hs.293441
2.00

unknown


92
203413_at
92
Hs.79389
2.04
NELL2
NEL-like 2 (chicken)


93
212074_at
93
Hs.7531
1.62
KIAA0810
KIAA0810 protein


94
220987_s_at
94
Hs.172012
1.62
DKFZP434J037
hypothetical protein








DKFZp434J037


95
212658_at
95
Hs.79299
1.66
LHFPL2
lipoma HMGIC fusion








partner-like 2


96
214467_at
96
Hs.131924
2.14
GPR65
G protein-coupled receptor








65


97
AFFX-DapX-
97
n/a
1.34

unknown



3_at


98
212812_at
98
Hs.288232
2.39

unknown


99
212579_at
99
Hs.8118
1.83
KIAA0650
KIAA0650 protein


100
206133_at
100
Hs.139262
1.86
HSXIAPAF1
XIAP associated factor-1


101
213797_at
101
Hs.17518
1.80
cig5
vipirin


102
213958_at
102
Hs.81226
1.55
CD6
CD6 antigen


103
204638_at
103
Hs.1211
1.66
ACP5
acid phosphatase 5, tartrate








resistant


104
202481_at
104
Hs.17144
1.69
SDR1
short-chain








dehydrogenase/reductase 1


105
204961_s_at
105
Hs.1583
1.95
NCF1
neutrophil cytosolic factor 1








(47 kD, chronic








granulomatous disease,








autosomal 1)


106
209448_at
106
Hs.90753
1.36
HTATIP2
HIV-1 Tat interactive protein








2, 30 kD


107
203290_at
107
Hs.198253
2.81
HLA-DQA1
major histocompatibility








complex, class II, DQ alpha 1


108
215275_at
108
n/a
2.10

unknown


109
221060_s_at
109
Hs.159239
1.60
TLR4
toll-like receptor 4


110
212573_at
110
Hs.167115
1.44
KIAA0830
KIAA0830 protein


111
213193_x_at
111
Hs.303157
1.89
TRB@
T cell receptor beta locus


112
205568_at
112
Hs.104624
3.54
AQP9
aquaporin 9


113
209281_s_at
113
Hs.78546
1.65
ATP2B1
ATPase, Ca++ transporting,








plasma membrane 1


114
204912_at
114
Hs.327
2.17
IL10RA
interleukin 10 receptor, alpha


115
219099_at
115
Hs.24792
1.39
C12orf5
chromosome 12 open








reading frame 5


116
211796_s_at
116
Hs.303157
2.06
TRB@
T cell receptor beta locus


117
221724_s_at
117
Hs.115515
1.84
CLECSF6
C-type (calcium dependent,








carbohydrate-recognition








domain) lectin, superfamily








member 6


118
219607_s_at
118
Hs.325960
1.56
MS4A4A
membrane-spanning 4-








domains, subfamily A,








member 4


119
218802_at
119
Hs.234149
1.91
FLJ20647
hypothetical protein








FLJ20647


120
221671_x_at
120
Hs.156110
2.19
IGKC
immunoglobulin kappa








constant


121
215121_x_at
121
Hs.8997
2.56
HSPA1A,
heat shock 70 kD protein 1A,







IGL@
immunoglobulin lambda locus


122
202147_s_at
122
Hs.7879
1.96
IFRD1
linterferon-related








developmental regulator 1


123
201739_at
123
Hs.296323
3.73
SGK
serum/glucocorticoid








regulated kinase


124
208014_x_at
124
Hs.129735
1.65
AD7C-NTP
neuronal thread protein


125
211339_s_at
125
Hs.211576
2.14
ITK
IL2-inducible T-cell kinase


126
211649_x_at
126
n/a
1.84

unknown


127
202643_s_at
127
Hs.211600
1.32
TNFAIP3
tumor necrosis factor, alpha-








induced protein 3


128
218829_s_at
128
n/a
1.95

unknown


129
204072_s_at
129
Hs.181304
1.33
13CDNA73
hypothetical protein CG003


130
211824_x_at
130
Hs.104305
1.38
DEFCAP
death effector filament-








forming Ced-4-like apoptosis








protein


131
209824_s_at
131
Hs.74515
2.15
ARNTL
aryl hydrocarbon receptor








nuclear translocator-like


132
213539_at
132
Hs.95327
1.81
CD3D
CD3D antigen, delta








polypeptide (TiT3 complex)


133
217143_s_at
133
Hs.2014
2.01
TRD@
T cell receptor delta locus


134
204479_at
134
Hs.95821
1.39
OSTF1
osteoclast stimulating factor 1


135
200628_s_at
135
Hs.374466
1.49
WARS
tryptophanyl-tRNA








synthetase


136
201694_s_at
136
Hs.326035
2.77
EGR1
early growth response 1


137
205821_at
137
Hs.74085
1.51
D12S2489E
DNA segment on








chromosome 12 (unique)








2489 expressed sequence


138
209138_x_at
138
Hs.181125
1.85
IGLJ3
immunoglobulin lambda








joining 3


139
215242_at
139
Hs.97375
1.40

unknown


140
211656_x_at
140
Hs.73931
1.87
HLA-DQB1
major histocompatibility








complex, class II, DQ beta 1


141
222221_x_at
141
Hs.155119
1.45
EHD1
EH-domain containing 1


142
208488_s_at
142
Hs.193716
1.70
CR1
complement component








(3b/4b) receptor 1, including








Knops blood group system


143
202437_s_at
143
Hs.154654
1.66
CYP1B1
cytochrome P450, subfamily I








(dioxin-inducible),








polypeptide 1 (glaucoma 3,








primary infantile)


144
212286_at
144
Hs.27973
1.45
KIAA0874
KIAA0874 protein


145
204959_at
145
Hs.153837
1.24
MNDA
myeloid cell nuclear








differentiation antigen


146
221651_x_at
146
Hs.156110
2.15
IGKC
immunoglobulin kappa








constant


147
201236_s_at
147
Hs.75462
1.81
BTG2
BTG family, member 2


148
211005_at
148
Hs.83496
1.52
LAT
linker for activation of T cells


149
208078_s_at
149
Hs.232068
2.27
TCF8
transcription factor 8








(represses interleukin 2








expression)


150
210018_x_at
150
Hs.180566
1.61
MALT1
mucosa associated lymphoid








tissue lymphoma








translocation gene 1


151
209273_s_at
151
Hs.177776
1.56
MGC4276
hypothetical protein








MGC4276 similar to CG8198


152
213624_at
152
Hs.42945
1.84
ASM3A
acid sphingomyelinase-like








phosphodiesterase


153
208075_s_at
153
Hs.251526
1.77
SCYA7
small inducible cytokine A7








(monocyte chemotactic








protein 3)


154
212154_at
154
Hs.1501
1.90
SDC2
syndecan 2 (heparan sulfate








proteoglycan 1, cell surface-








associated, fibroglycan)
















TABLE 3







Top 50 transcripts significantly elevated (p < 0.05)


at baseline in non-responder patient PBMCs













Affymetrix
SEQ



Fold Diff
p-value


ID
ID NO:
Name
Cyto Band
Unigene ID
(NR/R)
(unequal)
















209392_at
15
ectonucleotide
8q24.1
Hs.174185
2.64
4.91E−02




pyrophosphatase/phosphodiesterase




2 (autotaxin)


220974_x_at
25
similar to rat tricarboxylate
10q24.31
Hs.283844
2.10
1.71E−02




carrier-like protein


206480_at
66
leukotriene C4 synthase
5q35
Hs.456
2.05
4.90E−02


208581_x_at
1
metallothionein 1L,
16q13
Hs.278462
2.04
3.13E−02




metallothionein 1X


217165_x_at
28
unknown
n/a
n/a
2.02
3.54E−02


220668_s_at
39
DNA (cytosine-5-)-
20q11.2
Hs.251673
2.00
4.00E−02




methyltransferase 3 beta


212185_x_at
22
metallothionein 2A
16q13
Hs.118786
1.89
2.55E−02


209407_s_at
4
deformed epidermal
11p15.5
Hs.6574
1.88
2.01E−02




autoregulatory factor 1




(Drosophila)


37384_at
819
KIAA0015 gene product
22q11.22
Hs.278441
1.87
4.11E−02


203725_at
5
growth arrest and DNA-
1p31.2-p31.1
Hs.80409
1.84
4.70E−02




damage-inducible, alpha


202942_at
34
electron-transfer-flavoprotein,
19q13.3
Hs.74047
1.78
4.69E−02




beta polypeptide


216336_x_at
3
unknown
n/a
n/a
1.73
4.92E−02


212235_at
592
KIAA0620 protein
3q22.1
Hs.301685
1.69
4.00E−02


203089_s_at
284
protease, serine, 25
2p12
Hs.115721
1.67
2.23E−02


221504_s_at
77
ATPase, H+ transporting,
8p22-q22.3
Hs.19575
1.60
4.82E−02




lysosomal 50/57 kD V1 subunit H


220942_x_at
790
hypothetical protein, estradiol-
3q21.1
Hs.5243
1.57
2.85E−02




induced


214281_s_at
23
zinc finger protein 363
4q21.1
Hs.48297
1.56
2.43E−02


203091_at
285
far upstream element (FUSE)
1p31.1
Hs.118962
1.56
3.28E−02




binding protein 1


204050_s_at
50
clathrin, light polypeptide (Lca)
9p13
Hs.104143
1.54
4.99E−02


210093_s_at
14
mago-nashi homolog,
1p34-p33
Hs.57904
1.52
2.43E−04




proliferation-associated




(Drosophila)


217226_s_at
689
paired mesoderm homeo box
10q24.31,
Hs.155606
1.52
8.44E−03




1, similar to rat tricarboxylate
1q24




carrier-like protein


218807_at
26
vav 3 oncogene
1p13.2
Hs.267659
1.52
2.11E−02


200824_at
172
glutathione S-transferase pi
11q13
Hs.226795
1.51
2.96E−02


221923_s_at
805
nucleophosmin (nucleolar
5q35
Hs.9614
1.51
3.95E−03




phosphoprotein B23, numatrin)


202854_at
269
hypoxanthine
Xq26.1
Hs.82314
1.51
1.32E−02




phosphoribosyltransferase 1




(Lesch-Nyhan syndrome)


201241_at
197
DEAD/H (Asp-Glu-Ala-
2p24
Hs.78580
1.51
3.98E−02




Asp/His) box polypeptide 1


203720_s_at
305
excision repair cross-
19q13.2-q13.3
Hs.59544
1.49
2.55E−02




complementing rodent repair




deficiency, complementation




group 1 (includes overlapping




antisense sequence)


211941_s_at
578
prostatic binding protein
12q24.22
Hs.80423
1.48
5.88E−03


218049_s_at
31
mitochondrial ribosomal
8q22.1-q22.3
Hs.333823
1.48
4.24E−02




protein L13


218795_at
737
LPAP for lysophosphatidic
1q21
Hs.15871
1.48
4.03E−02




acid phosphatase


212749_s_at
606
zinc finger protein 363
4q21.1
Hs.48297
1.47
2.06E−02


200960_x_at
179
clathrin, light polypeptide (Lca)
9p13
Hs.104143
1.46
4.43E−02


201577_at
221
non-metastatic cells 1, protein
17q21.3
Hs.118638
1.46
3.31E−02




(NM23A) expressed in


205711_x_at
412
ATP synthase, H+
10q22-q23,
Hs.155433
1.44
2.59E−02




transporting, mitochondrial F1
8p22-p21.3




complex, gamma polypeptide




1, CCR4-NOT transcription




complex, subunit 7


213366_x_at
625
ATP synthase, H+
10q22-q23,
Hs.155433
1.44
4.59E−02




transporting, mitochondrial F1
8p22-p21.3




complex, gamma polypeptide




1, CCR4-NOT transcription




complex, subunit 7


217942_at
702
mitochondrial ribosomal
12p11
Hs.10724
1.44
3.24E−02




protein S35


208713_at
468
E1B-55 kDa-associated protein 5
19q13.31
Hs.155218
1.44
1.66E−02


201765_s_at
225
hexosaminidase A (alpha
15q23-q24
Hs.119403
1.43
4.74E−02




polypeptide)


216295_s_at
679
clathrin, light polypeptide (Lca)
9p13
Hs.348345
1.43
4.32E−02


202929_s_at
275
D-dopachrome tautomerase
22q11.23
Hs.180015
1.43
4.87E−02


217871_s_at
700
macrophage migration
22q11.23
Hs.73798
1.43
3.36E−02




inhibitory factor (glycosylation-




inhibiting factor)


218078_s_at
711
zinc finger, DHHC domain
3p21.32
Hs.14896
1.42
1.63E−02




containing 3


208870_x_at
474
ATP synthase, H+
10q22-q23,
Hs.155433
1.42
1.95E−02




transporting, mitochondrial F1
8p22-p21.3




complex, gamma polypeptide




1, CCR4-NOT transcription




complex, subunit 7


200822_x_at
171
triosephosphate isomerase 1
12p13
Hs.83848
1.42
4.53E−02


203103_s_at
286
nuclear matrix protein
11q12.2
Hs.173980
1.41
3.70E−02




NMP200 related to splicing




factor PRP19


213507_s_at
628
karyopherin (importin) beta 1
17q21
Hs.180446
1.41
1.07E−02


201231_s_at
195
enolase 1, (alpha)
1p36.3-p36.2
Hs.254105
1.40
2.89E−02


204905_s_at
376
eukaryotic translation
6p24.3-p25.1
Hs.298581
1.39
3.32E−02




elongation factor 1 epsilon 1


203177_x_at
19
transcription factor A,
10q21
Hs.75133
1.39
2.82E−02




mitochondrial


218154_at
714
hypothetical protein FLJ12150
8q24.3
Hs.118983
1.39
4.30E−02
















TABLE 4







Top 50 transcripts significantly elevated (p < 0.05) at


baseline in responder patient PBMCs













Affymetrix
SEQ ID



Fold Diff
p-value


ID
NO:
Name
Cyto Band
Unigene ID
(R/NR)
(unequal)
















218559_s_at
727
v-maf musculoaponeurotic
20q11.2-q13.1
Hs.169487
7.33
1.30E−02




fibrosarcoma oncogene




homolog B (avian)


209728_at
509
major histocompatibility
6p21.3
Hs.318720
6.49
5.81E−03




complex, class II, DR beta 4


204614_at
356
serine (or cysteine) proteinase
18q21.3
Hs.75716
4.11
4.20E−02




inhibitor, clade B (ovalbumin),




member 2


209671_x_at
85
T cell receptor alpha locus
14q11.2
Hs.74647
3.95
8.98E−03


210972_x_at
81
T cell receptor alpha locus
14q11.2
Hs.74647
3.89
6.39E−03


201739_at
123
serum/glucocorticoid
6q23
Hs.296323
3.73
5.87E−04




regulated kinase


219593_at
79
peptide transporter 3
11q13.1
Hs.237856
3.57
7.04E−04


205568_at
112
aquaporin 9
15q22.1-22.2
Hs.104624
3.54
8.87E−04


204885_s_at
372
mesothelin
16p13.12
Hs.155981
3.54
2.13E−02


211571_s_at
564
chondroitin sulfate
5q14.3
Hs.81800
3.45
4.23E−02




proteoglycan 2 (versican)


210655_s_at
545
forkhead box O3A
6q21
Hs.14845
3.36
5.20E−03


213338_at
622
Ras-induced senescence 1
3p21.3
Hs.35861
3.29
1.67E−02


213524_s_at
630
putative lymphocyte G0/G1
1q32.2-q41
Hs.95910
3.28
1.78E−03




switch gene


221602_s_at
798
regulator of Fas-induced
1q31.3
Hs.58831
3.19
8.83E−03




apoptosis


205220_at
82
putative chemokine receptor;
12q24.31
Hs.137555
3.11
7.86E−04




GTP-binding protein


208450_at
461
lectin, galactoside-binding,
22q13.1
Hs.113987
2.99
3.18E−02




soluble, 2 (galectin 2)


205898_at
416
chemokine (C—X3—C)
3p21.3
Hs.78913
2.98
2.29E−02




receptor 1


212099_at
584
ras homolog gene family,
2pter-p12
Hs.204354
2.96
3.05E−03




member B


218856_at
742
hypothetical protein
6p12.3, 6p21.1-12.2
Hs.65403
2.90
8.84E−03




LOC51323, tumor necrosis




factor receptor superfamily,




member 21


220088_at
775
complement component 5
19q13.3-q13.4
Hs.2161
2.86
6.44E−03




receptor 1 (C5a ligand)


221698_s_at
799
C-type (calcium dependent,
12p13.2-p12.3
Hs.161786
2.83
1.85E−03




carbohydrate-recognition




domain) lectin, superfamily




member 12


201743_at
224
CD14 antigen
5q31.1
Hs.75627
2.83
2.71E−02


212657_s_at
604
interleukin 1 receptor
2q14.2
Hs.81134
2.83
4.41E−03




antagonist


203290_at
107
major histocompatibility
6p21.3
Hs.198253
2.81
2.06E−02




complex, class II, DQ alpha 1


204588_s_at
354
solute carrier family 7 (cationic
14q11.2
Hs.194693
2.81
3.88E−03




amino acid transporter, y+




system), member 7


211506_s_at
561
interleukin 8
4q13-q21
Hs.624
2.80
1.47E−03


201694_s_at
136
early growth response 1
5q31.1
Hs.326035
2.77
1.04E−03


204890_s_at
373
lymphocyte-specific protein
1p34.3
Hs.1765
2.64
2.12E−02




tyrosine kinase


221558_s_at
88
lymphoid enhancer-binding
4q23-q25
Hs.44865
2.63
1.82E−02




factor 1


203907_s_at
90
KIAA0763 gene product
3p25.1
Hs.4764
2.63
1.45E−03


203066_at
282
B cell RAG associated protein
10q26
Hs.6079
2.61
1.90E−03


219434_at
87
triggering receptor expressed
6p21.1
Hs.283022
2.61
2.06E−02




on myeloid cells 1


216191_s_at
677
T cell receptor delta locus
14q11.2
Hs.2014
2.59
1.80E−02


205114_s_at
382
small inducible cytokine A3
17q11-q21
Hs.73817
2.57
3.76E−02


215223_s_at
668
superoxide dismutase 2,
6q25.3
Hs.372783
2.57
1.30E−03




mitochondrial


216491_x_at
682
unknown
n/a
n/a
2.55
4.12E−02


217739_s_at
695
pre-B-cell colony-enhancing
7q11.23
Hs.239138
2.53
1.04E−03




factor


201631_s_at
223
immediate early response 3
6p21.3
Hs.76095
2.47
2.21E−02


202086_at
238
myxovirus (influenza virus)
21q22.3
Hs.76391
2.47
1.04E−03




resistance 1, interferon-




inducible protein p78 (mouse)


204141_at
331
tubulin, beta polypeptide
6p21.3
Hs.336780
2.46
3.35E−02


209670_at
507
T cell receptor alpha locus
14q11.2
Hs.74647
2.46
3.71E−02


219528_s_at
762
B-cell CLL/lymphoma 11B
14q32.31-q32.32
Hs.57987
2.45
3.11E−02




(zinc finger protein)


206150_at
426
tumor necrosis factor receptor
12p13
Hs.180841
2.44
1.94E−02




superfamily, member 7


201506_at
213
transforming growth factor,
5q31
Hs.118787
2.42
4.20E−02




beta-induced, 68 kD


203939_at
314
5′-nucleotidase, ecto (CD73)
6q14-q21
Hs.153952
2.42
1.91E−02


205419_at
396
Epstein-Barr virus induced
13q32.3
Hs.784
2.39
1.56E−03




gene 2 (lymphocyte-specific G




protein-coupled receptor)


212812_at
98
unknown
n/a
Hs.288232
2.39
1.11E−04


217378_x_at
692
unknown
n/a
n/a
2.38
2.11E−02


211135_x_at
555
leukocyte immunoglobulin-like
19q13.4
Hs.105928
2.37
1.57E−02




receptor, subfamily B (with TM




and ITIM domains), member 3


204006_s_at
318
Fc fragment of IgG, low affinity
1q23
Hs.372679
2.36
4.30E−02




IIIa, receptor for (CD16), Fc




fragment of IgG, low affinity




IIIb, receptor for (CD16)










Genes Associated with the Onset of Veno-Occlusive Disease


Veno-occlusive disease (VOD) is one of the most serious complications following hematopoietic stem cell transplantation and is associated with a very high mortality in its severe form. Comparison of pretreatment PBMC profiles from the leukemia patients who experienced VOD with the PBMC profiles from the patients who did not experience VOD identifies significant transcripts that appear to be correlated with this serious adverse event prior to therapy.


To identify transcripts with significant differences in expression at baseline between the patients who experienced VOD and the non-VOD patients, average fold differences between VOD and non-VOD patient profiles were calculated by dividing the mean level of expression in the baseline VOD profiles by the mean level of expression in the baseline non-VOD profiles. A Student's t-test (two-sample, unequal variance) was used to assess the significance of the difference in expression between the groups.


Genes whose expression levels are significantly elevated (p<0.05) at baseline in VOD patients are shown in Table 5. Genes whose expression levels are significantly repressed (p<0.05) at baseline in VOD patients are shown in Table 6. Of interest, P-selectin ligand was one of the transcripts most significantly elevated at baseline in patients who experienced VOD. Without wishing to be bound by theory, the elevation in this transcript may be a biomarker indicative of endothelial damage which has been suggested to play a role in transplant-associated diseases such as graft-versus-host disease, sepsis, and VOD.









TABLE 5







Top 50 Transcripts significantly elevated (p < 0.05)


at baseline in VOD patient PBMCs














SEQ ID



Fold Diff
p-value


Affymetrix ID
NO::
Name
Cyto Band
Unigene ID
(VOD/non-VOD)
(unequal)
















204020_at
321
purine-rich element binding protein A
5q31
Hs.29117
2.096551724
0.025737029


202742_s_at
264
protein kinase, cAMP-dependent,
1p36.1
Hs.87773
2.031746032
0.023084697




catalytic, beta


209879_at
516
selectin P ligand
12q24
Hs.79283
2.02247191
0.024750558


AFFX-r2-
826
n/a
n/a
n/a
1.967450271
0.00094123


Hs28SrRNA-3_at


217986_s_at
704
bromodomain adjacent to zinc finger
14q12-q13
Hs.8858
1.948186528
0.040961702




domain, 1A


202322_s_at
247
geranylgeranyl diphosphate
1q43
Hs.55498
1.806451613
0.008621905




synthase 1


AFFX-
825
n/a
n/a
n/a
1.789173789
0.007668769


M27830_5_at


219974_x_at
772
uncharacterized hypothalamus
6q23.1
Hs.239218
1.741496599
0.026918594




protein HCDASE


201964_at
231
KIAA0625 protein
9q34.3
Hs.154919
1.739130435
0.025540988


202741_at
263
n/a
1p36.1
Hs.417060
1.737931034
0.003565502


203947_at
315
cleavage stimulation factor, 3′ pre-
11p12
Hs.180034
1.723076923
0.011499059




RNA, subunit 3, 77 kDa


218642_s_at
729
hypothetical protein MGC2217
8q11.22
Hs.323164
1.686486486
0.010323657


200860_s_at
173
KIAA1007 protein
16q21
Hs.279949
1.682403433
0.018297378


201027_s_at
185
translation initiation factor IF2
2p11.1-q11.1
Hs.158688
1.680672269
0.032120458


213361_at
624
tudor repeat associator with
9q22.33
Hs.283761
1.656804734
0.027072176




PCTAIRE 2


220956_s_at
791
egl nine homolog 2 (C. elegans)
19q13.2
Hs.324277
1.653631285
0.007996997


218646_at
730
hypothetical protein FLJ20534
4q32.3
Hs.44344
1.619047619
0.019526095


200604_s_at
156
protein kinase, cAMP-dependent,
17q23-q24
Hs.183037
1.608938547
0.040659084




regulatory, type I, alpha (tissue




specific extinguisher 1)


201989_s_at
233
cAMP responsive element binding
12p13
Hs.13313
1.608247423
0.042105857




protein-like 2


217993_s_at
706
methionine adenosyltransferase II,
5q34-q35.1
Hs.54642
1.597964377
0.002167131




beta


204613_at
355
phospholipase C, gamma 2
16q24.1
Hs.75648
1.592039801
0.012601371




(phosphatidylinositol-specific)


201142_at
191
eukaryotic translation initiation factor
14q23.3
Hs.151777
1.567010309
1.80074E−06




2, subunit 1 alpha, 35 kDa


219649_at
765
dolichyl-P-Glc: Man9GlcNAc2-PP-
1p31.3
Hs.80042
1.565217391
0.021274365




dolichylglucosyltransferase


209907_s_at
519
intersectin 2
2pter-p25.1
Hs.166184
1.5625
0.02410118


210502_s_at
540
peptidylprolyl isomerase E
1p32
Hs.379815
1.555555556
0.000233425




(cyclophilin E)


209903_s_at
517
ataxia telangiectasia and Rad3
3q22-q24
Hs.77613
1.551515152
0.016402019




related


212402_at
598
KIAA0853 protein
13q14.11
Hs.136102
1.543147208
1.96044E−06


202003_s_at
234
acetyl-Coenzyme A acyltransferase
18q21.1
Hs.356176
1.538461538
0.031540874




2 (mitochondrial 3-oxoacyl-




Coenzyme A thiolase)


220933_s_at
789
hypothetical protein FLJ13409
9q21
Hs.30732
1.536723164
0.030072848


208911_s_at
479
pyruvate dehydrogenase (lipoamide)
3p21.1-p14.2
Hs.979
1.531914894
0.020768712




beta


212697_at
605
n/a
n/a
Hs.432850
1.519832985
0.022783857


219940_s_at
770
hypothetical protein FLJ11305
13q34
Hs.7049
1.514403292
0.001555339


212754_s_at
607
KIAA1040 protein
12q13.13
Hs.9846
1.505882353
0.037849628


207614_s_at
453
cullin 1
7q34-q35
Hs.14541
1.496402878
0.049509373


209096_at
483
ubiquitin-conjugating enzyme E2
8q11.1
Hs.79300
1.493975904
0.047033925




variant 2


200802_at
167
seryl-tRNA synthetase
1p13.3-p13.1
Hs.144063
1.488372093
0.005291866


220408_x_at
779
transcription factor (p38 interacting
13q13.1-q13.2
Hs.376447
1.484848485
0.035433399




protein)


204780_s_at
364
tumor necrosis factor receptor
10q24.1
Hs.426662
1.476923077
0.000371305




superfamily, member 6


203879_at
310
phosphoinositide-3-kinase, catalytic,
1p36.2
Hs.162808
1.471406491
0.035824787




delta polypeptide


201384_s_at
204
membrane component,
17q21.1
Hs.277721
1.46875
0.009771907




chromosome 17, surface marker 2




(ovarian carcinoma antigen CA125)


212588_at
603
protein tyrosine phosphatase,
1q31-q32
Hs.170121
1.461700632
0.048016891




receptor type, C


219033_at
751
hypothetical protein FLJ21308
5q11.1
Hs.406232
1.459016393
0.02208168


203073_at
283
component of oligomeric golgi
1q42.13
Hs.82399
1.457489879
0.008447959




complex 2


206332_s_at
430
interferon, gamma-inducible protein
1q22
Hs.155530
1.455696203
0.027832428




16


202868_s_at
272
POP4 (processing of precursor,
19q13.11
Hs.82238
1.449275362
0.021497345





S. cerevisiae) homolog



218249_at
718
zinc finger, DHHC domain
10q26.11
Hs.22353
1.427509294
0.001378715




containing 6


212530_at
602
NIMA (never in mitosis gene a)-
1q31.3
Hs.24119
1.418719212
0.035013309




related kinase 7


218463_s_at
725
MUS81 endonuclease
11q13
Hs.288798
1.403508772
0.034273747


213115_at
613
n/a
n/a
n/a
1.398907104
0.038806001


218103_at
712
FtsJ homolog 3 (E. coli)
17q23
Hs.257486
1.393258427
5.58595E−05
















TABLE 6







Top 50 transcripts significantly repressed (p < 0.05)


at baseline in VOD patient PBMCs


















Fold Diff
p-value


Affymetrix ID
SEQ ID NO:
Name
Cyto Band
Unigene ID
(VOD/non-VOD)
(unequal)
















217023_x_at
688
tryptase beta 1, tryptase beta 2
16p13.3
Hs.294158, Hs.405479
0.131687243
0.000341


210084_x_at
525
tryptase beta 2, tryptase, alpha
16p13.3
Hs.294158
0.133828996
0.000347153


208029_s_at
58
lysosomal associated protein
8q22.1
Hs.296398
0.133891213
0.020766934




transmembrane 4 beta


213844_at
638
homeo box A5
7p15-p14
Hs.37034
0.148514851
0.003338613


215382_x_at
670
tryptase, alpha
16p13.3
Hs.334455
0.155477032
0.000156058


205683_x_at
411
tryptase beta 1, tryptase beta 2, tryptase,
16p13.3
Hs.405479
0.158102767
0.00154079




alpha


216474_x_at
681
tryptase beta 1, tryptase beta 2, tryptase,
16p13.3
Hs.334455
0.15954416
0.000338402




alpha


208789_at
470
polymerase I and transcript release factor
17q21.2
Hs.29759
0.172972973
0.004109481


202016_at
235
mesoderm specific transcript homolog
7q32
Hs.79284
0.176239182
0.001253864




(mouse)


207134_x_at
447
tryptase beta 1, tryptase beta 2, tryptase,
16p13.3
Hs.294158
0.180722892
0.002582561




alpha


214039_s_at
643
lysosomal associated protein
8q22.1
Hs.296398
0.221343874
0.015962264




transmembrane 4 beta


201015_s_at
184
junction plakoglobin
17q21
Hs.2340
0.227642276
2.96697E−06


202112_at
240
von Willebrand factor
12p13.3
Hs.110802
0.231884058
0.000771533


36711_at
817
v-maf musculoaponeurotic fibrosarcoma
22q13.1
Hs.51305
0.243093923
0.000110895




oncogene homolog F (avian)


207741_x_at
456
tryptase, alpha
16p13.3
Hs.334455
0.244741874
0.000539503


209395_at
495
chitinase 3-like 1 (cartilage glycoprotein-
1q31.1
Hs.75184
0.266666667
0.006968551




39)


205131_x_at
383
stem cell growth factor; lymphocyte
19q13.3
Hs.425339
0.266666667
0.01030592




secreted C-type lectin


201005_at
183
CD9 antigen (p24)
12p13.3
Hs.1244
0.270613108
0.001191345


215111_s_at
666
transforming growth factor beta-stimulated
13q14
Hs.114360
0.279957582
0.00118603




protein TSC-22


205624_at
409
carboxypeptidase A3 (mast cell)
3q21-q25
Hs.646
0.282225237
0.00249997


206067_s_at
423
Wilms tumor 1
11p13
Hs.1145
0.282352941
0.001463202


201596_x_at
222
glutamate receptor, ionotropic, N-methyl D-
12q13
Hs.406013
0.292358804
0.002605841




asparate-associated protein 1 (glutamate




binding), keratin 18


213479_at
627
neuronal pentraxin II
7q21.3-q22.1
Hs.3281
0.298507463
0.046185388


201324_at
201
epithelial membrane protein 1
12p12.3
Hs.79368
0.299065421
0.001554754


210783_x_at
549
stem cell growth factor; lymphocyte
19q13.3
Hs.425339
0.301886792
0.009424594




secreted C-type lectin


216202_s_at
678
serine palmitoyltransferase, long chain
14q24.3-q31
Hs.59403
0.306220096
0.000219065




base subunit 2


218880_at
744
FOS-like antigen 2
2p23-p22
Hs.301612
0.310679612
0.000328157


206461_x_at
435
metallothionein 1H
16q13
Hs.2667
0.310679612
0.001303906


204885_s_at
372
mesothelin
16p13.12
Hs.155981
0.310679612
0.021690405


220377_at
778
chromosome 14 open reading frame 110
14q32.33
Hs.128155
0.315789474
0.003681392


204011_at
319
sprouty homolog 2 (Drosophila)
13q22.2
Hs.18676
0.32
0.00124785


211948_x_at
579
KIAA1096 protein
1q23.3
Hs.69559
0.32
0.008446106


208886_at
476
H1 histone family, member 0
22q13.1
Hs.226117
0.321715818
0.00641406


215047_at
665
BIA2
1q44
Hs.51692
0.322147651
0.022774503


209905_at
518
homeo box A9
7p15-p14
Hs.127428
0.322496749
0.022921003


218332_at
721
brain expressed, X-linked 1
Xq21-q23
Hs.334370
0.325
0.026696331


203411_s_at
293
lamin A/C
1q21.2-q21.3
Hs.377973
0.329411765
0.000122251


209774_x_at
511
chemokine (C—X—C motif) ligand 1
4q21
Hs.75765
0.33256351
0.002389608




(melanoma growth stimulating activity,




alpha), chemokine (C—X—C motif)




ligand 2


209757_s_at
70
v-myc myelocytomatosis viral related
2p24.1
Hs.25960
0.333333333
0.0002004




oncogene, neuroblastoma derived (avian)


201830_s_at
227
neuroepithelial cell transforming gene 1
10p15
Hs.25155
0.335078534
0.000181408


219837_s_at
769
cytokine-like protein C17
4p16-p15
Hs.13872
0.347826087
0.009008447


205051_s_at
380
v-kit Hardy-Zuckerman 4 feline sarcoma
4q11-q12
Hs.81665
0.348993289
0.006943974




viral oncogene homolog


211709_s_at
566
stem cell growth factor; lymphocyte
19q13.3
Hs.425339
0.354948805
0.033343631




secreted C-type lectin


210665_at
75
tissue factor pathway inhibitor (lipoprotein-
2q31-q32.1
Hs.170279
0.355555556
0.001918239




associated coagulation inhibitor)


209301_at
491
carbonic anhydrase II
8q22
Hs.155097
0.355555556
0.003901677


204468_s_at
9
tyrosine kinase with immunoglobulin and
1p34-p33
Hs.78824
0.36036036
0.034680165




epidermal growth factor homology domains


208767_s_at
469
lysosomal associated protein
8q22.1
Hs.296398
0.361111111
0.022507793




transmembrane 4 beta


209183_s_at
485
decidual protein induced by progesterone
10q11.23
Hs.93675
0.363636364
0.0038473


213260_at
619


Hs.284186
0.366666667
0.030189907


209488_s_at
497
RNA-binding protein gene with multiple
8p12-p11
Hs.80248
0.367816092
0.013648398




splicing









Identification of Leukemia Diagnostic Genes

The above described methods can also be used to identify leukemia diagnostic genes (also referred to as disease genes). Each of these genes is differentially expressed in PBMCs of leukemia patients relative to PBMCs of leukemia-free or disease-free humans. In many cases, the average PBMC expression level of a leukemia disease gene in leukemia patients is statistically different from that in leukemia-free or disease-free humans. For example, the p-value of a Student's t-test for the observed difference can be no more than 0.05, 0.01, 0.005, 0.001, 0.0005, 0.0001, or less. In many other cases, the difference between the average PBMC expression levels of a leukemia disease gene in leukemia patients and that in leukemia-free humans is at least 2, 3, 4, 5, 10, 20, or more folds. The leukemia disease genes of the present invention can be used to detect the presence or absence, or monitor the development, progression or treatment of leukemia in a human of interest.


Leukemia disease genes can also be identified by correlating PBMC expression profiles with a class distinction under a class-based correlation metric (e.g., the nearest-neighbor analysis or the significance method of microarrays (SAM) method). The class distinction represents an idealized gene expression pattern in PBMCs of leukemia patients and disease-free humans. In many examples, the correlation between the PBMC expression profile of a leukemia disease gene and the class distinction is above the 1%, 5%, 10%, 25%, or 50% significance level under a permutation test. Gene classifiers can be constructed using the leukemia disease genes of the present invention. These classifiers can effectively predict class membership (e.g., leukemia versus leukemia-free) of a human of interest.


Identification of AML Diagnosis Genes Using HG-U133A Microarrays

As an example, AML-associated expression patterns in peripheral blood were identified by using the U133A gene chip platform. Mean levels of baseline gene expression in PBMCs from a group of disease-free volunteers (n=20) were compared with mean levels of corresponding baseline gene expression in PBMCs from AML patients (n=36). Transcripts showing elevated or decreased levels in PBMCs of AML patients relative to healthy controls were identified. Examples of these transcripts are depicted in Table 7. Each transcript in Table 7 has at least 2-fold difference in the mean level of expression between AML PBMCs and disease-free PBMCs (“AML/Disease-Free”). The p-value of the Student's t-test (unequal variances) for the observed difference (“P-Value”) is also shown in Table 7. “COV” refers to coefficient of variance.









TABLE 7







Example of AML Disease Genes Differentially Expressed in PBMCs of AML Patients Relative to Disease-Free Volunteers

















AML/


COV






SEQ ID
Disease-

COV
(Disease
Gene

Unigene


Qualifier
NO:
Free
P-Value
(AML)
Free)
Symbol
Gene Name
No.


















203948_s_at
316
46.69
4.63E−06
108.53%
33.68%
MPO
myeloperoxidase
Hs.1817


203949_at
317
35.14
1.19E−06
99.53%
29.31%
MPO
myeloperoxidase
Hs.1817


206310_at
429
22.75
3.86E−06


SPINK2
serine protease inhibitor, Kazal
Hs.98243









type, 2 (acrosin-trypsin









inhibitor)


209905_at
518
21.08
5.44E−05


HOXA9
homeo box A9
Hs.127428


214575_s_at
658
20.02
3.88E−04
145.25%
28.21%
AZU1
azurocidin 1 (cationic
Hs.72885









antimicrobial protein 37)


206871_at
444
18.41
1.23E−04
131.40%
48.57%
ELA2
elastase 2, neutrophil
Hs.99863


214651_s_at
660
16.25
5.98E−05
123.43%
21.22%
HOXA9
homeo box A9
Hs.127428


205653_at
410
14.76
1.24E−03
159.20%
28.58%
CTSG
cathepsin G
Hs.100764


210084_x_at
525
14.18
1.20E−04



tryptase beta 1, tryptase, alpha
Hs.347933


205683_x_at
411
13.92
4.32E−04



tryptase beta 1, tryptase beta
Hs.347933









2, tryptase, alpha


204798_at
368
12.95
7.41E−10
66.25%
24.66%
MYB
v-myb myeloblastosis viral
Hs.1334









oncogene homolog (avian)


206851_at
443
12.83
7.34E−03
194.31%
50.67%
RNASE3
ribonuclease, RNase A family,
Hs.73839









3 (eosinophil cationic protein)


217023_x_at
688
12.02
1.41E−04



tryptase beta 1, tryptase beta 2
Hs.294158,










Hs.347933


216474_x_at
681
11.06
8.25E−05



tryptase beta 1, tryptase beta 2
Hs.347933


202016_at
235
11.02
3.63E−04
138.17%
24.92%
MEST
mesoderm specific transcript
Hs.79284









homolog (mouse)


207134_x_at
447
10.94
6.98E−04
146.58%
35.48%
TPS1,
tryptase beta 1, tryptase beta
Hs.294158








TPSB1,
2, tryptase, alpha








TPSB2


215382_x_at
670
10.85
5.25E−05



tryptase beta 1, tryptase, alpha
Hs.347933


205950_s_at
420
10.85
5.23E−04


CA1
carbonic anhydrase I
Hs.23118


205051_s_at
380
10.24
2.37E−05
111.13%
30.96%
KIT
v-kit Hardy-Zuckerman 4 feline
Hs.81665









sarcoma viral oncogene









homolog


211709_s_at
566
10.06
1.23E−06
92.43%
24.57%
SCGF
stem cell growth factor;
Hs.425339,









lymphocyte secreted C-type
Hs.105927









lectin


205131_x_at
383
9.55
1.02E−04



stem cell growth factor;
Hs.105927









lymphocyte secreted C-type









lectin


219054_at
753
8.32
2.05E−06


FLJ14054
hypothetical protein FLJ14054
Hs.13528


204304_s_at
340
7.69
4.74E−07
84.71%
30.22%
PROML1
prominin-like 1 (mouse)
Hs.112360


206674_at
440
7.41
2.90E−07


FLT3
fms-related tyrosine kinase 3
Hs.385


207741_x_at
456
7.33
5.05E−05



tryptase, alpha
Hs.334455


202589_at
257
7.08
1.63E−05
103.09%
49.47%
TYMS
thymidylate synthetase
Hs.29475,










Hs.82962


210783_x_at
549
6.99
5.96E−05
112.68%
19.95%
SCGF
stem cell growth factor;
Hs.425339,









lymphocyte secreted C-type
Hs.105927









lectin


211922_s_at
576
6.71
1.13E−07
76.92%
32.08%
CAT
catalase
Hs.395771,










Hs.76359


203373_at
13
6.70
1.95E−02
208.35%
23.04%
STATI2
STAT induced STAT inhibitor-2
Hs.405946


201427_s_at
208
6.64
7.13E−04
137.31%
0.00%
SEPP1
selenoprotein P, plasma, 1
Hs.275775,










Hs.3314


206111_at
424
6.60
2.95E−05
106.04%
41.83%
RNASE2
ribonuclease, RNase A family,
Hs.728









2 (liver, eosinophil-derived









neurotoxin)


213844_at
638
6.60
2.86E−03
158.62%
46.12%
HOXA5
homeo box A5
Hs.37034


202503_s_at
255
6.39
2.92E−06


KIAA0101
KIAA0101 gene product
Hs.81892


205899_at
417
6.26
1.91E−03
150.19%
16.83%
CCNA1
cyclin A1
Hs.79378


220377_at
778
6.14
1.93E−04
120.57%
14.58%
HSPC053
HSPC053 protein
Hs.128155


201310_s_at
200
5.92
2.13E−09



P311 protein
Hs.142827


219672_at
767
5.86
9.81E−04
137.79%
96.37%
ERAF
erythroid associated factor
Hs.274309


208029_s_at
58
5.69
2.37E−02
208.96%
30.33%
LC27
putative integral membrane
Hs.296398









transporter


205624_at
409
5.66
9.30E−05
111.81%
43.05%
CPA3
carboxypeptidase A3 (mast
Hs.646









cell)


205609_at
407
5.59
1.49E−06
85.15%
34.40%
ANGPT1
angiopoietin 1
Hs.2463


206834_at
442
5.49
5.46E−05
106.29%
97.40%
HBD
hemoglobin, delta
Hs.36977


205557_at
402
5.28
1.42E−02
188.13%
75.52%
BPI
bactericidal/permeability-
Hs.89535









increasing protein


201162_at
192
5.25
3.09E−07
76.99%
53.67%
IGFBP7
insulin-like growth factor
Hs.119206









binding protein 7


201432_at
209
5.18
1.43E−09



catalase
Hs.76359


204430_s_at
8
5.17
6.73E−04
129.63%
30.33%
SLC2A5
solute carrier family 2
Hs.33084









(facilitated glucose/fructose









transporter), member 5


220416_at
780
5.16
1.24E−06
82.78%
18.42%
KIAA1939
KIAA1939 protein
Hs.182738


204030_s_at
322
5.06
2.43E−03
147.20%
34.79%
SCHIP1
schwannomin interacting
Hs.61490









protein 1


211743_s_at
568
4.95
7.28E−04
129.14%
32.90%
PRG2
proteoglycan 2, bone marrow
Hs.99962









(natural killer cell activator,









eosinophil granule major basic









protein)


201416_at
206
4.94
1.01E−04
109.06%
35.67%
MEIS3,
Meis1, myeloid ecotropic viral
Hs.83484








SOX4
integration site 1 homolog 3









(mouse), SRY (sex









determining region Y)-box 4


213150_at
617
4.90
3.44E−04
120.37%
26.79%
HOXA10
homeo box A10
Hs.110637


209543_s_at
502
4.88
6.90E−07
78.99%
30.30%
CD34,
CD34 antigen, FLJ00005
Hs.374990








FLJ00005
protein


213258_at
65
4.82
2.40E−07




Hs.288582


216667_at
684
4.79
3.15E−03
149.58%
27.72%


210664_s_at
546
4.73
8.77E−06
90.93%
34.92%
TFPI
tissue factor pathway inhibitor
Hs.170279









(lipoprotein-associated









coagulation inhibitor)


206067_s_at
423
4.72
2.81E−04


WT1
Wilms tumor 1
Hs.1145


209757_s_at
70
4.69
8.72E−06
90.78%
0.00%
MYCN
v-myc myelocytomatosis viral
Hs.25960









related oncogene,









neuroblastoma derived (avian)


213515_x_at
629
4.68
2.22E−05
95.77%
91.95%
GARS,
glycyl-tRNA synthetase,
Hs.356717,








HBG1,
hemoglobin, gamma A,
Hs.283108








HBG2
hemoglobin, gamma G


219837_s_at
769
4.60
2.68E−04
115.74%
34.92%
C17
cytokine-like protein C17
Hs.13872


218899_s_at
746
4.57
9.36E−04
129.54%
35.71%
BAALC
brain and acute leukemia,
Hs.169395









cytoplasmic


210665_at
75
4.55
5.86E−05
102.39%
28.60%
TFPI
tissue factor pathway inhibitor
Hs.170279









(lipoprotein-associated









coagulation inhibitor)


206478_at
436
4.52
1.57E−04
110.17%
39.54%
KIAA0125
KIAA0125 gene product
Hs.38365


201825_s_at
226
4.51
2.04E−07
72.49%
26.57%
LOC51097
CGI-49 protein
Hs.238126


202441_at
252
4.46
3.52E−09
59.64%
32.71%
KEO4
similar to Caenorhabditis
Hs.285818










elegans protein C42C1.9



209771_x_at
510
4.43
3.13E−02
206.78%
65.40%
CD24
CD24 antigen (small cell lung
Hs.375108









carcinoma cluster 4 antigen)


209160_at
484
4.38
3.56E−04
116.99%
34.40%
AKR1C3
aldo-keto reductase family 1,
Hs.78183









member C3 (3-alpha









hydroxysteroid









dehydrogenase, type II)


216379_x_at
680
4.38
2.65E−02
199.51%
62.52%
CD24,
CD24 antigen (small cell lung
Hs.381004








G22P1,
carcinoma cluster 4 antigen),








KIAA1919
KIAA1919 protein, thyroid









autoantigen 70 kD (Ku antigen)


206207_at
427
4.35
3.42E−02
209.28%
70.13%
CLC
Charot-Leyden crystal protein
Hs.889


204561_x_at
353
4.33
1.62E−02
182.63%
0.00%
APOC2
apolipoprotein C-II
Hs.75615


203372_s_at
16
4.33
4.22E−02
218.85%
18.42%
STATI2
STAT induced STAT inhibitor-2
Hs.405946


207269_at
448
4.30
9.46E−03
167.00%
84.09%
DEFA4
defensin, alpha 4, corticostatin
Hs.2582


218788_s_at
735
4.30
3.35E−06
83.45%
19.69%
FLJ21080
hypothetical protein FLJ21080
Hs.8109


211821_x_at
572
4.25
1.03E−03
128.12%
31.72%
GYPA
glycophorin A (includes MN
Hs.108694









blood group)


204419_x_at
347
4.25
5.06E−05
98.31%
100.03%
GARS,
glycyl-tRNA synthetase,
Hs.386655








HBG1,
hemoglobin, gamma A,








HBG2
hemoglobin, gamma G


213147_at
616
4.19
2.64E−05
94.35%
37.81%
HOXA10
homeo box A10
Hs.110637


221004_s_at
792
4.11
7.39E−06
86.29%
36.24%
ITM3
integral membrane protein 3
Hs.111577


204848_x_at
371
4.09
5.66E−05
97.77%
101.47%
HBG1,
hemoglobin, gamma A,
Hs.283108








HBG2
hemoglobin, gamma G


211560_s_at
563
4.08
9.01E−03
159.47%
191.88%
ALAS2
aminolevulinate, delta-,
Hs.381218









synthase 2









(sideroblastic/hypochromic









anemia)


206135_at
425
4.00
4.98E−02
221.44%
0.00%
ZNF387
zinc finger protein 387
Hs.151449


205366_s_at
6
3.87
2.03E−04
107.19%
30.33%
HOXB6
homeo box B6
Hs.98428


213110_s_at
612
3.87
2.06E−05
90.35%
32.83%
COL4A5
collagen, type IV, alpha 5
Hs.169825









(Alport syndrome)


219654_at
766
3.85
1.23E−06
75.89%
35.75%
PTPLA
protein tyrosine phosphatase-
Hs.114062









like (proline instead of catalytic









arginine), member a


201596_x_at
222
3.84
1.13E−03
125.06%
18.96%
KRT18
keratin 18
Hs.406013


220232_at
776
3.82
2.74E−07
69.76%
30.96%
FLJ21032
hypothetical protein FLJ21032
Hs.379191


207341_at
450
3.77
2.42E−03
134.65%
33.45%
PRTN3
proteinase 3 (serine
Hs.928









proteinase, neutrophil,









Wegener granulomatosis









autoantigen)


210746_s_at
547
3.73
7.35E−03
151.59%
136.15%
EPB42
erythrocyte membrane protein
Hs.733









band 4.2


201892_s_at
229
3.71
7.86E−08
64.85%
33.27%
IMPDH2
IMP (inosine monophosphate)
Hs.75432









dehydrogenase 2


214433_s_at
652
3.70
8.36E−03
153.06%
158.09%
SELENBP1
selenium binding protein 1
Hs.334841


218718_at
734
3.70
1.78E−06
76.48%
21.46%
PDGFC
platelet derived growth factor C
Hs.43080


213479_at
627
3.64
2.60E−02
187.19%
14.58%
NPTX2
neuronal pentraxin II
Hs.3281


201459_at
210
3.61
4.46E−07
70.09%
40.13%
RUVBL2
RuvB-like 2 (E. coli)
Hs.6455


218313_s_at
720
3.60
6.70E−07
71.60%
22.51%
GALNT7
UDP-N-acetyl-alpha-D-
Hs.246315









galactosamine:polypeptide N-









acetylgalactosaminyltransferase









7 (GalNAc-T7)


207459_x_at
451
3.59
3.58E−05
91.28%
28.85%
GYPA,
glycophorin A (includes MN
Hs.372513








GYPB
blood group), glycophorin B









(includes Ss blood group)


214407_x_at
651
3.58
2.91E−04
107.39%
22.02%
GYPA,
glycophorin A (includes MN
Hs.372513








GYPB
blood group), glycophorin B









(includes Ss blood group)


202502_at
254
3.58
1.42E−07
65.88%
20.33%
ACADM
acyl-Coenzyme A
Hs.79158









dehydrogenase, C-4 to C-12









straight chain


201418_s_at
207
3.55
7.35E−07
71.24%
61.97%
MEIS3,
Meis1, myeloid ecotropic viral
Hs.83484








SOX4
integration site 1 homolog 3









(mouse), SRY (sex









determining region Y)-box 4


209790_s_at
512
3.49
4.47E−05
91.75%
25.40%
CASP6
caspase 6, apoptosis-related
Hs.3280









cysteine protease


204069_at
325
3.48
3.01E−04
106.42%
25.85%
MEIS1
Meis1, myeloid ecotropic viral
Hs.170177









integration site 1 homolog









(mouse)


203502_at
295
3.46
5.36E−04
110.86%
77.38%
BPGM
2,3-bisphosphoglycerate
Hs.198365









mutase


206726_at
441
3.45
9.57E−03
155.35%
30.96%
PGDS
prostaglandin D2 synthase,
Hs.128433









hematopoietic


209813_x_at
513
3.42
9.06E−04
116.74%
46.61%
TRG@
T cell receptor gamma locus
Hs.112259


218332_at
721
3.40
1.19E−02
159.40%
27.69%
BEX1
brain expressed, X-linked 1
Hs.334370


219218_at
757
3.37
2.70E−05
87.16%
34.79%
FLJ23058
hypothetical protein FLJ23058
Hs.98968


211144_x_at
556
3.37
1.07E−03
117.91%
41.76%
TRG@
T cell receptor gamma locus
Hs.112259


202444_s_at
253
3.31
2.44E−10
47.88%
12.86%
KEO4
similar to Caenorhabditis
Hs.285818










elegans protein C42C1.9



201193_at
194
3.29
4.31E−05
89.35%
22.26%
IDH1
isocitrate dehydrogenase 1
Hs.11223









(NADP+), soluble


212175_s_at
587
3.28
2.59E−08
58.54%
25.74%
AK2
adenylate kinase 2
Hs.334802


205513_at
400
3.28
1.70E−03
122.27%
42.32%
TCN1
transcobalamin I (vitamin B12
Hs.2012









binding protein, R binder









family)


205592_at
403
3.25
3.97E−03
131.52%
121.76%
SLC4A1
solute carrier family 4, anion
Hs.432645









exchanger, member 1









(erythrocyte membrane protein









band 3, Diego blood group)


205769_at
413
3.24
1.32E−05
81.73%
33.71%
FACVL1
fatty-acid-Coenzyme A ligase,
Hs.11729









very long-chain 1


212141_at
586
3.19
7.85E−05
92.20%
0.00%
MCM4
MCM4 minichromosome
Hs.154443









maintenance deficient 4 (S. cerevisiae)


213541_s_at
631
3.17
2.40E−09
51.84%
32.90%
ERG
v-ets erythroblastosis virus
Hs.45514









E26 oncogene like (avian)


204468_s_at
9
3.17
1.48E−02
160.05%
0.00%
TIE
tyrosine kinase with
Hs.78824









immunoglobulin and epidermal









growth factor homology









domains


222036_s_at
807
3.16
1.44E−04
96.14%
7.37%
MCM4
MCM4 minichromosome
Hs.319215









maintenance deficient 4 (S. cerevisiae)


220668_s_at
39
3.15
2.45E−07
64.13%
20.33%
DNMT3B
DNA (cytosine-5-)-
Hs.251673









methyltransferase 3 beta


218847_at
741
3.15
2.96E−12
40.44%
50.24%
IMP-2
IGF-II mRNA-binding protein 2
Hs.30299


217294_s_at
691
3.14
2.68E−08
57.40%
44.65%
ENO1
enolase 1, (alpha)
Hs.381397


213779_at
636
3.12
5.52E−07
66.61%
27.57%
LOC129080
putative emu1
Hs.289106


218825_at
738
3.12
7.45E−07
67.61%
35.39%
LOC51162
NEU1 protein
Hs.91481


218858_at
743
3.09
1.82E−05
81.78%
17.08%
FLJ12428
hypothetical protein FLJ12428
Hs.87729


216153_x_at
676
3.08
8.64E−06
77.60%
35.89%
RECK
reversion-inducing-cysteine-
Hs.29640









rich protein with kazal motifs


204467_s_at
351
3.08
3.20E−02
176.33%
158.31%
SNCA
synuclein, alpha (non A4
Hs.76930









component of amyloid









precursor)


204409_s_at
345
3.08
8.03E−04
109.25%
66.65%
EIF1AY
eukaryotic translation initiation
Hs.155103









factor 1A, Y chromosome


205202_at
384
3.05
2.34E−05
82.67%
22.02%
PCMT1
protein-L-isoaspartate (D-
Hs.79137









aspartate) O-









methyltransferase


205382_s_at
394
3.05
2.83E−05
83.59%
34.99%
DF
D component of complement
Hs.155597









(adipsin)


209576_at
503
3.04
7.79E−04
109.41%
14.58%
GNAI1
guanine nucleotide binding
Hs.203862









protein (G protein), alpha









inhibiting activity polypeptide 1


211546_x_at
562
3.03
6.29E−03
136.16%
91.15%
SNCA
synuclein, alpha (non A4
Hs.76930









component of amyloid









precursor)


212115_at
585
3.02
4.78E−04
103.69%
45.78%
FLJ13092
hypothetical protein FLJ13092
Hs.172035


211820_x_at
571
3.01
6.29E−04
106.39%
33.71%
GYPA
glycophorin A (includes MN
Hs.108694









blood group)


210254_at
530
2.98
6.65E−03
137.19%
59.25%
MS4A3
membrane-spanning 4-
Hs.99960









domains, subfamily A, member









3 (hematopoietic cell-specific)


210829_s_at
550
2.97
2.80E−05
82.60%
20.75%
SSBP2
single-stranded DNA binding
Hs.424652









protein 2


200923_at
177
2.97
1.47E−04
93.21%
32.12%
LGALS3BP
lectin, galactoside-binding,
Hs.79339









soluble, 3 binding protein


204900_x_at
375
2.96
1.38E−04
92.64%
31.39%
SAP30
sin3-associated polypeptide,
Hs.20985









30 kD


202845_s_at
268
2.95
1.36E−07
59.80%
60.88%
RALBP1
ralA binding protein 1
Hs.75447


203787_at
307
2.94
3.89E−05
83.97%
20.55%
SSBP2
single-stranded DNA binding
Hs.169833









protein 2


206622_at
437
2.93
4.83E−02
193.09%
26.43%
TRH
thyrotropin-releasing hormone
Hs.182231


201413_at
205
2.93
5.86E−08
57.63%
26.79%
HSD17B4
hydroxysteroid (17-beta)
Hs.75441









dehydrogenase 4


201054_at
189
2.91
2.70E−07
62.01%
29.74%
HNRPA0
heterogeneous nuclear
Hs.77492









ribonucleoprotein A0


204647_at
360
2.90
2.54E−04
96.25%
29.14%
HOMER-3
Homer, neuronal immediate
Hs.424053









early gene, 3


219789_at
768
2.89
4.95E−06
72.67%
26.79%
NPR3
natriuretic peptide receptor
Hs.123655









C/guanylate cyclase C









(atrionatriuretic peptide









receptor C)


204011_at
319
2.88
7.38E−04
105.71%
21.81%
SPRY2
sprouty homolog 2
Hs.18676









(Drosophila)


204391_x_at
343
2.87
4.74E−11
42.14%
25.33%
TIF1
transcriptional intermediary
Hs.183858









factor 1


205844_at
415
2.85
9.58E−03
141.91%
32.83%
VNN1
vanin 1
Hs.12114


209183_s_at
485
2.85
1.07E−03
108.94%
19.95%
DEPP
decidual protein induced by
Hs.93675









progesterone


214657_s_at
661
2.82
1.23E−06
66.05%
31.54%
MEN1
multiple endocrine neoplasia I
Hs.434021


200615_s_at
157
2.81
6.19E−08
56.39%
39.24%
AP2B1
adaptor-related protein
Hs.74626









complex 2, beta 1 subunit


204466_s_at
350
2.80
1.14E−02
141.03%
106.77%
SNCA
synuclein, alpha (non A4
Hs.76930









component of amyloid









precursor)


215537_x_at
672
2.80
1.10E−06
65.18%
41.33%
DDAH2
dimethylarginine
Hs.247362









dimethylaminohydrolase 2


206480_at
66
2.79
4.45E−05
82.52%
19.95%
LTC4S
leukotriene C4 synthase
Hs.456


222067_x_at
809
2.77
5.86E−06
71.70%
31.83%
H2BFB
H2B histone family, member B
Hs.180779


204173_at
333
2.77
4.04E−12
37.74%
23.97%
MLC1SA
myosin light chain 1 slow a
Hs.90318


204885_s_at
372
2.77
2.56E−02
164.20%
19.95%
MSLN
mesothelin
Hs.155981


212268_at
593
2.75
5.30E−08
55.45%
22.18%
SERPINB1
serine (or cysteine) proteinase
Hs.183583









inhibitor, clade B (ovalbumin),









member 1


215182_x_at
667
2.75
2.81E−08
53.77%
25.51%


Hs.274511


201037_at
188
2.75
1.97E−06
66.97%
23.73%
PFKP
phosphofructokinase, platelet
Hs.99910


205900_at
418
2.75
2.10E−02
151.32%
152.69%
KRT1
keratin 1 (epidermolytic
Hs.80828









hyperkeratosis)


214236_at
648
2.74
4.55E−04
98.32%
26.79%


Hs.343877


210644_s_at
544
2.74
4.64E−08
54.96%
29.13%
LAIR1
leukocyte-associated Ig-like
Hs.115808









receptor 1


201563_at
217
2.73
1.24E−06
64.94%
22.33%
SORD
sorbitol dehydrogenase
Hs.878


210395_x_at
535
2.72
1.04E−02
139.39%
52.16%
MYL4
myosin, light polypeptide 4,
Hs.356717









alkali; atrial, embryonic


213301_x_at
621
2.72
5.42E−10
45.00%
23.44%
TIF1
transcriptional intermediary
Hs.183858









factor 1


218039_at
709
2.71
1.12E−06
64.37%
23.77%
ANKT
nucleolar protein ANKT
Hs.279905


218069_at
710
2.70
1.77E−05
75.65%
39.91%
MGC5627
hypothetical protein MGC5627
Hs.237971


203588_s_at
300
2.69
2.26E−06
66.62%
29.27%
TFDP2
transcription factor Dp-2 (E2F
Hs.379018









dimerization partner 2)


218883_s_at
745
2.68
1.49E−05
74.69%
22.08%
FLJ23468
hypothetical protein FLJ23468
Hs.38178


209360_s_at
493
2.67
3.42E−07
59.70%
35.04%
RUNX1
runt-related transcription factor
Hs.129914









1 (acute myeloid leukemia 1;









aml1 oncogene)


201503_at
212
2.66
4.32E−05
80.08%
23.20%
G3BP
Ras-GTPase-activating protein
Hs.220689









SH3-domain-binding protein


200696_s_at
160
2.65
2.10E−08
51.86%
26.02%
GSN
gelsolin (amyloidosis, Finnish
Hs.290070









type)


216054_x_at
675
2.63
6.99E−03
128.94%
51.23%
MYL4
myosin, light polypeptide 4,
Hs.433562









alkali; atrial, embryonic


218342_s_at
722
2.62
1.78E−08
51.17%
29.01%
FLJ23309
hypothetical protein FLJ23309
Hs.87128


209825_s_at
514
2.62
1.18E−07
55.95%
20.26%
UMPK
uridine monophosphate kinase
Hs.95734


217975_at
24
2.60
3.93E−05
78.27%
30.22%
LOC51186
pp21 homolog
Hs.15984


217791_s_at
697
2.60
3.00E−08
52.16%
27.47%
PYCS
pyrroline-5-carboxylate
Hs.114366









synthetase (glutamate









gamma-semialdehyde









synthetase)


203662_s_at
302
2.60
3.81E−03
115.58%
96.82%
TMOD
tropomodulin
Hs.374849


208967_s_at
481
2.59
1.23E−09
45.20%
19.58%
AK2
adenylate kinase 2
Hs.294008


202371_at
249
2.59
4.15E−06
67.51%
23.93%
FLJ21174
hypothetical protein FLJ21174
Hs.194329


212055_at
583
2.59
1.69E−06
63.82%
35.39%
DKFZP586M1523
DKFZP586M1523 protein
Hs.22981


200703_at
161
2.58
6.22E−05
80.36%
34.35%
PIN
dynein, cytoplasmic, light
Hs.5120









polypeptide


202262_x_at
245
2.57
1.20E−07
55.38%
30.08%
DDAH2
dimethylarginine
Hs.247362









dimethylaminohydrolase 2


209200_at
487
2.56
5.08E−04
95.07%
35.56%
MEF2C
MADS box transcription
Hs.78995









enhancer factor 2, polypeptide









C (myocyte enhancer factor









2C)


213572_s_at
632
2.56
6.00E−07
60.04%
24.71%
SERPINB1
serine (or cysteine) proteinase
Hs.183583









inhibitor, clade B (ovalbumin),









member 1


210762_s_at
548
2.56
1.07E−04
83.59%
21.67%
DLC1
deleted in liver cancer 1
Hs.8700


200658_s_at
159
2.56
1.37E−06
62.62%
33.60%
PHB
prohibitin
Hs.75323


201325_s_at
202
2.56
1.02E−03
101.41%
34.91%
EMP1
epithelial membrane protein 1
Hs.79368


210999_s_at
554
2.56
4.21E−06
67.09%
10.66%
GRB10
growth factor receptor-bound
Hs.81875









protein 10


205518_s_at
401
2.55
7.90E−09
48.51%
21.91%
CMAH
cytidine monophosphate-N-









acetylneuraminic acid









hydroxylase (CMP-N-









acetylneuraminate









monooxygenase)


217809_at
698
2.55
6.77E−09
48.13%
20.59%
HSPC028
HSPC028 protein
Hs.5216


210088_x_at
526
2.54
1.55E−02
142.11%
53.21%
MYL4
myosin, light polypeptide 4,
Hs.433562









alkali; atrial, embryonic


220725_x_at
785
2.54
1.18E−07
54.83%
20.23%
FLJ23558
hypothetical protein FLJ23558
Hs.288552


208857_s_at
472
2.54
7.84E−06
69.20%
24.21%
PCMT1
protein-L-isoaspartate (D-
Hs.79137









aspartate) O-









methyltransferase


210401_at
536
2.53
1.55E−09
45.09%
36.41%
P2RX1
purinergic receptor P2X,
Hs.41735









ligand-gated ion channel, 1


201555_at
215
2.53
9.94E−06
70.17%
23.11%
MCM3
MCM3 minichromosome
Hs.179565









maintenance deficient 3 (S. cerevisiae)


202708_s_at
260
2.53
1.43E−04
84.55%
34.53%
H2BFQ
H2B histone family, member Q
Hs.2178


208651_x_at
464
2.53
2.33E−02
151.82%
55.28%
CD24
CD24 antigen (small cell lung
Hs.375108









carcinoma cluster 4 antigen)


201951_at
230
2.52
5.47E−05
78.34%
35.71%
ALCAM
activated leucocyte cell
Hs.10247









adhesion molecule


201564_s_at
218
2.52
9.43E−05
81.60%
35.59%
SNL
singed-like (fascin homolog,
Hs.118400









sea urchin) (Drosophila)


220807_at
787
2.51
1.86E−02
142.62%
100.98%
HBQ1
hemoglobin, theta 1
Hs.247921


201005_at
183
2.51
1.68E−03
104.10%
68.43%
CD9
CD9 antigen (p24)
Hs.1244


205801_s_at
414
2.50
5.77E−03
121.93%
35.56%
GRP3
guanine nucleotide exchange
Hs.24024









factor for Rap1


221521_s_at
797
2.50
6.08E−03
123.19%
14.58%
LOC51659
HSPC037 protein
Hs.433180


208690_s_at
467
2.50
5.11E−07
58.47%
25.48%
PDLIM1
PDZ and LIM domain 1 (elfin)
Hs.75807


201015_s_at
184
2.48
1.26E−04
81.37%
61.73%
JUP
junction plakoglobin
Hs.2340


203661_s_at
301
2.47
4.13E−03
114.18%
73.79%
TMOD
tropomodulin
Hs.374849


266_s_at
814
2.46
3.21E−02
159.03%
38.81%
CD24
CD24 antigen (small cell lung
Hs.375108









carcinoma cluster 4 antigen)


209409_at
496
2.46
2.57E−06
63.47%
10.66%
GRB10
growth factor receptor-bound
Hs.81875









protein 10


203560_at
299
2.46
1.44E−04
83.27%
16.83%
GGH
gamma-glutamyl hydrolase
Hs.78619









(conjugase,









folylpolygammaglutamyl









hydrolase)


213170_at
618
2.45
5.82E−10
42.28%
21.81%
CL683
weakly similar to glutathione
Hs.43728









peroxidase 2


205227_at
11
2.45
6.61E−05
77.91%
32.30%
IL1RAP
interleukin 1 receptor
Hs.173880









accessory protein


218927_s_at
747
2.44
1.69E−05
70.44%
42.51%
C4S-2
chondroitin 4-O-
Hs.25204









sulfotransferase 2


209318_x_at
492
2.44
7.63E−06
67.41%
20.62%
PLAGL1
pleiomorphic adenoma gene-
Hs.75825









like 1


214106_s_at
645
2.43
4.48E−03
116.13%
23.65%
GMDS
GDP-mannose 4,6-
Hs.105435









dehydratase


213346_at
623
2.43
8.55E−06
67.73%
20.13%
LOC93081
hypothetical protein BC015148
Hs.13413


205418_at
395
2.43
2.60E−04
86.33%
37.54%
FES
feline sarcoma oncogene
Hs.7636


220051_at
773
2.43
2.32E−02
148.56%
15.25%
PRSS21
protease, serine, 21 (testisin)
Hs.72026


202107_s_at
239
2.43
8.20E−05
78.99%
21.20%
MCM2
MCM2 minichromosome
Hs.57101









maintenance deficient 2,









mitotin (S. cerevisiae)


202862_at
271
2.42
3.03E−07
55.80%
20.78%
FAH
fumarylacetoacetate hydrolase
Hs.73875









(fumarylacetoacetase)


204086_at
327
2.42
4.35E−02
167.93%
24.76%
PRAME
preferentially expressed
Hs.30743









antigen in melanoma


212526_at
601
2.42
2.71E−06
62.96%
7.37%
KIAA0610
KIAA0610 protein
Hs.118087


210358_x_at
533
2.42
1.91E−06
61.37%
32.70%
GATA2,
GATA binding protein 2,
Hs.760








MGC2306
hypothetical protein MGC2306


220615_s_at
782
2.41
7.40E−04
94.63%
30.22%
FLJ10462
hypothetical protein FLJ10462
Hs.100895


205612_at
408
2.40
3.50E−02
159.14%
23.65%
MMRN
multimerin
Hs.268107


200648_s_at
158
2.39
5.01E−04
89.77%
52.01%
GLUL
glutamate-ammonia ligase
Hs.170171









(glutamine synthase)


201277_s_at
198
2.39
4.92E−06
64.59%
19.32%
HNRPAB
heterogeneous nuclear
Hs.81361









ribonucleoprotein A/B


210044_s_at
522
2.39
2.22E−09
43.75%
45.66%
LYL1
lymphoblastic leukemia
Hs.46446









derived sequence 1


214501_s_at
656
2.38
2.15E−08
48.45%
21.49%
H2AFY
H2A histone family, member Y
Hs.75258


201240_s_at
196
2.37
6.69E−07
56.91%
36.63%
KIAA0102
KIAA0102 gene product
Hs.77665


208626_s_at
463
2.36
2.87E−08
48.71%
24.12%
VATI
vesicle amine transport protein 1
Hs.157236


205349_at
393
2.35
2.52E−05
70.03%
46.83%
GNA15
guanine nucleotide binding
Hs.73797









protein (G protein), alpha 15









(Gq class)


216833_x_at
686
2.35
4.00E−04
87.94%
12.86%
GYPB,
glycophorin B (includes Ss
Hs.372513








GYPE
blood group), glycophorin E


218026_at
707
2.34
5.33E−06
63.97%
21.95%
HSPC009
HSPC009 protein
Hs.16059


211464_x_at
560
2.34
2.51E−06
60.85%
35.12%
CASP6
caspase 6, apoptosis-related
Hs.3280









cysteine protease


208677_s_at
466
2.34
1.72E−08
47.26%
31.21%
BSG
basigin (OK blood group)
Hs.74631


203744_at
306
2.34
2.96E−13
31.01%
19.36%
HMG4
high-mobility group
Hs.19114









(nonhistone chromosomal)









protein 4


212358_at
596
2.34
2.49E−02
146.05%
33.71%
CLIPR-59
CLIP-170-related protein
Hs.7357


201036_s_at
187
2.33
1.53E−05
68.07%
19.36%
HADHSC
L-3-hydroxyacyl-Coenzyme A
Hs.8110









dehydrogenase, short chain


205600_x_at
404
2.33
1.45E−07
51.99%
32.81%
HOXB5
homeo box B5
Hs.22554


219007_at
750
2.31
1.48E−05
67.23%
30.35%
FLJ13287
hypothetical protein FLJ13287
Hs.53263


201069_at
190
2.31
3.71E−03
109.02%
24.70%
MMP2
matrix metalloproteinase 2
Hs.111301









(gelatinase A, 72 kD









gelatinase, 72 kD type IV









collagenase)


201231_s_at
195
2.30
5.73E−10
40.37%
18.11%
ENO1
enolase 1, (alpha)
Hs.254105


218409_s_at
724
2.29
1.56E−03
98.22%
22.49%
DNAJL1
hypothetical protein similar to
Hs.13015









mouse Dnajl1


221471_at
795
2.29
1.27E−08
45.85%
23.06%
TDE1
tumor differentially expressed 1
Hs.272168


216705_s_at
685
2.28
8.43E−07
56.23%
28.91%
ADA
adenosine deaminase
Hs.1217


205601_s_at
405
2.28
3.00E−05
70.06%
24.09%
HOXB5
homeo box B5
Hs.22554


209208_at
489
2.28
3.02E−07
53.16%
28.79%
MPDU1
mannose-P-dolichol utilization
Hs.6710









defect 1


218188_s_at
716
2.27
2.80E−08
47.33%
21.04%
TIMM13
translocase of inner
Hs.23410









mitochondrial membrane 13









homolog (yeast)


200983_x_at
182
2.27
8.67E−06
64.32%
25.73%
CD59
CD59 antigen p18-20 (antigen
Hs.278573









identified by monoclonal









antibodies 16.3A5, EJ16,









EJ30, EL32 and G344)


208964_s_at
480
2.27
3.72E−10
39.28%
19.16%
FADS1
fatty acid desaturase 1
Hs.132898


217274_x_at
690
2.27
2.17E−03
99.73%
56.76%
MYL4
myosin, light polypeptide 4,
Hs.433562









alkali; atrial, embryonic


210365_at
534
2.27
1.71E−05
66.55%
41.85%
RUNX1
runt-related transcription factor
Hs.129914









1 (acute myeloid leukemia 1;









aml1 oncogene)


214455_at
653
2.27
2.04E−03
100.36%
21.81%
H2BFA,
H2B histone family, member
Hs.356901








H2BFL
A, H2B histone family,









member L


220741_s_at
786
2.27
1.33E−06
57.27%
31.33%
SID6-306
inorganic pyrophosphatase
Hs.375016


218585_s_at
728
2.25
6.54E−04
88.37%
35.75%
RAMP
RA-regulated nuclear matrix-
Hs.126774









associated protein


205608_s_at
406
2.25
3.35E−08
47.27%
23.20%
ANGPT1
angiopoietin 1
Hs.2463


205453_at
397
2.24
9.34E−05
74.65%
34.31%
HOXB2
homeo box B2
Hs.2733


201890_at
228
2.24
5.28E−03
111.27%
22.47%
RRM2
ribonucleotide reductase M2
Hs.75319









polypeptide


204386_s_at
342
2.23
2.36E−07
51.76%
22.35%
MRP63
mitochondrial ribosomal
Hs.182695









protein 63


210052_s_at
523
2.23
9.78E−07
55.82%
20.14%
C20orf1
chromosome 20 open reading
Hs.9329









frame 1


208898_at
477
2.23
1.62E−07
50.69%
23.80%
ATP6V1D
ATPase, H+ transporting,
Hs.272630









lysosomal 34 kD, V1 subunit D


200821_at
170
2.22
5.72E−08
47.87%
26.92%
LAMP2
lysosomal-associated
Hs.8262









membrane protein 2


207719_x_at
455
2.21
2.09E−13
29.62%
22.01%
KIAA0470
KIAA0470 gene product
Hs.25132


204438_at
21
2.21
2.04E−03
98.49%
17.08%
MRC1
mannose receptor, C type 1
Hs.75182


209199_s_at
486
2.21
5.25E−05
70.69%
35.75%
MEF2C
MADS box transcription
Hs.78995









enhancer factor 2, polypeptide









C (myocyte enhancer factor









2C)


214500_at
655
2.21
5.45E−04
85.81%
30.19%
H2AFY
H2A histone family, member Y
Hs.75258


201028_s_at
186
2.21
3.32E−06
59.25%
21.39%
MIC2
antigen identified by
Hs.433387









monoclonal antibodies 12E7,









F21 and O13


209395_at
495
2.21
3.51E−02
148.36%
52.07%
CHI3L1
chitinase 3-like 1 (cartilage
Hs.75184









glycoprotein-39)


216554_s_at
683
2.20
5.42E−13
30.22%
18.05%
ENO1
enolase 1, (alpha)
Hs.381397


222294_s_at
812
2.20
2.12E−04
78.67%
31.23%


Hs.432533


203688_at
303
2.20
3.64E−06
59.34%
25.67%
PKD2
polycystic kidney disease 2
Hs.82001









(autosomal dominant)


200728_at
163
2.20
2.37E−12
32.00%
25.79%
ACTR2
ARP2 actin-related protein 2
Hs.396278









homolog (yeast)


201562_s_at
216
2.20
1.75E−14
27.69%
29.44%
SORD
sorbitol dehydrogenase
Hs.878


211714_x_at
567
2.19
5.66E−07
53.34%
16.95%
FKBP1A
FK506 binding protein 1A
Hs.179661









(12 kD)


206057_x_at
422
2.19
7.42E−12
33.11%
25.12%
SPN
sialophorin (gpL115,
Hs.80738









leukosialin, CD43)


207761_s_at
457
2.19
8.33E−06
62.25%
19.69%
DKFZP586A0522
DKFZP586A0522 protein
Hs.288771


200769_s_at
165
2.18
1.09E−07
48.80%
26.93%
MAT2A
methionine
Hs.77502









adenosyltransferase II, alpha


206665_s_at
439
2.18
4.65E−03
106.39%
44.14%
BCL2L1
BCL2-like 1
Hs.305890


208858_s_at
473
2.17
2.26E−07
50.14%
37.12%
KIAA0747
KIAA0747 protein
Hs.8309


205239_at
386
2.17
3.39E−02
144.04%
72.62%
AREG
amphiregulin (schwannoma-
Hs.270833









derived growth factor)


205919_at
419
2.17
4.72E−03
105.44%
54.93%
HBE1
hemoglobin, epsilon 1
Hs.117848


203253_s_at
288
2.17
1.36E−08
44.04%
22.47%
KIAA0433
KIAA0433 protein
Hs.26179


210549_s_at
542
2.17
8.57E−04
88.61%
0.00%
SCYA23
small inducible cytokine
Hs.169191









subfamily A (Cys-Cys),









member 23


201329_s_at
203
2.16
5.35E−04
82.28%
57.70%
ETS2
v-ets erythroblastosis virus
Hs.85146









E26 oncogene homolog 2









(avian)


204429_s_at
348
2.16
1.40E−05
63.30%
28.97%
SLC2A5
solute carrier family 2
Hs.33084









(facilitated glucose/fructose









transporter), member 5


218136_s_at
713
2.15
3.01E−02
137.41%
93.36%
LOC51312
mitochondrial solute carrier
Hs.283716


200806_s_at
168
2.15
1.71E−06
55.72%
20.60%
HSPD1
heat shock 60 kD protein 1
Hs.79037









(chaperonin)


212296_at
594
2.15
9.97E−09
43.04%
17.60%
POH1
26S proteasome-associated
Hs.178761









pad1 homolog


218160_at
715
2.14
4.05E−06
58.42%
24.57%
NDUFA8
NADH dehydrogenase
Hs.31547









(ubiquinone) 1 alpha









subcomplex, 8 (19 kD, PGIV)


204039_at
323
2.14
7.35E−04
85.48%
36.46%
CEBPA
CCAAT/enhancer binding
Hs.76171









protein (C/EBP), alpha


200727_s_at
162
2.14
4.97E−11
34.77%
36.28%
ACTR2
ARP2 actin-related protein 2
Hs.393201









homolog (yeast)


48808_at
823
2.13
4.23E−02
151.12%
14.58%
DHFR
dihydrofolate reductase
Hs.83765


222037_at
808
2.13
3.35E−04
79.27%
35.71%
MCM4
MCM4 minichromosome
Hs.319215









maintenance deficient 4 (S. cerevisiae)


202345_s_at
248
2.13
8.72E−04
86.92%
27.92%
FABP5
fatty acid binding protein 5
Hs.153179









(psoriasis-associated)


210036_s_at
521
2.12
1.28E−03
90.00%
31.48%
KCNH2
potassium voltage-gated
Hs.188021









channel, subfamily H (eag-









related), member 2


200812_at
169
2.12
1.07E−05
61.36%
26.73%
CCT7
chaperonin containing TCP1,
Hs.108809









subunit 7 (eta)


202974_at
277
2.12
2.27E−04
75.68%
43.58%
MPP1
membrane protein,
Hs.1861









palmitoylated 1 (55 kD)


201577_at
221
2.11
1.31E−07
47.86%
22.32%
NME1
non-metastatic cells 1, protein
Hs.118638









(NM23A) expressed in


202201_at
241
2.11
1.87E−03
92.07%
49.52%
BLVRB
biliverdin reductase B (flavin
Hs.76289









reductase (NADPH))


210849_s_at
552
2.11
1.31E−10
35.54%
31.11%
VPS41
vacuolar protein sorting 41
Hs.180941









(yeast)


209365_s_at
494
2.10
3.90E−06
56.91%
34.40%
ECM1
extracellular matrix protein 1
Hs.81071


217988_at
705
2.10
8.48E−06
60.04%
23.33%
HEI10
enhancer of invasion 10
Hs.107003


203904_x_at
313
2.10
4.53E−08
45.10%
27.01%
KAI1
kangai 1 (suppression of
Hs.323949









tumorigenicity 6, prostate;









CD82 antigen (R2 leukocyte









antigen, antigen detected by









monoclonal and antibody IA4))


200986_at
35
2.09
1.08E−04
71.48%
22.84%
SERPING1
serine (or cysteine) proteinase
Hs.151242









inhibitor, clade G (C1









inhibitor), member 1,









(angioedema, hereditary)


201491_at
211
2.09
7.56E−06
59.51%
18.40%
C14orf3
chromosome 14 open reading
Hs.204041









frame 3


200942_s_at
178
2.09
1.47E−08
42.77%
22.51%
HSBP1
heat shock factor binding
Hs.250899









protein 1


200973_s_at
181
2.09
8.67E−08
46.27%
30.93%
TSPAN-3
tetraspan 3
Hs.100090


207943_x_at
459
2.09
2.78E−09
39.76%
25.61%
PLAGL1
pleiomorphic adenoma gene-
Hs.75825









like 1


208899_x_at
478
2.09
3.61E−09
40.15%
27.32%
ATP6V1D
ATPase, H+ transporting,
Hs.272630









lysosomal 34 kD, V1 subunit D


204187_at
334
2.09
3.03E−02
133.16%
94.60%
GMPR
guanosine monophosphate
Hs.1435









reductase


220240_s_at
777
2.08
2.48E−07
48.85%
18.46%
FLJ20623
hypothetical protein FLJ20623
Hs.27337


218966_at
749
2.08
3.83E−05
65.76%
27.14%
MYO5C
myosin 5C
Hs.111782


214321_at
649
2.07
4.28E−02
146.79%
35.71%
NOV
nephroblastoma
Hs.235935









overexpressed gene


211769_x_at
570
2.07
2.26E−09
39.09%
24.73%
TDE1
tumor differentially expressed 1
Hs.272168


202990_at
279
2.07
1.72E−04
73.21%
26.24%
PYGL
phosphorylase, glycogen; liver
Hs.771









(Hers disease, glycogen









storage disease type VI)


202429_s_at
251
2.06
5.39E−06
57.32%
26.50%
PPP3CA
protein phosphatase 3
Hs.272458









(formerly 2B), catalytic subunit,









alpha isoform (calcineurin A









alpha)


209215_at
490
2.06
2.44E−05
62.66%
37.86%
TETRAN
tetracycline transporter-like
Hs.157145









protein


217949_s_at
703
2.06
9.23E−06
59.41%
20.57%
IMAGE3455200
hypothetical protein
Hs.324844









IMAGE3455200


205330_at
392
2.06
9.95E−03
112.06%
45.65%
MN1
meningioma (disrupted in
Hs.268515









balanced translocation) 1


218027_at
708
2.06
7.08E−08
45.38%
19.16%
MRPL15
mitochondrial ribosomal
Hs.18349









protein L15


219479_at
761
2.06
6.63E−04
82.11%
23.65%
MGC5302
endoplasmic reticulum
Hs.44970









resident protein 58;









hypothetical protein MGC5302


215416_s_at
671
2.06
1.08E−10
34.37%
18.21%
STOML2
stomatin (EPB72)-like 2
Hs.3439


221479_s_at
796
2.06
9.03E−03
110.65%
34.64%
BNIP3L
BCL2/adenovirus E1B 19 kD
Hs.132955









interacting protein 3-like


215285_s_at
669
2.05
1.83E−03
90.98%
18.13%
PHTF1
putative homeodomain
Hs.123637









transcription factor 1


219559_at
763
2.05
9.10E−10
37.29%
24.99%
C20orf59
chromosome 20 open reading
Hs.353013









frame 59


211342_x_at
557
2.05
4.07E−08
42.42%
51.95%
TNRC11
trinucleotide repeat containing
Hs.211607









11 (THR-associated protein,









230 kD subunit)


210298_x_at
71
2.05
4.94E−03
101.70%
26.72%
FHL1
four and a half LIM domains 1
Hs.239069


217724_at
694
2.04
6.51E−07
50.51%
16.73%
PAI-RBP1
PAI-1 mRNA-binding protein
Hs.165998


208817_at
471
2.04
1.23E−08
41.49%
24.81%
COMT
catechol-O-methyltransferase
Hs.240013


204040_at
324
2.04
1.37E−05
60.01%
30.27%
KIAA0161
KIAA0161 gene product
Hs.78894


213854_at
639
2.04
4.56E−07
49.43%
20.27%
SYNGR1
synaptogyrin 1
Hs.6139


200729_s_at
164
2.04
1.28E−11
31.75%
24.98%
ACTR2
ARP2 actin-related protein 2
Hs.393201









homolog (yeast)


201970_s_at
232
2.04
3.64E−04
76.63%
31.58%
NASP
nuclear autoantigenic sperm
Hs.380400









protein (histone-binding)


203021_at
280
2.03
3.92E−04
76.95%
33.19%
SLPI
secretory leukocyte protease
Hs.251754









inhibitor (antileukoproteinase)


200900_s_at
175
2.03
8.48E−06
58.01%
25.64%
M6PR
mannose-6-phosphate
Hs.134084









receptor (cation dependent)


203800_s_at
308
2.03
7.24E−07
50.35%
21.68%
MRPS14
mitochondrial ribosomal
Hs.247324









protein S14


212320_at
595
2.02
2.59E−07
47.68%
15.36%


Hs.179661


217892_s_at
701
2.02
1.64E−10
34.53%
25.93%
ARL4,
ADP-ribosylation factor-like 4,
Hs.10706








EPLIN
epithelial protein lost in









neoplasm beta


218270_at
719
2.02
2.16E−05
61.02%
34.29%
MRPL24
mitochondrial ribosomal
Hs.9265









protein L24


201302_at
199
2.02
1.45E−05
59.43%
31.19%
ANXA4
annexin A4
Hs.77840


214113_s_at
61
2.02
4.98E−06
56.07%
12.21%
RBM8A
RNA binding motif protein 8A
Hs.10283


206438_x_at
434
2.01
2.03E−11
31.90%
26.02%
FLJ12975
hypothetical protein FLJ12975
Hs.167165


205505_at
399
2.01
1.77E−05
60.46%
21.22%
GCNT1
glucosaminyl (N-acetyl)
Hs.159642









transferase 1, core 2 (beta-









1,6-N-









acetylglucosaminyltransferase)


209515_s_at
499
2.01
6.79E−05
66.13%
27.14%
RAB27A
RAB27A, member RAS
Hs.50477









oncogene family


221831_at
802
2.01
1.72E−04
69.36%
52.04%


Hs.348515


221942_s_at
806
2.01
1.14E−07
44.95%
33.24%
GUCY1A3
guanylate cyclase 1, soluble,
Hs.75295









alpha 3


213797_at
101
2.01
4.76E−04
77.51%
26.86%
cig5
vipirin
Hs.17518


209517_s_at
500
2.00
4.18E−09
38.85%
19.12%
ASH2L
ash2 (absent, small, or
Hs.6856









homeotic)-like (Drosophila)


213617_s_at
634
2.00
2.38E−09
37.89%
23.87%
DKFZP586M1523
DKFZP586M1523 protein
Hs.22981


214390_s_at
650
2.00
1.54E−02
116.91%
34.44%
BCAT1
branched chain
Hs.317432









aminotransferase 1, cytosolic


219423_x_at
760
0.50
8.47E−11
61.84%
27.11%
TNFRSF12
tumor necrosis factor receptor
Hs.180338









superfamily, member 12









(translocating chain-









association membrane protein)


35626_at
816
0.50
1.86E−06
91.46%
39.11%
SGSH
N-sulfoglucosamine
Hs.31074









sulfohydrolase (sulfamidase)


211984_at
581
0.50
2.35E−15
48.17%
17.35%


Hs.374441


200965_s_at
180
0.50
6.00E−07
96.72%
24.80%
ABLIM
actin binding LIM protein
Hs.158203


201531_at
214
0.50
7.92E−11
59.64%
30.26%
ZFP36
zinc finger protein 36, C3H
Hs.343586









type, homolog (mouse)


205022_s_at
379
0.49
3.82E−12
26.84%
36.11%
CHES1
checkpoint suppressor 1
Hs.211773


207697_x_at
454
0.49
3.04E−09
78.11%
19.85%
LILRB1,
leukocyte immunoglobulin-like
Hs.22405








LILRB2
receptor, subfamily B (with TM









and ITIM domains), member 1,









leukocyte immunoglobulin-like









receptor, subfamily B (with TM









and ITIM domains), member 2


205019_s_at
378
0.49
1.92E−10
62.69%
30.88%
VIPR1
vasoactive intestinal peptide
Hs.348500









receptor 1


210845_s_at
551
0.49
1.37E−07
66.07%
46.38%
PLAUR
plasminogen activator,
Hs.179657









urokinase receptor


213831_at
637
0.49
1.63E−03
90.56%
91.29%
HLA-DQA1
major histocompatibility
Hs.198253









complex, class II, DQ alpha 1


203341_at
292
0.49
6.80E−17
34.29%
25.70%
CBF2
CCAAT-box-binding
Hs.184760









transcription factor


209657_s_at
506
0.49
6.13E−14
51.61%
24.06%
HSF2
heat shock transcription factor 2
Hs.158195


220684_at
784
0.49
7.01E−09
71.86%
34.98%
TBX21
T-box 21
Hs.272409


211924_s_at
577
0.49
4.60E−05
82.81%
65.29%
PLAUR
plasminogen activator,
Hs.179657









urokinase receptor


32032_at
815
0.49
5.45E−18
33.09%
24.48%
DGSI
DiGeorge syndrome critical
Hs.154879









region gene DGSI; likely









ortholog of mouse expressed









sequence 2 embryonic lethal


212914_at
610
0.49
6.70E−09
76.90%
30.67%
PKP4
plakophilin 4
Hs.356416


204847_at
370
0.49
2.64E−20
37.08%
18.34%
ZNF-
zinc finger protein
Hs.301956








U69274


218559_s_at
727
0.49
3.58E−03
191.41%
42.94%
MAFB
v-maf musculoaponeurotic
Hs.169487









fibrosarcoma oncogene









homolog B (avian)


213587_s_at
633
0.49
5.00E−10
60.46%
35.98%


Hs.351612


203547_at
297
0.48
8.38E−13
57.70%
24.56%
CD4
CD4 antigen (p55)
Hs.17483


214696_at
662
0.48
1.43E−08
82.10%
29.38%
MGC14376
hypothetical protein
Hs.417157









MGC14376


220088_at
775
0.48
1.73E−04
116.92%
60.98%
C5R1
complement component 5
Hs.2161









receptor 1 (C5a ligand)


202724_s_at
262
0.48
5.23E−11
63.15%
29.60%
FOXO1A
forkhead box O1A
Hs.170133









(rhabdomyosarcoma)


200788_s_at
166
0.48
1.43E−12
61.50%
19.94%
PEA15
phosphoprotein enriched in
Hs.194673









astrocytes 15


213376_at
626
0.48
1.04E−14
49.81%
24.43%


Hs.372699


204621_s_at
357
0.48
1.11E−08
79.04%
32.70%
NR4A2
nuclear receptor subfamily 4,
Hs.82120









group A, member 2


214945_at
664
0.48
3.42E−07
63.69%
51.89%
KIAA0752
KIAA0752 protein
Hs.126779


221757_at
801
0.48
5.42E−11
69.15%
23.27%
MGC17330
hypothetical protein
Hs.26670









MGC17330


211985_s_at
582
0.48
3.30E−12
62.39%
23.79%


Hs.374441


200871_s_at
174
0.48
1.63E−09
81.31%
16.45%
PSAP
prosaposin (variant Gaucher
Hs.406455









disease and variant









metachromatic









leukodystrophy)


202842_s_at
267
0.48
2.16E−14
52.79%
23.79%
DNAJB9
DnaJ (Hsp40) homolog,
Hs.6790









subfamily B, member 9


219155_at
756
0.48
8.61E−16
47.62%
23.40%
RDGBB
retinal degeneration B beta
Hs.333212


203234_at
287
0.48
2.03E−07
89.59%
37.67%
UP
uridine phosphorylase
Hs.77573


219040_at
752
0.48
6.47E−10
42.85%
43.00%
FLJ22021
hypothetical protein FLJ22021
Hs.7258


214714_at
663
0.48
2.31E−17
47.52%
14.02%
FLJ12298
hypothetical protein FLJ12298
Hs.284168


219279_at
758
0.47
4.42E−11
68.97%
25.55%
FLJ20220
hypothetical protein FLJ20220
Hs.21126


40420_at
822
0.47
4.30E−19
39.97%
20.91%
STK10
serine/threonine kinase 10
Hs.16134


214467_at
96
0.47
8.57E−09
86.65%
24.10%
GPR65
G protein-coupled receptor 65
Hs.131924


202518_at
256
0.47
4.27E−19
42.88%
17.86%
BCL7B
B-cell CLL/lymphoma 7B
Hs.16269


204224_s_at
338
0.47
4.35E−15
53.97%
19.72%
GCH1
GTP cyclohydrolase 1 (dopa-
Hs.86724









responsive dystonia)


203045_at
281
0.47
3.33E−07
92.08%
40.13%
NINJ1
ninjurin 1
Hs.11342


39582_at
821
0.47
1.97E−11
70.10%
20.79%


Hs.26295


210225_x_at
529
0.47
3.53E−07
98.45%
34.82%
LILRB3
leukocyte immunoglobulin-like
Hs.105928









receptor, subfamily B (with TM









and ITIM domains), member 3


204891_s_at
374
0.47
5.17E−05
128.95%
45.60%
LCK
lymphocyte-specific protein
Hs.1765









tyrosine kinase


218711_s_at
733
0.47
1.60E−12
34.72%
36.28%
SDPR
serum deprivation response
Hs.26530









(phosphatidylserine binding









protein)


205254_x_at
388
0.47
4.07E−07
104.29%
28.42%
TCF7
transcription factor 7 (T-cell
Hs.169294









specific, HMG-box)


204396_s_at
344
0.47
4.98E−11
72.12%
23.82%
GPRK5
G protein-coupled receptor
Hs.211569









kinase 5


204369_at
341
0.47
1.47E−14
47.33%
28.81%
PIK3CA
phosphoinositide-3-kinase,
Hs.85701









catalytic, alpha polypeptide


212998_x_at
611
0.47
3.46E−09
72.57%
38.15%
HLA-DQB1
major histocompatibility
Hs.73931









complex, class II, DQ beta 1


204588_s_at
354
0.47
1.36E−06
111.56%
31.06%
SLC7A7
solute carrier family 7 (cationic
Hs.194693









amino acid transporter, y+









system), member 7


208881_x_at
475
0.47
2.85E−21
33.87%
21.20%
IDI1
isopentenyl-diphosphate delta
Hs.76038









isomerase


202861_at
270
0.47
1.34E−08
76.10%
40.36%
PER1
period homolog 1 (Drosophila)
Hs.68398


218828_at
739
0.46
5.31E−06
70.98%
62.75%
PLSCR3
phospholipid scramblase 3
Hs.103382


202388_at
250
0.46
2.71E−11
71.26%
25.16%
RGS2
regulator of G-protein
Hs.78944









signalling 2, 24 kD


219118_at
755
0.46
4.33E−09
60.48%
44.50%
FKBP11
FK506 binding protein 11 (19 kDa)
Hs.24048


213906_at
640
0.46
2.86E−06
109.54%
42.47%
MYBL1
v-myb myeloblastosis viral
Hs.300592









oncogene homolog (avian)-like 1


202880_s_at
273
0.46
9.28E−17
51.09%
19.25%
PSCD1
pleckstrin homology, Sec7 and
Hs.1050









coiled/coil domains









1(cytohesin 1)


201631_s_at
223
0.46
2.35E−04
129.87%
65.59%
IER3
immediate early response 3
Hs.76095


213758_at
635
0.46
1.89E−14
53.82%
26.63%


Hs.373513


209616_s_at
505
0.46
1.05E−06
93.94%
48.20%
CES1
carboxylesterase 1
Hs.76688









(monocyte/macrophage serine









esterase 1)


205281_s_at
390
0.46
1.44E−16
51.93%
20.24%
PIGA
phosphatidylinositol glycan,
Hs.51









class A (paroxysmal nocturnal









hemoglobinuria)


204215_at
337
0.46
1.33E−13
57.29%
27.83%
MGC4175
hypothetical protein MGC4175
Hs.322404


212812_at
98
0.46
6.01E−10
72.92%
35.84%


Hs.288232


207826_s_at
458
0.45
2.92E−06
63.43%
63.90%
ID3
inhibitor of DNA binding 3,
Hs.76884









dominant negative helix-loop-









helix protein


202072_at
237
0.45
5.57E−04
111.63%
84.78%
HNRPL
heterogeneous nuclear
Hs.2730









ribonucleoprotein L


210439_at
538
0.45
2.90E−06
112.93%
44.33%
ICOS
inducible T-cell co-stimulator
Hs.56247


203320_at
290
0.45
3.65E−15
55.50%
24.57%
LNK
lymphocyte adaptor protein
Hs.13131


204440_at
349
0.45
1.79E−10
68.74%
36.26%
CD83
CD83 antigen (activated B
Hs.79197









lymphocytes, immunoglobulin









superfamily)


211458_s_at
559
0.45
1.95E−10
69.84%
35.88%
GABARAPL3
GABA(A) receptors associated
Hs.334497









protein like 3


212769_at
608
0.45
1.48E−10
56.88%
40.54%
TLE3
transducin-like enhancer of
Hs.287362









split 3 (E(sp1) homolog,










Drosophila)



221841_s_at
803
0.45
9.97E−06
134.32%
33.96%
KLF4
Kruppel-like factor 4 (gut)
Hs.376206


217784_at
696
0.45
1.90E−12
60.94%
31.98%
YKT6
SNARE protein Ykt6
Hs.296244


202782_s_at
265
0.45
2.24E−14
51.88%
30.16%
SKIP
skeletal muscle and kidney
Hs.178347









enriched inositol phosphatase


220987_s_at
94
0.45
9.43E−16
56.70%
21.86%
DKFZP434J037
hypothetical protein
Hs.172012









DKFZp434J037


218708_at
732
0.45
2.34E−14
39.15%
33.34%
NXT1
NTF2-like export factor 1
Hs.24563


215785_s_at
674
0.45
6.95E−10
68.97%
40.16%
CYFIP2
cytoplasmic FMR1 interacting
Hs.258503









protein 2


202969_at
276
0.45
2.29E−16
49.47%
26.00%


Hs.432856


207000_s_at
445
0.45
1.12E−13
66.37%
20.02%
PPP3CC
protein phosphatase 3
Hs.75206









(formerly 2B), catalytic subunit,









gamma isoform (calcineurin A









gamma)


203555_at
298
0.45
2.68E−15
46.47%
29.83%
PTPN18
protein tyrosine phosphatase,
Hs.278597









non-receptor type 18 (brain-









derived)


202928_s_at
274
0.45
6.61E−13
54.32%
33.85%
PHF1
PHD finger protein 1
Hs.166204


204627_s_at
359
0.45
4.89E−05
142.91%
47.23%
ITGB3
integrin, beta 3 (platelet
Hs.87149









glycoprotein IIIa, antigen









CD61)


209674_at
508
0.44
4.83E−10
74.94%
36.71%
CRY1
cryptochrome 1 (photolyase-
Hs.151573









like)


204158_s_at
332
0.44
2.24E−09
60.61%
45.60%
TCIRG1
T-cell, immune regulator 1,
Hs.46465









ATPase, H+ transporting,









lysosomal V0 protein a isoform 3


204731_at
362
0.44
3.88E−08
89.75%
41.63%
TGFBR3
transforming growth factor,
Hs.342874









beta receptor III (betaglycan,









300 kD)


222315_at
813
0.44
1.83E−08
61.85%
50.17%


Hs.292853


214617_at
659
0.44
3.89E−05
132.11%
54.52%
PRF1
perforin 1 (pore forming
Hs.411106









protein)


211429_s_at
558
0.44
1.47E−08
99.17%
28.25%
SERPINA1
serine (or cysteine) proteinase
Hs.297681









inhibitor, clade A (alpha-1









antiproteinase, antitrypsin),









member 1


211919_s_at
575
0.44
1.78E−13
66.91%
23.29%
CXCR4
chemokine (C—X—C motif),
Hs.89414









receptor 4 (fusin)


212508_at
600
0.44
2.82E−20
45.20%
19.28%
MAP-1
modulator of apoptosis 1
Hs.24719


213193_x_at
111
0.44
7.58E−07
118.46%
35.66%
TRB@
T cell receptor beta locus
Hs.303157


215275_at
108
0.44
8.07E−11
85.22%
17.38%


205070_at
381
0.44
1.03E−13
42.45%
35.11%
ING3
inhibitor of growth family,
Hs.143198









member 3


220890_s_at
788
0.44
6.68E−25
36.96%
16.82%
LOC51202
hqp0256 protein
Hs.284288


210606_x_at
543
0.44
1.80E−08
92.09%
39.34%
KLRD1
killer cell lectin-like receptor
Hs.41682









subfamily D, member 1


204491_at
352
0.44
9.84E−15
57.70%
27.77%
PDE4D
phosphodiesterase 4D, cAMP-
Hs.172081









specific (phosphodiesterase









E3 dunce homolog,










Drosophila)



220066_at
774
0.44
2.04E−10
77.28%
35.18%
CARD15
caspase recruitment domain
Hs.135201









family, member 15


218964_at
748
0.44
1.85E−15
43.77%
31.13%
DRIL2
dead ringer (Drosophila)-like 2
Hs.10431









(bright and dead ringer)


204019_s_at
320
0.44
2.32E−07
96.30%
47.51%
DKFZP586F1318
hypothetical protein
Hs.432325









DKFZP586F1318


212400_at
597
0.43
1.01E−10
83.88%
27.30%


Hs.349755


219947_at
771
0.43
2.91E−09
85.16%
39.01%
CLECSF6
C-type (calcium dependent,
Hs.115515









carbohydrate-recognition









domain) lectin, superfamily









member 6


204912_at
114
0.43
2.36E−13
71.20%
22.28%
IL10RA
interleukin 10 receptor, alpha
Hs.327


204951_at
377
0.43
6.62E−13
68.70%
29.59%
ARHH
ras homolog gene family,
Hs.109918









member H


214049_x_at
644
0.43
7.17E−11
78.15%
33.94%
CD7
CD7 antigen (p41)
Hs.36972


218831_s_at
740
0.43
7.63E−09
101.10%
30.44%
FCGRT
Fc fragment of IgG, receptor,
Hs.111903









transporter, alpha


205992_s_at
421
0.43
4.36E−14
40.54%
35.31%
IL15
interleukin 15
Hs.168132


60084_at
824
0.43
4.04E−19
48.64%
22.69%
CYLD
cylindromatosis (turban tumor
Hs.18827









syndrome)


207460_at
452
0.42
3.62E−14
59.33%
30.98%
GZMM
granzyme M (lymphocyte metase
Hs.268531









1)


215666_at
673
0.42
2.16E−03
118.92%
106.86%
HLA-DRB4
major histocompatibility
Hs.318720









complex, class II, DR beta 4


217838_s_at
699
0.42
3.55E−09
98.35%
32.55%
RNB6
RNB6
Hs.241471


202833_s_at
266
0.42
3.54E−08
110.50%
32.29%
SERPINA1
serine (or cysteine) proteinase
Hs.297681









inhibitor, clade A (alpha-1









antiproteinase, antitrypsin),









member 1


210915_x_at
553
0.42
1.97E−06
135.65%
35.59%
TRB@
T cell receptor beta locus
Hs.303157


207339_s_at
449
0.42
1.22E−06
126.75%
42.23%
LTB
lymphotoxin beta (TNF
Hs.890









superfamily, member 3)


221724_s_at
117
0.42
1.32E−10
85.44%
33.28%
CLECSF6
C-type (calcium dependent,
Hs.115515









carbohydrate-recognition









domain) lectin, superfamily









member 6


221059_s_at
793
0.42
6.90E−15
68.88%
20.17%
CHST6
carbohydrate (N-
Hs.157439









acetylglucosamine 6-O)









sulfotransferase 6


209201_x_at
488
0.42
1.63E−15
65.60%
21.71%
CXCR4
chemokine (C—X—C motif),
Hs.89414









receptor 4 (fusin)


212501_at
599
0.42
8.81E−12
84.93%
22.86%
CEBPB
CCAAT/enhancer binding
Hs.99029









protein (C/EBP), beta


201739_at
123
0.42
1.15E−07
102.88%
46.70%
SGK
serum/glucocorticoid regulated
Hs.296323









kinase


207072_at
446
0.42
9.05E−10
77.08%
43.43%
IL18RAP
interleukin 18 receptor
Hs.158315









accessory protein


200920_s_at
176
0.42
1.24E−10
72.36%
40.91%
BTG1
B-cell translocation gene 1,
Hs.77054









anti-proliferative


203334_at
291
0.41
9.88E−18
53.89%
25.03%
DDX8
DEAD/H (Asp-Glu-Ala-
Hs.171872









Asp/His) box polypeptide 8









(RNA helicase)


204622_x_at
358
0.41
1.60E−09
93.16%
37.30%
NR4A2
nuclear receptor subfamily 4,
Hs.82120









group A, member 2


212231_at
591
0.41
1.45E−19
51.15%
21.95%
FBXO21
F-box only protein 21
Hs.184227


202637_s_at
258
0.41
2.23E−11
72.25%
38.03%
ICAM1
intercellular adhesion molecule
Hs.168383









1 (CD54), human rhinovirus









receptor


213539_at
132
0.41
2.78E−08
106.66%
39.69%
CD3D
CD3D antigen, delta
Hs.95327









polypeptide (TiT3 complex)


205291_at
391
0.41
1.22E−11
67.18%
38.85%
IL2RB
interleukin 2 receptor, beta
Hs.75596


202723_s_at
261
0.41
2.90E−12
55.21%
39.67%
FOXO1A
forkhead box O1A
Hs.170133









(rhabdomyosarcoma)


206343_s_at
431
0.41
5.98E−10
55.18%
48.19%
NRG1
neuregulin 1
Hs.172816


203543_s_at
296
0.41
1.87E−10
92.09%
32.00%
BTEB1
basic transcription element
Hs.150557









binding protein 1


202644_s_at
259
0.41
5.67E−12
86.22%
23.66%
TNFAIP3
tumor necrosis factor, alpha-
Hs.211600









induced protein 3


219622_at
764
0.41
1.13E−10
85.10%
35.95%
RAB20
RAB20, member RAS
Hs.179791









oncogene family


219528_s_at
762
0.41
2.09E−08
118.86%
24.30%
BCL11B
B-cell CLL/lymphoma 11B
Hs.57987









(zinc finger protein)


217591_at
693
0.41
2.28E−10
51.94%
47.24%


Hs.272108


204838_s_at
369
0.41
2.59E−10
38.33%
48.54%
MLH3
mutL homolog 3 (E. coli)
Hs.279843


213915_at
641
0.41
4.26E−08
113.63%
38.58%
NKG7
natural killer cell group 7
Hs.10306









sequence


213142_x_at
615
0.40
3.38E−14
72.90%
26.61%
LOC54103
hypothetical protein
Hs.12969


203888_at
312
0.40
1.09E−05
125.03%
63.75%
THBD
thrombomodulin
Hs.2030


211841_s_at
574
0.40
1.02E−12
83.08%
25.18%
TNFRSF12
tumor necrosis factor receptor
Hs.180338









superfamily, member 12









(translocating chain-









association membrane protein)


204118_at
330
0.40
9.75E−15
74.10%
14.40%
CD48
CD48 antigen (B-cell
Hs.901









membrane protein)


212841_s_at
609
0.40
1.41E−07
48.10%
62.68%
PPFIBP2
PTPRF interacting protein,
Hs.12953









binding protein 2 (liprin beta 2)


205255_x_at
389
0.40
4.07E−10
91.84%
38.82%
TCF7
transcription factor 7 (T-cell
Hs.169294









specific, HMG-box)


209871_s_at
515
0.40
4.73E−09
98.50%
42.93%
APBA2
amyloid beta (A4) precursor
Hs.26468









protein-binding, family A,









member 2 (X11-like)


209536_s_at
501
0.39
6.76E−15
55.98%
33.99%
EHD4
EH-domain containing 4
Hs.4943


203708_at
304
0.39
3.49E−11
95.00%
30.17%
PDE4B
phosphodiesterase 4B, cAMP-
Hs.188









specific (phosphodiesterase









E4 dunce homolog,










Drosophila)



202048_s_at
236
0.39
5.89E−16
63.65%
28.85%
CBX6
chromobox homolog 6
Hs.107374


218205_s_at
717
0.39
4.03E−18
34.91%
30.54%
MKNK2
MAP kinase-interacting
Hs.261828









serine/threonine kinase 2


209824_s_at
131
0.38
2.79E−13
73.55%
35.30%
ARNTL
aryl hydrocarbon receptor
Hs.74515









nuclear translocator-like


213958_at
102
0.38
4.17E−10
111.46%
28.16%
CD6
CD6 antigen
Hs.81226


221558_s_at
88
0.38
8.56E−10
109.99%
35.27%
LEF1
lymphoid enhancer-binding
Hs.44865









factor 1


208622_s_at
462
0.38
4.22E−16
67.21%
29.57%
VIL2
villin 2 (ezrin)
Hs.155191


218345_at
723
0.38
9.04E−07
111.02%
62.99%
HCA112
hepatocellular carcinoma-
Hs.12126









associated antigen 112


204777_s_at
363
0.38
5.40E−10
101.33%
41.03%
MAL
mal, T-cell differentiation
Hs.80395









protein


213300_at
620
0.37
9.54E−10
49.97%
53.43%
KIAA0404
KIAA0404 protein
Hs.105850


210054_at
524
0.37
1.89E−18
65.35%
23.26%
MGC4701
hypothetical protein MGC4701
Hs.116771


219117_s_at
754
0.37
2.29E−10
97.73%
40.82%
FKBP11
FK506 binding protein 11 (19 kDa)
Hs.24048


204244_s_at
339
0.37
6.56E−18
60.46%
27.96%
ASK
activator of S phase kinase
Hs.152759


222142_at
810
0.37
2.29E−22
50.09%
22.95%
CYLD
cylindromatosis (turban tumor
Hs.18827









syndrome)


205241_at
387
0.37
3.84E−12
78.99%
39.96%
SCO2
SCO cytochrome oxidase
Hs.278431









deficient homolog 2 (yeast)


202320_at
246
0.37
5.08E−09
41.96%
57.92%
GTF3C1
general transcription factor
Hs.331









IIIC, polypeptide 1 (alpha









subunit, 220 kD)


204103_at
328
0.37
6.82E−04
106.80%
109.56%
SCYA4
small inducible cytokine A4
Hs.75703


211583_x_at
565
0.37
3.06E−13
50.67%
41.55%
LY117
lymphocyte antigen 117
Hs.88411


211962_s_at
580
0.37
1.52E−16
74.42%
25.97%
ZFP36L1
zinc finger protein 36, C3H
Hs.85155









type-like 1


204411_at
346
0.37
1.46E−12
70.01%
41.24%
KIAA0449
KIAA0449 protein
Hs.169182


208657_s_at
465
0.36
6.92E−19
66.29%
23.55%
MSF
MLL septin-like fusion
Hs.181002


219593_at
79
0.36
4.65E−11
108.68%
31.98%
PHT2
peptide transporter 3
Hs.237856


222150_s_at
811
0.36
6.54E−15
71.48%
34.24%
LOC54103
hypothetical protein
Hs.12969


201425_at
51
0.36
1.85E−12
103.39%
24.19%
ALDH2
aldehyde dehydrogenase 2
Hs.195432









family (mitochondrial)


201565_s_at
219
0.36
1.22E−16
71.93%
28.77%
ID2
inhibitor of DNA binding 2,
Hs.180919









dominant negative helix-loop-









helix protein


209501_at
498
0.36
1.08E−20
57.82%
25.10%
CDR2
cerebellar degeneration-
Hs.75124









related protein (62 kD)


221890_at
804
0.36
6.50E−11
58.22%
49.64%
ZNF335
zinc finger protein 335
Hs.165983


211840_s_at
573
0.35
4.46E−15
59.93%
37.12%
PDE4D
phosphodiesterase 4D, cAMP-
Hs.172081









specific (phosphodiesterase









E3 dunce homolog,










Drosophila)



218486_at
726
0.35
5.27E−22
58.11%
23.19%
TIEG2
TGFB inducible early growth
Hs.12229









response 2


212196_at
590
0.35
1.52E−18
72.60%
23.80%


Hs.71968


219359_at
759
0.35
1.37E−12
82.00%
41.21%
FLJ22635
hypothetical protein FLJ22635
Hs.353181


204655_at
361
0.34
2.21E−09
116.09%
47.89%
SCYA5
small inducible cytokine A5
Hs.241392









(RANTES)


206366_x_at
432
0.34
7.78E−08
129.93%
55.60%
SCYC1,
small inducible cytokine
Hs.3195








SCYC2
subfamily C, member 1









(lymphotactin), small inducible









cytokine subfamily C, member 2


214146_s_at
646
0.34
1.46E−10
122.42%
36.27%
PPBP
pro-platelet basic protein
Hs.2164









(includes platelet basic protein,









beta-thromboglobulin,









connective tissue-activating









peptide III, neutrophil-









activating peptide-2)


38037_at
820
0.34
1.33E−07
135.13%
56.83%
DTR
diphtheria toxin receptor
Hs.799









(heparin-binding epidermal









growth factor-like growth









factor)


209062_x_at
482
0.34
9.87E−21
65.89%
24.70%
NCOA3
nuclear receptor coactivator 3
Hs.225977


213524_s_at
630
0.33
2.99E−10
105.05%
47.78%
G0S2
putative lymphocyte G0/G1
Hs.432132









switch gene


213135_at
614
0.33
1.80E−16
89.95%
22.91%


Hs.82141


210479_s_at
539
0.33
1.86E−16
83.74%
29.89%
RORA
RAR-related orphan receptor A
Hs.2156


210279_at
531
0.33
2.25E−08
123.27%
56.47%
GPR18
G protein-coupled receptor 18
Hs.88269


1405_i_at
155
0.33
2.64E−09
135.74%
44.48%
SCYA5
small inducible cytokine A5
Hs.241392









(RANTES)


210321_at
532
0.33
3.67E−03
326.10%
90.79%
CTLA1
similar to granzyme B
Hs.348264









(granzyme 2, cytotoxic T-









lymphocyte-associated serine









esterase 1) (H. sapiens)


201566_x_at
220
0.33
2.67E−14
79.78%
38.73%
ID2
inhibitor of DNA binding 2,
Hs.180919









dominant negative helix-loop-









helix protein


204198_s_at
336
0.33
1.17E−13


RUNX3
runt-related transcription factor 3
Hs.170019


218696_at
731
0.32
2.48E−23


EIF2AK3
eukaryotic translation initiation
Hs.102506









factor 2-alpha kinase 3


213624_at
152
0.32
1.74E−09



acid sphingomyelinase-like
Hs.42945









phosphodiesterase


218793_s_at
736
0.32
1.17E−18


SCML1
sex comb on midleg-like 1
Hs.109655









(Drosophila)


204197_s_at
335
0.32
3.00E−17


RUNX3
runt-related transcription factor 3
Hs.170019


209728_at
509
0.32
2.53E−04
163.58%
101.38%
HLA-DRB4
major histocompatibility
Hs.318720









complex, class II, DR beta 4


202206_at
242
0.32
1.53E−15
89.61%
32.16%
ARL7
ADP-ribosylation factor-like 7
Hs.111554


212195_at
589
0.32
3.87E−17
90.97%
24.26%


Hs.71968


206296_x_at
428
0.32
1.58E−10
59.76%
54.60%
MAP4K1
mitogen-activated protein
Hs.95424,









kinase kinase kinase kinase 1
Hs.86575


201189_s_at
193
0.32
3.76E−16
98.75%
23.89%
ITPR3
inositol 1,4,5-triphosphate
Hs.77515









receptor, type 3


219099_at
115
0.32
1.10E−20
66.40%
27.62%
C12orf5
chromosome 12 open reading
Hs.24792









frame 5


210113_s_at
527
0.31
9.95E−18


NALP1
death effector filament-forming
Hs.104305









Ced-4-like apoptosis protein


212187_x_at
588
0.31
1.65E−11
72.81%
50.49%
PTGDS
prostaglandin D2 synthase
Hs.8272









(21 kD, brain)


209604_s_at
504
0.31
7.32E−17
83.69%
32.25%
GATA3
GATA binding protein 3
Hs.169946


204794_at
367
0.31
3.14E−15
98.27%
32.11%
DUSP2
dual specificity phosphatase 2
Hs.1183


204790_at
365
0.31
3.37E−12
53.77%
49.07%
MADH7
MAD, mothers against
Hs.100602









decapentaplegic homolog 7









(Drosophila)


202208_s_at
244
0.31
2.85E−11
97.48%
48.91%
ARL7
ADP-ribosylation factor-like 7
Hs.111554


203821_at
309
0.30
2.38E−09
132.98%
52.56%
DTR
diphtheria toxin receptor
Hs.799









(heparin-binding epidermal









growth factor-like growth









factor)


214567_s_at
657
0.30
7.72E−12
65.03%
50.48%
SCYC1,
small inducible cytokine
Hs.174228








SCYC2
subfamily C, member 1









(lymphotactin), small inducible









cytokine subfamily C, member 2


203887_s_at
311
0.30
1.57E−07
136.61%
66.32%
THBD
thrombomodulin
Hs.2030


206655_s_at
438
0.30
5.47E−11
69.52%
53.78%
GP1BB
glycoprotein lb (platelet), beta
Hs.283743









polypeptide


214219_x_at
647
0.30
2.94E−10
70.71%
57.65%
MAP4K1
mitogen-activated protein
Hs.95424,









kinase kinase kinase kinase 1
Hs.86575


211748_x_at
569
0.29
6.29E−11



prostaglandin D2 synthase
Hs.8272









(21 kD, brain)


202988_s_at
278
0.29
6.99E−06


RGS1
regulator of G-protein
Hs.75256









signalling 1


202207_at
243
0.29
9.60E−22


ARL7
ADP-ribosylation factor-like 7
Hs.111554


204793_at
366
0.29
2.70E−18
97.58%
22.06%
KIAA0443
KIAA0443 gene product
Hs.113082


214470_at
654
0.29
1.86E−17
94.96%
29.59%
KLRB1
killer cell lectin-like receptor
Hs.169824









subfamily B, member 1


210164_at
528
0.29
1.45E−11
128.23%
43.90%
GZMB
granzyme B (granzyme 2,
Hs.1051









cytotoxic T-lymphocyte-









associated serine esterase 1)


221756_at
800
0.29
1.38E−20
80.93%
27.52%
MGC17330
hypothetical protein
Hs.26670









MGC17330


206390_x_at
433
0.28
3.02E−11


PF4
platelet factor 4
Hs.81564


208146_s_at
460
0.28
1.04E−17


CPVL
carboxypeptidase, vitellogenic-
Hs.95594









like


214032_at
642
0.27
4.56E−16
102.92%
36.01%
ZAP70
zeta-chain (TCR) associated
Hs.234569









protein kinase (70 kD)


216834_at
687
0.27
9.67E−08
107.30%
73.61%
RGS1
regulator of G-protein
Hs.385701,









signalling 1
Hs.75256


210426_x_at
537
0.26
4.55E−19
95.05%
31.13%
RORA
RAR-related orphan receptor A
Hs.2156


220646_s_at
783
0.25
4.98E−14
136.06%
39.89%
KLRF1
killer cell lectin-like receptor
Hs.183125









subfamily F, member 1


203414_at
294
0.25
5.84E−28
65.64%
23.41%
MMD
monocyte to macrophage
Hs.79889









differentiation-associated


210512_s_at
541
0.25
6.16E−11
77.66%
58.76%
VEGF
vascular endothelial growth
Hs.73793









factor


203271_s_at
289
0.24
1.08E−20
57.24%
33.16%
UNC119
unc-119 homolog (C. elegans)
Hs.81728


204081_at
326
0.24
1.14E−16
60.84%
40.84%
NRGN
neurogranin (protein kinase C
Hs.26944









substrate, RC3)


204115_at
329
0.23
8.80E−16


GNG11
guanine nucleotide binding
Hs.83381









protein 11


37145_at
818
0.23
3.86E−12
161.44%
48.15%
GNLY
granulysin
Hs.105806


205495_s_at
398
0.22
1.07E−11
153.17%
52.73%
GNLY
granulysin
Hs.105806


205237_at
385
0.22
1.12E−17
131.65%
33.86%
FCN1
ficolin (collagen/fibrinogen
Hs.252136









domain containing) 1


210031_at
520
0.22
1.72E−21
106.54%
30.59%
CD3Z
CD3Z antigen, zeta
Hs.97087









polypeptide (TiT3 complex)


220532_s_at
781
0.21
3.51E−07
129.47%
85.67%
LR8
LR8 protein
Hs.190161


221211_s_at
794
0.20
6.63E−15
44.22%
46.84%
C21orf7
chromosome 21 open reading
Hs.41267









frame 7


201506_at
213
0.14
2.13E−27
140.21%
27.11%
TGFBI
transforming growth factor,
Hs.118787









beta-induced, 68 kD









Each HG-U133A qualifier represents an oligonucleotide probe set on the HG-U133A gene chip. The RNA transcript(s) of a gene that corresponds to a HG-U133A qualifier can hybridize under nucleic acid array hybridization conditions to at least one oligonucleotide probe (PM or perfect match probe) of the qualifier. Preferably, the RNA transcript(s) of the gene does not hybridize under nucleic acid array hybridization conditions to a mismatch probe (MM) of the PM probe. A mismatch probe is identical to the corresponding PM probe except for a single, homomeric substitution at or near the center of the mismatch probe. For a 25-mer PM probe, the MM probe has a homomeric base change at the 13th position.


In many cases, the RNA transcript(s) of a gene that corresponds to a HG-U133A qualifier can hybridize under nucleic acid array hybridization conditions to at least 50%, 60%, 70%, 80%, 90% or 100% of all of the PM probes of the qualifier, but not to the mismatch probes of these PM probes. In many other cases, the discrimination score (R) for each of these PM probes, as measured by the ratio of the hybridization intensity difference of the corresponding probe pair (i.e., PM−MM) over the overall hybridization intensity (i.e., PM+MM), is no less than 0.015, 0.02, 0.05, 0.1, 0.2, 0.3, 0.4, 0.5 or greater. In one example, the RNA transcript(s) of the gene, when hybridized to the HG-U133A gene chip according to the manufacturer's instructions, produces a “present” call under the default settings, i.e., the threshold Tau is 0.015 and the significance level α1 is 0.4. See GeneChip® Expression Analysis—Data Analysis Fundamentals (Part No. 701190 Rev. 2, Affymetrix, Inc., 2002), the entire content of which is incorporated herein by reference.


The sequences of each PM probe on the HG-U133A gene chip, and the corresponding target sequences from which the PM probes are derived, can be obtained from Affymetrix's sequence databases. See, for example, www.affymetrix.com/support/technical/byproduct.affx?product=hgu133. All of these target and oligonucleotide probe sequences are incorporated herein by reference.


In addition, genes whose expression levels are significantly elevated (p<0.001) in PBMCs of AML patients relative to disease-free subjects are shown in Table 8. Genes whose expression levels are significantly lowered (p<0.001) in PBMCs of AML patients relative to disease-free subjects are shown in Table 9.


Each gene described in Tables 7, 8 and 9 and the corresponding unigene(s) are identified based on HG-U133A genechip annotations. A unigene is composed of a non-redundant set of gene-oriented clusters. Each unigene cluster is believed to include sequences that represent a unique gene. Information for each gene listed in Table 7, 8 and 9 and its corresponding unigene(s) can also be obtained from the Entrez Gene and Unigene databases at National Center for Biotechnology Information (NCBI), Bethesda, Md.


In addition to Affymetrix annotations, gene(s) that corresponds to a HG-U133A qualifier can be identified by BLAST searching the target sequence of the qualifier against a human genome sequence database. Human genome sequence databases suitable for this purpose include, but are not limited to, the NCBI human genome database. NCBI also provides BLAST programs, such as “blastn,” for searching its sequence databases. In one embodiment, the BLAST search of the NCBI human genome database is performed by using an unambiguous segment (e.g., the longest unambiguous segment) of the target sequence of the qualifier. Gene(s) that aligns to the unambiguous segment with significant sequence identity can be identified. In many cases, the identified gene(s) has at least 95%, 96%, 97%, 98%, 99%, or more sequence identity to the unambiguous segment.


As used herein, genes listed in all the Tables encompasse not only the genes that are explicitly depicted, but also genes that are not listed in the table but nonetheless corresponds to a qualifier in the table. All of these genes can be used as biological markers for the diagnosis or monitoring the development, progression or treatment of AML.









TABLE 8







Top 50 transcripts at significantly elevated levels (p < 0.001)


in PBMCs of AML patients relative to disease-free subjects




















AML
Normal









Average
Average
Fold Diff
p-value


Affymetrix ID
SEQ ID NO:
Name
Cyto Band
Unigene ID
(ppm)
(ppm)
AML/Norm
(unequal)


















203948_s_at
316
myeloperoxidase
17q23.1
Hs.1817
83.00
1.78
46.69
4.63E−06


203949_at
317
myeloperoxidase
17q23.1
Hs.1817
74.97
2.13
35.14
1.19E−06


206310_at
429
serine protease inhibitor, Kazal type,
4q11
Hs.98243
43.47
1.91
22.75
3.86E−06




2 (acrosin-trypsin inhibitor)


209905_at
518
homeo box A9
7p15-p14
Hs.127428
21.08
1.00
21.08
5.44E−05


214575_s_at
658
azurocidin 1 (cationic antimicrobial
19p13.3
Hs.72885
36.92
1.84
20.02
3.88E−04




protein 37)


206871_at
444
elastase 2, neutrophil
19p13.3
Hs.99863
35.58
1.93
18.41
1.23E−04


214651_s_at
660
homeo box A9
7p15-p14
Hs.127428
29.61
1.82
16.25
5.98E−05


210084_x_at
525
tryptase beta 1, tryptase, alpha
16p13.3
Hs.347933
14.50
1.02
14.18
1.20E−04


205683_x_at
411
tryptase beta 1, tryptase beta 2,
16p13.3
Hs.347933
20.42
1.47
13.92
4.32E−04




tryptase, alpha


204798_at
368
v-myb myeloblastosis viral oncogene
6q22-q23
Hs.1334
35.69
2.76
12.95
7.41E−10




homolog (avian)


217023_x_at
688
tryptase beta 1, tryptase beta 2
16p13.3
Hs.294158,
13.08
1.09
12.02
1.41E−04






Hs.347933


216474_x_at
681
tryptase beta 1, tryptase beta 2
16p13.3
Hs.347933
18.92
1.71
11.06
8.25E−05


202016_at
235
mesoderm specific transcript
7q32
Hs.79284
34.28
3.11
11.02
3.63E−04




homolog (mouse)


207134_x_at
447
tryptase beta 1, tryptase beta 2,
16p13.3
Hs.294158
17.75
1.62
10.94
6.98E−04




tryptase, alpha


215382_x_at
670
tryptase beta 1, tryptase, alpha
16p13.3
Hs.347933
15.19
1.40
10.85
5.25E−05


205950_s_at
420
carbonic anhydrase I
8q13-q22.1
Hs.23118
101.03
9.31
10.85
5.23E−04


205051_s_at
380
v-kit Hardy-Zuckerman 4 feline
4q11-q12
Hs.81665
16.39
1.60
10.24
2.37E−05




sarcoma viral oncogene homolog


211709_s_at
566
stem cell growth factor; lymphocyte
19q13.3
Hs.105927
32.19
3.20
10.06
1.23E−06




secreted C-type lectin


205131_x_at
383
stem cell growth factor; lymphocyte
19q13.3
Hs.105927
12.31
1.29
9.55
1.02E−04




secreted C-type lectin


219054_at
753
hypothetical protein FLJ14054
5p13.2
Hs.13528
14.61
1.76
8.32
2.05E−06


204304_s_at
340
prominin-like 1 (mouse)
4p15.33
Hs.112360
12.47
1.62
7.69
4.74E−07


206674_at
440
fms-related tyrosine kinase 3
13q12
Hs.385
15.97
2.16
7.41
2.90E−07


207741_x_at
456
tryptase, alpha
16p13.3
Hs.334455
14.33
1.96
7.33
5.05E−05


202589_at
257
thymidylate synthetase
18p11.32
Hs.82962
32.89
4.64
7.08
1.63E−05


210783_x_at
549
stem cell growth factor; lymphocyte
19q13.3
Hs.105927
7.31
1.04
6.99
5.96E−05




secreted C-type lectin


211922_s_at
576
catalase
11p13
Hs.76359
38.47
5.73
6.71
1.13E−07


201427_s_at
208
selenoprotein P, plasma, 1
5q31
Hs.3314
6.64
1.00
6.64
7.13E−04


206111_at
424
ribonuclease, RNase A family, 2
14q24-q31
Hs.728
63.06
9.56
6.60
2.95E−05




(liver, eosinophil-derived neurotoxin)


202503_s_at
255
KIAA0101 gene product
15q22.1
Hs.81892
25.86
4.04
6.39
2.92E−06


220377_at
778
HSPC053 protein
14q32.33
Hs.128155
6.28
1.02
6.14
1.93E−04


201310_s_at
200
P311 protein
5q21.3
Hs.142827
29.44
4.98
5.92
2.13E−09


219672_at
767
erythroid associated factor
16p11.1
Hs.274309
28.78
4.91
5.86
9.81E−04


205624_at
409
carboxypeptidase A3 (mast cell)
3q21-q25
Hs.646
20.11
3.56
5.66
9.30E−05


205609_at
407
angiopoietin 1
8q22.3-q23
Hs.2463
6.83
1.22
5.59
1.49E−06


206834_at
442
hemoglobin, delta
11p15.5
Hs.36977
183.31
33.40
5.49
5.46E−05


201162_at
192
insulin-like growth factor binding
4q12
Hs.119206
17.72
3.38
5.25
3.09E−07




protein 7


201432_at
209
catalase
11p13
Hs.76359
121.17
23.38
5.18
1.43E−09


204430_s_at
8
solute carrier family 2 (facilitated
1p36.2
Hs.33084
5.86
1.13
5.17
6.73E−04




glucose/fructose transporter),




member 5


220416_at
780
KIAA1939 protein
15q15.2
Hs.182738
9.64
1.87
5.16
1.24E−06


211743_s_at
568
proteoglycan 2, bone marrow
11q12
Hs.99962
7.58
1.53
4.95
7.28E−04




(natural killer cell activator,




eosinophil granule major basic




protein)


201416_at
206
Meis1, myeloid ecotropic viral
17p11.2,
Hs.83484
30.64
6.20
4.94
1.01E−04




integration site 1 homolog 3
6p22.3




(mouse), SRY (sex determining




region Y)-box 4


213150_at
617
homeo box A10
7p15-p14
Hs.110637
8.39
1.71
4.90
3.44E−04


209543_s_at
502
CD34 antigen, FLJ00005 protein
15, 1q32
Hs.367690
11.39
2.33
4.88
6.90E−07


213258_at
65
unknown

Hs.288582
5.25
1.09
4.82
2.40E−07


210664_s_at
546
tissue factor pathway inhibitor
2q31-q32.1
Hs.170279
5.89
1.24
4.73
8.77E−06




(lipoprotein-associated coagulation




inhibitor)


206067_s_at
423
Wilms tumor 1
11p13
Hs.1145
4.72
1.00
4.72
2.81E−04


209757_s_at
70
v-myc myelocytomatosis viral related
2p24.1
Hs.25960
4.69
1.00
4.69
8.72E−06




oncogene, neuroblastoma derived




(avian)


213515_x_at
629
glycyl-tRNA synthetase, hemoglobin,
11p15.5, 7p15
Hs.283108
345.06
73.71
4.68
2.22E−05




gamma A, hemoglobin, gamma G


219837_s_at
769
cytokine-like protein C17
4p16-p15
Hs.13872
5.72
1.24
4.60
2.68E−04


218899_s_at
746
brain and acute leukemia,
8q22.3
Hs.169395
6.19
1.36
4.57
9.36E−04




cytoplasmic
















TABLE 9







Top 50 transcripts at significantly lower levels (p < 0.001)


in PBMCs of AML patients relative to disease-free subjects




















AML
Normal









Average
Average
Fold Diff
p-value


Affymetrix
SEQ ID NO:
Name
Cyto Band
Unigene ID
(ppm)
(ppm)
Norm/AML
(unequal)


















201506_at
213
transforming growth factor, beta-
5q31
Hs.118787
6.56
47.31
7.22
2.13E−27




induced, 68 kD


221211_s_at
794
chromosome 21 open reading
21q22.3
Hs.41267
2.44
11.93
4.88
6.63E−15




frame 7


220532_s_at
781
LR8 protein
7q35
Hs.190161
3.00
14.02
4.67
3.51E−07


210031_at
520
CD3Z antigen, zeta polypeptide
1q22-q23
Hs.97087
11.72
53.98
4.60
1.72E−21




(TiT3 complex)


205237_at
385
ficolin (collagen/fibrinogen domain
9q34
Hs.252136
29.56
132.64
4.49
1.12E−17




containing) 1


205495_s_at
398
granulysin
2p12-q11
Hs.105806
12.86
57.69
4.49
1.07E−11


37145_at
818
granulysin
2p12-q11
Hs.105806
14.22
62.47
4.39
3.86E−12


204115_at
329
guanine nucleotide binding protein
7q31-q32
Hs.83381
2.75
11.80
4.29
8.80E−16




11


204081_at
326
neurogranin (protein kinase C
11q24
Hs.26944
7.83
32.69
4.17
1.14E−16




substrate, RC3)


203271_s_at
289
unc-119 homolog (C. elegans)
17q11.2
Hs.81728
1.58
6.60
4.17
1.08E−20


210512_s_at
541
vascular endothelial growth factor
6p12
Hs.73793
3.00
12.18
4.06
6.16E−11


203414_at
294
monocyte to macrophage
17q
Hs.79889
7.78
31.47
4.05
5.84E−28




differentiation-associated


220646_s_at
783
killer cell lectin-like receptor
12p12.3-13.2
Hs.183125
4.36
17.51
4.02
4.98E−14




subfamily F, member 1


210426_x_at
537
RAR-related orphan receptor A
15q21-q22
Hs.2156
4.17
15.78
3.79
4.55E−19


216834_at
687
regulator of G-protein signalling 1
1q31
Hs.75256
10.50
38.56
3.67
9.67E−08


214032_at
642
zeta-chain (TCR) associated protein
2q12
Hs.234569
4.78
17.49
3.66
4.56E−16




kinase (70 kD)


206390_x_at
433
platelet factor 4
4q12-q21
Hs.81564
16.11
58.53
3.63
3.02E−11


208146_s_at
460
carboxypeptidase, vitellogenic-like
7p15-p14
Hs.95594
10.75
38.51
3.58
1.04E−17


221756_at
800
hypothetical protein MGC17330
22q11.2-q22
Hs.26670
13.81
47.98
3.48
1.38E−20


210164_at
528
granzyme B (granzyme 2, cytotoxic
14q11.2
Hs.1051
8.28
28.60
3.46
1.45E−11




T-lymphocyte-associated serine




esterase 1)


211748_x_at
569
prostaglandin D2 synthase (21 kD,
9q34.2-q34.3
Hs.8272
5.36
18.47
3.44
6.29E−11




brain)


202988_s_at
278
regulator of G-protein signalling 1
1q31
Hs.75256
2.58
8.89
3.44
6.99E−06


202207_at
243
ADP-ribosylation factor-like 7
2q37.2
Hs.111554
20.22
69.47
3.44
9.60E−22


214470_at
654
killer cell lectin-like receptor
12p13
Hs.169824
18.14
61.67
3.40
1.86E−17




subfamily B, member 1


204793_at
366
KIAA0443 gene product
Xq22.1
Hs.113082
4.81
16.31
3.39
2.70E−18


214219_x_at
647
mitogen-activated protein kinase
19q13.1-q13.4
Hs.86575
2.00
6.78
3.39
2.94E−10




kinase kinase kinase 1


206655_s_at
438
glycoprotein lb (platelet), beta
22q11.21
Hs.283743
2.36
7.82
3.31
5.47E−11




polypeptide


203887_s_at
311
thrombomodulin
20p12-cen
Hs.2030
4.28
14.13
3.30
1.57E−07


214567_s_at
657
small inducible cytokine subfamily
1q23, 1q23-q25
Hs.174228
1.39
4.58
3.30
7.72E−12




C, member 1 (lymphotactin), small




inducible cytokine subfamily C,




member 2


203821_at
309
diphtheria toxin receptor (heparin-
5q23
Hs.799
11.81
38.84
3.29
2.38E−09




binding epidermal growth factor-like




growth factor)


202208_s_at
244
ADP-ribosylation factor-like 7
2q37.2
Hs.111554
8.67
28.07
3.24
2.85E−11


204790_at
365
MAD, mothers against
18q21.1
Hs.100602
2.81
9.07
3.23
3.37E−12




decapentaplegic homolog 7




(Drosophila)


210113_s_at
527
death effector filament-forming Ced-
17p13
Hs.104305
3.61
11.64
3.22
9.95E−18




4-like apoptosis protein


204794_at
367
dual specificity phosphatase 2
2q11
Hs.1183
7.64
24.51
3.21
3.14E−15


209604_s_at
504
GATA binding protein 3
10p15
Hs.169946
7.36
23.60
3.21
7.32E−17


212187_x_at
588
prostaglandin D2 synthase (21 kD,
9q34.2-q34.3
Hs.8272
4.03
12.91
3.21
1.65E−11




brain)


219099_at
115
chromosome 12 open reading
12p13.3
Hs.24792
3.78
11.96
3.16
1.10E−20




frame 5


201189_s_at
193
inositol 1,4,5-triphosphate receptor,
6p21
Hs.77515
2.94
9.31
3.16
3.76E−16




type 3


206296_x_at
428
mitogen-activated protein kinase
19q13.1-q13.4
Hs.86575
2.86
8.96
3.13
1.58E−10




kinase kinase kinase 1


212195_at
589
Unknown
N/a
Hs.71968
8.11
25.33
3.12
3.87E−17


218696_at
731
eukaryotic translation initiation
2p12
Hs.102506
6.86
21.42
3.12
2.48E−23




factor 2-alpha kinase 3


213624_at
152
acid sphingomyelinase-like
6
Hs.42945
2.19
6.82
3.11
1.74E−09




phosphodiesterase


202206_at
242
ADP-ribosylation factor-like 7
2q37.2
Hs.111554
14.14
43.80
3.10
1.53E−15


209728_at
509
major histocompatibility complex,
6p21.3
Hs.318720
11.25
34.69
3.08
2.53E−04




class II, DR beta 4


218793_s_at
736
sex comb on midleg-like 1
Xp22.2-p22.1
Hs.109655
2.03
6.24
3.08
1.17E−18




(Drosophila)


204197_s_at
335
runt-related transcription factor 3
1p36
Hs.170019
19.69
60.64
3.08
3.00E−17


201566_x_at
220
inhibitor of DNA binding 2,
2p25
Hs.180919
5.64
17.31
3.07
2.67E−14




dominant negative helix-loop-helix




protein


204198_s_at
336
runt-related transcription factor 3
1p36
Hs.170019
12.08
37.00
3.06
1.17E−13


1405_i_at
155
small inducible cytokine A5
17q11.2-q12
Hs.241392
11.69
35.67
3.05
2.64E−09




(RANTES)


210279_at
531
G protein-coupled receptor 18
13q32
Hs.88269
4.28
13.02
3.04
2.25E−08









Prognosis, Diagnosis and Selection of Treatment of AML or Other Leukemias

The prognostic genes of the present invention can be used for the prediction of clinical outcome of a leukemia patient of interest. The prediction typically involves comparison of the peripheral blood expression profile of one or more prognostic genes in the leukemia patient of interest to at least one reference expression profile. Each prognostic gene employed in the present invention is differentially expressed in peripheral blood samples of leukemia patients who have different clinical outcomes.


In one embodiment, the prognostic genes employed for the outcome prediction are selected such that the peripheral blood expression profile of each prognostic gene is correlated with a class distinction under a class-based correlation analysis (such as the nearest-neighbor analysis), where the class distinction represents an idealized expression pattern of the selected genes in peripheral blood samples of leukemia patients who have different clinical outcomes. In many cases, the selected prognostic genes are correlated with the class distinction at above the 50%, 25%, 10%, 5%, or 1% significance level under a random permutation test.


The prognostic genes can also be selected such that the average expression profile of each prognostic gene in peripheral blood samples of one class of leukemia patients is statistically different from that in another class of leukemia patients. For instance, the p-value under a Student's t-test for the observed difference can be no more than 0.05, 0.01, 0.005, 0.001, or less. In addition, the prognostic genes can be selected such that the average peripheral blood expression level of each prognostic gene in one class of patients is at least 2-, 3-, 4-, 5-, 10-, or 20-fold different from that in another class of patients.


The expression profile of a patient of interest can be compared to one or more reference expression profiles. The reference expression profiles can be determined concurrently with the expression profile of the patient of interest. The reference expression profiles can also be predetermined or prerecorded in electronic or other types of storage media.


The reference expression profiles can include average expression profiles, or individual profiles representing peripheral blood gene expression patterns in particular patients. In one embodiment, the reference expression profiles include an average expression profile of the prognostic gene(s) in peripheral blood samples of reference leukemia patients who have known or determinable clinical outcome. Any averaging method may be used, such as arithmetic means, harmonic means, average of absolute values, average of log-transformed values, or weighted average. In one example, the reference leukemia patients have the same clinical outcome. In another example, the reference leukemia patients can be divided into at least two classes, each class of patients having a different respective clinical outcome. The average peripheral blood expression profile in each class of patients constitutes a separate reference expression profile, and the expression profile of the patient of interest is compared to each of these reference expression profiles.


In another embodiment, the reference expression profiles includes a plurality of expression profiles, each of which represents the peripheral blood expression pattern of the prognostic gene(s) in a particular leukemia patient whose clinical outcome is known or determinable. Other types of reference expression profiles can also be used in the present invention. In yet another embodiment, the present invention uses a numerical threshold as a control level.


The expression profile of the patient of interest and the reference expression profile(s) can be constructed in any form. In one embodiment, the expression profiles comprise the expression level of each prognostic gene used in outcome prediction. The expression levels can be absolute, normalized, or relative levels. Suitable normalization procedures include, but are not limited to, those used in nucleic acid array gene expression analyses or those described in Hill, et al., GENOME BIOL, 2:research0055.1-0055.13 (2001). In one example, the expression levels are normalized such that the mean is zero and the standard deviation is one. In another example, the expression levels are normalized based on internal or external controls, as appreciated by those skilled in the art. In still another example, the expression levels are normalized against one or more control transcripts with known abundances in blood samples. In many cases, the expression profile of the patient of interest and the reference expression profile(s) are constructed using the same or comparable methodologies.


In another embodiment, each expression profile being compared comprises one or more ratios between the expression levels of different prognostic genes. An expression profile can also include other measures that are capable of representing gene expression patterns.


The peripheral blood samples used in the present invention can be either whole blood samples, or samples comprising enriched PBMCs. In one example, the peripheral blood samples used for preparing the reference expression profile(s) comprise enriched or purified PBMCs, and the peripheral blood sample used for preparing the expression profile of the patient of interest is a whole blood sample. In another example, all of the peripheral blood samples employed in outcome prediction comprise enriched or purified PBMCs. In many cases, the peripheral blood samples are prepared from the patient of interest and reference patients using the same or comparable procedures.


Other types of blood samples can also be employed in the present invention, and the gene expression profiles in these blood samples are statistically significantly correlated with patient outcome.


The peripheral blood samples used in the present invention can be isolated from respective patients at any disease or treatment stage, and the correlation between the gene expression patterns in these peripheral blood samples and clinical outcome is statistically significant. In many embodiments, clinical outcome is measured by patients' response to a therapeutic treatment, and all of the blood samples used in outcome prediction are isolated prior to the therapeutic treatment. The expression profiles derived from these blood samples are therefore baseline expression profiles for the therapeutic treatment.


Construction of the expression profiles typically involves detection of the expression level of each prognostic gene used in the outcome prediction. Numerous methods are available for this purpose. For instance, the expression level of a gene can be determined by measuring the level of the RNA transcript(s) of the gene. Suitable methods include, but are not limited to, quantitative RT-PCT, Northern Blot, in situ hybridization, slot-blotting, nuclease protection assay, and nucleic acid array (including bead array). The expression level of a gene can also be determined by measuring the level of the polypeptide(s) encoded by the gene. Suitable methods include, but are not limited to, immunoassays (such as ELISA, RIA, FACS, or Western blot), 2-dimensional gel electrophoresis, mass spectrometry, or protein arrays.


In one aspect, the expression level of a prognostic gene is determined by measuring the RNA transcript level of the gene in a peripheral blood sample. RNA can be isolated from the peripheral blood sample using a variety of methods. Exemplary methods include guanidine isothiocyanate/acidic phenol method, the TRIZOL® Reagent (Invitrogen), or the Micro-FastTrack™ 2.0 or FastTrack™ 2.0 mRNA Isolation Kits (Invitrogen). The isolated RNA can be either total RNA or mRNA. The isolated RNA can be amplified to cDNA or cRNA before subsequent detection or quantitation. The amplification can be either specific or non-specific. Suitable amplification methods include, but are not limited to, reverse transcriptase PCR(RT-PCR), isothermal amplification, ligase chain reaction, and Qbeta replicase.


In one embodiment, the amplification protocol employs reverse transcriptase. The isolated mRNA can be reverse transcribed into cDNA using a reverse transcriptase, and a primer consisting of oligo (dT) and a sequence encoding the phage T7 promoter. The cDNA thus produced is single-stranded. The second strand of the cDNA is synthesized using a DNA polymerase, combined with an RNase to break up the DNA/RNA hybrid. After synthesis of the double-stranded cDNA, T7 RNA polymerase is added, and cRNA is then transcribed from the second strand of the doubled-stranded cDNA. The amplified cDNA or cRNA can be detected or quantitated by hybridization to labeled probes. The cDNA or cRNA can also be labeled during the amplification process and then detected or quantitated.


In another embodiment, quantitative RT-PCR (such as TaqMan, ABI) is used for detecting or comparing the RNA transcript level of a prognostic gene of interest. Quantitative RT-PCR involves reverse transcription (RT) of RNA to cDNA followed by relative quantitative PCR(RT-PCR).


In PCR, the number of molecules of the amplified target DNA increases by a factor approaching two with every cycle of the reaction until some reagent becomes limiting. Thereafter, the rate of amplification becomes increasingly diminished until there is not an increase in the amplified target between cycles. If a graph is plotted on which the cycle number is on the X axis and the log of the concentration of the amplified target DNA is on the Y axis, a curved line of characteristic shape can be formed by connecting the plotted points. Beginning with the first cycle, the slope of the line is positive and constant. This is said to be the linear portion of the curve. After some reagent becomes limiting, the slope of the line begins to decrease and eventually becomes zero. At this point the concentration of the amplified target DNA becomes asymptotic to some fixed value. This is said to be the plateau portion of the curve.


The concentration of the target DNA in the linear portion of the PCR is proportional to the starting concentration of the target before the PCR is begun. By determining the concentration of the PCR products of the target DNA in PCR reactions that have completed the same number of cycles and are in their linear ranges, it is possible to determine the relative concentrations of the specific target sequence in the original DNA mixture. If the DNA mixtures are cDNAs synthesized from RNAs isolated from different tissues or cells, the relative abundances of the specific mRNA from which the target sequence was derived may be determined for the respective tissues or cells. This direct proportionality between the concentration of the PCR products and the relative mRNA abundances is true in the linear range portion of the PCR reaction.


The final concentration of the target DNA in the plateau portion of the curve is determined by the availability of reagents in the reaction mix and is independent of the original concentration of target DNA. Therefore, in one embodiment, the sampling and quantifying of the amplified PCR products are carried out when the PCR reactions are in the linear portion of their curves. In addition, relative concentrations of the amplifiable cDNAs can be normalized to some independent standard, which may be based on either internally existing RNA species or externally introduced RNA species. The abundance of a particular mRNA species may also be determined relative to the average abundance of all mRNA species in the sample.


In one embodiment, the PCR amplification utilizes internal PCR standards that are approximately as abundant as the target. This strategy is effective if the products of the PCR amplifications are sampled during their linear phases. If the products are sampled when the reactions are approaching the plateau phase, then the less abundant product may become relatively over-represented. Comparisons of relative abundances made for many different RNA samples, such as is the case when examining RNA samples for differential expression, may become distorted in such a way as to make differences in relative abundances of RNAs appear less than they actually are. This can be improved if the internal standard is much more abundant than the target. If the internal standard is more abundant than the target, then direct linear comparisons may be made between RNA samples.


A problem inherent in clinical samples is that they are of variable quantity or quality. This problem can be overcome if the RT-PCR is performed as a relative quantitative RT-PCR with an internal standard in which the internal standard is an amplifiable cDNA fragment that is larger than the target cDNA fragment and in which the abundance of the mRNA encoding the internal standard is roughly 5-100 fold higher than the mRNA encoding the target. This assay measures relative abundance, not absolute abundance of the respective mRNA species.


In another embodiment, the relative quantitative RT-PCR uses an external standard protocol. Under this protocol, the PCR products are sampled in the linear portion of their amplification curves. The number of PCR cycles that are optimal for sampling can be empirically determined for each target cDNA fragment. In addition, the reverse transcriptase products of each RNA population isolated from the various samples can be normalized for equal concentrations of amplifiable cDNAs. While empirical determination of the linear range of the amplification curve and normalization of cDNA preparations are tedious and time-consuming processes, the resulting RT-PCR assays may, in certain cases, be superior to those derived from a relative quantitative RT-PCR with an internal standard.


In yet another embodiment, nucleic acid arrays (including bead arrays) are used for detecting or comparing the expression profiles of a prognostic gene of interest. The nucleic acid arrays can be commercial oligonucleotide or cDNA arrays. They can also be custom arrays comprising concentrated probes for the prognostic genes of the present invention. In many examples, at least 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, or more of the total probes on a custom array of the present invention are probes for leukemia prognostic genes. These probes can hybridize under stringent or nucleic acid array hybridization conditions to the RNA transcripts, or the complements thereof, of the corresponding prognostic genes.


As used herein, “stringent conditions” are at least as stringent as, for example, conditions G-L shown in Table 10. “Highly stringent conditions” are at least as stringent as conditions A-F shown in Table 10. Hybridization is carried out under the hybridization conditions (Hybridization Temperature and Buffer) for about four hours, followed by two 20-minute washes under the corresponding wash conditions (Wash Temp. and Buffer).









TABLE 10







Stringency Conditions












Poly-
Hybrid
Hybridization



Stringency
nucleotide
Length
Temperature and
Wash Temp.


Condition
Hybrid
(bp)1
BufferH
and BufferH





A
DNA:DNA
>50
65° C.; 1xSSC -or-
65° C.;





42° C.; 1xSSC, 50%
0.3xSSC





formamide


B
DNA:DNA
<50
TB*; 1xSSC
TB*; 1xSSC


C
DNA:RNA
>50
67° C.; 1xSSC -or-
67° C.;





45° C.; 1xSSC, 50%
0.3xSSC





formamide


D
DNA:RNA
<50
TD*; 1xSSC
TD*; 1xSSC


E
RNA:RNA
>50
70° C.; 1xSSC -or-
70° C.;





50° C.; 1xSSC, 50%
0.3xSSC





formamide


F
RNA:RNA
<50
TF*; 1xSSC
Tf*; 1xSSC


G
DNA:DNA
>50
65° C.; 4xSSC -or-
65° C.; 1xSSC





42° C.; 4xSSC, 50%





formamide


H
DNA:DNA
<50
TH*; 4xSSC
TH*; 4xSSC


I
DNA:RNA
>50
67° C.; 4xSSC -or-
67° C.; 1xSSC





45° C.; 4xSSC, 50%





formamide


J
DNA:RNA
<50
TJ*; 4xSSC
TJ*; 4xSSC


K
RNA:RNA
>50
70° C.; 4xSSC -or-
67° C.; 1xSSC





50° C.; 4xSSC, 50%





formamide


L
RNA:RNA
<50
TL*; 2xSSC
TL*; 2xSSC






1The hybrid length is that anticipated for the hybridized region(s) of the hybridizing polynucleotides. When hybridizing a polynucleotide to a target polynucleotide of unknown sequence, the hybrid length is assumed to be that of the hybridizing polynucleotide. When polynucleotides of known sequence are hybridized, the hybrid length can be determined by aligning the sequences of the polynucleotides and identifying the region or regions of optimal sequence complementarity.




HSSPE (1x SSPE is 0.15M NaCl, 10 mM NaH2PO4, and 1.25 mM EDTA, pH 7.4) can be substituted for SSC (1x SSC is 0.15M NaCl and 15 mM sodium citrate) in the hybridization and wash buffers.



TB*-TR*: The hybridization temperature for hybrids anticipated to be less than 50 base pairs in length should be 5-10° C. less than the melting temperature (Tm) of the hybrid, where Tm is determined according to the following equations. For hybrids less than 18 base pairs in length, Tm(° C.) = 2(# of A + T bases) + 4(# of G + C bases). For hybrids between 18 and 49 base pairs in length, Tm (° C.) = 81.5 + 16.6(log10[Na+]) + 0.41(% G + C) − (600/N), where N is the number of bases in the hybrid, and [Na+] is the molar concentration of sodium ions in the hybridization buffer ([Na+] for 1x SSC = 0.165 M).






In one example, a nucleic acid array of the present invention includes at least 2, 5, 10, or more different probes. Each of these probes is capable of hybridizing under stringent or nucleic acid array hybridization conditions to a different respective prognostic gene of the present invention. Multiple probes for the same prognostic gene can be used on the same nucleic acid array. The probe density on the array can be in any range.


The probes for a prognostic gene of the present invention can be a nucleic acid probe, such as, DNA, RNA, PNA, or a modified form thereof. The nucleotide residues in each probe can be either naturally occurring residues (such as deoxyadenylate, deoxycytidylate, deoxyguanylate, deoxythymidylate, adenylate, cytidylate, guanylate, and uridylate), or synthetically produced analogs that are capable of forming desired base-pair relationships. Examples of these analogs include, but are not limited to, aza and deaza pyrimidine analogs, aza and deaza purine analogs, and other heterocyclic base analogs, wherein one or more of the carbon and nitrogen atoms of the purine and pyrimidine rings are substituted by heteroatoms, such as oxygen, sulfur, selenium, and phosphorus. Similarly, the polynucleotide backbones of the probes can be either naturally occurring (such as through 5′ to 3′ linkage), or modified. For instance, the nucleotide units can be connected via non-typical linkage, such as 5′ to 2′ linkage, so long as the linkage does not interfere with hybridization. For another instance, peptide nucleic acids, in which the constitute bases are joined by peptide bonds rather than phosphodiester linkages, can be used.


The probes for the prognostic genes can be stably attached to discrete regions on a nucleic acid array. By “stably attached,” it means that a probe maintains its position relative to the attached discrete region during hybridization and signal detection. The position of each discrete region on the nucleic acid array can be either known or determinable. All of the methods known in the art can be used to make the nucleic acid arrays of the present invention.


In another embodiment, nuclease protection assays are used to quantitate RNA transcript levels in peripheral blood samples. There are many different versions of nuclease protection assays. The common characteristic of these nuclease protection assays is that they involve hybridization of an antisense nucleic acid with the RNA to be quantified. The resulting hybrid double-stranded molecule is then digested with a nuclease that digests single-stranded nucleic acids more efficiently than double-stranded molecules. The amount of antisense nucleic acid that survives digestion is a measure of the amount of the target RNA species to be quantified. Examples of suitable nuclease protection assays include the RNase protection assay provided by Ambion, Inc. (Austin, Tex.).


Hybridization probes or amplification primers for the prognostic genes of the present invention can be prepared by using any method known in the art. For prognostic genes whose genomic locations have not been determined or whose identities are solely based on EST or mRNA data, the probes/primers for these genes can be derived from the target sequences of the corresponding qualifiers, or the corresponding EST or mRNA sequences.


In one embodiment, the probes/primers for a prognostic gene significantly diverge from the sequences of other prognostic genes. This can be achieved by checking potential probe/primer sequences against a human genome sequence database, such as the Entrez database at the NCBI. One algorithm suitable for this purpose is the BLAST algorithm. This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold. The initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence to increase the cumulative alignment score. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. These parameters can be adjusted for different purposes, as appreciated by those skilled in the art.


In another embodiment, the probes for prognostic genes can be polypeptide in nature, such as, antibody probes. The expression levels of the prognostic genes of the present invention are thus determined by measuring the levels of polypeptides encoded by the prognostic genes. Methods suitable for this purpose include, but are not limited to, immunoassays such as ELISA, RIA, FACS, dot blot, Western Blot, immunohistochemistry, and antibody-based radioimaging. In addition, high-throughput protein sequencing, 2-dimensional SDS-polyacrylamide gel electrophoresis, mass spectrometry, or protein arrays can be used.


In one embodiment, ELISAs are used for detecting the levels of the target proteins. In an exemplifying ELISA, antibodies capable of binding to the target proteins are immobilized onto selected surfaces exhibiting protein affinity, such as wells in a polystyrene or polyvinylchloride microtiter plate. Samples to be tested are then added to the wells. After binding and washing to remove non-specifically bound immunocomplexes, the bound antigen(s) can be detected. Detection can be achieved by the addition of a second antibody which is specific for the target proteins and is linked to a detectable label. Detection can also be achieved by the addition of a second antibody, followed by the addition of a third antibody that has binding affinity for the second antibody, with the third antibody being linked to a detectable label. Before being added to the microtiter plate, cells in the samples can be lysed or extracted to separate the target proteins from potentially interfering substances.


In another exemplifying ELISA, the samples suspected of containing the target proteins are immobilized onto the well surface and then contacted with the antibodies. After binding and washing to remove non-specifically bound immunocomplexes, the bound antigen detected. Where the initial antibodies are linked to a detectable label, the immunocomplexes can be detected directly. The immunocomplexes can also be detected using a second antibody that has binding affinity for the first antibody, with the second antibody being linked to a detectable label.


Another exemplary ELISA involves the use of antibody competition in the detection. In this ELISA, the target proteins are immobilized on the well surface. The labeled antibodies are added to the well, allowed to bind to the target proteins, and detected by means of their labels. The amount of the target proteins in an unknown sample is then determined by mixing the sample with the labeled antibodies before or during incubation with coated wells. The presence of the target proteins in the unknown sample acts to reduce the amount of antibody available for binding to the well and thus reduces the ultimate signal.


Different ELISA formats can have certain features in common, such as coating, incubating or binding, washing to remove non-specifically bound species, and detecting the bound immunocomplexes. For instance, in coating a plate with either antigen or antibody, the wells of the plate can be incubated with a solution of the antigen or antibody, either overnight or for a specified period of hours. The wells of the plate are then washed to remove incompletely adsorbed material. Any remaining available surfaces of the wells are then “coated” with a nonspecific protein that is antigenically neutral with regard to the test samples. Examples of these nonspecific proteins include bovine serum albumin (BSA), casein and solutions of milk powder. The coating allows for blocking of nonspecific adsorption sites on the immobilizing surface and thus reduces the background caused by nonspecific binding of antisera onto the surface.


In ELISAs, a secondary or tertiary detection means can be used. After binding of a protein or antibody to the well, coating with a non-reactive material to reduce background, and washing to remove unbound material, the immobilizing surface is contacted with the control or clinical or biological sample to be tested under conditions effective to allow immunocomplex (antigen/antibody) formation. These conditions may include, for example, diluting the antigens and antibodies with solutions such as BSA, bovine gamma globulin (BGG) and phosphate buffered saline (PBS)/Tween and incubating the antibodies and antigens at room temperature for about 1 to 4 hours or at 4° C. overnight. Detection of the immunocomplex is facilitated by using a labeled secondary binding ligand or antibody, or a secondary binding ligand or antibody in conjunction with a labeled tertiary antibody or third binding ligand.


Following all incubation steps in an ELISA, the contacted surface can be washed so as to remove non-complexed material. For instance, the surface may be washed with a solution such as PBS/Tween, or borate buffer. Following the formation of specific immunocomplexes between the test sample and the originally bound material, and subsequent washing, the occurrence of the amount of immunocomplexes can be determined.


To provide a detecting means, the second or third antibody can have an associated label to allow detection. In one embodiment, the label is an enzyme that generates color development upon incubating with an appropriate chromogenic substrate. Thus, for example, one may contact and incubate the first or second immunocomplex with a urease, glucose oxidase, alkaline phosphatase or hydrogen peroxidase-conjugated antibody for a period of time and under conditions that favor the development of further immunocomplex formation (e.g., incubation for 2 hours at room temperature in a PBS-containing solution such as PBS-Tween).


After incubation with the labeled antibody, and subsequent washing to remove unbound material, the amount of label can be quantified, e.g., by incubation with a chromogenic substrate such as urea and bromocresol purple or 2,2′-azido-di-(3-ethyl)-benzthiazoline-6-sulfonic acid (ABTS) and H2O2, in the case of peroxidase as the enzyme label. Quantitation can be achieved by measuring the degree of color generation, e.g., using a spectrophotometer.


Another method suitable for detecting polypeptide levels is RIA (radioimmunoassay). An exemplary RIA is based on the competition between radiolabeled-polypeptides and unlabeled polypeptides for binding to a limited quantity of antibodies. Suitable radiolabels include, but are not limited to, I125. In one embodiment, a fixed concentration of I125-labeled polypeptide is incubated with a series of dilution of an antibody specific to the polypeptide. When the unlabeled polypeptide is added to the system, the amount of the I125-polypeptide that binds to the antibody is decreased. A standard curve can therefore be constructed to represent the amount of antibody-bound I125-polypeptide as a function of the concentration of the unlabeled polypeptide. From this standard curve, the concentration of the polypeptide in unknown samples can be determined. Protocols for conducting RIA are well known in the art.


Suitable antibodies for the present invention include, but are not limited to, polyclonal antibodies, monoclonal antibodies, chimeric antibodies, humanized antibodies, single chain antibodies, Fab fragments, or fragments produced by a Fab expression library. Neutralizing antibodies (i.e., those which inhibit dimer formation) can also be used. Methods for preparing these antibodies are well known in the art. In one embodiment, the antibodies of the present invention can bind to the corresponding prognostic gene products or other desired antigens with binding affinities of at least 104 M−1, 105 M−1, 106 M−1, 107 M−1, or more.


The antibodies of the present invention can be labeled with one or more detectable moieties to allow for detection of antibody-antigen complexes. The detectable moieties can include compositions detectable by spectroscopic, enzymatic, photochemical, biochemical, bioelectronic, immunochemical, electrical, optical or chemical means. The detectable moieties include, but are not limited to, radioisotopes, chemiluminescent compounds, labeled binding proteins, heavy metal atoms, spectroscopic markers such as fluorescent markers and dyes, magnetic labels, linked enzymes, mass spectrometry tags, spin labels, electron transfer donors and acceptors, and the like.


The antibodies of the present invention can be used as probes to construct protein arrays for the detection of expression profiles of the prognostic genes. Methods for making protein arrays or biochips are well known in the art. In many embodiments, a substantial portion of probes on a protein array of the present invention are antibodies specific for the prognostic gene products. For instance, at least 10%, 20%, 30%, 40%, 50%, or more probes on the protein array can be antibodies specific for the prognostic gene products.


In yet another aspect, the expression levels of the prognostic genes are determined by measuring the biological functions or activities of these genes. Where a biological function or activity of a gene is known, suitable in vitro or in vivo assays can be developed to evaluate the function or activity. These assays can be subsequently used to assess the level of expression of the prognostic gene.


After the expression level of each prognostic gene is determined, numerous approaches can be employed to compare expression profiles. Comparison of the expression profile of a patient of interest to the reference expression profile(s) can be conducted manually or electronically. In one example, comparison is carried out by comparing each component in one expression profile to the corresponding component in a reference expression profile. The component can be the expression level of a prognostic gene, a ratio between the expression levels of two prognostic genes, or another measure capable of representing gene expression patterns. The expression level of a gene can have an absolute or a normalized or relative value. The difference between two corresponding components can be assessed by fold changes, absolute differences, or other suitable means.


Comparison of the expression profile of a patient of interest to the reference expression profile(s) can also be conducted using pattern recognition or comparison programs, such as the k-nearest-neighbors algorithm as described in Armstrong, et al., NATURE GENETICS, 30:41-47 (2002), or the weighted voting algorithm as described below. In addition, the serial analysis of gene expression (SAGE) technology, the GEMTOOLS gene expression analysis program (Incyte Pharmaceuticals), the GeneCalling and Quantitative Expression Analysis technology (Curagen), and other suitable methods, programs or systems can be used to compare expression profiles.


Multiple prognostic genes can be used in the comparison of expression profiles. For instance, 2, 4, 6, 8, 10, 12, 14, or more prognostic genes can be used. In addition, the prognostic gene(s) used in the comparison can be selected to have relatively small p-values (e.g., two-sided p-values). In many examples, the p-values indicate the statistical significance of the difference between gene expression levels in different classes of patients. In many other examples, the p-values suggest the statistical significance of the correlation between gene expression patterns and clinical outcome. In one embodiment, the prognostic genes used in the comparison have p-values of no greater than 0.05, 0.01, 0.001, 0.0005, 0.0001, or less. Prognostic genes with p-values of greater than 0.05 can also be used. These genes may be identified, for instance, by using a relatively small number of blood samples.


Similarity or difference between the expression profile of a patient of interest and a reference expression profile is indicative of the class membership of the patient of interest. Similarity or difference can be determined by any suitable means. The comparison can be qualitative, quantitative, or both.


In one example, a component in a reference profile is a mean value, and the corresponding component in the expression profile of the patient of interest falls within the standard deviation of the mean value. In such a case, the expression profile of the patient of interest may be considered similar to the reference profile with respect to that particular component. Other criteria, such as a multiple or fraction of the standard deviation or a certain degree of percentage increase or decrease, can be used to measure similarity.


In another example, at least 50% (e.g., at least 60%, 70%, 80%, 90%, or more) of the components in the expression profile of the patient of interest are considered similar to the corresponding components in a reference profile. Under these circumstances, the expression profile of the patient of interest may be considered similar to the reference profile. Different components in the expression profile may have different weights for the comparison. In some cases, lower percentage thresholds (e.g., less than 50% of the total components) are used to determine similarity.


The prognostic gene(s) and the similarity criteria can be selected such that the accuracy of outcome prediction (the ratio of correct calls over the total of correct and incorrect calls) is relatively high. For instance, the accuracy of prediction can be at least 50%, 60%, 70%, 80%, 90%, or more.


The effectiveness of outcome prediction can also be assessed by sensitivity and specificity. The prognostic genes and the comparison criteria can be selected such that both the sensitivity and specificity of outcome prediction are relatively high. For instance, the sensitivity and specificity can be at least 50%, 60%, 70%, 80%, 90%, 95%, or more. As used herein, “sensitivity” refers to the ratio of correct positive calls over the total of true positive calls plus false negative calls, and “specificity” refers to the ratio of correct negative calls over the total of true negative calls plus false positive calls.


Moreover, peripheral blood expression profile-based outcome prediction can be combined with other clinical evidence or prognostic methods to improve the effectiveness or accuracy of outcome prediction.


In many embodiments, the expression profile of a patient of interest is compared to at least two reference expression profiles. Each reference expression profile can include an average expression profile, or a set of individual expression profiles each of which represents the peripheral blood gene expression pattern in a particular AML patient or disease-free human. Suitable methods for comparing one expression profile to two or more reference expression profiles include, but are not limited to, the weighted voting algorithm or the k-nearest-neighbors algorithm. Softwares capable of performing these algorithms include, but are not limited to, GeneCluster 2 software. GeneCluster 2 software is available from MIT Center for Genome Research at Whitehead Institute (e.g., wwwgenome.wi.mit.edu/cancer/software/genecluster2/gc2.html).


Both the weighted voting and k-nearest-neighbors algorithms employ gene classifiers that can effectively assign a patient of interest to an outcome class. By “effectively,” it means that the class assignment is statistically significant. In one example, the effectiveness of class assignment is evaluated by leave-one-out cross validation or k-fold cross validation. The prediction accuracy under these cross validation methods can be, for instance, at least 50%, 60%, 70%, 80%, 90%, 95%, or more. The prediction sensitivity or specificity under these cross validation methods can also be at least 50%, 60%, 70%, 80%, 90%, 95%, or more. Prognostic genes or class predictors with low assignment sensitivity/specificity or low cross validation accuracy, such as less than 50%, can also be used in the present invention.


Under one version of the weighted voting algorithm, each gene in a class predictor casts a weighted vote for one of the two classes (class 0 and class 1). The vote of gene “g” can be defined as vg=ag (xg−bg), wherein ag equals to P(g,c) and reflects the correlation between the expression level of gene “g” and the class distinction between the two classes, bg is calculated as bg=[x0(g)+x1(g)]/2 and represents the average of the mean logs of the expression levels of gene “g” in class 0 and class 1, and xg is the normalized log of the expression level of gene “g” in the sample of interest. A positive vg indicates a vote for class 0, and a negative vg indicates a vote for class 1. V0 denotes the sum of all positive votes, and V1 denotes the absolute value of the sum of all negative votes. A prediction strength PS is defined as PS=(V0−V1)/(V0+V1). Thus, the prediction strength varies between −1 and 1 and can indicate the support for one class (e.g., positive PS) or the other (e.g., negative PS). A prediction strength near “0” suggests narrow margin of victory, and a prediction strength close to “1” or “−1” indicates wide margin of victory. See Slonim, et al., PROCS. OF THE FOURTH ANNUAL INTERNATIONAL CONFERENCE ON COMPUTATIONAL MOLECULAR BIOLOGY, Tokyo, Japan, April 8-11, p 263-272 (2000); and Golub, et al., SCIENCE, 286: 531-537 (1999).


Suitable prediction strength (PS) thresholds can be assessed by plotting the cumulative cross-validation error rate against the prediction strength. In one embodiment, a positive predication is made if the absolute value of PS for the sample of interest is no less than 0.3. Other PS thresholds, such as no less than 0.1, 0.2, 0.4 or 0.5, can also be selected for class prediction. In many embodiments, a threshold is selected such that the accuracy of prediction is optimized and the incidence of both false positive and false negative results is minimized.


Any class predictor constructed according to the present invention can be used for the class assignment of a leukemia patient of interest. In many examples, a class predictor employed in the present invention includes n prognostic genes identified by the neighborhood analysis, where n is an integer greater than 1. A half of these prognostic genes has the largest P(g,c) scores, and the other half has the largest −P(g,c) scores. The number n therefore is the only free parameter in defining the class predictor.


The expression profile of a patient of interest can also be compared to two or more reference expression profiles by other means. For instance, the reference expression profiles can include an average peripheral blood expression profile for each class of patients. The fact that the expression profile of a patient of interest is more similar to one reference profile than to another suggests that the patient of interest is more likely to have the clinical outcome associated with the former reference profile than that associated with the latter reference profile.


In one particular embodiment, the present invention features prediction of clinical outcome of an AML patient of interest. AML patients can be divided into at least two classes based on their responses to a specified treatment regime. One class of patients (responders) has complete remission in response to the treatment, and the other class of patients (non-responders) has non-remission or partial remission in response to the treatment. AML prognostic genes that are correlated with a class distinction between these two classes of patients can be identified and then used to assign the patient of interest to one of these two outcome classes. Examples of AML prognostic genes suitable for this purpose are depicted in Tables 1 and 2.


In one example, the treatment regime includes administration of at least one chemotherapy agent (e.g., daunorubicin or cytarabine) and an anti-CD33 antibody conjugated with a cytotoxic agent (e.g., gemtuzumab ozogamicin), and the expression profile of an AML patient of interest is compared to two or more reference expression profiles by using a weighted voting or k-nearest-neighbors algorithm. All of these expression profiles are baseline profiles representing peripheral blood gene expression patterns prior to the treatment regime. A classifier including at least one gene selected from Table 1 and at least one gene selected from Table 2 can be employed for the outcome prediction. For instance, a classifier can include at least 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75 or more genes selected from Table 1, and at least 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75 or more genes selected from Table 2. The total number of genes selected from Table 1 can be equal to, or different from, that selected from Table 2.


Prognostic genes or class predictors capable of distinguishing three or more outcome classes can also be employed in the present invention. These prognostic genes can be identified using multi-class correlation metrics. Suitable programs for carrying out multi-class correlation analysis include, but are not limited to, GeneCluster 2 software (MIT Center for Genome Research at Whitehead Institute, Cambridge, Mass.). Under the analysis, patients having a specified type of leukemia are divided into at least three classes, and each class of patients has a different respective clinical outcome. The prognostic genes identified under multi-class correlation analysis are differentially expressed in PBMCs of one class of patients relative to PBMCs of other classes of patients. In one embodiment, the identified prognostic genes are correlated with a class distinction at above the 1%, 5%, 10%, 25%, or 50% significance level under a permutation test. The class distinction represents an idealized expression pattern of the identified genes in peripheral blood samples of patients who have different clinical outcomes.


For example, FIGS. 1A and 1B illustrate the identification and cross validation of gene classifiers for distinction of PBMCs from patients who did or did not respond to Mylotarg combination therapy. FIG. 1A shows the relative expression levels of 98 class-correlated genes. As graphically presented, 49 genes were elevated in responding patient PBMCs relative to non-responding patient PBMCs and the other 49 genes were elevated in non-responding patient PBMCs relative to responding patient PBMCs. FIG. 1B demonstrates cross validation results for each sample using a class predictor consisting of the 154 genes depicted in Tables 1 and 2. A leave-one out cross validation was performed and the prediction strengths were calculated for each sample. Samples are ordered in the same order as the nearest neighbor analysis in FIG. 1A.


The 154-gene classifier exhibited a sensitivity of 82%, correctly identifying 24 of the 28 true responders in the study. The gene classifier also exhibited a specificity of 75%, correctly identifying 6 of the 8 true non-responders in the study. Similar sensitivities, specificities and overall accuracies were observed with optimal gene classifiers identified by 10-fold and leave-one-out cross validation approaches.


The above investigation evaluated expression patterns in peripheral blood samples of AML patients prior to therapy and identified transcriptional signatures correlated with initial response to therapy. The result of this study demonstrates that pharmacogenomic peripheral blood profiling strategies enable identification of patients with high likelihoods of positive or negative outcomes in response to GO combination therapy.


Diagnosis or Monitoring the Development, Progression or Treatment of AML

The above described methods, including preparation of blood samples, assembly of class predictors, and construction and comparison of expression profiles, can be readily adapted for the diagnosis or monitoring the development, progression or treatment of AML. This can be achieved by comparing the expression profile of one or more AML disease genes in a subject of interest to at least one reference expression profile of the AML disease gene(s). The reference expression profile(s) can include an average expression profile, or a set of individual expression profiles each of which represents the peripheral blood gene expression of the AML disease gene(s) in a particular AML patient or disease-free human. Similarity between the expression profile of the subject of interest and the reference expression profile(s) is indicative of the presence or absence or the disease state of AML. In many embodiments, the disease genes employed for AML diagnosis are selected from Table 7.


One or more AML disease genes selected from Table 7 can be used for AML diagnosis or disease monitoring. In one embodiment, each AML disease gene has a p-value of less than 0.01, 0.005, 0.001, 0.0005, 0.0001, or less. In another embodiment, the AML disease genes comprise at least one gene having an “AML/Disease-Free” ratio of no less than 2 and at least one gene having an “AML/Disease-Free” ratio of no more than 0.5.


The leukemia disease genes of the present invention can be used alone, or in combination with other clinical tests, for leukemia diagnosis or disease monitoring. Conventional methods for detecting or diagnosing leukemia include, but are not limited to, bone marrow aspiration, bone marrow biopsy, blood tests for abnormal levels of white blood cells, platelets or hemoglobin, cytogenetics, spinal tap, chest X-ray, or physical exam for swelling of the lymph nodes, spleen and liver. Any of these methods, as well as any other conventional or nonconventional method, can be used, in addition to the methods of the present invention, to improve the accuracy of leukemia diagnosis.


The present invention also features electronic systems useful for the prognosis, diagnosis or selection of treatment of AML or other leukemias. These systems include an input or communication device for receiving the expression profile of a patient of interest or the reference expression profile(s). The reference expression profile(s) can be stored in a database or other media. The comparison between expression profiles can be conducted electronically, such as through a processor or a computer. The processor or computer can execute one or more programs which compare the expression profile of the patient of interest to the reference expression profile(s). The programs can be stored in a memory or downloaded from another source, such as an internet server. In one example, the programs include a k-nearest-neighbors or weighted voting algorithm. In another example, the electronic system is coupled to a nucleic acid array and can receive or process expression data generated by the nucleic acid array.


Kits for Prognosis, Diagnosis or Selection of Treatment of Leukemia

In addition, the present invention features kits useful for the prognosis, diagnosis or selection of treatment of AML or other leukemias. Each kit includes or consists essentially of at least one probe for a leukemia prognosis or disease gene (e.g., a gene selected from Tables 1, 2, 3, 4, 5, 6, 7, 8 or 9). Reagents or buffers that facilitate the use of the kit can also be included. Any type of probe can be using in the present invention, such as hybridization probes, amplification primers, or antibodies.


In one embodiment, a kit of the present invention includes or consists essentially of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more polynucleotide probes or primers. Each probe/primer can hybridize under stringent conditions or nucleic acid array hybridization conditions to a different respective leukemia prognosis or disease gene. As used herein, a polynucleotide can hybridize to a gene if the polynucleotide can hybridize to an RNA transcript, or the complement thereof, of the gene. In another embodiment, a kit of the present invention includes one or more antibodies, each of which is capable of binding to a polypeptide encoded by a different respective leukemia prognosis or disease gene.


In one example, a kit of the present invention includes or consists essentially of probes (e.g., hybridization or PCR amplification probes or antibodies) for at least 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75 or more genes selected from Table 2a, and probes for at least 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75 or more genes selected from Table 2b. The total number of probes for the genes selected from Table 2a can be identical to, or different from, that for the genes selected from Table 2b.


The probes employed in the present invention can be either labeled or unlabeled. Labeled probes can be detectable by spectroscopic, photochemical, biochemical, bioelectronic, immunochemical, electrical, optical, chemical, or other suitable means. Exemplary labeling moieties for a probe include radioisotopes, chemiluminescent compounds, labeled binding proteins, heavy metal atoms, spectroscopic markers, such as fluorescent markers and dyes, magnetic labels, linked enzymes, mass spectrometry tags, spin labels, electron transfer donors and acceptors, and the like.


The kits of the present invention can also have containers containing buffer(s) or reporter means. In addition, the kits can include reagents for conducting positive or negative controls. In one embodiment, the probes employed in the present invention are stably attached to one or more substrate supports. Nucleic acid hybridization or immunoassays can be directly carried out on the substrate support(s). Suitable substrate supports for this purpose include, but are not limited to, glasses, silica, ceramics, nylons, quartz wafers, gels, metals, papers, beads, tubes, fibers, films, membranes, column matrices, or microtiter plate wells. The kits of the present invention may also contain one or more controls, each representing a reference expression level of a prognostic or diagnostic gene detectable by one or more probes contained in the kits.


The present invention also allows for personalized treatment of AML or other leukemias. Numerous treatment options or regimes can be analyzed according to the present invention to identify prognostic genes for each treatment regime. The peripheral blood expression profiles of these prognostic genes in a patient of interest are indicative of the clinical outcome of the patient and, therefore, can be used for the selection of treatments that have favorable prognoses for the patient. As used herein, a “favorable” prognosis is a prognosis that is better than the prognoses of the majority of all other available treatments for the patient of interest. The treatment regime with the best prognosis can also be identified.


Treatment selection can be conducted manually or electronically. Reference expression profiles or gene classifiers can be stored in a database. Programs capable of performing algorithms such as the k-nearest-neighbors or weighted voting algorithms can be used to compare the peripheral blood expression profile of a patient of interest to the database to determine which treatment should be used for the patient.


It should be understood that the above-described embodiments and the following examples are given by way of illustration, not limitation. Various changes and modifications within the scope of the present invention will become apparent to those skilled in the art from the present description.


EXAMPLES
Example 1
Clinical Trial and Data Collection
Experimental Design

AML patients (13 females and 23 males) were exclusively of Caucasian descent and had a median age of 45 years (range of 19-66 years). Inclusion criteria for AML patients included blasts in excess of 20% in the bone marrow, morphologic diagnosis of AML according to the FAB classification system and flow cytometry analysis indicating positive CD33+ status. Participation in the clinical trial required concordant pathological diagnosis of AML by both an onsite pathologist following histological evaluation of bone marrow aspirates. A summary of the cytogenetic characteristics of the patients is presented in Table 11.









TABLE 11







Cytogenetic characteristics of PG consented AML patients


contributing baseline samples in 0903B1-206-US.











PG Consented



Cytogenetic Characteristic(s)
(n = 36)*







Normal karyotype
12 (33%)



Complex karyotype (>3 abnormalities)
 6 (17%)



Other
 6 (17%)



 +8
 4 (11%)



not determined
3 (8%)



 −7
3 (8%)



inv (16)
3 (8%)



−5q
2 (6%)



−7q
1 (3%)



−5q
1 (3%)



t (11; 17)
1 (3%)



+11
1 (3%)



11q23 aberration
1 (3%)










All patients received the following standard course of induction chemotherapy and were then evaluated at 36 days. On Days 1 through 7, patients received continuous infusion cytarabine at 100 mg/m2/day. Daunorubicin was given intravenously (IV bolus) on Days 1 through 3 at 45 mg/m2. On Day 4, gemtuzumab ozogamicin (6 mg/m2) was administered over approximately 2 hours as an IV infusion.


Purification and Storage of PBMCs

All disease-free and AML peripheral blood samples were shipped overnight and processed to PBMCs by a Ficoll-gradient purification. Cell counts in whole blood and in the isolated PBMC pellets were measured by hematology analyzers and isolated PBMCs were stored at −80° C. until the RNA was extracted from these samples.


RNA Extraction

RNA extraction was performed according to a modified RNeasy mini kit method (Qiagen, Valencia, Calif., USA). Briefly, PBMC pellets were digested in RLT lysis buffer containing 0.1% beta-mercaptoethanol and processed for total RNA isolation using the RNeasy mini kit. A phenol:chloroform extraction was then performed, and the RNA was repurified using the Rneasy mini kit reagents. Eluted RNA was quantified using a Spectramax 96 well plate UV reader (Molecular Devices, Sunnyvale, Calif., USA) monitoring A260/280 OD values. The quality of each RNA sample was assessed by gel electrophoresis.


RNA Amplification and Generation of GeneChip Hybridization Probe

Labeled targets for oligonucleotide arrays were prepared according to a standard laboratory method. In brief, two micrograms of total RNA were converted to cDNA using an oligo-(dT)24 primer containing a T7 DNA polymerase promoter at the 5′ end. The cDNA was used as the template for in vitro transcription using a T7 DNA polymerase kit (Ambion, Woodlands, Tex., USA) and biotinylated CTP and UTP (Enzo, Farmingdale, N.Y., USA). Labeled cRNA was fragmented in 40 mM Tris-acetate pH 8.0, 100 mM KOAc, 30 mM MgOAc for 35 min at 94° C. in a final volume of 40 mL. Ten micrograms of labeled target were diluted in 1×MES buffer with 100 mg/mL herring sperm DNA and 50 mg/mL acetylated BSA. In vitro synthesized transcripts of 11 bacterial genes were included in each hybridization reaction. The abundance of these transcripts ranged from 1:300000 (3 ppm) to 1:1000 (1000 ppm) stated in terms of the number of control transcripts per total transcripts. Labeled probes were denatured at 99° C. for 5 min and then 45° C. for 5 min and hybridized to HG_U133A oligonucleotide arrays comprised of over 22000 human genes (Affymetrix, Santa Clara, Calif., USA) according to the Affymetrix GeneChip Analysis Suite User Guide (Affymetrix). Arrays were hybridized for 16 h at 45° C. with rotation at 60 rpm. After hybridization, the hybridization mixtures were removed and stored, and the arrays were washed and stained with streptavidin R-phycoerythrin (Molecular Probes) using the GeneChip Fluidics Station 400 (Affymetrix) and scanned with an HP GeneArray Scanner (Hewlett Packard, Palo Alto, Calif., USA) following the manufacturer's instructions. These hybridization and wash conditions are collectively referred to as “nucleic acid array hybridization conditions.”


Generation of Affymetrix Signals

Array images were processed using the Affymetrix MicroArray Suite (MAS5) software such that raw array image data (.dat) files produced by the array scanner were reduced to probe feature-level intensity summaries (.cel files) using the desktop version of MAS5. Using the Gene Expression Data System (GEDS) as a graphical user interface, users provided a sample description to the Expression Profiling Information and Knowledge System (EPIKS) Oracle database and associated the correct .cel file with the description. The database processes then invoked the MAS5 software to create probeset summary values; probe intensities were summarized for each sequence using the Affymetrix Affy Signal algorithm and the Affymetrix Absolute Detection metric (Absent, Present, or Marginal) for each probeset. MAS5 was also used for the first pass normalization by scaling the trimmed mean to a value of 100. The “average difference” values for each transcript were normalized to “frequency” values using the scaled frequency normalization method (Hill, et al., Genome Biol., 2(12):research0055.1-0055.13 (2001)) in which the average differences for 11 control cRNAs with known abundance spiked into each hybridization solution were used to generate a global calibration curve. This calibration was then used to convert average difference values for all transcripts to frequency estimates, stated in units of parts per million ranging from 1:300,000 (3 parts per million (ppm)) to 1:1000 (1000 ppm) The database processes also calculated a series of chip quality control metrics and stored all the raw data and quality control calculations in the database. Only hybridized samples passing QC criteria were included in the analysis.


Example 2
Disease-Associated Transcripts in AML PBMCs

U133A-derived transcriptional profiles of the 36 AML PBMC samples were co-normalized using the scaled frequency normalization method with 20 MDS PBMC and 45 healthy volunteer PBMC. A total of 7879 transcripts were detected in one or more profiles with a maximal frequency greater than or equal to 10 ppm (denoted as 1 P, 1≧10 ppm) across the profiles.


To identify AML-associated transcripts, average fold differences between AML and normal PBMCs were calculated by dividing the mean level of expression in the AML profiles by the mean level of expression in normal profiles. A Student's t-test (two-sample, unequal variance) was used to assess the significance of the difference in expression between the groups.


For unsupervised hierarchical clustering, the 7879 transcripts meeting the expression filter 1P, 1≧10 ppm were used. Data were log transformed and gene expression values were median centered, and profiles were clustered using an average linkage clustering approach with an uncentered correlation similarity metric.


Unsupervised analysis using hierarchical clustering demonstrated that PBMCs from AML, MDS and normal healthy individuals clustered into two main clusters, with the first subgroup composed exclusively of normal PBMCs and a second subgroup composed of AML, MDS and normal PBMCs (FIG. 2). The second subgroup broke further into two distinguishable subclusters composed of an AML-like cluster populated mainly with AML PBMC profiles, an MDS-like cluster populated mainly with MDS PBMC profiles.


AML-associated transcripts in peripheral blood were identified by comparing mean levels of expression in PBMCs from the group of healthy volunteers (n=45) with mean levels of expression in PBMCs from the AML patients (n=36). The numbers of transcripts exhibiting at least a 2-fold average difference between normal and AML PBMCs at increasing levels of significance are presented in Table 12. A total of 660 transcripts possessed at least an average 2-fold difference between the AML profiles and normal PBMC profiles and a significance in an unpaired Student's t-test less than 0.001. These transcripts are presented in Table 7, above. Of these, 382 transcripts exhibited a mean elevated level of expression 2 fold or higher in AML and the fifty genes with the greatest fold elevation are presented in Table 8. A total of 278 transcripts exhibited a mean reduced level of expression 2-fold or lower in AML and the fifty genes with the greatest fold reduction in AML are presented in Table 9.









TABLE 12







Numbers of two-fold changed genes between AML and


disease-free PBMCs meeting increasing levels of significance











No. of transcripts with average 2-fold



Significance Level
change in AML PBMCs







p < 1 × 10-3
660



p < 1 × 10-4
575



p < 1 × 10-5
491



p < 1 × 10-6
407



p < 1 × 10-7
319



p < 1 × 10-8
264



p < 1 × 10-9
218










In these studies a total of 382 transcripts possessed significantly higher levels of expression in AML PBMCs. Elevated levels of expression may be due to 1) increased transcriptional activation in circulating PBMCs or 2) elevated levels of certain subtypes of cells in circulating PBMCs. Many of the transcripts that are elevated in AML PBMCs in this study appear to be contributed by leukemic blasts present in the peripheral circulation of these patients. Many of the transcripts are known to be specifically expressed and/or linked to disease-processes in immature or leukemic blasts (myeloperoxidase, v-myb myeloblastosis proto-oncogene, v-kit proto-oncogene, fms-related tyrosine kinase 3, CD34). In addition, many of the transcripts with the highest level of expression in AML PBMCs are at undetectable or extremely low levels in purified populations of monocytes, B-cells, T-cells, and neutrophils (data not shown) and were classified as low expressors in a healthy volunteer observational study. Thus the majority of transcripts observed to present in higher quantitites in AML PBMCs do not appear to be mainly due to transcriptional activation but rather due to the presence of leukemic blasts in the circulation of AML patients.


Conversely, disease-associated transcripts at significantly lower levels in AML PBMCs appear to be transcripts exhibiting high levels of expression in one or more of the normal types of cells typically isolated by cell-purification tubes (monocytes, B-cells, T-cells, and copurifying neutrophils). For instance, eight of the top ten transcripts at lower levels in AML PBMCs possess average levels of expression in their respective purified cell type of greater than 50 ppm, and were classified as high expressors in a healthy volunteer observational study. Thus the majority of transcripts observed to be present in lower quantities in AML PBMCs do not appear to be mainly due to transcriptional repression but rather due to the decreased presence of normal mononuclear cells in the blast-rich circulation of patients with AML.


Example 3
Transcriptional Effects of Therapy

A total of 27 AML patients provided evaluable baseline and Day 36 post-treatment PBMC samples. The U133A-derived transcriptional profiles of the 27 paired AML PBMC samples were co-normalized using the scaled frequency normalization method. A total of 8809 transcripts were detected in one or more profiles with a maximal frequency greater than or equal to 10 ppm (denoted as 1P, 1≧10 ppm) across the profiles.


To identify transcripts altered during the course of therapy, average fold differences between Day 0 and Day 36 PBMC profiles were calculated by dividing the mean level of expression in the baseline Day 0 profiles by the mean level of expression in the post-treatment Day 36 profiles. A Student's t-test (two-sample, unequal variance) was used to assess the significance of the difference in expression between the groups.


GO-based therapy-associated transcripts in peripheral blood were identified by comparing mean levels of expression in PMBCs from baseline samples (n=27) with mean levels of expression in PBMCs from the paired post-treatment samples (n=27) from the same AML patients. The numbers of transcripts exhibiting at least a 2-fold average difference between baseline and post-treatment PBMCs with increasing levels of significance are presented in Table 13. A total of 607 transcripts possessed at least an average 2-fold difference between the baseline and post-treatment samples, and significance in a paired Student's t-test of less than 0.001. Of these, 348 transcripts exhibited a mean reduced level of expression 2-fold or greater over the course of therapy and the fifty genes with the greatest fold reduction following GO therapy are presented in Table 14. A total of 259 transcripts exhibited a mean elevated level of expression 2-fold or greater over the course of therapy and the fifty genes with the greatest fold elevation following GO therapy are presented in Table 15. The genes most strongly altered over the course of therapy (mean induction or repression of 3-fold or greater) were annotated with respect to their cellular functions according to their Gene Ontology annotation and the percent of transcripts in each category are presented in FIG. 3.









TABLE 13







Numbers of two-fold changed genes between Day 0 (baseline) and


Day 36 (final visit) meeting increasing levels of significance











No. of transcripts with average




2-fold change between



Significance Level
baseline (Day 0) and final visit (Day 36)














p < 1 × 10-3
607



p < 1 × 10-4
451



p < 1 × 10-5
272



p < 1 × 10-6
122



p < 1 × 10-7
38



p < 1 × 10-8
16



p < 1 × 10-9
5

















TABLE 14







Top 50 transcripts significantly repressed (p < 0.001)


in AML PBMCs following 36-day therapy regimen
















Fold Diff







(Final/
p-value


Affymetrix ID
Name
Cyto Band
Unigene ID
Baseline)
(unequal)





205051_s_at
v-kit Hardy-Zuckerman 4
4q11-q12
Hs.81665
0.13
3.02E−06



feline sarcoma viral



oncogene homolog


206310_at
serine protease inhibitor,
4q11
Hs.98243
0.14
1.06E−04



Kazal type, 2 (acrosin-



trypsin inhibitor)


209905_at
homeo box A9
7p15-p14
Hs.127428
0.14
6.28E−04


209160_at
aldo-keto reductase
10p15-p14
Hs.78183
0.15
1.71E−04



family 1, member C3 (3-



alpha hydroxysteroid



dehydrogenase, type II)


215382_x_at
tryptase beta 1, tryptase,
16p13.3
Hs.347933
0.15
8.80E−04



alpha


204798_at
v-myb myeloblastosis
6q22-q23
Hs.1334
0.16
4.65E−07



viral oncogene homolog



(avian)


207741_x_at
tryptase, alpha
16p13.3
Hs.334455
0.16
7.19E−04


214651_s_at
homeo box A9
7p15-p14
Hs.127428
0.16
2.12E−04


205131_x_at
stem cell growth factor;
19q13.3
Hs.105927
0.16
3.08E−05



lymphocyte secreted C-



type lectin


211709_s_at
stem cell growth factor;
19q13.3
Hs.105927
0.16
3.85E−06



lymphocyte secreted C-



type lectin


219054_at
hypothetical protein
5p13.2
Hs.13528
0.17
1.19E−05



FLJ14054


203948_s_at
myeloperoxidase
17q23.1
Hs.1817
0.17
1.36E−04


203949_at
myeloperoxidase
17q23.1
Hs.1817
0.17
2.81E−05


204304_s_at
prominin-like 1 (mouse)
4p15.33
Hs.112360
0.17
3.79E−05


201892_s_at
IMP (inosine
3p21.2
Hs.75432
0.18
8.66E−07



monophosphate)



dehydrogenase 2


219837_s_at
cytokine-like protein C17
4p16-p15
Hs.13872
0.18
5.00E−04


206674_at
fms-related tyrosine
13q12
Hs.385
0.18
1.01E−06



kinase 3


201416_at
Meis1, myeloid ecotropic
17p11.2,
Hs.83484
0.18
8.38E−04



viral integration site 1
6p22.3



homolog 3 (mouse), SRY



(sex determining region



Y)-box 4


221004_s_at
integral membrane
2q37
Hs.111577
0.20
6.77E−05



protein 3


211743_s_at
proteoglycan 2, bone
11q12
Hs.99962
0.20
9.21E−04



marrow (natural killer cell



activator, eosinophil



granule major basic



protein)


205609_at
angiopoietin 1
8q22.3-q23
Hs.2463
0.21
3.50E−05


210783_x_at
stem cell growth factor;
19q13.3
Hs.105927
0.22
8.73E−05



lymphocyte secreted C-



type lectin


218788_s_at
hypothetical protein
1q44
Hs.8109
0.22
3.92E−06



FLJ21080


209790_s_at
caspase 6, apoptosis-
4q25
Hs.3280
0.23
2.24E−04



related cysteine protease


202589_at
thymidylate synthetase
18p11.32
Hs.82962
0.24
3.96E−04


201418_s_at
Meis1, myeloid ecotropic
17p11.2,
Hs.83484
0.24
7.62E−05



viral integration site 1
6p22.3



homolog 3 (mouse), SRY



(sex determining region



Y)-box 4


201459_at
RuvB-like 2 (E. coli)
19q13.3
Hs.6455
0.24
8.40E−06


209757_s_at
v-myc myelocytomatosis
2p24.1
Hs.25960
0.25
1.59E−04



viral related oncogene,



neuroblastoma derived



(avian)


213258_at
unknown
N/A
Hs.288582
0.25
1.55E−05


212115_at
hypothetical protein
16p13.11
Hs.172035
0.25
3.00E−04



FLJ13092


204040_at
KIAA0161 gene product
2p25.3
Hs.78894
0.26
4.12E−07


218858_at
hypothetical protein
8q12.2
Hs.87729
0.26
5.84E−04



FLJ12428


205899_at
cyclin A1
13q12.3-q13
Hs.79378
0.26
4.58E−04


201310_s_at
P311 protein
5q21.3
Hs.142827
0.26
2.90E−06


206589_at
growth factor
1p22
Hs.73172
0.27
1.28E−05



independent 1


222036_s_at
MCM4 minichromosome
8q12-q13
Hs.154443
0.28
4.13E−04



maintenance deficient 4



(S. cerevisiae)


201596_x_at
keratin 18
12q13
Hs.65114
0.28
5.76E−04


201162_at
insulin-like growth factor
4q12
Hs.119206
0.28
2.51E−06



binding protein 7


203787_at
single-stranded DNA
5q14.1
Hs.169833
0.29
7.97E−05



binding protein 2


219218_at
hypothetical protein
17q25.3
Hs.98968
0.29
1.32E−04



FLJ23058


220416_at
KIAA1939 protein
15q15.2
Hs.182738
0.29
5.92E−05


201307_at
hypothetical protein
4q13.3
Hs.8768
0.29
1.17E−05



FLJ10849


201841_s_at
heat shock 27 kD protein 1
7p12.3
Hs.76067
0.30
7.13E−04


209360_s_at
runt-related transcription
21q22.3
Hs.129914
0.30
1.79E−05



factor 1 (acute myeloid



leukemia 1; aml1



oncogene)


202502_at
acyl-Coenzyme A
1p31
Hs.79158
0.31
1.62E−06



dehydrogenase, C-4 to



C-12 straight chain


202503_s_at
KIAA0101 gene product
15q22.1
Hs.81892
0.31
3.51E−04


201930_at
MCM6 minichromosome
2q21
Hs.155462
0.31
1.36E−05



maintenance deficient 6



(MIS5 homolog, S. pombe)



(S. cerevisiae)


201417_at
unknown
N/A
N/A
0.31
1.07E−04


202746_at
unknown
N/A
N/A
0.32
6.07E−04


212009_s_at
stress-induced-
11q13
Hs.75612
0.32
4.03E−06



phosphoprotein 1



(Hsp70/Hsp90-



organizing protein)
















TABLE 15







Top 50 transcripts significantly elevated (p < 0.001) in AML PBMCs following


36-day therapy regimen
















Fold Diff





Cyto

(Final/
p-value


Affymetrix ID
Name
Band
Unigene ID
Baseline)
(unequal)





201506_at
transforming growth
5q31
Hs.118787
7.89
9.88E−09



factor, beta-induced,



68 kD


210244_at
cathelicidin antimicrobial
3p21.3
Hs.51120
7.53
2.43E−05



peptide


203887_s_at
thrombomodulin
20p12-cen
Hs.2030
6.84
3.15E−07


202437_s_at
cytochrome P450,
2p21
Hs.154654
6.25
1.56E−04



subfamily I (dioxin-



inducible), polypeptide 1



(glaucoma 3, primary



infantile)


212531_at
lipocalin 2 (oncogene
9q34
Hs.204238
6.05
6.81E−05



24p3)


206343_s_at
neuregulin 1
8p21-p12
Hs.172816
5.25
1.02E−06


203888_at
thrombomodulin
20p12-cen
Hs.2030
5.12
1.46E−06


210512_s_at
vascular endothelial
6p12
Hs.73793
5.05
3.55E−07



growth factor


202436_s_at
cytochrome P450,
2p21
Hs.154654
4.93
2.11E−04



subfamily I (dioxin-



inducible), polypeptide 1



(glaucoma 3, primary



infantile)


203821_at
diphtheria toxin receptor
5q23
Hs.799
4.89
2.64E−07



(heparin-binding



epidermal growth factor-



like growth factor)


206881_s_at
leukocyte
19q13.4
Hs.113277
4.76
2.08E−06



immunoglobulin-like



receptor, subfamily A



(without TM domain),



member 3


205237_at
ficolin
9q34
Hs.252136
4.64
1.21E−08



(collagen/fibrinogen



domain containing) 1


208146_s_at
carboxypeptidase,
7p15-p14
Hs.95594
4.53
9.53E−09



vitellogenic-like


220532_s_at
LR8 protein
7q35
Hs.190161
4.51
6.60E−04


38037_at
diphtheria toxin receptor
5q23
Hs.799
4.36
1.13E−06



(heparin-binding



epidermal growth factor-



like growth factor)


201566_x_at
inhibitor of DNA binding
2p25
Hs.180919
4.31
1.15E−08



2, dominant negative



helix-loop-helix protein


203435_s_at
membrane metallo-
3q25.1-q25.2
Hs.1298
4.20
9.64E−04



endopeptidase (neutral



endopeptidase,



enkephalinase, CALLA,



CD10)


213524_s_at
putative lymphocyte
1q32.2-q41
Hs.95910
4.17
7.96E−08



G0/G1 switch gene


205174_s_at
glutaminyl-peptide
2p22.3
Hs.79033
4.11
2.91E−10



cyclotransferase



(glutaminyl cyclase)


204115_at
guanine nucleotide
7q31-q32
Hs.83381
4.10
1.06E−05



binding protein 11


221211_s_at
chromosome 21 open
21q22.3
Hs.41267
3.99
7.25E−06



reading frame 7


202018_s_at
lactotransferrin
3q21-q23
Hs.105938
3.98
2.62E−04


211924_s_at
plasminogen activator,
19q13
Hs.179657
3.86
2.20E−07



urokinase receptor


204006_s_at
Fc fragment of IgG, low
1q23
Hs.372679
3.75
1.62E−04



affinity IIIa, receptor for



(CD16), Fc fragment of



IgG, low affinity IIIb,



receptor for (CD16)


201565_s_at
inhibitor of DNA binding
2p25
Hs.180919
3.68
4.06E−10



2, dominant negative



helix-loop-helix protein


206130_s_at
asialoglycoprotein
17p
Hs.1259
3.65
1.56E−05



receptor 2


203979_at
cytochrome P450,
2q33-qter
Hs.82568
3.57
3.78E−04



subfamily XXVIIA (steroid



27-hydroxylase,



cerebrotendinous



xanthomatosis),



polypeptide 1


206390_x_at
platelet factor 4
4q12-q21
Hs.81564
3.57
9.97E−06


210146_x_at
leukocyte
19q13.4
Hs.22405
3.49
5.04E−08



immunoglobulin-like



receptor, subfamily B



(with TM and ITIM



domains), member 2


204112_s_at
histamine N-
2q21.1
Hs.81182
3.49
1.30E−06



methyltransferase


211135_x_at
leukocyte
19q13.4
Hs.105928
3.49
4.18E−07



immunoglobulin-like



receptor, subfamily B



(with TM and ITIM



domains), member 3


208601_s_at
tubulin, beta 1
20q13.32
Hs.303023
3.45
3.68E−04


210845_s_at
plasminogen activator,
19q13
Hs.179657
3.42
1.72E−09



urokinase receptor


211527_x_at
vascular endothelial
6p12
Hs.73793
3.40
1.08E−05



growth factor


221210_s_at
chromosome 1 open
1q25
Hs.23756
3.40
2.18E−07



reading frame 13


201393_s_at
insulin-like growth factor
6q26
Hs.76473
3.40
1.75E−06



2 receptor


205568_at
aquaporin 9
15q22.1-22.2
Hs.104624
3.33
3.73E−05


221698_s_at
C-type (calcium
12p13.2-p12.3
Hs.161786
3.33
1.08E−06



dependent,



carbohydrate-recognition



domain) lectin,



superfamily member 12


204081_at
neurogranin (protein
11q24
Hs.26944
3.31
2.29E−05



kinase C substrate, RC3)


206359_at
suppressor of cytokine
17q25.3
Hs.345728
3.28
1.70E−07



signaling 3


219593_at
peptide transporter 3
11q13.1
Hs.237856
3.27
6.44E−07


204007_at
Fc fragment of IgG, low
1q23
Hs.176663
3.26
3.24E−04



affinity IIIa, receptor for



(CD16)


201739_at
serum/glucocorticoid
6q23
Hs.296323
3.21
9.28E−08



regulated kinase


203645_s_at
CD163 antigen
12p13.3
Hs.74076
3.20
3.41E−04


203414_at
monocyte to macrophage
17q
Hs.79889
3.16
5.41E−09



differentiation-associated


214696_at
hypothetical protein
17p13.3
Hs.29206
3.16
4.12E−08



MGC14376


210225_x_at
leukocyte
19q13.4
Hs.105928
3.13
1.37E−06



immunoglobulin-like



receptor, subfamily B



(with TM and ITIM



domains), member 3


203561_at
Fc fragment of IgG, low
1q23
Hs.78864
3.11
1.83E−06



affinity IIa, receptor for



(CD32)


218454_at
hypothetical protein
12p13.31
Hs.178470
3.10
1.67E−07



FLJ22662


221724_s_at
C-type (calcium
12p13
Hs.115515
3.08
1.10E−08



dependent,



carbohydrate-recognition



domain) lectin,



superfamily member 6









Comparison of pre- and post-treatment PBMC profiles from AML patients revealed a large number of differences in transcript levels over the course of therapy. Annotation of the genes apparently repressed over the course of therapy using Gene Ontology annotation (see FIG. 3) demonstrated that many of the transcripts at lower levels following therapy fell into an uncharacterized category. Further evaluation revealed that the vast majority of these transcripts were disease associated and were present at lower quantities in post-treatment samples due to the disappearance of leukemic blasts in these patients following therapy. Consistent with this observation, forty-five of the top 50 transcripts down-regulated following the GO regimen were disease (blast)-associated genes. Thus the down-regulation of v-kit, tryptase, aldo-keto reductase 1C3, homeobox A9, meis1, myeloperoxidase, and the majority of other transcripts exhibiting the greatest fold reduction appear to be due to the disappearance of leukemic blasts in the circulation, rather than direct transcriptional effects of the chemotherapy regimen.


Evaluation of the transcripts in PBMCs at higher levels following therapy revealed the opposite trend and showed that the vast majority of these transcripts were associated with normal PBMC expression and were present at higher quantities in post-treatment samples due to the reappearance of normal mononuclear cells in the majority of treated patients. A total of thirty-one of the top 50 transcripts up-regulated following the GO regimen were transcripts associated with normal mononuclear cell expression. Thus the up-regulation of the TGF-beta induced protein (68 kDa), thrombomodulin, putative lymphocyte G0/G1 switch gene, and the majority of other transcripts are likely due to the disappearance of leukemic blasts and repopulation of normal cells in the circulation, rather than direct transcriptional effects of the chemotherapy regimen.


For a smaller number of genes, transcriptional activation or repression may be the cause for differences in transcript levels. For instance, cytochrome P4501A1 (CYP1A1) is induced following therapy but is not significantly associated with normal mononuclear cell expression (i.e., CYP1A1 was not significantly repressed in AML PBMCs compared to normal PBMCs). CYP1A1 is involved in the metabolism of daunorubicin, and daunorubicin is a mechanism-based inactivator of CYP1A1 activity. Thus the elevation of CYP1A1mRNA may represent a feedback transcriptional response to the present therapeutic regimen. Interferon-inducible proteins were also elevated during the course of therapy (interferon-inducible protein 30, interferon-induced transmembrane protein 2), and these effects may also represent transcriptional inductions of interferon-dependent signaling pathways activated during the course of therapy.


Whether due to disappearance of blasts, elevations in normal cell counts or actual transcriptional activation or repression, alterations in several of the PBMC transcripts may have functional consequences on the progression of AML. TGF-beta induces cell cycle arrest and antagonizes FLT3-induced proliferation of leukemic cells, and a TGF-beta induced protein was the most strongly upregulated transcript (>7 fold elevated) in PBMCs during the course of therapy.


Example 4
Pretreatment Expression Patterns Associated with Veno-Occlusive Disease

U133A-derived transcriptional profiles of the 36 AML PBMC samples were co-normalized using the scaled frequency normalization method. A total of 7405 transcripts were detected in one or more profiles with a maximal frequency greater than or equal to 10 ppm (denoted as 1P, 1≧10 ppm) across the profiles.


Veno-occlusive disease (VOD) is one of the most serious complications following hematopoietic stem cell transplantation and is associated with a very high mortality in its severe form. To identify transcripts with significant differences in expression at baseline between the four patients who eventually experienced VOD and the thirty-two non-VOD patients, average fold differences between VOD and non-VOD patient profiles were calculated by dividing the mean level of expression in the four baseline VOD profiles by the mean level of expression in the 32 baseline non-VOD profiles. A Student's t-test (two-sample, unequal variance) was used to assess the significance of the difference in expression between the groups.


Transcripts in baseline PBMCs significantly associated with the onset of VOD were identified by comparing mean levels of expression in PMBCs from the VOD baseline samples (n=4) with mean levels of expression in PBMCs from the non-VOD baseline samples (n=32). The numbers of transcripts exhibiting at least a 2-fold average difference between VOD and non-VOD baseline PBMCs with increasing levels of significance are presented in Table 16. A total of 161 transcripts possessed at least an average 2-fold difference between the baseline VOD and non-VOD samples, and significance in a paired Student's t-test of less than 0.05. Of the 161 transcripts, only 3 transcripts exhibited a mean elevated level of expression 2-fold or greater in VOD PBMCs at baseline. These and forty-seven other transcripts showing less than 2-fold but exhibiting the greatest fold elevation in VOD patients at baseline are presented in Table 5. The levels of p-selectin ligand, a potentially biologically relevant transcript that appeared to be significantly elevated in PBMCs of patients who eventually experienced VOD, are presented in FIG. 4.









TABLE 16







Numbers of two-fold changed genes between baseline samples


of VOD patients (n = 4) and non-VOD patients (n = 32)


meeting increasing levels of significance









No. of transcripts with average 2-fold change


Significance Level
between baseline (Day 0) and final visit (Day 36)











p < 0.05
161


p < 0.01
98


p < 1 × 10-3
42


p < 1 × 10-4
10


p < 1 × 10-5
4


p < 1 × 10-6
2









The remaining 158 transcripts exhibited a mean reduced level of expression 2-fold or greater in VOD PBMCs at baseline, and the fifty genes with the greatest fold reduction in VOD patient PBMCs at baseline are presented in Table 6. Evaluation of this set of transcripts revealed a majority of leukemic blast-associated markers. This unanticipated finding by microarray analysis actually suggests that patients with lower peripheral blast counts may be more susceptible to VOD in the context of GO-based therapy.


Example 5
Pretreatment Transcriptional Patterns Associated with Clinical Response

As in the preceding Example, 7405 transcripts detected with a maximal frequency greater than or equal to 10 ppm in one or more profiles were selected for further evaluation.


To identify transcripts with significant differences in expression at baseline between the 8 patients who were non-responders (NR) and the 28 patients who were responders (R), average fold differences between NR and R patient profiles were calculated by dividing the mean level of expression in the eight baseline NR profiles by the mean level of expression in the 28 baseline R profiles. A Student's t-test (two-sample, unequal variance) was used to assess the significance of the difference in expression between the groups. The numbers of transcripts exhibiting at least a 2-fold average difference between R and NR baseline PBMCs with increasing levels of significance are presented in Table 17. A total of 113 transcripts possessed at least an average 2-fold difference between the baseline R and NR samples, and significance in a paired Student's t-test of less than 0.05. Of the 113 transcripts, 6 transcripts exhibited a mean elevated level of expression 2-fold or higher in non-responder PBMCs at baseline. These and forty-four other transcripts showing less than 2-fold but exhibiting the greatest fold elevation in responding patients at baseline are presented in Table 3. A total of 107 transcripts exhibited a mean reduced level of expression 2-fold or greater in non-responder PBMCs at baseline, and the fifty genes with the greatest fold reduction are presented in Table 4.









TABLE 17







Numbers of two-fold changed genes between baseline


samples of non-responding patients (n = 8) and responding


patients (n = 28) meeting increasing levels of significance











No. of transcripts with average 2-fold




change between NR and R at



Significance Level
baseline














p < 0.05
113



p < 0.01
45



p < 1 × 10-3
7



p < 1 × 10-4
1










Pretreatment levels of transcripts encoded by genes with potential roles in the metabolism or mechanism of action of GO were specifically interrogated as well. Levels of the MDR1 drug efflux transporter were low in all PBMC samples and were not significantly distinct between responders and non-responders at baseline (FIG. 5). The remaining members of the ABC transporter family contained on the Affymetrix U133A gene chip were also interrogated in the event that another ABC transporter might be differentially expressed, but none of the ABC transporters were significantly distinct between responder and non-responder PBMCs at baseline (FIG. 6). Levels of transcripts encoding the CD33 cell surface receptor were detected at generally higher levels in the AML PBMCs, but like MDR1, the CD33 transcript was also not significantly distinct between R and NR PBMCs at baseline (FIG. 7).


To identify a gene classifier capable of classifying responder and non-responders on the basis of baseline gene expression patterns, gene selection and supervised class prediction were performed using Genecluster version 2.0 previously described and available at (http://www.genome.wi.mit.edu/cancer/software/genecluster2.html). For nearest neighbor analysis, expression profiles for 36 baseline AML PMBCs from were co-normalized using the scale frequency method with 14 baseline AML PBMCs from an independent clinical trial of GO in combination with daunorubicin. All expression data were z-score normalized prior to analysis. A total of 11382 sequences were used in this analysis, based on inclusion of all transcripts with frequencies possessing at least one value of greater than or equal to 5 ppm across the baseline profiles. The 36 PBMC baseline profiles from were treated as a training set, and models containing increasing numbers of features (transcript sequences) were built using a one versus all approach with a S2N similarity metric that used median values for the class estimate. All comparisons were binary distinctions, and each model (with increasing numbers of features) was evaluated in the 36 PBMC profiles by 10-fold cross validation. The optimally predictive model arising from the 10-fold cross validation of the 36 PBMC profiles was then applied to the 14 co-normalized profiles from the other clinical trial to evaluate the gene classifiers accuracy in an independent set of clinical samples taken from AML patients prior to therapy.


A 10-gene classifier was found to yield the highest overall prediction accuracy (78%) by 10-fold cross validation on the peripheral blood AML profiles in the present study (FIG. 8 and Table 18). This gene classifier exhibited a sensitivity of 86%, a specificity of 50%, a positive predictive value of 86% and a negative predictive value of 50%. This classifier was also applied to the 14 untested profiles from the independent study in which GO plus daunorubicin composed the therapy regimen; the results are presented in FIG. 9. For those 14 profiles, the ten gene classifier demonstrated an overall prediction accuracy of 78%, a sensitivity of 100%, a specificity of 57%, a positive predictive value of 70% and a negative predictive value of 100%.









TABLE 18







Transcripts in the 10-gene classifier associated with elevated PBMC levels in


responders (top panel) or non-responders (bottom panel) prior to therapy.












Top S2N







Transcripts

Affymetrix


Elevated in:
Rank
ID
Name
Cyto Band
Unigene ID





R
1
203739_at
zinc finger protein 217
20q13.2
Hs.155040


R
2
219593_at
peptide transporter 3
11q13.1
Hs.237856


R
3
204132_s_at
forkhead box O3A
6q21
Hs.14845


R
4
210972_x_at
T cell receptor alpha
14q11.2
Hs.74647





locus


R
5
205220_at
putative chemokine
12q24.31
Hs.137555





receptor; GTP-binding





protein


NR
1
208581_x_at
metallothionein 1L,
16q13
Hs.278462





metallothionein 1X


NR
2
208963_x_at
fatty acid desaturase 1
11q12.2-q13.1
Hs.132898


NR
3
216336_x_at
uncharacterized
n/a
n/a


NR
4
209407_s_at
deformed epidermal
11p15.5
Hs.6574





autoregulatory factor 1





(Drosophila)


NR
5
203725_at
growth arrest and DNA-
1p31.2-p31.1
Hs.80409





damage-inducible, alpha









Some pharmacogenomic co-diagnostics developed in the future will likely rely on qRT-PCR based assays that can utilize small (pair-wise or greater) combinations of genes that enable accurate classification. To identify a smaller classifier the Affymetrix-based expression levels of two genes (Table 19), metallothionein 1X/1L and serum glucocorticoid regulated kinase, which were overexpressed in AML PBMCs from non-responders and responders respectively, were plotted to determine whether a pair-wise combination of transcripts could enable classification (FIG. 10, panel A). The two gene classifier employing metallothionein 1X/1L and serum glucocorticoid regulated kinase was selected on the basis of their 1) significantly elevated or repressed fold differences between responder and non-responder categories, respectively; and 2) known annotation. The individual expression values (in terms of ppm) of each transcript in each baseline AML sample were plotted to identify cutoffs for expression that gave the highest sensitivity and specificity for class assignment. From the original 36 patients, six of the eight non-responders had serum glucocorticoid regulated kinase levels <30 ppm and metallothionein 1X/1L levels>30 ppm. Only 2 of the 28 responders possessed similar levels of gene expression. For these 36 sample, the 2-gene classifier therefore exhibited an apparent 88% overall accuracy, a sensitivity of 93%, a specificity of 75%, a positive predictive value of 93% and a negative predictive value of 75%.


Table 19. Transcripts in the 2-Gene Classifier Associated with Elevated Levels in Responders (Serum/Gluclocorticoid Regulated Kinase) or Non-Responders (metallothionein 1L,1X) Prior to Therapy









TABLE 19







Transcripts in the 2-gene classifier associated with elevated levels


in responders (serum/gluclocorticoid regulated kinase) or non-


responders (metallothionein 1L, 1X) prior to therapy.












Cyto
Unigene


Affymetrix ID
Name
Band
ID





201739_at
serum/glucocorticoid
6q23
Hs•296323



regulated kinase


208581_x_at
metallothionein 1L,
16q13
Hs•278462



metallothionein 1X









This 2-gene classifier (serum glucocorticoid regulated kinase <30 ppm, metallothionein 1X,1L>30 ppm) was also applied to the 14 untested profiles from the independent clinical trial in which GO plus daunorubicin composed the therapy regimen (FIG. 10, panel B). In that study, the 2-gene classifier demonstrated identical overall performance as the 10-gene classifier, with an overall prediction accuracy of 78%, a sensitivity of 100%, a specificity of 57%, a positive predictive value of 70% and a negative predictive value of 100%.


Apparent performance characteristics of both the 10-gene and 2-gene classifiers for the first dataset of 36 samples and actual performance characteristics of both classifiers in the evaluation of the 14 independent samples are listed in Table 20.









TABLE 20







Performance characteristics of the 2-gene and 10-gene classifiers


by cross-validation and in a test set.










10 gene classifier
2 gene classifier











Cross-validation











Accuracy
78%
88%



Sensitivity
86%
93%



Specificity
50%
75%



Positive predictive value
86%
93%



Negative predictive value
50%
75%







Test set











Accuracy
78%
78%



Sensitivity
100%
100%



Specificity
57%
57%



Positive predictive value
70%
70%



Negative predictive value
100%
100%










In this analysis transcriptional profiling was applied to baseline peripheral blood samples to characterize transcriptional patterns that might provide insights into, or biomarkers for, AML patients' abilities to respond or fail to respond to a GO combination chemotherapy regimen. The largest percentage of patients in this study possessed a normal karyotype (33%), while other chromosomal abnormalities were relatively evenly distributed among the remaining patients. This heterogeneity of cytogenetic backgrounds allowed us to analyze the entire group of AML profiles without segregating them into karyotype-based groups, which in turn enabled us to search for transcriptional patterns that might be correlated with response to the GO combination regimen regardless of the molecular abnormalities involved in this complex disease. Despite the recent description of expression signatures associated with various chromosomal abnormalities in AML, it is clear that expression of many of the individual transcripts in the hallmark signatures are not unique to specific karyotypes. In addition, Bullinger et al. (2004) N. Engl. J. Med. 350:1605-16, importantly demonstrated in their recent study that relatively homogeneous transcriptional patterns correlated with overall survival were detectable in AML samples from patients despite their diverse cytogenetic backgrounds, and these prognostic profiles segregated samples from a test set of patients into good and poor outcome categories that possessed significant differences in overall survival.


An objective of the present study was not necessarily to identify generally prognostic profiles associated with overall survival, but rather to identify a transcriptional pattern in peripheral blood that, if validated, could allow identification of patients who would or would not benefit (i.e., achieve initial remission) from a GO combination chemotherapy regimen. Comparison of responder (i.e. remission) and non-responder profiles at baseline identified a number of transcripts significantly altered between the groups.


Transcripts present at higher levels in responding patients prior to therapy included T-cell receptor alpha locus, serum/glucocorticoid regulated kinase, aquaporin 9, forkhead box 03, IL8, TOSO (regulator of fas-induced apoptosis), IL1 receptor antagonist, p21/cip1, a specific subset of IFN-inducible transcripts, and other regulatory molecules. The list of transcripts elevated in responder peripheral blood appears to contain markers of both normal peripheral blood cells (lymphocytes, monocytes and neutrophils) and blast-specific transcripts alike. A higher percentage of pro-apoptotic related molecules were elevated in peripheral blood of patients who ultimately responded to therapy. FOX03 is a critical pro-apoptotic molecule that is inactivated during IL2-mediated T-cell survival and has recently been shown to be inactivated during FLT3-induced, PI3Kinase dependent stimulation of proliferation in myeloid cells. The finding that FOX03 is elevated in peripheral blood of AML patients that ultimately responded to GO combination therapy supports the theory that apoptotically “primed” cells will be more sensitive to the effects of GO based therapy regimens and possibly other chemotherapies as well. Levels of FOX01A are positively correlated with survival in AML patients receiving two different regimens.


A number of transcripts were also elevated in blood samples of AML patients who failed to respond to therapy. A comparison was made between transcripts associated with failure to respond to the current GO combination regimen and transcripts recently reported as predictive of poor outcome with respect to overall survival. Elevation in homeobox B6 levels in peripheral blood samples of non-responders in this study was consistent with the overexpression of multiple homeobox genes in patients with poor outcomes related to survival. Homeobox B6 is elevated during normal granulocytopoiesis and monocytopoiesis, but is normally turned off following cell maturation. Homeobox B6 was found to be dysregulated in a substantial percentage of AML samples and has been proposed to play a role in leukemogenesis.


The present analyses also identified several families of transcripts where overexpression appears to be correlated with failure to respond to the GO combination regimen and do not appear to be correlated with overall survival. Several metallothionein isoforms were elevated in peripheral blood samples of patients who failed to respond to the GO combination regimen. Based on the mechanism of action of GO, elevated antioxidant defenses would be expected to adversely impact the efficacy of the chalechiamicin-directed cytotoxic conjugate. These findings however contrast with those reported by Goasguen et al. (1996) Leuk. Lymphoma. 23(5-6):567-76, who identified metallothionein overexpression as strongly associated with complete remission in the context of the absence or presence of other drug-resistance phenotypes in patients with leukemias. Metallothionein isoform overexpression has recently been characterized as a hallmark of the t(15;17) chromosomal translocation in AML but none of the patients in the present study were characterized as possessing this cytogenetic abnormality. However, in that study metallothionein isoform overexpression was not specific to the t(15;17) translocation, occurring in several other karyotypes as well.


The foregoing description of the present invention provides illustration and description, but is not intended to be exhaustive or to limit the invention to the precise one disclosed. Modifications and variations are possible consistent with the above teachings or may be acquired from practice of the invention. Thus, it is noted that the scope of the invention is defined by the claims and their equivalents.

Claims
  • 1. A method for predicting a clinical outcome in response to a treatment of a leukemia, the method comprising the steps of: (1) measuring expression levels of one or more prognostic genes of the leukemia in a peripheral blood mononuclear cell sample derived from a patient prior to the treatment; and(2) comparing each of the expression levels to a corresponding control level,wherein the result of the comparison is predictive of a clinical outcome.
  • 2. The method of claim 1, wherein the one or more prognostic genes comprise at least a first gene selected from a first class and a second gene selected from a second class, wherein the first class comprises genes having higher expression levels in peripheral blood mononuclear cells in patients predicted to have a less desirable clinical outcome in response to the treatment and the second class comprises genes having higher expression levels in peripheral blood mononuclear cells in patients predicted to have a more desirable clinical outcome in response to the treatment.
  • 3. The method of claim 2, wherein the first gene is selected from Table 3 and the second gene is selected from Table 4.
  • 4. The method of claim 2, wherein the first gene is selected from the group consisting of zinc finger protein 217, peptide transporter 3, forkhead box O3A, T cell receptor alpha locus and putative chemokine receptor/GTP-binding protein, and the second gene is selected from the group consisting of metallothionein, fatty acid desaturase 1, uncharacterized gene corresponding to Affymetrix ID 216336, deformed epidermal autoregulatory factor 1 and growth arrest and DNA-damage-inducible alpha.
  • 5. The method of claim 2, wherein the first gene is serum glucocorticoid regulated kinase and the second gene is metallothionein 1X/1L.
  • 6. The method of claim 1, wherein the clinical outcome is development of an adverse event.
  • 7. The method of claim 6, wherein the adverse event is veno-occlusive disease.
  • 8. The method of claim 7, wherein the one or more prognostic genes comprise one or more genes selected from Table 5 or Table 6.
  • 9. The method of claim 8, wherein the one or more prognostic genes comprise p-selectin ligand.
  • 10. The method of any one of the preceding claims, wherein the treatment comprises a gemtuzumab ozogamicin (GO) combination therapy.
  • 11. The method of any one of the preceding claims, wherein the corresponding control level is a numerical threshold.
  • 12. A method for predicting a clinical outcome of a leukemia, the method comprising the steps of: (1) generating a gene expression profile from a peripheral blood sample of a patient having the leukemia; and(2) comparing the gene expression profile to one or more reference expression profiles,wherein the gene expression profile and the one or more reference expression profiles comprise expression patterns of one or more prognostic genes of the leukemia in peripheral blood mononuclear cells, and wherein the difference or similarity between the gene expression profile and the one or more reference expression profiles is indicative of the clinical outcome for the patient.
  • 13. The method of claim 12, wherein the leukemia is acute leukemia, chronic leukemia, lymphocytic leukemia or nonlymphocytic leukemia.
  • 14. The method of claim 13, wherein the leukemia is acute myeloid leukemia (AML).
  • 15. The method of any one of claims 12-14, wherein the clinical outcome is measured by a response to an anti-cancer therapy.
  • 16. The method of claim 15, wherein the anti-cancer therapy comprises administering one or more compounds selected from the group consisting of an anti-CD33 antibody, a daunorubicin, a cytarabine, a gemtuzumab ozogamicin, an anthracycline, and a pyrimidine or purine nucleotide analog.
  • 17. The method of any one of claims 12-16, wherein the one or more prognostic genes comprise one or more genes selected from Table 3 or Table 4.
  • 18. The method of claim 17, wherein the one or more prognostic genes comprise ten or more genes selected from Table 3 or Table 4.
  • 19. The method of claim 18, wherein the one or more prognostic genes comprise twenty or more genes selected from Table 3 or Table 4.
  • 20. The method of any one of claims 12-19, wherein step (2) comprises comparing the gene expression profile to the one or more reference expression profiles by a k-nearest neighbor analysis or a weighted voting algorithm.
  • 21. The method of any one of claims 12-19, wherein the one or more reference expression profiles represent known or determinable clinical outcomes.
  • 22. The method of any one of claims 12-19, wherein step (2) comprises comparing the gene expression profile to at least two reference expression profiles, each of which represents a different clinical outcome.
  • 23. The method of claim 22, wherein each reference expression profile represents a different clinical outcome selected from the group consisting of remission to less than 5% blasts in response to the anti-cancer therapy; remission to no less than 5% blasts in response to the anti-cancer therapy; and non-remission in response to the anti-cancer therapy.
  • 24. The method of any one of claims 12-19, wherein the one or more reference expression profiles comprise a reference expression profile representing a leukemia-free human.
  • 25. The method of any one claims 12-19, wherein step (1) comprises generating the gene expression profile using a nucleic acid array.
  • 26. The method of claim 15, wherein step (1) comprises generating the gene expression profile from the peripheral blood sample of the patient prior to the anti-cancer therapy.
  • 27. A method for selecting a treatment for a leukemia patient, the method comprising the steps of: (1) generating a gene expression profile from a peripheral blood sample derived from the leukemia patient;(2) comparing the gene expression profile to a plurality of reference expression profiles, each representing a clinical outcome in response to one of a plurality of treatments; and(3) selecting from the plurality of treatments a treatment which has a favorable clinical outcome for the leukemia patient based on the comparison in step (2),wherein the gene expression profile and the one or more reference expression profiles comprise expression patterns of one or more prognostic genes of the leukemia in peripheral blood mononuclear cells.
  • 28. The method of claim 27, wherein the one or more prognostic genes comprise one or more genes selected from Table 3 or Table 4.
  • 29. The method of claim 28, wherein the one or more prognostic genes comprise ten or more genes selected from Table 3 or Table 4.
  • 30. The method of claim 29, wherein the one or more prognostic genes comprise twenty or more genes selected from Table 3 or Table 4.
  • 31. The method of any one of claims 27-30, wherein step (2) comprises comparing the gene expression profile to the plurality of reference expression profiles by a k-nearest neighbor analysis or a weighted voting algorithm.
  • 32. A method for diagnosis, or monitoring the occurrence, development, progression or treatment, of a leukemia, the method comprising the steps of: (1) generating a gene expression profile from a peripheral blood sample of a patient having the leukemia; and(2) comparing the gene expression profile to one or more reference expression profiles,wherein the gene expression profile and the one or more reference expression profiles comprise the expression patterns of one or more diagnostic genes of the leukemia in peripheral blood mononuclear cells, and wherein the difference or similarity between the gene expression profile and the one or more reference expression profiles is indicative of the presence, absence, occurrence, development, progression, or effectiveness of treatment of the leukemia in the patient.
  • 33. The method of claim 32, wherein the leukemia is AML.
  • 34. The method of claim 33, wherein the one or more diagnostic genes comprise one or more genes selected from Table 7.
  • 35. The method of claim 33, wherein the one or more diagnostic genes comprise one or more genes selected from Table 8 or Table 9.
  • 36. The method of claim 33, wherein the one or more diagnostic genes comprise ten or more genes selected from Table 7.
  • 37. The method of claim 33, wherein the one or more diagnostic genes comprise ten or more genes selected from Table 8 or Table 9.
  • 38. The method of claim 32, wherein the one or more reference expression profiles comprise a reference expression profile representing a disease-free human.
  • 39. An array for use in a method for predicting a clinical outcome for an AML patient comprising a substrate having a plurality of addresses, each address comprising a distinct probe disposed thereon, wherein at least 15% of the plurality of addresses have disposed thereon probes that can specifically detect prognostic genes of AML in peripheral blood mononuclear cells.
  • 40. The array of claim 39, wherein at least 30% of the plurality of addresses have disposed thereon probes that can specifically detect prognostic genes of AML in peripheral blood mononuclear cells.
  • 41. The array of claim 39, wherein at least 50% of the plurality of addresses have disposed thereon probes that can specifically detect prognostic genes of AML in peripheral blood mononuclear cells.
  • 42. The array of any one of claims 39-41, wherein the prognostic genes are selected from Tables 3, 4, 5 or 6.
  • 43. The array of any one of claims 39-41, wherein the probe is a nucleic acid probe.
  • 44. The array of any one of claims 39-41, wherein the probe is an antibody probe.
  • 45. An array for use in a method for diagnosis of AML comprising a substrate having a plurality of addresses, each address comprising a distinct probe disposed thereon, wherein at least 15% of the plurality of addresses have disposed thereon probes that can specifically detect diagnostic genes of AML in peripheral blood mononuclear cells.
  • 46. The array of claim 45, wherein at least 30% of the plurality of addresses have disposed thereon probes that can specifically detect diagnostic genes of AML in peripheral blood mononuclear cells.
  • 47. The array of claim 45, wherein at least 50% of the plurality of addresses have disposed thereon probes that can specifically detect diagnostic genes of AML in peripheral blood mononuclear cells.
  • 48. The array of any one of claims 45-47, wherein the diagnostic genes are selected from Table 7.
  • 49. The array of any one of claims 45-47, wherein the probe is a nucleic acid probe.
  • 50. The array of any one of claims 45-47, wherein the probe is an antibody probe.
  • 51. A computer-readable medium comprising a digitally-encoded expression profile comprising a plurality of digitally-encoded expression signals, wherein each of the plurality of digitally-encoded expression signals comprises a value representing the expression of a prognostic gene of AML in a peripheral blood mononuclear cell.
  • 52. The computer-readable medium of claim 51, wherein the prognostic gene is selected from Tables 3, 4, 5 or 6.
  • 53. The computer-readable medium of claim 51, wherein the value represents the expression of the prognostic gene of AML in a peripheral blood mononuclear cell of a patient with a known or determinable clinical outcome.
  • 54. The computer-readable medium of claim 51, wherein the digitally-encoded expression profile comprises at least ten digitally-encoded expression signals.
  • 55. A computer-readable medium comprising a digitally-encoded expression profile comprising a plurality of digitally-encoded expression signals, wherein each of the plurality of digitally-encoded expression signals comprises a value representing the expression of a diagnostic gene of AML in a peripheral blood mononuclear cell.
  • 56. The computer-readable medium of claim 55, wherein the diagnostic gene is selected from Table 7.
  • 57. The computer-readable medium of claim 55, wherein the value represents the expression of the diagnostic gene of AML in a peripheral blood mononuclear cell of an AML-free human.
  • 58. The computer-readable medium of claim 55, wherein the digitally-encoded expression profile comprises at least ten digitally-encoded expression signals.
  • 59. A kit for prognosis of AML, the kit comprising: a) one or more probes that can specifically detect prognostic genes of AML in peripheral blood mononuclear cells; and b) one or more controls, each representing a reference expression level of a prognostic gene detectable by the one or more probes.
  • 60. The kit of claim 59, wherein the prognostic genes are selected from Tables 3, 4, 5 or 6.
  • 61. A kit for diagnosis of AML, the kit comprising: a) one or more probes that can specifically detect diagnostic genes of AML in peripheral blood mononuclear cells; and b) one or more controls, each representing a reference expression level of a prognostic gene detectable by the one or more probes.
  • 62. The kit of claim 61, wherein the diagnostic genes are selected from Table 7.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Ser. No. 60/653,117, filed Feb. 16, 2005.

PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US06/05855 2/16/2006 WO 00 4/24/2008
Provisional Applications (1)
Number Date Country
60653117 Feb 2005 US