Methods of identifying patients at risk of developing encephalitis following immunotherapy for Alzheimer's disease

Abstract
The present invention generally relates to a method for an improved treatment for Alzheimer's disease (AD) using immunotherapy, e.g., immunotherapy targeting β amyloid (Aβ), e.g., immunotherapy based on AN1792. In one embodiment, the method allows for predicting an adverse clinical response, and therefore allows for an improved safety profile of AN1792. In another embodiment, the method allows for predicting a favorable clinical response, and therefore allows for an improved efficacy profile of AN1792. The methods of the present invention may be combined to predict a favorable clinical response and the lack of an adverse clinical response.
Description
BACKGROUND OF THE INVENTION

1. Field of the Invention


The present invention generally relates to methods for an improved treatment for Alzheimer's disease. The methods employ pharmacogenomic information to develop an immunotherapy targeted against Aβ peptide, e.g., an immunotherapy based on AN1792, that exhibits a reduction in adverse clinical responses and/or an increased incidence of favorable clinical responses to such immunotherapy resulting in its improved safety and efficacy.


2. Related Background Art


Alzheimer's disease (AD) is a progressive degenerative disease of the brain primarily associated with aging. Clinical presentation of AD is characterized by loss of memory, cognition, reasoning, judgment, and orientation. As the disease progresses, motor, sensory, and linguistic abilities are also affected until there is global impairment of multiple cognitive functions. These cognitive losses may occur gradually, but typically lead to severe impairment and eventual death in the range of four to twelve years.


Alzheimer's disease is characterized by major pathologic observations in the brain: neurofibrillary tangles, the accumulation of β-amyloid (or neuritic) plaques (comprised predominantly of an aggregate of a peptide fragment known as Aβ), and by increased rates of neuronal atrophy. Individuals with AD exhibit characteristic β-amyloid deposits in the brain (β-amyloid plaques), cerebral blood vessels (β-amyloid angiopathy), and neurofibrillary tangles. Neurofibrillary tangles occur not only in AD but also in other dementia-inducing disorders. On autopsy, large numbers of these lesions are generally found in areas of the human brain important for memory and cognition.


Smaller numbers of these lesions in a more restricted anatomical distribution are found in the brains of most elderly humans who do not have clinical AD. Amyloidogenic plaques and vascular amyloid angiopathy also characterize the brains of individuals with trisomy 21 (Down syndrome), hereditary cerebral hemorrhage with amyloidosis of the Dutch-type (HCHWA-D), and other neurodegenerative disorders. β-amyloid is a defining feature of AD, and is now believed to be a causative precursor or factor in the development of disease. Deposition of Aβ in areas of the brain responsible for cognitive activities is a major factor in the development of AD. β-amyloid plaques predominantly are composed of amyloid β peptide (Aβ, also sometimes designated β-A/4). Aβ peptide is derived by proteolysis of the amyloid precursor protein (APP). Several proteases called secretases are involved in the processing of APP.


Cleavage of APP at the N-terminus of the Aβ peptide by β-secretase and at the C-terminus by one or more γ-secretases constitutes the α-amyloidogenic pathway, i.e., the pathway by which Aβ is formed. Cleavage of APP by α-secretase produces α-sAPP, a secreted form of APP that does not result in β-amyloid plaque formation. This alternate pathway precludes the formation of Aβ peptide. A description of the proteolytic processing fragments of APP is found, for example, in U.S. Pat. Nos. 5,441,870; 5,721,130; and 5,942,400.


Several lines of evidence indicate that progressive cerebral deposition of β-amyloid peptide (Aβ) plays an influential role in the pathogenesis of AD and can precede cognitive symptoms by years or decades (see, e.g., Selkoe (1991) Neuron 6 (4):487-98). Release of Aβ from neuronal cells grown in culture and the presence of Aβ in cerebrospinal fluid of both normal individuals and AD patients has been demonstrated (see, e.g., Seubert et al. (1992) Nature 359:325-27).


At present there is no effective treatment for preventing, slowing, arresting, and/or reversing the progression of AD. Therefore, there is an urgent need for pharmaceutical agents capable of preventing, slowing, arresting and/or, reversing the progression of AD.


One problem with finding a treatment for AD is that, in general, there is great heterogeneity in the way that humans respond to medications. Currently, empirical methods are typically used to find the appropriate drug therapy for an individual patient. However, such empirical strategies run the risk that a patient will receive a drug that is ineffective, thus delaying effective therapy, or that a patient may develop an adverse clinical response or side effect to the drug. When the subset of patients at risk of the development of an adverse clinical response cannot be identified prior to the administration of a given drug, the development of that drug may be terminated; thus, the possibility of benefiting from therapy involving that drug may be denied to those patients who are not susceptible to an adverse clinical response to that drug.


One such adverse drug reaction was seen with AN1792, a peptide immunogen consisting of Aβ1-42, the section of amyloid recognized as a major component of AD-related plaques (Iwatsubo et al. (1994) Neuron 13:45-53). Administration of AN1792 is an experimental therapeutic strategy against AD based on the theory that administration of β-amyloid might activate the immune system to raise its own protective anti-amyloid antibodies that “recognize” and attack the β-amyloid plaques that are a hallmark of AD brain abnormality (Schenk et al (2000) Arch. Neurol. 57:934-36).


In 1999, the first preclinical animal studies with AN1792 were reported (see Schenk et al. (1999) Nature 400:173-77). Studies in transgenic mouse models of cognitive impairment and amyloid plaque-associated CNS pathology demonstrated that immunization with AN1792 resulted in improved cognitive function and inhibited the development of AD-like amyloid plaques, neuritic dystrophy, and gliosis in mice (Games et al. (1995) Nature 373:523-27; Schenk et al. (1999) Nature 400:173-77; Morgan et al. (2000) Nature 408:982-85; Janus et al. (2000) Nature 408:979-82; DeMattos et al. (2001) Proc. Natl. Acad. Sci. USA 98:8850-55; McLaurin et al. (2002) Nat. Med. 8:1263-69). The mice treated with AN1792, and not those treated with placebo, had improved performance in memory tests. Based on these preclinical results, both the U.S. Food and Drug Administration and the U.K. Medicines Control Agency permitted Phase I human trials of AN1792 to assess its safety and tolerability in people with mild to moderate AD.


The U.K. trial enrolled about 80 patients and the U.S. trial enrolled about 24 patients with mild to moderate AD for the Phase I trials. Results from the Phase I trials were announced in 2000 and indicated that AN1792 was well tolerated in human recipients and that a portion of the participants developed amyloid antibodies, as was seen in the preclinical animal studies (Klocinski and Karlawish (2002) University of Pennsylvania Memory Disorders Clinic News Letter, 1 (4):5-8; Bayer et al (2005) Neurology 64:94-101). Based on these outcomes, in late 2001, a small Phase Ia double-blind, placebo-controlled, multi-centered trial began in the United States and Europe enrolling 372 patients with mild to moderate AD to evaluate safety, tolerability and pilot-efficacy of AN1792 administered with QS-21 adjuvant (Fox et al. (2005) Neurology 64:1563-72; Gilman et al. (2005) Neurology 64:1553-62; Orgogozo et al. (2003) Neurology 61:46-54). For the Phase Ia trials, 300 patients were randomly selected to receive six immunizations of AN1792 and 72 patients were randomly selected to receive placebo (Gilman, supra). Four of the participants developed signs of meningoencephalitis at an early phase of the clinical trial, and the trial was suspended. Soon after the suspension, 14 more patients developed signs of meningoencephalitis; the Safety Monitoring Committee concluded that dosing with the immunotherapeutic AN1792 should be discontinued. At the time the treatments were discontinued, the maximum number of immunization received was three (by 24 patients), with the majority of patients receiving two immunizations (274 patients) and two patients receiving one immunization. Ultimately, meningoencephalitis was reported in 18 of 300 immunized patients (Orgogozo (2003) supra). All 18 patients had received AN1792, whereas no patient in the placebo group developed encephalitis (Orgogozo (2003) supra).


Trial researchers continued to follow all participants, i.e., cognitive function, memory and executive function, and anti-AN1792 antibody, CSF tau, and CSF Aβ1-42 levels were assessed to the conclusion of the original follow-up period. Antibody responders were compared to placebo controls. Two sets of measurements, levels of CSF tau and a battery of neuropsychological tests, gave results favoring patients with a positive IgG titer (Gilman, supra). However, the exact cause of the brain inflammation, i.e., meningoencephalitis, in some subjects is not yet known. The follow-up studies showed that the participants who suffered from meningoencephalitis developed antibodies to β-amyloid but that there did not appear to be any correlation between antibody levels and the risk of developing brain inflammation. An autopsy of one participant who died of causes unrelated to treatment showed signs of brain inflammation. Interestingly, significant areas of the brain lacked the β-amyloid plaques targeted by the immunotherapeutic, a phenomenon not seen in the brains of patients diagnosed with AD. Whole-trial analysis remains ongoing (Gilman, supra).


In order for AN1792 to be considered a possible therapy for AD, it is desirable to understand how the immune system responds to AN1792 such that the complications associated with the therapy, e.g., inflammation leading to, e.g., meningoencephalitis, may be reduced. Pharmacogenomics may allow the identification of predictive biomarkers of responsiveness to the immunotherapeutic, e.g., for the identification of patients, prior to therapy, who are most likely to develop a favorable clinical response, e.g., a protective immune response, (e.g., an antibody response) and/or least likely to develop an adverse clinical response, e.g., inflammation that may result in, e.g., encephalitis (e.g., meningoencephalitis).


Pharmacogenomics seeks to investigate and identify genomic factors that contribute to drug response variation(s) among individuals with seemingly equivalent disease symptoms. Recent advances in the sequencing of the human genome have enabled researchers to more efficiently and effectively link certain genomic variations to particular diseases. Pharmacogenomics has the potential to revolutionize treatment strategies and to aid in the development of clinical in vitro diagnostics, which would be far superior to empirical treatment. Increasing knowledge about the interactions between genes and drug treatment should create a proportionate demand for rapid and reliable pretreatment diagnostic tests to ensure the safest and most effective treatment possible.


By utilizing the tools of pharmacogenomics, the present invention overcomes the inadequacies of AN1792 immunotherapy by providing an effective method for optimizing both the efficacy and safety of AN1792. The present invention draws correlations between gene expression patterns and clinical responses to a treatment for AD (particularly administration of AN1792), provides methods for predicting clinical and pathological responses, and provides methods for using this information to improve the clinical response profile of AN1792 and to develop a therapeutic product for patients preselected for optimal safety and efficacy (e.g., a “genomically guided” therapeutic product).


SUMMARY OF THE INVENTION

The present invention is directed to a method of using pharmacogenomic information to predict a clinical response in an AD patient to a treatment for AD. In one embodiment of the invention, the treatment is an immunotherapeutic, e.g., an active immunotherapeutic. In particular, the present invention is directed to active immunotherapy targeting Aβ peptide, e.g., an immunotherapy based on AN1792.


Accordingly, the invention provides methods of compiling pharmacogenomic information to associate a unique gene expression pattern of a patient sample with a particular clinical response to a treatment for AD. Generally, the methods for compiling pharmacogenomic information to associate a unique gene expression pattern of a patient sample with a particular clinical response to a treatment for AD comprise the steps of procuring at least one patient sample from a patient of a first population of patients and at least one patient sample from a patient of a second population of patients (wherein the first population consists of one or more patients who developed the particular clinical response to the treatment for AD and wherein the second population consists of one or more patients who did not develop the particular response to the treatment for AD); acquiring a gene expression pattern from each procured patient sample; and determining whether at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population; wherein a determination that at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population results in associating the unique gene expression pattern with the particular clinical response to the treatment for AD. In one embodiment of the invention, the particular clinical response is one that is neither favorable nor adverse (e.g., antibody nonresponsiveness). In some embodiments, the particular clinical response is either a favorable clinical response or an adverse clinical response. In other embodiments, the particular clinical response is both a favorable and adverse clinical response. For example, the particular clinical response may be inflammation, and said inflammation may encompass development of both an IgG response and encephalitis.


The invention thus provides a method for compiling pharmacogenomic information to associate a unique gene expression pattern of a patient sample with a favorable clinical response to a treatment for AD comprising the steps of procuring at least one patient sample from a patient of a first population of patients and at least one patient sample from a patient of a second population of patients (wherein the first population consists of one or more patients who developed the favorable clinical response to the treatment for AD and wherein the second population consists of one or more patients who did not develop the favorable response to the treatment for AD); acquiring a gene expression pattern from each procured patient sample; and determining whether at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population; wherein a determination that at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population results in associating the unique gene expression pattern with the favorable clinical response to the treatment for AD. In one embodiment of the invention, the second population consists of one or more patients who did not develop the favorable clinical response to the treatment and also developed an adverse clinical response. In another embodiment of the invention, the method further comprises excluding patients who also developed an adverse clinical response to the treatment for AD from the first population of patients.


The present invention also provides a method of compiling pharmacogenomic information to associate a unique gene expression pattern of a patient sample with an adverse clinical response to a treatment for AD comprising the steps of procuring at least one patient sample from a patient of a first population of patients and at least one patient sample from a patient of a second population of patients (wherein the first population consists of one or more patients who developed the adverse clinical response to the treatment for AD and wherein the second population consists of one or more patients who did not develop the adverse response to the treatment for AD); acquiring a gene expression pattern from each procured patient sample; and determining whether at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population, wherein a determination that at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population results in associating the unique gene expression pattern with the adverse clinical response to the treatment for AD. In one embodiment of the invention, the second population consists of one or more patients who did not develop the adverse clinical response to the treatment and also developed a favorable clinical response. In another embodiment of the invention, the method further comprises excluding patients who also developed a favorable response to the treatment for AD from the first population of patients. In some embodiments, selected genes or groups of genes are excluded before acquiring a gene expression pattern to improve the accuracy of statistical findings, e.g., genes identified as significant covariates.


In some methods of compiling pharmacogenomic information, samples are placed under a certain culture condition(s) prior to acquisition of gene expression patterns. In some embodiments, the clinical response that is neither favorable nor adverse is low to undetectable antibody production. In some embodiments, the favorable clinical response is a protective immune response. In some embodiments, the favorable clinical response is an antibody response, e.g., an IgG response. In some embodiments, the adverse clinical response is an inflammatory response. In some embodiments, the inflammatory response leads to encephalitis, e.g., meningoencephalitis. In some embodiments of the methods of compiling pharmacogenomic information, the patient samples are peripheral blood mononuclear cells. In some embodiments, the gene expression pattern is selected from the group consisting of protein expression patterns and RNA expression patterns.


In some embodiments of the invention, the methods of compiling pharmacogenomic information are used to associate a unique gene expression pattern of a patient sample with a particular clinical response to administration of AN1792. Accordingly, the invention also provides methods of compiling pharmacogenomic information to associate a unique gene expression pattern of a patient sample taken from a patient treated with AN1792 with a clinical response to the administration of AN1792. In one embodiment of the invention, gene expression patterns are acquired from unstimulated samples. In another embodiment of the invention, gene expression patterns are acquired from stimulated (e.g., cultured) samples.


The invention also provides a method for determining a unique gene expression pattern for predicting whether a candidate AD patient is likely to develop an IgG response to administration of AN1792 comprising referring to nucleic acid samples from a patient population previously exposed to AN1792, wherein the patient population includes IgG responders and IgG nonresponders, and wherein IgG expression is developed in response to administration of AN1792; and comparing the nucleic acid samples of the IgG responders with the nucleic acid samples of the IgG nonresponders to determine the unique gene expression pattern associated with IgG responders. Also provided is a method for determining a unique gene expression pattern for predicting whether a candidate AD patient is likely to not develop an IgG response to administration of AN1792 comprising referring to nucleic acid samples from a patient population previously exposed to AN1792, wherein the patient population includes IgG responders and IgG nonresponders, and wherein IgG expression is developed in response to administration of AN1792; and comparing the nucleic acid samples of the IgG nonresponders with the nucleic acid samples of the IgG responders to determine the unique gene expression pattern associated with the IgG nonresponders. The invention also provides a method for determining a unique gene expression pattern for predicting whether a candidate AD patient is likely to develop inflammation in response to administration of AN1792 comprising referring to nucleic acid samples from a patient population previously exposed to AN1792, wherein the patient population includes inflammation developers and inflammation nondevelopers, and wherein inflammation is developed in response to administration of AN1792; and comparing the nucleic acid samples of the inflammation developers with the nucleic acid samples of the inflammation nondevelopers to determine the unique gene expression pattern associated with inflammation developers.


The invention also provides a method for determining a unique gene expression pattern for predicting whether a candidate AD patient is likely to develop an IgG response to administration of AN1792 comprising acquiring gene expression patterns from a patient population previously exposed to AN1792, wherein the patient population includes IgG responders and IgG nonresponders, and wherein IgG expression is developed in response to administration of AN1792; and comparing the gene expression patterns of the IgG responders to the gene expression patterns of the IgG nonresponders to determine the unique gene expression pattern associated with the IgG responders. The invention also provides a method for determining a unique gene expression pattern for predicting whether a candidate AD patient is likely to not develop an IgG response to administration of AN1792 comprising acquiring gene expression patterns from a patient population previously exposed to AN1792, wherein the patient population includes IgG nonresponders and IgG responders, and wherein IgG expression is developed in response to administration of AN1792; and comparing the gene expression patterns of the IgG nonresponders to the gene expression patterns of the IgG responders to determine the unique gene expression pattern associated with the IgG nonresponders. Additionally, the invention provides a method for determining a unique gene expression pattern for predicting whether a candidate AD patient is likely to develop inflammation in response to administration of AN1792 comprising acquiring gene expression patterns from a patient population previously exposed to AN1792, wherein the patient population includes inflammation developers and inflammation nondevelopers, and wherein inflammation is developed in response to administration of AN1792; and comparing the gene expression patterns of the inflammation developers to the gene expression patterns of the inflammation nondevelopers to determine the unique gene expression pattern associated with the inflammation developers.


The invention also provides a method for determining a unique gene expression pattern for predicting whether a candidate AD patient is likely to develop an IgG response to administration of AN1792 comprising procuring blood samples from a patient population previously exposed to AN1792, wherein the patient population includes IgG responders and IgG nonresponders, and wherein IgG expression is developed in response to administration of AN1792; purifying total RNA from the blood samples, thereby producing RNA samples; assaying RNA expression levels from the RNA samples to obtain gene expression patterns for the IgG responders and the IgG nonresponders; and comparing the gene expression patterns of the IgG responders to the gene expression patterns of the IgG nonresponders to determine the unique gene expression pattern associated with the IgG responders. Also provided is a method for determining a unique gene expression pattern for predicting whether a candidate AD patient is likely to not develop an IgG response to administration of AN1792 comprising procuring blood samples from a patient population previously exposed to AN1792, wherein the patient population includes IgG nonresponders and IgG responders, and wherein IgG expression is developed in response to administration of AN1792; purifying total RNA from the blood samples, thereby producing RNA samples; assaying RNA expression levels from the RNA samples to obtain gene expression patterns for the IgG nonresponders and the IgG responders; and comparing the gene expression patterns of the IgG nonresponders to the gene expression patterns of the IgG responders to determine the unique gene expression pattern associated with the IgG nonresponders. The invention also provides a method for determining a unique gene expression pattern for predicting whether a candidate AD patient is likely to develop inflammation in response to administration of AN1792 comprising procuring blood samples from a patient population previously exposed to AN1792, wherein the patient population includes inflammation developers and inflammation nondevelopers, and wherein inflammation is developed in response to administration of AN1792; purifying total RNA from the blood samples, thereby producing RNA samples; assaying RNA expression levels from the RNA samples to obtain gene expression patterns for the inflammation developers and the inflammation nondevelopers; and comparing the gene expression patterns of the inflammation developers to the gene expression patterns of the inflammation nondevelopers to determine the unique gene expression pattern associated with the inflammation developers.


In some embodiments of methods of determining a unique gene expression pattern, the gene expression pattern is selected from the group consisting of protein gene expression patterns and RNA gene expression patterns. In other embodiments of methods of determining a unique gene expression pattern, the methods further comprise assaying total RNA expression levels from an RNA sample obtained from the patient population to acquire the gene expression pattern. Other embodiments of methods of determining a unique gene expression pattern further comprise assaying total protein expression levels from a protein sample obtained from the patient population to acquire the gene expression pattern.


The invention also provides unique gene expression patterns that are associated with a particular response to a treatment for AD. In some embodiments, a gene expression pattern of the invention is a protein gene expression pattern. In other embodiments, a gene expression pattern of the invention is an RNA gene expression pattern. In some embodiments, the unique gene expression pattern comprises the expression level of one gene that may be considered individually. In other embodiments, the invention provides a unique gene expression pattern that comprises expression levels of a panel of genes, wherein the expression levels are or will be measured, e.g., by the measurement of gene products (e.g., RNA, proteins, etc.). In one embodiment, a panel of the invention may comprise 2-5, 5-15, 15-35, 35-50, 50-100, or more than 100 genes. In one embodiment, a panel may comprise 15-20 genes. In another embodiment, a panel may comprise two genes.


The invention also provides kits, e.g., a kit comprising one or more polynucleotides, each capable of hybridizing under stringent conditions to an RNA transcript, or the complement thereof, of a gene differentially expressed in a unique gene expression pattern of the invention; and/or one or more antibodies, each capable of binding to a polypeptide encoded by a gene differentially expressed in a unique gene expression pattern of the invention. In some embodiments, a gene differentially expressed in a unique gene expression pattern of the invention is a gene differentially expressed in PBMCs of AD patients likely to develop a particular clinical response when treated with AN1792 as compared to PBMCs of AD patients likely not to develop the particular clinical response when treated with AN1792. For example, in some embodiments, the particular clinical response may be an antibody response (e.g., an IgG response). In other embodiments, the particular clinical response is inflammation, e.g., encephalitis (e.g., meningoencephalitis). In some embodiments, the polynucleotides and/or antibodies of a kit of the invention are coupled to a solid support.


In one embodiment, a panel or kit of the invention comprises genes selected from one of Tables 10-12, 18, and 24-37. In another embodiment, a panel or kit of the invention comprises a combination of genes selected from those listed in Tables 10-12, 18, and 24-37. In a further embodiment, a panel or kit of the invention comprises genes listed in Table 36. In another embodiment, a panel or kit of the invention comprises a pair of genes, e.g., any of the pairs of genes listed in Table 37.


It is an object of the invention to use unique gene expression patterns associated with particular clinical responses to predict the clinical response of a candidate patient to a treatment for AD. Thus the invention also provides methods of predicting whether a candidate patient who has not been previously exposed to a treatment for AD will develop a particular clinical response to a treatment for AD, the methods generally comprising the steps of associating at least one unique gene expression pattern of a patient sample with a particular clinical response to the treatment for AD; procuring a test sample from the candidate patient who has not been previously exposed to the treatment for AD; and determining that the test sample procured from the candidate patient who has not been previously exposed to the treatment for AD has a test gene expression pattern that is substantially similar to the at least one gene expression pattern associated with the particular clinical response to the treatment for AD (i.e., the at least one reference gene expression pattern), wherein if it is determined that the test sample has a test gene expression pattern that is substantially similar to the at least one gene expression pattern associated with the particular clinical response, it may be predicted that the candidate patient will develop the particular clinical response. In one embodiment, the particular clinical response is neither favorable nor adverse. In another embodiment, the particular clinical response is either a favorable or adverse clinical response. In an additional embodiment, the particular clinical response is both a favorable and adverse clinical response.


In a specific embodiment, the methods of the present invention include obtaining and/or determining a first population of patients that develops a particular clinical response (wherein the particular clinical response is, e.g., the development of an inflammatory response, particularly encephalitis, and/or the development of an IgG response, but may be any other particular clinical response, such as decrease in plaque formation, to a treatment for AD (e.g., an immunotherapeutic-based treatment for AD, e.g., AN1792)), and a second population of patients that does not develop the particular clinical response. The method of the present invention further comprises examining the gene expression patterns of the first population to discover whether there are any specific gene expression patterns associated with the particular clinical response. Phenotypic characteristics may further define genomic populations and result in further improved response profiles of treatments for AD, e.g., immunotherapeutics, including but not limited to AN1792; for example, in some treatments, females may have a greater degree of adverse clinical responses than males. The method then comprises associating a unique gene expression pattern with the particular clinical response(s), wherein the unique gene expression pattern defines a population having, e.g., an improved therapeutic response profile to a treatment. The gene expression pattern predicts patients, for example, who may develop inflammation and/or who may have or develop a certain level of IgG response.


In a further aspect of the invention, there is provided a system comprising a computer readable memory which stores at least one reference gene expression pattern of one or more genes wherein each of the one or more genes is differentially expressed in patient samples procured from AD patients who are likely to develop a particular clinical response to a therapy for AD, e.g., AN1792 treatment, compared to patient samples procured from AD patients who are not likely to develop the particular clinical response to the therapy for AD; a program capable of comparing a test gene expression pattern to the reference gene expression pattern and a processor capable of executing the program are also provided in the system.


When such computer readable memory and program exist, i.e., where there already exists reference gene expression patterns (e.g., wherein the reference gene expression pattern is associated with a particular response to the treatment for AD by any method of compiling pharmacogenomic information), the methods of predicting a clinical response of a candidate patient comprise the steps of procuring a test sample from the candidate patient not previously exposed to the treatment for AD, and determining whether the test sample from the candidate patient has a test gene expression pattern that is substantially similar to a reference gene expression pattern that has been previously associated with a particular clinical response, wherein if it is determined that the test sample has a test gene expression pattern that is substantially similar to a reference gene expression pattern that has been previously associated with the particular clinical response, it may be predicted that the candidate patient will develop the particular clinical response. In some embodiments, the particular clinical response is neither a favorable nor an adverse clinical response. In other embodiments, the particular clinical response is a favorable or an adverse clinical response.


In particular, the invention provides methods for predicting whether an AD patient is likely to benefit from treatment for AD comprising the steps of collecting a blood sample from the patient; isolating blood cells from the sample; purifying total RNA from the cells, thereby producing an RNA sample; assaying RNA expression levels from the RNA sample to obtain a gene expression pattern; and comparing the gene expression pattern of the patient with the gene expression pattern of patients who benefited from the treatment, whereby a substantial similarity between the gene expression patterns indicates the patient is likely to benefit from the treatment for AD. For example, the invention provides a method for predicting whether an AD patient is likely to develop an immune response to an immunotherapy treatment for AD comprising collecting a blood sample from the patient; isolating blood cells from the sample; purifying total RNA from the cells, thereby producing an RNA sample; assaying RNA expression levels from the RNA sample to obtain a gene expression pattern; and comparing the gene expression pattern of the patient with the gene expression pattern of patients who developed an immune response to the immunotherapy, whereby a substantial similarity between the gene expression patterns indicates the patient is likely to develop an immune response to the immunotherapy treatment for AD. In some embodiments of the invention, the particular immune response is neither a favorable nor an adverse clinical response, e.g., the clinical response may be undetectable to low IgG production. In other embodiments, the clinical response is both favorable and adverse. In another embodiment, the clinical response is an immune response, e.g., an IgG response. In other embodiments, the clinical response is the development of inflammation, e.g., meningoencephalitis.


Additionally, the invention provides a method for predicting whether an AD patient is likely to develop an adverse reaction in response to a treatment for AD comprising collecting a blood sample from the patient; isolating blood cells from the sample; purifying total RNA from the cells, thereby producing an RNA sample; assaying RNA expression levels from the RNA sample to obtain a gene expression pattern; and comparing the gene expression pattern of the patient with the gene expression pattern of patients who developed an adverse reaction in response to the treatment, whereby a substantial similarity between the gene expression patterns indicates the patient is likely to develop an adverse reaction in response to the treatment for AD. For example, the invention provides a method for predicting whether an AD patient is likely to develop an adverse reaction in response to an immunotherapy treatment for AD comprising collecting a blood sample from the patient; isolating blood cells from the sample; purifying total RNA from the cells, thereby producing an RNA sample; assaying RNA expression levels from the RNA sample to obtain a gene expression pattern; and comparing the gene expression pattern of the patient with the gene expression pattern of patients who developed an adverse reaction in response to the immunotherapy, whereby a substantial similarity between the gene expression patterns indicates the patient is likely to develop an adverse reaction in response to the immunotherapy treatment for AD.


In some embodiments, a candidate patient's clinical response to AN1792 is predicted. Therefore the present invention relates to a method of predicting whether a candidate patient will develop a particular clinical response when administered AN1792 comprising the steps of compiling pharmacogenomic information to associate at least one unique gene expression pattern of a preimmunization patient sample procured from a patient who has been treated with AN1792 with a particular clinical response, procuring a test sample from the candidate patient, and determining whether the test sample has a test gene expression pattern that is substantially similar to the at least one unique gene expression pattern, wherein if the test sample has a test gene expression pattern that is substantially similar to the at least one gene expression pattern associated with the particular clinical response, it may be predicted that the candidate patient will develop the particular clinical response. In some embodiments, the step of determining is performed with unstimulated patient samples. In other embodiments, the step of determining is performed with in vitro cultured patient samples. In one embodiment, the particular clinical response is neither favorable nor adverse, e.g., nonresponsiveness. In another embodiment, the particular clinical response to AN1792 is a favorable clinical response, e.g., a protective immune response, e.g., an IgG antibody response. In another embodiment, the particular clinical response to AN1792 is an adverse clinical response, e.g., an inflammatory response, e.g., encephalitis. Thus, the invention also provides methods of identifying an AD patient who is likely not to develop an IgG response when treated with AN1792, comprising the steps of providing at least one test patient sample of a candidate AD patient; and comparing a test gene expression pattern of one or more genes to at least one reference gene expression pattern, wherein each of the one or more genes of the reference gene expression pattern is differentially expressed in patient samples procured from AD patients who are likely not to develop an IgG response when treated with AN1792 as compared to patient samples procured from AD patients who are likely to develop an IgG response when treated with AN1792. The invention also provides a method of identifying an AD patient who is likely to develop inflammation when treated with AN1792, comprising the steps of providing at least one test patient sample of a candidate AD patient; and comparing a test gene expression pattern of one or more genes in the at least one test patient sample to at least one reference gene expression pattern from an AD patient treated with AN1792, wherein each of the one or more genes is differentially expressed in patient samples procured from patients who are likely to develop inflammation when treated with AN1792 as compared to in patient samples procured from patients who are not likely to develop inflammation when treated with AN1792. In the methods of identifying an AD patient unlikely or likely to develop a particular clinical response when treated with AN1792, the patient sample may comprise enriched PBMCs. In some embodiments, the patient sample is a whole blood sample. In some embodiments, the gene expression pattern is determined using quantitative RT-PCR or an immunoassay.


In some embodiments, the clinical response of a candidate patient to treatment with AN1792 may be predicted, and/or AD patients may be identified using gene expression patterns, kits, and systems of the invention. In some embodiments, a gene expression pattern described in Table 10-12, 18, or 24-37 is used.


Also provided by the invention is a method for increasing the chances that an AD patient develops a favorable clinical response to a therapeutic administration of a treatment for AD, such as AN1792, by determining, prior to treatment, whether the patient has a unique gene expression pattern associated with the development of a favorable clinical response to the treatment.


Accordingly, the present invention provides a method for predicting whether a candidate AD patient is likely to develop a favorable clinical response, particularly a favorable immune response (e.g., an antibody response), to administration of a treatment for AD, particularly AN1792, comprising determining whether the candidate AD patient has a unique gene expression pattern associated with development of a favorable immune response, particularly the development of IgG antibodies, to the treatment. In some embodiments, the method further comprises referring to an AD patient population previously exposed to AN1792, wherein the patient population includes IgG responders and IgG nonresponders, and the unique gene expression is associated with a favorable immune response (e.g., IgG responders). In some embodiments, the presence of the unique gene expression pattern associated with a favorable immune response in the candidate AD patient predicts that the patient is likely to develop an IgG response to the administration of AN1792.


In some embodiments of the invention, the gene expression pattern of IgG responders is acquired from unstimulated patient samples and includes a moderate to high level of expression of at least one of the genes listed in Table 24 as having higher average expression in IgG responders (i.e., the odds ratio is greater than 1), and/or a low level of at least one of the genes listed in Table 24 as having lower average expression in IgG responders (i.e., the odds ration is less than 1). In other embodiments, the gene expression pattern of IgG responders is acquired from in vitro stimulated patient samples and includes a moderate to high level of expression of at least one of the genes listed in Table 18 as having higher average expression in IgG responders, and/or a low level of at least one of the genes listed in Table 18 as having lower average expression in IgG responders


Also provided by the invention is a method for reducing the risk that an AD patient develops meningoencephalitis, or another form of inflammation, or another adverse clinical response to the therapeutic administration of a treatment for AD, including but not limited to AN1792, by determining, prior to treatment, whether the patient has a unique gene expression pattern associated with the development of an adverse clinical response, e.g., an inflammatory response, including but not limited to the development of encephalitis (e.g., meningoencephalitis), to the treatment.


Accordingly, the present invention provides a method for predicting whether a candidate AD patient is likely to develop an adverse clinical response, e.g., an inflammatory response, particularly encephalitis, to administration of a treatment for AD, particularly AN1792, comprising determining whether the candidate AD patient has a unique gene expression pattern associated with development of an adverse clinical response, e.g., an inflammatory response, particularly encephalitis, to the treatment. In one embodiment, the method further comprises referring to an AD patient population previously exposed to AN1792, wherein the patient population includes inflammation developers and inflammation nondevelopers, and the unique gene expression pattern is associated with inflammation developers. In some embodiments, the presence of the unique gene expression pattern associated with inflammation developers in the candidate AD patient predicts that the patient is likely to develop inflammation in response to administration of AN1792.


In another embodiment, the gene expression pattern associated with an adverse clinical response is procured from an unstimulated sample and includes a moderate to high level of expression at least one of the genes listed in Tables 32-36 as having a higher average expression in encephalitis developers and/or a low level of expression of at least one of the genes listed in Tables 32-36 as having lower average expression in encephalitis developers (i.e., higher-odds ratio>1; lower-odds ratio<1). In other embodiments, the gene expression pattern associated with an adverse clinical response is procured from an in vitro stimulated sample and includes a moderate to high level of expression at least one of the genes listed in Tables 10 and 11 as having a higher or increased expression in meningoencephalitis (inflammation) developers and/or a low level of expression of at least one of the genes listed in Tables 10 and 12 as having lower expression in meningoencephalitis (inflammation) developers.


Another aspect of the invention relates to a method comprising the steps of providing at least one peripheral blood sample of an AD patient; and comparing a unique gene expression pattern of one or more genes in the at least one peripheral blood sample to at least one reference gene expression pattern of the one or more genes from an AD patient(s) treated with AN1792. Each of the genes is differentially expressed in peripheral blood mononuclear cells (PBMCs) of AD patients who, e.g., developed encephalitis, or did not develop an IgG response, or both, in response to AN1792 treatment as compared to AD patients who, e.g., did not develop encephalitis, or did develop an IgG response, or both, respectively, in response to AN1792 treatment. The method may be used to predict whether an AD patient is likely to develop an IgG response to AN1792, is likely not to develop an IgG response to AN1792, or is likely or not likely to develop inflammation in response to AN1792. In some embodiments, the step of providing at least one peripheral blood sample of an AD patient comprises the steps of collecting a blood sample form the patient; isolating blood cells from the sample; culturing the cells in the absence of AN1792; purifying total RNA fro the cells, thereby producing an RNA sample; and assaying RNA expression levels from the RNA sample to obtain a gene expression pattern. In other embodiments, assaying RNA expression levels from the RNA sample to obtain a gene expression pattern, wherein the expression levels comprise expression levels of one or more genes listed in, e.g., Tables 10-12 with a predictive strength ≧0.95, predicts that the AD patient is likely to develop inflammation. In another embodiment, assaying RNA expression levels from the RNA sample to obtain a gene expression pattern, wherein the expression levels comprise expression levels of one or more genes listed in, e.g., Table 18 with a predictive strength ≧0.95, predicts that the AD patient is likely not to develop an IgG response.


The invention is also directed to a method for using pharmacogenomics and/or other assays that measure gene expression levels to develop an improved, genomically guided AN1792 therapeutic product or therapy for treating AD having improved efficacy and/or safety profiles. The methods of the present invention are based on the utilization of gene expression patterns in a patient(s) with mild to moderate AD and the therapeutic response profiles to AN1792 in the patient(s).


Thus, the present invention provides methods for improving a response profile of a treatment for AD by increasing the chances that an AD patient develops a favorable and/or nonadverse clinical response to the treatment for AD, comprising the steps of determining that the AD patient, e.g., has a unique gene expression pattern associated with a favorable clinical response to the treatment for AD and/or does not have a unique gene expression pattern associated with an adverse clinical response, and administering the treatment for AD to the AD patient. The present invention also provides methods for improving a response profile of a treatment for AD by decreasing the chances that an AD patient develops an adverse clinical response to the treatment for AD, comprising determining that the AD patient has a unique gene expression pattern associated with an adverse clinical response to the treatment for AD, and not administering the treatment for AD to the AD patient.


The present invention also seeks to improve a response profile of a treatment for AD by regulating the expression levels of one or more genes of a patient sample procured from a candidate patient to be substantially similar to the expression levels of the same one or more genes that are involved in a unique gene expression pattern associated with a favorable clinical response (or associated with the lack of an adverse clinical response). In one embodiment of the invention, regulation of such expression levels is effected by the use of agents, e.g., inhibitory polynucleotides. Administration of such a therapeutic regulatory agent may regulate the aberrant expression of at least one gene that is part of a unique gene expression pattern, and therefore may be used to increase the chances for a favorable clinical response and/or decrease the risk of an adverse clinical response to a treatment for AD. Accordingly, the present invention also provides methods of improving the efficacy of a clinical trial of a treatment for AD, the methods generally comprising the steps of collecting blood samples from candidate patients; isolating blood cells from the samples; purifying total RNA from the cells, thereby producing an RNA sample; assaying RNA expression levels from the RNA samples to obtain gene expression patterns; and comparing the gene expression patterns of the candidate patients with the gene expression patterns of individuals who developed a particular clinical response to the treatment. In some embodiments, candidate patients with a substantially similar gene expression pattern to the gene expression pattern of individuals who developed a favorable clinical response to the treatment are included in the clinical trial of the treatment for AD. In other embodiments, candidate patients with a substantially similar gene expression pattern to the gene expression pattern of individuals who did not respond to the treatment are not included in the clinical trial of the treatment for AD. In another embodiment, candidate patients with a substantially similar gene expression pattern to the gene expression pattern of individuals who developed an adverse clinical response to the treatment are not included in the clinical trial of the treatment for AD; the method of this embodiment may also be used to improve the safety of a clinical trial of a treatment for AD.


Additionally, the present invention is directed to a method for treating AD comprising determining that an AD patient has a unique gene expression pattern previously determined to be associated with the development of a favorable clinical response, e.g., a favorable immune response, e.g., IgG antibodies, to a treatment for AD, including but not limited to AN1792, and administering the treatment for AD to the AD patient. The present invention is also directed to a method for treating AD comprising determining that an AD patient does not have a unique gene expression pattern previously determined to be associated with the development of an adverse clinical response, e.g., inflammation, to administration of, e.g., AN1792, and administering a treatment for AD to the AD patient. In one embodiment, the inflammation is encephalitis and the treatment is AN1792. In another embodiment, the invention provides a method for treating AD comprising determining that an AD patient does not have a unique gene expression pattern previously determined to be associated with the lack of a development of a favorable clinical response and administering the treatment, e.g., AN1792, to the AD patient. In another method of treating embodied in the invention, an AD patient who has a gene expression pattern associated with the lack of a development of a favorable clinical response, e.g., a gene expression pattern associated with a poor IgG response, is administered the treatment in combination with an agent that enhances a favorable clinical response.


The present invention is also directed to a new genomically guided AN1792 for treating AD that is developed using the methods of the present invention, and methods for developing such genomically guided AN1792. The genomically guided AN1792 includes AN1792 having an improved therapeutic response profile for an individual or a group of individuals belonging to a genomically defined population selected from a nongenomically defined population having AD, wherein the genomically defined population is preidentified as having or not having a particular gene expression pattern(s), and wherein the particular gene expression pattern(s) is associated with an improved response to AN1792. The compositions of the present invention are administered to at least one individual of the genomically defined population and are capable of treating AD in the genomically defined population more effectively or safely than treating a nongenomically defined population of individuals having AD. The genomically defined population would typically be identified as part of the indication by information printed on the label or packaging of the genomically guided therapeutic product or composition, e.g., genomically guided AN1792, but any means of communicating the relevant information is contemplated. A skilled artisan will recognize that a genomically guided version of another therapy for Alzheimer's disease (i.e., a therapy other than AN1792) can be developed by using the methods of the present invention, and is also contemplated as part of the present invention.


In some embodiments, a unique gene expression pattern of the invention comprises different expression levels in inflammation developers, as compared to inflammation nondevelopers, of one or more genes selected from the group consisting of TPR, NKTR, XTP2, SRPK2, THOC2, PSME3, DAB2, SCAP2, furin, and CD54. In other embodiments, the one or more genes are selected from the group consisting of ASRGL1, TPR, and SRPK2. In another embodiment, a unique gene expression pattern comprises high expression levels of at least one gene selected from the group consisting of FCGRT and granulin and/or low expression levels of at least one gene selected from the group consisting of IARS and MCM3.




BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a schematic diagram summarizing the design of the pharmacogenomics study of the present invention.



FIG. 2 shows the efficiency of removal of neutrophils by CPT fractionation.



FIG. 3 provides an overview of the samples generated and the samples selected for pharmacogenomic analysis.



FIG. 4 shows the gene expression frequency pattern for TPR.



FIG. 5 shows the gene expression frequency pattern for NKTR.



FIG. 6 shows the gene expression frequency pattern for XTP2.



FIG. 7 shows the gene expression frequency pattern for SRPK2.



FIG. 8 shows the gene expression frequency pattern for THOC2.



FIG. 9 shows the gene expression frequency pattern for PSME3.



FIG. 10 shows the gene expression frequency pattern for DAB2.



FIG. 11 shows the gene expression frequency pattern for SCAP2.



FIG. 12 shows the gene expression frequency pattern for furin.



FIG. 13 shows the gene expression frequency pattern for ICAM1 (CD54).



FIG. 14 shows the gene expression levels of IARS.



FIG. 15 shows the gene expression levels of FCGRT.



FIG. 16 shows the gene expression levels of granulin.



FIG. 17 shows the gene expression levels of MCM3.



FIG. 18 shows the disposition of patients from whom samples were analyzed in Example 2. The asterisk (*) represents that in 14 of 167 cases, pharmacogenomic data are not available for patients who consented to participate in the study. In 6 of these 14 cases, shipping time exceeded specifications. In the remaining 8 cases, yield of RNA or amplification product (IVT) was insufficient for chip hybridization



FIG. 19 shows the ratio of monocytes to lymphocytes for each of the 123 immunized patients.



FIG. 20 shows a classification by GeneCluster of the five encephalitis patients and a representative 30 (of 118) nonencephalitis patients using the optimal classifier set of 8 genes selected by GeneCluster.



FIG. 21 shows the gene expression frequencies of 123 immunized patients (X encephalitis developers and ●=nonencephalitis developers): a) frequencies of AKAP13 and NPukP68, the top ranked pairwise combination identified by logistic models for classification of encephalitis patients; and b) frequencies of STAT1 and TPR, the top ranked pairwise combination containing STAT1 (third ranked pairwise combination overall) identified by logistic models for classification of encephalitis patients. Solid lines indicate decision boundaries where encephalitis and nonencephalitis classes were equiprobable (i.e., log odds ratio=0) in the logistic models.



FIG. 22 shows the gene expression frequencies in 123 immunized patients (X=encephalitis developers and ●=nonencephalitis developers) for 18 other pairs of genes. The number of the graph indicates the pair's rank among pairwise combinations of genes identified by logistic models for classification of encephalitis patients (as shown in Table 37): (2) 213064_at (NPukP68) and 211962_at (ZFP36L1); (4) 212152_x_at (ARID1A) and 209969_s_at (STAT1); (5) 213064_at (NPukP68) and 221753_at (SSH1); (6) 211960_s_at (RAB7) and 209969_s_at (STAT1); (7) 213064_at (NPukP68) and 202469_s_at (CPSF6); (8) 213064_at (NPukP68) and 21010_x_at (HNRPH3); (9) 208657_s_at (MSF) and 209969_s_at (STAT1); (10) 213064_at (NPukP68) and 205281_s_at (PIGA); (11) 221753_at (SSH1) and 209969_s_at (STAT1); (12) 211960_s_at (RAB7) and 213064_at (NPukP68); (13) 202270_at (GBP1) and 215823_x_at (PABPC1); (14) 209969_s_at (STAT1) and 201394_s_at (RBM5); (15) 203159_at (GLS) and 209969_sat (STAT1); (16) 202256_at (CD2BP2) and 209969_s_at (STAT1); (17) 209484_s_at (DC8) and 202256_at (CD2BP2); (18) 214911_s_at (BRD2) and 209969_s_at (STAT1); (19) 205988_at (CD84) and 209969_s_at (STAT1); and (20) 200626_s_at (MATR3) and 213064_at (NPukP68). Solid lines indicate decision boundaries where encephalitis and nonencephalitis classes were equiprobable (i.e., log odds ratio=0) in the logistic models.




DETAILED DESCRIPTION OF THE INVENTION

In order that the present invention may be more readily understood, certain terms are first defined. Additional definitions are set forth throughout the detailed description.


The term “adjuvant” refers to one or more biological immunomodulators that enhance antigen-specific immune responses.


The term “ApoE4” refers to apolipoprotein E, allele 4.


The term “cell saturation ratio” refers to the number of saturated features divided by the total number of features on the array.


The term “chip sensitivity” refers to the concentration level, in ppm, at which there is a 70% probability of obtaining a Present call, as calculated using Microarray Suite 5.0 (MAS 5.0; Affymetrix, Inc., Santa Clara, Calif.).


The term “cRNA” refers to complementary RNA.


The term “defect on visual inspection” refers to patterns in chip fluorescence visible after the chip has been run that reveal scratches, uneven staining, or other defects.


The term “EPIKS” refers to the Wyeth Expression Profiling Information and Knowledge System, an Oracle database (Oracle Corporation, Redwood Shores, Calif.) containing probe intensities and Absent/Present calls for each gene.


The term “final dataset” refers to the raw dataset which has been processed, and from which chips and genes not meeting various criteria have been filtered.


The term “FDR” refers to false discovery rate, an estimate of the percentage of genes that are false positive in a set of statistically significant genes.


The term “GEDS” refers to a graphical user interface that allows users to manually provide sample descriptions to EPIKS.


The term “GeneChip®” refers to an Affymetrix high-density array (Affymetrix, Inc., Santa Clara, Calif.) containing oligonucleotides of defined sequences that probe the cRNA derived from a target sample.


The term “GeneCluster” refers to an academic software application from the Whitehead Institute for Biomedical Research (Cambridge, Mass.) that chooses marker genes based on a signal-to-noise metric, and evaluates them by their ability to predict a given response parameter using a weighted voting algorithm.


The term “gene frequency” refers to a quantitative representation of the amount of gene present in a target sample, expressed as ppm.


The term “GLP” refers to Good Laboratory Practice.


The term “IVT” refers to in vitro transcription (used to generate the probe for hybridization to a gene chip).


The term “mitogen” refers to a compound with the property of inducing mitosis in culture.


The term “number of outliers across the array” refers to the capability of Affymetrix MAS 5.0 to detect outlier features. The MAS 5.0 manual indicates “outliers are probe cells that are obscured or nonuniform in intensity.” High numbers of outliers can indicate a poorly placed grid or a poorly aligned scanner. The MAS 5.0 software determines this number.


The term “PBMC” refers to peripheral blood mononuclear cells.


The term “PHA” refers to phytohemagglutinin, a T cell mitogen.


The term “ppm” refers to parts per million.


The term “probeset” refers to the oligonucleotides tiled on the gene chip representing a particular gene.


The term “QC” refers to quality control.


The term “QCP probability average difference” refers to the signal value for which there is a 70% probability of a Present call, as determined by the MAS 5.0 software.


The term “QCP probability frequency” refers to the QCP probability average difference expressed in ppm units.


The term “raw dataset” refers to the original gene expression and chip QC data, as stored on EPIKS.


The term “raw Q” refers to a measure of the noise level of the array. It is the degree of pixel-to-pixel variation among the probe cells used to calculate the background. Raw Q is an Affymetrix QC metric, which is determined by the MAS 5.0 software.


The term “scale factor” refers to the value required to obtain a trimmed mean intensity indicated by the target value. For all data in this study, the target value was set to a value of 100 and the scale factor was determined by dividing the trimmed mean of all probesets by the target value.


The term “U133A” refers to the commercial Affymetrix GeneChip® (Affymetrix, Inc., Santa Clara, Calif.) used in this study, which has been tiled with approximately 22,000 human probesets.


Generally, the present invention provides methods for predicting a clinical response of an AD patient to a treatment for AD to increase the chances for a favorable clinical response and/or reduce the risk of an adverse clinical response in an AD patient to a treatment for AD. The methods provided herein employ pharmacogenomic information to determine gene expression patterns associated with particular clinical responses. In one embodiment, the treatment is an immunotherapeutic, such as an active immunotherapeutic. The immunotherapeutic or immunotherapeutic agent is sometimes also termed an immunogen or immunogenic agent (see, e.g., WO 99/27944, to Schenk, incorporated by reference herein in its entirety). In another embodiment, the immunotherapeutic targets Aβ peptide. An example of such an immunotherapeutic is AN1792. In one embodiment of the invention, a favorable clinical response is the development of a protective immune response; in some embodiments, the protective immune response involves protective antibodies, e.g., IgG antibodies. In another embodiment, an adverse clinical response is the development of inflammation, e.g., encephalitis, e.g., meningoencephalitis. Methods for associating a gene expression pattern with a particular clinical response


Accordingly, the invention provides methods of compiling pharmacogenomic information to associate a unique gene expression pattern of a patient sample with a particular clinical response to a treatment for AD. Generally, the methods for compiling pharmacogenomic information to associate a unique gene expression pattern of a patient sample with a particular clinical response to a treatment for AD comprise the following steps: (1) procuring at least one patient sample from a patient of a first population of patients and at least one patient sample from a patient of a second population of patients, wherein the first population consists of one or more patients who developed the particular clinical response to the treatment for AD and wherein the second population consists of one or more patients who did not develop the particular response to the treatment for AD; (2) acquiring a gene expression pattern from each procured patient sample; and (3) determining whether at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population, wherein a determination that at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population results in associating the unique gene expression pattern with the particular clinical response to the treatment for AD. In one embodiment of the invention, the particular clinical response is one that is neither favorable nor adverse (e.g., antibody nonresponsiveness). In some embodiments, the particular clinical response is either a favorable clinical response or an adverse clinical response. In other embodiments, the particular clinical response is both a favorable and adverse clinical response.


For example, the invention also provides a method for compiling pharmacogenomic information to associate a unique gene expression pattern of a patient sample with a favorable clinical response to a treatment for AD comprising the following steps: (1) procuring at least one patient sample from a patient of a first population of patients and at least one patient sample from a patient of a second population of patients, wherein the first population consists of one or more patients who developed the favorable clinical response to the treatment for AD and wherein the second population consists of one or more patients who did not develop the favorable response to the treatment for AD; (2) acquiring a gene expression pattern from each procured patient sample; and (3) determining whether at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population, wherein a determination that at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population results in associating the unique gene expression pattern with the favorable clinical response to the treatment for AD.


In one embodiment of the invention, the second population consists of one or more patients who did not develop the favorable clinical response to the treatment and also developed an adverse clinical response. In another embodiment of the invention, the method further comprises excluding patients who also developed an adverse clinical response to the treatment for AD from the first population of patients.


The present invention also provides a method of compiling pharmacogenomic information to associate a unique gene expression pattern of a patient sample with an adverse clinical response to a treatment for AD comprising the following steps: (1) procuring at least one patient sample from a patient of a first population of patients and at least one patient sample from a patient of a second population of patients, wherein the first population consists of one or more patients who developed the adverse clinical response to the treatment for AD and wherein the second population consists of one or more patients who did not develop the adverse response to the treatment for AD; (2) acquiring a gene expression pattern from each procured patient sample; and (3) determining whether at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population, wherein a determination that at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population results in associating the unique gene expression pattern with the adverse clinical response to the treatment for AD. In one embodiment of the invention, the second population consists of one or more patients who did not develop the adverse clinical response to the treatment and also developed a favorable adverse clinical response. In another embodiment of the invention, the method further comprises excluding patients who also developed a favorable clinical response from the first population of patients.


Although the inventors were able to associate unique gene expression patterns to either favorable or adverse clinical responses to the AD treatment comprising administration of AN1792, a skilled artisan will recognize that the methods of compiling pharmacogenomic information provided herein may be used to associate unique gene expression profiles with either, neither, or both favorable or adverse clinical responses to any treatment for AD, e.g., including, but not limited to, immunotherapies, i.e., active or passive immunotherapies. In one embodiment, the treatment for AD comprises administration of AN1792.


A skilled artisan will recognize that a unique gene expression pattern may be defined as the pattern created by the differential, i.e., increased or decreased, expression level(s) of one or more genes in at least most patient samples from one population compared to expression level(s) of the same one or more genes in at least most patient samples from a second population. As used herein, an increased or decreased expression level relates to any statistically significant increase or decrease. Additionally, one of skill in the art will recognize that a unique gene expression pattern may consist of (1) the upregulation of one or more genes, (2) the downregulation of one or more genes, or (3) the upregulation of one or more genes and the downregulation of one or more other genes. Finally, a skilled artisan will recognize that a gene expression pattern may be considered unique when it can be used to differentiate the clinical response(s) of at least most of one patient population from the clinical response(s) of at least most of a second patient population, i.e., when it is associated with either a favorable or adverse clinical response, with both a favorable and adverse clinical response, or with neither favorable nor an adverse clinical response.


Methods of procuring a patient sample and what would constitute an appropriate patient sample are well known in the art. Additionally in the provided methods of compiling pharmacogenomic information, a patient sample may be taken before, during, or after the patient is treated with a treatment for AD, as long as the patient sample may be correlated with the final clinical response developed by the patient from which the sample was procured. In one embodiment of the invention, the patient sample is a PBMC fraction. In another embodiment, the patient sample is procured prior to the patient being treated with a treatment for AD. In another embodiment of the invention, the sample may be further processed, e.g., stimulated (e.g., placed under a certain in vitro culture condition), prior to the acquisition of its gene expression pattern, and the gene expression pattern of the sample cultured under a certain culture condition may be associated with either a favorable or adverse clinical response to a treatment for AD. For example, a sample may be placed under culture conditions that mimic the treatment for AD, e.g., incubated with an immunotherapeutic that is administered as a treatment for AD. A skilled artisan will be able to determine appropriate culture conditions, e.g., media, temperature, atmosphere, etc., for this type of analysis, and will know to include appropriate control conditions, e.g., the absence of the immunotherapeutic, the presence of a cell activator, etc.


To determine whether a gene expression pattern is unique, i.e., may be associated with a particular clinical response to a treatment for AD, a comparison must be made between gene expression patterns of samples procured from patients who developed a particular clinical response to a treatment for AD and gene expression patterns of samples procured from patients who did not develop the particular clinical response to the same treatment for AD. Consequently, patient samples must be procured from at least one patient of a first patient population consisting of one or more patients who developed the particular clinical response and from at least one patient of a second patient population consisting of one or more patients who did not develop the particular clinical response, such that a comparison of the gene expression patterns of the two populations may be made. Additionally, the patient populations must comprise patients who have been treated with the treatment for AD or will be treated with the treatment for AD (e.g., if the patient sample is taken before the treatment for begins) so that the patients will have a clinical response to the treatment. A skilled artisan will recognize that the association of a unique gene expression pattern with a favorable or adverse clinical response will be stronger if more AD patients are within the patient populations. Additionally, a skilled artisan will recognize that, in addition to patients who did not develop a favorable and/or adverse clinical response to the treatment for AD, samples may be procured from patients who developed a clinical response to a treatment for AD that is neither favorable nor adverse, AD patients who were given a placebo, and/or patients who do not have AD, e.g., healthy patients, etc. A skilled artisan will recognize that the phrase “AD patient” may also refer to candidates for AD therapy, e.g., individuals not presently diagnosed with AD, for example, patients only at risk of developing AD, or patients (e.g., elderly patients) presently in good health. Gene expression patterns from such patients may serve to corroborate the association of a unique gene expression pattern with a particular clinical response, as controls, etc. For example, where the favorable and adverse clinical responses are at opposite ends of the spectrum of one response, or where the clinical response may be graduated (e.g., an immune response) the gene expression pattern of a sample procured from an AD patient who developed a clinical response that is neither favorable nor adverse may prove to be one that is in between, or intermediate compared to, the expression levels(s) of the gene(s) involved in the a unique gene expression pattern associated with a favorable clinical response and the expression levels(s) of the gene(s) involved in a unique gene expression pattern associated with an adverse clinical response.


Since an object of the invention is to provide methods by which a unique gene expression pattern may be associated with either a favorable or an adverse clinical response, the clinical responses of each patient from whom a sample was procured should be monitored and recorded. A skilled artisan will recognize that, generally, a favorable clinical response to a treatment for AD may include the prevention, slowing down, arrest, and/or reversal of the development of AD, and may include the biological responses that promote the prevention, slowing down, arrest, and/or reversal of the development of AD (e.g., a protective immune response, e.g., an antibody response). A skilled artisan will also recognize that an adverse clinical response (1) is more than the natural progression of AD despite of the treatment for AD, (2) generally involves responses to the treatment for AD, and (3) is harmful to the patient. In other words, an adverse clinical response may be considered a harmful side effect of the treatment for AD and may include the biological responses that cause the side effects. For example, an adverse clinical response to a treatment for AD may be encephalitis, e.g., meningoencephalitis, and/or the inflammatory response that leads to encephalitis. Thus, in some instances, it may be that what constitutes a favorable clinical response only can be determined after the patient population has been treated and a favorable clinical response(s) is observed. Similarly, in some instances, it may be that what constitutes an adverse clinical response only can be determined after the patient population has been treated and an adverse clinical response(s) is observed. In this situation, it becomes clear why procurement of a patient sample prior to treating the patient with a treatment for AD is preferable. Thus, the methods provided herein may be used to associate a unique gene expression pattern with a favorable clinical response, e.g., a protective immune response, to a treatment for AD. In one embodiment, the favorable clinical response is an antibody response. In a more specific embodiment, the favorable clinical response is an IgG antibody response. The methods provided herein may also be used to associate a unique gene expression pattern with an adverse clinical response. In one embodiment, the adverse clinical response is inflammation, e.g., encephalitis, e.g., meningoencephalitis.


A skilled artisan will recognize the well-known methods for acquiring a gene expression pattern from a patient sample, e.g., methods of using preexisting gene expression patterns of a patient sample (e.g., those that may be stored in a database), and methods for detecting gene products (e.g., mRNA, proteins, etc.) such as, but not limited to, RT-PCR, in situ hybridization, slot-blotting, nuclease protection assays, Southern blot analysis, Northern blot analysis, microarray analysis, ELISA, RIA, FACS, dot blot analysis, Western blot analysis, immunohistochemistry, etc. In one embodiment of the invention, the patient sample is a PBMC fraction. In another embodiment, gene expression patterns are measured using RNA isolated from a patient sample. In another embodiment, a gene expression pattern is acquired by methods of microarray hybridization and microarray data analyses. In another embodiment, gene expression patterns are measured using protein isolated from a patient sample.


In the methods of compiling pharmacogenomic information that will determine an association between a unique gene expression pattern of a patient sample with a particular clinical response to a treatment for AD, all that is required for the association is that at least most of the patient samples procured from patients that developed a particular clinical response have a unique gene expression pattern that is not found in at least most of the patient samples procured from patients who did not develop the particular response. At least most encompasses at least 51%. In one embodiment, at least most means at least 75%. In another embodiment, at least most means at least 80%. Additionally, a skilled artisan will recognize that cross-validation studies of the association between a gene expression and a clinical response will serve to corroborate the association.


A skilled artisan will recognize that the step of excluding patients from a first population of patients may encompass, but is not limited to, the following: 1) excluding patient samples procured from patients prior to the step of acquiring a gene expression pattern from each procured patient sample, and/or 2) excluding from the unique gene expression pattern genes that are part of a gene expression pattern associated with another clinical response. For example, as described below, treatment with AN1792 led to some patients developing only the favorable IgG response and some patients developing both the favorable IgG response and encephalitis. Thus, a unique gene expression pattern may be associated with a favorable clinical response by excluding patient samples, procured from patients who also developed an adverse clinical response, prior to acquiring a gene expression pattern from each procured sample, and/or by excluding from the unique gene expression pattern to be associated with the favorable clinical response one or more genes that may also be associated with an adverse clinical response. Similarly, a unique gene expression pattern may be associated with an adverse clinical response by excluding patient samples, procured from patients who also developed a favorable clinical response, prior to acquiring a gene expression pattern from each procured sample, and/or excluding from the gene expression pattern genes to be associated with the adverse clinical response one or more genes that may also be associated with a favorable clinical response.


As noted above, AN1792 is considered a promising treatment for AD. However, although a subset of patients developed a favorable clinical response to AN1792 that correlated with a protective immune response, e.g., the development of antibodies, a smaller subset of AD patients developed an adverse clinical response, e.g., inflammation leading to encephalitis, and the immunotherapeutic dosing was discontinued. The information obtained during the clinical trials and the availability of samples from patients who participated in the study has allowed for the pharmacogenomic studies disclosed herein. In other words, the methods of compiling pharmacogenomic information as provided herein were used to associate at least one gene expression pattern of a sample procured from an AD patient treated with AN1792 with a favorable or adverse clinical response to AN1792.


In one embodiment, blood samples were taken from participants in the AN1792 phase II clinical trial (see Examples 1 and 2). For each sample, the peripheral blood mononuclear cell (PBMC) fraction was purified by CPT (cell preparation tube) fractionation. However, the PBMCs may be purified by flotation or density barrier, or any other means known in the art. After the PBMCs have been purified from the total cell population, which increases the percentage of neutrophils in the remaining cell population, some of the PBMCs were cultured, e.g., with AN1792 (see Example 1). However a skilled artisan will recognize that samples may be cultured by any means known in the art, and also that gene expression patterns may be acquired from unstimulated samples (see, e.g., Example 2). After culture, the nonadherent cultured cells were harvested and removed from the culture media by centrifugation and the RNA was purified by conventional means, specifically by QIAshredders and Qiagen RNeasy mini-kits (Qiagen Inc., Valencia, Calif.); the same purification steps were used for unstimulated cells. Any method known in the art for purifying RNA may be used. The purified RNA was then amplified by in vitro translation amplification with biotinylated nucleotides, to make biotinylated cRNA. The biotinylated cRNA was then hybridized to known sequences to determine which sequences are present or absent in the RNA sample. For example, the amplified, biotinylated cRNA was hybridized to the Affymetrix human U133A oligonucleotide GeneChip, which interrogates the RNA levels of over 22,000 sequences. The GeneChip was then washed to remove unhybridized cRNA, stained with streptavidin, and scanned to produce array images that were processed with the Affymetrix MicroArray Suite (MAS 5.0) software and was further processed to create probeset summary values. Probe intensities were summarized for each message using the Affymetrix Signal algorithm and the Affymetrix Absolute Detection metric (Absent, Present, or Marginal) for each probeset. Normalization, filtering, and identification and reporting of outlier samples were then performed. The data was then statistically analyzed using, e.g., analysis of variance (ANOVA) and signal-to-noise metrics to determine a unique gene expression patterns of cultured or unstimulated patient samples associated with encephalitis, IgG responsiveness, and/or IgG nonresponsiveness, as noted in Example 1. Other well-known combinations of computer programs, databases, and/or statistical algorithms, including, but not limited to, Affymetrix programs (e.g., MAS 5.0, SAS, etc.), the EPIKS database, determination of Pearson correlation coefficients (r2), analysis of covariance (ANCOVA), analysis of variance (ANOVA), Benjamini and Hochberg's False Discovery Rate (FDR) procedure, logistic regression, Ingenuity pathways analysis, GeneCluster analysis, etc., may be used to associate gene expression patterns with particular clinical outcomes (see also, e.g., Example 2). The skilled artisan will recognize that other means may be used to analyze the data from the hybridizations and acquire a gene expression profile from a procured sample.


Accordingly, the invention also provides methods of compiling pharmacogenomic information to associate a unique gene expression pattern of a patient sample taken from a patient treated with AN1792 with a clinical response to the administration of AN1792. In one embodiment of the invention, gene expression patterns are acquired from unstimulated samples. In another embodiment, samples are placed under a certain culture condition prior to acquisition of gene expression patterns. In one embodiment, the favorable clinical response is a protective immune response. In another embodiment, the favorable clinical response is an antibody response, e.g., an IgG response. In another embodiment, the adverse clinical response is an inflammatory response. In one embodiment, the inflammatory response leads to encephalitis, e.g., meningoencephalitis. A skilled artisan will recognize that the term “inflammation,” or “inflammatory response” refers to an innate immune response that results in an adverse clinical response when used regarding or in the context of discussing encephalitis (or other adverse inflammatory side effects, e.g., vasculitis, cellulitis, nephritis, etc.) and/or results in absence of a favorable response. A skilled artisan also will recognize that, as described above, a favorable or adverse clinical response to AN1792 may be chosen from a variety of responses, including but not limited to the prevention, slowing down, arrest and/or reversal of the development of AD (e.g., a protective immune response) or an adverse drug response (e.g., an inflammatory response).


Applying the methods of compiling pharmacogenomic information as provided herein to at least one patient of a first patient population consisting of one or more patients who developed a particular clinical response and at least one patient of a second patient population consisting of one ore more patients who did not develop the particular clinical response to AN1792, several unique gene expression patterns were obtained that may be associated with a particular clinical response to AN1792, e.g., IgG responders, IgG partial responders, IgG nonresponders, encephalitis developers, and/or encephalitis nondevelopers.


In practicing the methods of compiling pharmacogenomic information, the inventors were able to associate gene expression patterns of cultured patient samples, e.g., patient samples incubated with AN1792, with a particular response (e.g., encephalitis developers, IgG nonresponders) to AN1792. The genes of expression patterns of stimulated samples that may be associated with either a favorable or adverse clinical response to AN1792 are listed in Tables 10-12 and 18. Additionally, the inventors were able to associate unique gene expression patterns of unstimulated samples with a particular clinical response to AN1792 (e.g., IgG responders and/or encephalitis developers). The gene expression patterns of unstimulated samples that may be associated with either a favorable or adverse clinical response to AN1792 are listed in Tables 24-37.


The genes listed in Table 10 (and discussed in Example 1) are associated with the development of encephalitis and are either upregulated or downregulated in cultured patient samples procured from encephalitis developers, i.e., encephalitis developers may have increased or decreased levels of these genes as compared to encephalitis nondevelopers.


In one embodiment, increased gene expression levels of one or more of the genes listed in Table 11 (and discussed in Example 1) in a cultured patient sample are associated with the development of encephalitis.


In another embodiment, decreased gene expression levels of one or more of the genes listed in Table 12 (and discussed in Example 1) in a cultured patient sample are associated with the development of encephalitis.


In another embodiment of the invention, the differential expression levels in encephalitis developers as compared to encephalitis nondevelopers for at least one or more of the following genes in a cultured patient sample is associated with the development of encephalitis, as further illustrated in FIGS. 4-13: TPR; NKTR; XTP2; SRPK2; THOC2; PSME3; DAB2; SCAP2; furin; and ICAM1 (CD54). In another embodiment of the invention, the difference in expression levels in encephalitis developers as compared to encephalitis nondevelopers for at least one or more of the following genes in a cultured patient sample is associated with the development of encephalitis: TPR; NKTR; SRPK2; DAB2; SCAP2; and furin (PACE).


In another embodiment, the differential expression levels of one or more genes in cultured patient samples are associated with neither a favorable or adverse clinical response, i.e., these genes are upregulated or downregulated in cultured patient samples procured from AD patients who did not develop an IgG antibody response, i.e., IgG nonresponders, compared to those in cultured patient samples procured from AD patients who did develop an IgG response. Preferably, the gene expression pattern of IgG nonresponders includes a moderate to high level of expression of at least one of the genes listed in Table 18 in cultured patient samples as having “higher” average expression in IgG nonresponders, and/or a low level of at least one of the genes listed in Table 18 as having “lower” average expression in IgG nonresponders. As used herein, moderate to high levels of expression means any statistically significant increase in expression in IgG nonresponders as compared to IgG responders, and low levels means any statistically significant decrease in expression in IgG nonresponders as compared to IgG responders. More preferably, the gene expression pattern of IgG nonresponders includes a moderate to high level of expression of at least one of the genes selected from the group consisting of granulin and FCGRT, and/or a low level of expression of at least one of the genes selected from the group consisting of IARS and MCM3.


The genes listed in Table 24 (and discussed in Example 2.3.2) are associated with the development of a favorable clinical response, i.e., a protective immune response, particularly an IgG antibody response, and have an odds ratio for IgG association (as calculated with meningoencephalitics) of at least three-fold between IgG responders and others, and are either upregulated (e.g., have an odds ratio ≧3) or downregulated (e.g., have an odds ratio ≦0.33) in unstimulated patient samples procured from AD patients who developed an IgG antibody response to administration of AN1792 (i.e., IgG responders), as compared to unstimulated patient samples procured from AD patients who did not develop an IgG antibody response (IgG nonresponders) or patient samples procured from AD patients who developed an IgG antibody response but also developed an adverse clinical response, particularly inflammation leading to encephalitis (i.e., IgG responder and meningoencephalitic). In other words, IgG responders may have increased or decreased expression levels of these genes compared to IgG nonresponders and/or IgG responders and meningoencephalitics.


In one embodiment, increased gene expression levels of one or more of the genes listed in Tables 25-27 having a three-fold increase in odds ratios (e.g., genes listed in Tables 25-27 as having an odds ratio ≧3) in an unstimulated patient sample are associated with the development of a protective IgG response (see Example 2.3.3). In another embodiment, decreased gene expression levels of one or more of the genes listed in Tables 25-27 having a three-fold decrease in odds ratio (e.g., genes listed in Tables 25-27 as having an odds ratio≦0.33) are associated with the development of a favorable protective IgG response (see Example 2.3.3).


In another embodiment of the invention, the differential expression levels in patients who developed an IgG antibody response to AN1792 as compared to patients who did not develop an IgG antibody response or who did develop an IgG antibody response but also developed an adverse clinical response, e.g., inflammation leading to encephalitis, for at least one of the genes listed in Tables 28 and 30 in an unstimulated patient sample is associated with the development of a favorable IgG immune response. In other words, the upregulation of expression of one or more genes listed in Tables 28-31 listed as having an odds ratio ≧3) and/or the downregulation of expression of one or more genes in Tables 28 and 30 listed as having an odds ratio ≦0.33) in an unstimulated patient sample may be associated with a favorable IgG immune response.


The genes listed in Table 32 (and discussed in Example 2.3.5) are associated with the development of encephalitis and are either upregulated (i.e., have an odds ratio for association with encephalitis ≧3) or downregulated (i.e., have an odds ratio for association with encephalitis ≦0.33) in unstimulated patient samples procured from encephalitis developers.


In one embodiment, increased gene expression levels of one or more of the genes listed in Table 34 (including the subset of genes listed in Table 35), e.g., genes listed in Table 34 or 35 as having an odds ratio for association with encephalitis ≧3, in an unstimulated patient sample are associated with the development of encephalitis (see Example 2.3.6). In another embodiment, decreased gene expression levels of one or more of the genes listed in Table 34 (including the subset of genes listed in Table 35), e.g., genes listed in Table 34 or 35 as having an odds ratio for association with encephalitis ≦0.33, are associated with the development of encephalitis (see Example 2.3.6).


In another embodiment of the invention, the differential expression levels in encephalitis developers as compared to encephalitis nondevelopers for at least one or more of the genes listed in Table 36 in an unstimulated patient sample is associated with the development of encephalitis (see also FIG. 20). In other words, an upregulated expression of one or more genes listed in Table 36 as having an odds ratio for encephalitis ≧3, and/or a downregulated expression of one or more genes listed in Table 36 as having an odds ratio for encephalitis ≦0.33, in a patient sample may be associated with the development of encephalitis.


In another embodiment of the invention, the differential expression level of one or more pairs of genes, e.g., those pairs listed in Table 37, in a patient sample distinguishes encephalitis developers from encephalitis nondevelopers (see Example 2.3.7). As depicted in FIGS. 21 and 22, whether the differential expression levels of one or more pairs of genes is associated with encephalitis development or encephalitis nondevelopment in a patient is dependent on where the expression levels of the two genes within a pair of genes (e.g., as noted on the X and Y axes of the graphs in FIGS. 21 and 22) are in relation to the decision boundary (e.g., the solid line in a graph in FIG. 21 or FIG. 22) for that pair.


Polynucleotides of the Invention


Polynucleotides encoding the genes involved with unique gene expression patterns of the present invention may be used as hybridization probes and primers to identify and isolate nucleic acids having sequences identical to or similar to the disclosed genes. Hybridization methods for identifying and isolating nucleic acids include polymerase chain reaction (PCR), Southern hybridizations, in situ hybridization and Northern hybridization, and are well known to those skilled in the art.


Hybridization reactions can be performed under conditions of different stringency. The stringency of a hybridization reaction includes the difficulty with which any two nucleic acid molecules will hybridize to one another. Preferably, each hybridizing polynucleotide hybridizes to its corresponding polynucleotide under reduced stringency conditions, more preferably stringent conditions, and most preferably highly stringent conditions. Examples of stringency conditions are shown in Table 1 below: highly stringent conditions are those that are at least as stringent as, for example, conditions A-F; stringent conditions are at least as stringent as, for example, conditions G-L; and reduced stringency conditions are at least as stringent as, for example, conditions M-R.


Polynucleotides associated with genes of the present invention may be used as hybridization probes and primers to identify and isolate DNA having sequences encoding allelic variants of the disclosed genes. Allelic variants are naturally occurring alternative forms of polynucleotides that encode polypeptides that are identical to or have significant similarity to the polypeptides encoded by the polynucleotides associated with the disclosed genes. Preferably, allelic variants have at least 90% sequence identity (more preferably, at least 95% identity; most preferably, at least 99% identity) with the polynucleotides associated with the disclosed genes.


Polynucleotides associated with the disclosed genes of the present invention may also be used as hybridization probes and primers to identify and isolate DNAs having sequences encoding polypeptides homologous to the disclosed genes. These homologs are polynucleotides and polypeptides isolated from a different species than that of the polypeptides and polynucleotides associated with the disclosed genes, or within the same species, but with significant sequence similarity to the polynucleotides and polypeptides associated with the disclosed genes. Preferably, polynucleotide homologs have at least 50% sequence identity (more preferably, at least 75% identity; most preferably, at least 90% identity) with the polynucleotides associated with the disclosed genes, whereas polypeptide homologs have at least 30% sequence identity (more preferably, at least 45% identity; most preferably, at least 60% identity) with the polypeptides associated with the disclosed genes. Preferably, homologs of the polynucleotides and polypeptides associated with the disclosed genes are those isolated from mammalian species. Polynucleotides associated with the disclosed genes of the present invention may also be used as hybridization probes and primers to identify cells and tissues that express polypeptides associated with the disclosed genes of the present invention and the conditions under which they are expressed.


Panels and Kits


A unique gene expression pattern may comprise the expression level of one gene that may be considered individually, although it is within the scope of the invention that a unique gene expression pattern may comprise the expression levels of two or more genes to increase the confidence of the analysis. In one embodiment, the invention provides a unique gene expression pattern that comprises a panel of genes. A panel may comprise 2-5, 5-15, 15-35, 35-50, 50-100, or more than genes. In one embodiment, a panel may comprise 15-20 genes.


In another embodiment, panels of genes are selected such that the genes within any one panel share certain features. As a nonlimiting example, the genes of a first panel may each have high expression levels in a unique gene expression pattern associated with a particular clinical response. Alternatively, genes of a second panel may each exhibit differential expression as compared to a first panel. Similarly, different panels of genes may be composed of genes that are from different functional categories (i.e., proteolysis, signal transduction, transcription, etc.), or may be selected to represent different stages of, e.g., an immune response. Panels of genes may be made by selecting genes involved in a unique gene expression pattern associated with a particular clinical response. As a nonlimiting example, a panel may comprise genes selected from, e.g., Table 24. Panels may also be made by combining genes selected from those listed in Table 10-12, 18, and 24-37. In one embodiment, a panel comprises genes listed in Table 36. In another embodiment, a panel comprises a pair of genes, e.g., any of the pairs of genes listed in Table 37.


In addition to providing unique gene expression patterns that may comprise one gene or a panel of genes, it is within the scope of the invention to provide kits for detecting one or a panel of genes involved in a unique gene expression pattern of the invention. These kits may comprise one or more polynucleotides, each capable of hybridizing under stringent conditions to an RNA transcript, or the complement thereof, of a gene differentially expressed in a unique gene expression pattern of the invention; and/or one or more antibodies, each capable of binding to a polynucleotide encoded by a gene differentially expressed in a unique gene expression of the invention.


Additionally, the kits of the invention may comprise one or more polynucleotides and/or one or more antibodies for the detection of one or more genes involved in a gene expression pattern of the invention, wherein the one or more polynucleotides and/or antibodies are conveniently coupled to a solid support. For example, polynucleotides of genes involved in a unique gene expression pattern of the invention may be coupled to an array (e.g., a biochip for hybridization analysis), to a resin (e.g., a resin that can be packed into a column for column chromatography), or a matrix (e.g., a nitrocellulose matrix for Northern blot analysis). By providing such support, discrete analysis of the expression level(s) of each gene selected for the panel may be detected. For example, in an array, polynucleotides complementary to each gene of a unique gene expression pattern comprising a panel of gene may be individually attached to different known locations on the array. The array may be hybridized with, for example, polynucleotides extracted from a sample (e.g., a blood sample) from a subject. The hybridization of polynucleotides from the sample with the array at any location on the array can be detected, and thus the expression level of the gene in the sample can be ascertained. Thus, not only tissue specificity, but also the level of expression of a panel of genes in the tissue is ascertainable. In one embodiment, an array based on a biochip is employed. Similarly, ELISA analyses may be performed on immobilized antibodies specific for different polypeptide biomarkers hybridized to a protein sample from a subject. Methods of making and using such arrays, including the use of such arrays with computer readable media comprising genes of the invention and/or databases, e.g., a relational database, are well known in the art.


In another embodiment, a reporter nucleic acid is utilized to detect the expression of one or more genes involved in a unique gene expression pattern. Such a reporter nucleic acid can be useful for high-throughput screens for agents that alter the expression profiles of peripheral blood mononuclear cells. The construction and use of such reporter assays are well known.


For example, the construction of a reporter for transcriptional regulation of a gene involved in a unique gene expression pattern of the invention generally requires a regulatory sequence of the gene, typically the promoter. The promoter can be obtained by a variety of routine methods. For example, a genomic library can be hybridized with a labeled probe consisting of the coding region of the nucleic acid to identify genomic library clones containing promoter sequences. The isolated clones can be sequenced to identify sequences upstream from the coding region. Another method is an amplification reaction using a primer that anneals to the 5′ end of the coding region of a polynucleotide for the gene. The amplification template can be, for example, restricted genomic nucleic acid to which anchor bubble adaptors have been ligated.


To construct the reporter, the promoter of the selected gene may be operably linked to the reporter nucleic acid, e.g., without utilizing the reading frame of the polynucleotide sequence of the selected gene. The nucleic acid construct is transformed into tissue culture cells, e.g., peripheral blood mononuclear cells, by a transfection protocol to generate reporter cells.


Many of the well-known reporter nucleic acids may be used. In one embodiment, the reporter nucleic acid is green fluorescent protein. In a second embodiment, the reporter is β-galactosidase. In other embodiments, the reporter nucleic acid is alkaline phosphatase, β-lactamase, luciferase, or chloramphenicol acetyltransferase. The reporter nucleic acid construct may be maintained on an episome or inserted into a chromosome by, for example, using targeted homologous recombination. Methods of making and using such reporter nucleic acids and others are well known.


Methods of Using a Gene Expression Pattern Associated with a Particular Clinical Response


Once at least one unique gene expression pattern of a patient sample is associated with a particular clinical response to a treatment for AD, the at least one unique gene expression pattern may be used to predict whether a patient will develop the particular clinical response to the treatment for AD, even if the AD patient had not been previously exposed to the treatment for AD. Thus the invention also provides methods of predicting whether a candidate patient who has not been previously exposed to a treatment for AD will develop a particular clinical response to a treatment for AD, the methods generally comprising (1) associating at least one unique gene expression pattern of a patient sample with a particular clinical response to the treatment for AD by methods of compiling pharmacogenomic information (2) procuring a test sample from the candidate patient who has not been previously exposed to the treatment for AD, and (3) determining whether the test sample procured from the candidate patient who has not been previously exposed to the treatment for AD has a test gene expression pattern that is substantially similar to the at least one gene expression pattern associated with the particular clinical response to the treatment for AD, wherein if it is determined that the test sample has a test gene expression pattern that is substantially similar to the at least one gene expression pattern associated with the particular clinical response, it may be predicted that the candidate patient will develop the particular clinical response. In one embodiment, the particular clinical response is neither favorable nor adverse. In one embodiment, the particular clinical response is either a favorable or adverse clinical response. In another embodiment, the particular clinical response is both a favorable and adverse clinical response.


In some embodiments, a database of unique gene expression patterns that are each associated with a particular clinical response to a treatment for AD will have been previously established. In such a case, the methods of predicting a clinical response of a candidate patient comprises the steps procuring a test sample from the candidate patient not previously exposed to the treatment for AD, and determining whether the test sample from the candidate patient not previously exposed to the treatment for AD has a test gene expression pattern that is substantially similar to a reference gene expression pattern that has been previously associated with a particular clinical response, wherein if it is determined that the test sample has a test gene expression pattern that is substantially similar to the reference gene expression pattern that has been previously associated with the particular clinical response, it may be predicted that the candidate patient will develop the particular clinical response. A skilled artisan will recognize that a particular clinical response may be a favorable clinical response, e.g., a protective immune response, an adverse clinical response, e.g., an inflammatory response, a clinical response that is neither favorable nor adverse, e.g., nonresponsiveness, or any combination of the three.


A skilled artisan will recognize that in the above-described methods of predicting the clinical response of a candidate AD patient, the test sample should be procured from the candidate AD patient in the same manner, or as close as possible to the same manner, as the procurement of the reference sample (i.e., the sample of which the gene expression pattern is associated a particular clinical response) from the reference AD patient. Additionally, a skilled artisan will recognize that in determining whether the test sample has a test gene expression pattern that is substantially similar to a reference gene expression pattern, i.e., a gene expression pattern that has been previously associated with a particular clinical response to the treatment for AD, a test gene expression pattern must be acquired from the test sample. Also, the test gene expression pattern should be acquired in a similar manner as the gene expression pattern that has been previously associated with a particular clinical response. Such methods of procuring a sample (or test sample) and acquiring a gene expression pattern (or test gene expression pattern) are well known in the art, as described above.


As a nonlimiting example, if the gene expression pattern associated with a particular clinical response was acquired via microarray analysis of a PBMC sample procured from an patient treated with a treatment for AD prior to the patient being exposed to the treatment for AD, the test gene expression pattern would also be acquired via microarray analysis of a PBMC sample procured from a candidate patient prior to the candidate patient being exposed to the treatment for AD. As another nonlimiting example, if the gene expression pattern previously associated with a particular clinical response was acquired from a patient sample that was placed under certain culture conditions after its procurement, the test gene expression pattern would be acquired from a test sample placed under similar culture conditions after its procurement. In other words, the timing of procuring a sample and a test sample in relation to exposure to a treatment for AD, the conditions in which the sample and the test sample are processed (e.g., unstimulated, cultured, etc.), the methods used to acquire the gene expression pattern previously associated with a particular clinical response and the test gene expression pattern, and the treatment administered to the AD patient treated with the treatment and the treatment for which candidate AD patient is a candidate, ideally would be similar or as similar as possible.


Since part of the invention associates unique gene expression patterns with particular clinical responses to AN1792 by AD patients to treatment with AN1792, the clinical response of a candidate patient to treatment with AN1792 may be predicted using the gene expression patterns described in Tables 10-12, 18, and 24-37. Therefore the present invention relates to a method of predicting whether a candidate patient will develop a particular clinical response when administered AN1792 by (1) compiling pharmacogenomic information to associate at least one unique gene expression pattern of a preimmunization patient sample procured from a patient who has been treated with AN1792 with a particular clinical response, (2) procuring a test sample from the candidate patient, and (3) determining whether the test sample has a test gene expression pattern that is substantially similar to the at least one unique gene expression pattern, wherein if the test sample has a test gene expression pattern that is substantially similar to the at least one gene expression pattern associated with the particular clinical response, it may be predicted that the candidate patient will develop the particular clinical response. In one embodiment, the particular clinical response is neither favorable nor adverse, e.g., nonresponsiveness. In another embodiment, the particular clinical response to AN1792 is a favorable clinical response, e.g., a protective immune response, e.g., an IgG antibody response. In another embodiment, the particular clinical response to AN1792 is an adverse clinical response, e.g., an inflammatory response, e.g., encephalitis.


For example, the invention is therefore further directed to a method for predicting whether a candidate AD patient will have an IgG response. Preferably, an AD patient treated with a treatment for AD, such as an immunotherapeutic, e.g., AN1792, will have a moderate to high level of IgG expression and will not develop an inflammatory response, such as encephalitis. As noted above, AN1792 is an immunotherapeutic for patients with AD. It presumably works by stimulating the immune system to “recognize” and attack the β-amyloid plaques in patients with AD, and does so by causing the production of antibodies against the β-amyloid protein. Therefore, a good IgG response after administration of AN1792 is desired. Accordingly, the present invention provides a method for predicting whether a candidate AD patient is likely to mount a moderate to high IgG response, either by determining that a test sample procured from the candidate AD patient does not express a unique gene expression pattern associated with nonresponsiveness or determining that a test sample procured from the candidate AD patient has another unique gene expression pattern associated with IgG responsiveness. Generally, the method comprises (1) obtaining a patient population previously exposed to AN1792, wherein the patient population includes IgG responders and IgG nonresponders and wherein IgG expression is associated with administration of AN1792, (2) determining whether there is a unique gene expression pattern associated with patient samples procured from IgG nonresponders that is not found in patient samples procured from IgG responders, and (3) determining whether a test patient sample procured from the candidate patient does not have the unique gene expression pattern associated with IgG nonresponders, wherein if the test sample does not have a test gene expression pattern that is substantially similar to the at least one gene expression pattern associated with IgG nonresponders, it may be predicted that the candidate patient will not be an IgG nonresponder, i.e., will be an IgG responder. More specifically, the method comprises (1) collecting blood from a patient population previously exposed to AN1792, wherein the patient population includes patients who mount a moderate to high IgG response to AN1792 and patients who mount a low or undetectable IgG response, i.e., IgG responders and IgG nonresponders, respectively, (2) purifying, e.g., total RNA from the blood sample, (3) assaying RNA expression levels to obtain gene expression patterns for the IgG responders and IgG nonresponders, (4) comparing the gene expression patterns of the IgG responders and IgG nonresponders to obtain a unique gene expression pattern for IgG nonresponders, and (5) determining whether a candidate patient not previously exposed to AN1792 has the unique gene expression pattern for IgG nonresponders, wherein the presence of the unique gene expression pattern in the candidate patient predicts a likelihood that the candidate patient will not mount an IgG response. If the candidate patient does not have the unique gene expression pattern associated with a poor IgG response, it is possible that the patient is a good candidate for treatment with AN1792. Similarly to the disclosure involving predicting whether a candidate patient will be an encephalitis developer or nondeveloper, IgG responders and nonresponders can also be predicted by assaying protein expression levels to obtain gene expression patterns. One of ordinary skill in the art will appreciate that the general disclosure related to treatment with AN1792 may also be used for treatments for Alzheimer's disease other than AN1792.


Preferably, the gene expression pattern of IgG nonresponders includes a moderate to high level of expression of at least one of the genes listed in Table 18 in cultured cells as having “higher” average expression in IgG nonresponders, and/or a low level of at least one of the genes listed in Table 18 as having “lower” average expression in IgG nonresponders. As used herein, moderate to high levels of expression means any statistically significant increase in expression in IgG nonresponders as compared to IgG responders, and low levels means any statistically significant decrease in expression in IgG nonresponders as compared to IgG responders. More preferably, the gene expression pattern of IgG nonresponders includes a moderate to high level of expression of at least one of the genes selected from the group consisting of granulin and FCGRT, and/or a low level of expression of at least one of the genes selected from the group consisting of IARS and MCM3.


A unique gene expression pattern may also be associated with a favorable clinical response, e.g., the production of antibodies, particularly IgG antibodies. The invention is thus further directed to methods for predicting that a candidate AD patient will have a favorable clinical response to treatment with AN1792, the method comprising (1) associating at least one gene expression pattern of a sample with a favorable clinical response to AN1792 by methods of compiling pharmacogenomic information, as described above, (2) procuring a test sample from the candidate AD patient not previously exposed to AN1792, and (3) determining that the test sample procured from the candidate AD patient not previously exposed to AN1792 has a test gene expression pattern that is substantially similar to the at least one gene expression pattern associated with a favorable clinical response AN1792. In one embodiment of the invention, a favorable clinical response to AN1792 includes a protective immune response. In another embodiment, a favorable clinical response to AN1792 includes the development of antibodies, e.g., IgG. Preferably, the gene expression pattern of IgG responders is acquired from unstimulated patient samples and includes a moderate to high level of expression of at least one of the genes listed in Tables 24-31 as having “higher” average expression in IgG responders, and/or a low level of at least one of the genes listed in Tables 24-31 as having “lower” average expression in IgG responders. As used herein, moderate to high levels of expression means any statistically significant increase in expression in IgG nonresponders as compared to IgG responders, and low levels means any statistically significant decrease in expression in IgG nonresponders as compared to IgG responders.


Along the same lines, the present invention provides a method for predicting whether a candidate patient is likely to develop inflammation in response to the administration of a treatment for AD comprising determining whether the candidate patient has a unique gene expression pattern associated with the development of inflammation in response to the treatment.


In one embodiment of the invention, the method predicts the likelihood of whether a candidate AD patient not previously exposed to a particular treatment for AD, such as AN1792, will develop an inflammatory response, such as encephalitis, to AN1792. In this embodiment, the method comprises (1) obtaining a nucleic acid sample from a patient population previously exposed to the treatment, wherein the patient population includes inflammation developers and inflammation nondevelopers, (2) using the nucleic acid sample to determine whether the inflammation developers of the patient population have a unique gene expression pattern not found in the inflammation nondevelopers, and (3) determining whether a candidate patient not previously exposed to the treatment has the unique gene expression pattern, wherein the presence of the unique gene expression pattern in the candidate patient predicts a likelihood that the candidate patient will develop inflammation. While inflammation is the adverse effect in this embodiment, any adverse effect is contemplated by the present invention.


In another embodiment of the invention, the method predicts that a candidate AD patient not previously exposed to AN1792 will develop an adverse clinical response to AN1792. In this embodiment, the method comprises (1) associating at least one gene expression pattern of a sample with an adverse clinical response to AN1792 by methods of compiling pharmacogenomic information, as described above, (2) procuring a test sample from the candidate AD patient not previously exposed to AN1792, and (3) determining that the test sample procured from the candidate AD patient not previously exposed to AN1792 has a test gene expression pattern that is substantially similar to the at least one gene expression pattern associated with an adverse clinical response AN1792. In one embodiment of the invention, an adverse clinical response to AN1792 includes an inflammatory response. In another embodiment, an adverse clinical response to AN1792 includes the development of encephalitis, e.g., meningoencephalitis. In another embodiment, the gene expression pattern associated with an adverse clinical response is procured from an unstimulated sample and includes a moderate to high level of expression at least one of the genes listed in Tables 32-37 as having a higher average expression in encephalitis developers and/or a low level of expression of at least one of the genes listed in Tables 32-37 as having lower expression in encephalitis developers.


The determination of gene expression patterns associated with the encephalitis response in AD patients to AN1792 is useful for predicting the likelihood that a patient will develop encephalitis. Therefore, the present invention relates to a method of predicting whether a patient will develop encephalitis when administered AN1792 by (1) determining whether patients who developed encephalitis during clinical trials have a unique (preimmunization) gene expression pattern associated with encephalitis, and (2) determining whether a candidate patient has the unique gene expression pattern, wherein the presence of the unique gene expression pattern indicates that the candidate patient is not a good candidate for AN1792 treatment and the absence of the unique gene expression pattern indicates that that candidate patient is (or may be) a good candidate for AN1792 treatment.


In one embodiment, the method comprises comparing gene expression patterns of AD patients who develop encephalitis in response to AN1792 treatment (encephalitis developers) and AD patients who do not develop encephalitis in response to AN1792 treatment (encephalitis nondevelopers) to define a unique gene expression pattern for encephalitis developers, and determining whether a candidate AD patient not previously exposed to AN1792 has the unique gene expression pattern, wherein the presence of the unique gene expression pattern in the candidate AD patient predicts a likelihood that the patient will develop encephalitis. Gene expression patterns may be determined by any means known in the art, including, but not limited to determining protein and/or RNA expression patterns in a sample, as described above. In another embodiment of the invention, the method comprises (1) assaying RNA expression levels to obtain gene expression patterns for the encephalitis developers and encephalitis nondevelopers, (2) comparing the gene expression patterns of the encephalitis developers and encephalitis nondevelopers to define a unique gene expression pattern for encephalitis developers, and (3) determining whether a candidate AD patient not previously exposed to AN1792 has the unique gene expression pattern, wherein the presence of the unique gene expression pattern in the candidate AD patient predicts a likelihood that the patient will develop encephalitis. If the candidate AD patient does not have the unique gene expression pattern associated with encephalitis, the patient is (or may be) a good candidate for treatment with AN1792. The method may further comprise collecting blood from a patient population previously exposed to AN1792, wherein the patient population includes encephalitis developers and encephalitis nondevelopers, and purifying total RNA from the blood sample. In another embodiment of the invention, the method comprises (1) assaying protein expression levels to obtain gene expression patterns for the encephalitis developers and encephalitis nondevelopers, (2) comparing the gene expression patterns of the encephalitis developers and encephalitis nondevelopers to define a unique gene expression pattern for encephalitis developers, and (3) determining whether a candidate AD patient not previously exposed to AN1792 has the unique gene expression pattern, wherein the presence of the unique gene expression pattern in the candidate AD patient predicts a likelihood that the patient will develop encephalitis. If the candidate AD patient does not have the unique gene expression pattern associated with encephalitis, the patient is (or may be) a good candidate for treatment with AN1792. Protein expression levels may be assayed by any means known in the art. The method may further comprise collecting blood from a patient population previously exposed to AN1792, wherein the patient population includes encephalitis developers and encephalitis nondevelopers, and obtaining protein from the blood sample.


Methods to Improve the Safety and Efficacy of a Treatment for AD


A skilled artisan will recognize that the ability to predict the clinical response of an AD patient to treatment for AD will enable methods to improve the safety and efficacy of the treatment for AD. Such methods include, but are not limited to, providing a treatment for AD to only candidate AD patients predicted to have favorable clinical response(s) to the treatment, modifying the gene expression pattern of a sample taken from a candidate AD patient to resemble a gene expression pattern associated with a favorable clinical response (i.e., modifying the ‘gene expression pattern’ of the patient to have the gene expression pattern of a later-procured sample resemble a gene expression pattern associated with a favorable clinical response), developing a genomically guided therapeutic product, etc.


I. Improving Clinical Response Profiles of Treatments for AD


Accordingly, the present invention provides methods for improving a response profile of a treatment for AD by increasing the chances that an AD patient develops a favorable clinical response to the treatment for AD, comprising (1) determining that the AD patient has a unique gene expression pattern associated with a favorable clinical response to the treatment for AD, and (2) administering the treatment for AD to the AD patient.


The present invention provides methods for improving a response profile of a treatment for AD by reducing the risk that an AD patient will develop an adverse clinical response to the treatment for AD, comprising (1) determining that the patient has a unique gene expression pattern associated with an adverse clinical response to the treatment for AD, and (2) not administering the treatment for AD to the AD patient. In one embodiment of the invention, the methods improve the response profile of treating AD with AN1792.


Accordingly, the present invention is also directed to an improved treatment for AD comprising administering AN1792 to a patient population, wherein the patient population has a gene expression pattern associated with a favorable clinical response and/or lacks another gene expression pattern associated with an adverse clinical response.


By targeting a population of AD patients who develop a favorable clinical response to AN1792, e.g., patients who are IgG responders (thus avoiding a population of AD patients who are IgG nonresponders), i.e., patients from whom patient samples that have at least one unique gene expression profile associated with a favorable clinical response to AN1792 are procured, the efficacy of AN1792 as a treatment for AD may be improved. Therefore, the present invention provides an improved method of treatment of AD comprising treating a population of AD patients with AN1792, wherein samples procured from the population of AD patients have a unique gene expression pattern associated with a favorable clinical response. Alternatively, it may be that the samples, e.g., after culture, do not express an appropriate level(s) of one or more of the above-indicated genes that is associated with IgG nonresponsiveness in Table 18. This method of treatment results in a reduction or elimination of AD patients who are treated with AN1792 that do not mount an IgG response, and thus improves the efficacy of AN1792.


In accordance with the invention, there is also provided a method for treating a population of AD patients with AN1792, wherein the population of patients does not express a gene expression pattern associated with an adverse clinical response, e.g., expresses different expression levels of one or more of the above-indicated genes as compared to encephalitis nondevelopers. The treatment results in a reduction or elimination of the incidence of adverse clinical responses, e.g., encephalitis, in the population of AD patients and improves the safety of AN1792.


The present invention also contemplates a method of targeting candidate AD patients who are not likely to develop an adverse clinical response, e.g., encephalitis, to AN1792 and are likely to develop a favorable clinical response, e.g., a protective immune (e.g., IgG) response to AN1792. The method comprises determining a unique gene expression pattern associated with patients who develop adverse or nonfavorable clinical responses, e.g., encephalitis developers and/or IgG nonresponders, respectively, and then determining whether the candidate AD patient has this unique gene expression pattern(s). Similarly, the invention relates to a method for treating an AD patient with AN1792, wherein the AN1792 has improved safety and efficacy profiles, comprising administering AN1792 to the candidate patient not having a gene expression pattern(s) associated with an adverse or a nonfavorable clinical response, e.g., an encephalitis developer and/or an IgG nonresponder, respectively.


II. Altering a Gene Expression Pattern Associated with an Adverse Clinical Response.


One or more genes included as part of a unique gene expression pattern may also be useful as a therapeutic agent(s) or a target(s) for a treatment. Therefore, without limitation as to mechanism, some of the methods of the invention are based, in part, on the principle that regulation of the expression level(s) of one or more genes involved in a unique expression pattern associated with a particular clinical response may promote a favorable clinical response to a treatment for AD when expressed at levels similar or substantially similar in patient samples isolated from patients who develop a favorable response to a treatment for AD. The discovery of these unique expression patterns for individual or panels of genes that may be associated with a favorable or clinical response allows for screening of test compounds with the goal of regulating a unique gene expression pattern associated with a particular clinical response; for example, screening can be done for compounds that will convert a unique gene expression pattern associated with an adverse clinical response to a unique gene expression pattern associated with a favorable clinical response.


For example, in relation to these embodiments, a unique gene expression pattern may comprise genes that are determined to have modulated activity or expression in response to a therapy regime. Alternatively, the modulation of the activity or expression of a unique gene expression pattern, or one or more genes of the gene expression pattern, may be correlated with a particular clinical outcome to a treatment for AD. In addition, regulatory agents affecting the expression level of at least one gene that is part of a unique gene expression pattern (associated polynucleotides and/or polypeptides, related associated polynucleotides and/or polypeptides (e.g., inhibitory polynucleotides, inhibitory polypeptides (e.g., antibodies), small molecules, etc.) may be administered as therapeutic drugs. In another embodiment of the invention, regulatory agents of the invention may be used in combination with one or more other therapeutic compositions of the invention. Formulation of such compounds into pharmaceutical compositions is described below. Administration of such a therapeutic regulatory agent may regulate the aberrant expression of at least one gene that is part of a unique gene expression pattern, and therefore may be used to increase the chances for a favorable clinical response and/or decrease the risk of an adverse clinical response to a treatment for AD.


Altered expression of the genes of the present invention may be achieved in a cell or organism through the use of various inhibitory polynucleotides, such as antisense polynucleotides and ribozymes that bind and/or cleave the mRNA transcribed from the genes involved in a unique gene expression pattern of the invention (see, e.g., Galderisi et al. (1999) J. Cell Physiol. 181:251-57; Sioud (2001) Curr. Mol. Med. 1:575-88). Such inhibitory polynucleotides may be useful in preventing or treating inflammation and similar or related disorders.


The antisense polynucleotides or ribozymes of the invention can be complementary to an entire coding strand of a gene of the invention, or to only a portion thereof. Alternatively, antisense polynucleotides or ribozymes can be complementary to a noncoding region of the coding strand of a gene of the invention. The antisense polynucleotides or ribozymes can be constructed using chemical synthesis and enzymatic ligation reactions using procedures well known in the art. The nucleoside linkages of chemically synthesized polynucleotides can be modified to enhance their ability to resist nuclease-mediated degradation, as well as to increase their sequence specificity. Such linkage modifications include, but are not limited to, phosphorothioate, methylphosphonate, phosphoroamidate, boranophosphate, morpholino, and peptide nucleic acid (PNA) linkages (Galderisi et al., supra; Heasman (2002) Dev. Biol. 243:209-14; Micklefield (2001) Curr. Med. Chem. 8:1157-79). Alternatively, these molecules can be produced biologically using an expression vector into which a polynucleotide of the present invention has been subcloned in an antisense (i.e., reverse) orientation.


The inhibitory polynucleotides of the present invention also include triplex-forming oligonucleotides (TFOs) that bind in the major groove of duplex DNA with high specificity and affinity (Knauert and Glazer (2001) Hum. Mol. Genet. 10:2243-51). Expression of the genes of the present invention can be inhibited by targeting TFOs complementary to the regulatory regions of the genes (i.e., the promoter and/or enhancer sequences) to form triple helical structures that prevent transcription of the genes.


In one embodiment of the invention, the inhibitory polynucleotides of the present invention are short interfering RNA (siRNA) molecules. These siRNA molecules are short (preferably 19-25 nucleotides; most preferably 19 or 21 nucleotides), double-stranded RNA molecules that cause sequence-specific degradation of target mRNA. This degradation is known as RNA interference (RNAi) (e.g., Bass (2001) Nature 411:428-29). Originally identified in lower organisms, RNAi has been effectively applied to mammalian cells and has recently been shown to prevent fulminant hepatitis in mice treated with siRNA molecules targeted to Fas mRNA (Song et al. (2003) Nature Med. 9:347-51). In addition, intrathecally delivered siRNA has recently been reported to block pain responses in two models (agonist-induced pain model and neuropathic pain model) in the rat (Dorn et al. (2004) Nucleic Acids Res. 32 (5):e49).


These siRNA molecules can be generated by annealing two complementary single-stranded RNA molecules together (one of which matches a portion of the target mRNA) (Fire et al., U.S. Pat. No. 6,506,559) or through the use of a single hairpin RNA molecule that folds back on itself to produce the requisite double-stranded portion (Yu et al. (2002) Proc. Natl. Acad. Sci. USA 99:6047-52). The siRNA molecules can be chemically synthesized (Elbashir et al. (2001) Nature 411:494-98) or produced by in vitro transcription using single-stranded DNA templates (Yu et al., supra). Alternatively, the siRNA molecules can be produced biologically, either transiently (Yu et al., supra; Sui et al. (2002) Proc. Natl. Acad. Sci. USA 99:5515-20) or stably (Paddison et al. (2002) Proc. Natl. Acad. Sci. USA 99:1443-48), using an expression vector(s) containing the sense and antisense siRNA sequences. Recently, reduction of levels of target mRNA in primary human cells, in an efficient and sequence-specific manner, was demonstrated using adenoviral vectors that express hairpin RNAs, which are further processed into siRNAs (Arts et al. (2003) Genome Res. 13:2325-32).


The siRNA molecules targeted to polynucleotides associated with the disclosed genes of the present invention can be designed based on criteria well known in the art (e.g., Elbashir et al. (2001) EMBO J. 20:6877-88). For example, the target segment of the target mRNA preferably should begin with AA (most preferred), TA, GA, or CA; the GC ratio of the siRNA molecule preferably should be 45-55%; the siRNA molecule preferably should not contain three of the same nucleotides in a row; the siRNA molecule preferably should not contain seven mixed G/Cs in a row; and the target segment preferably should be in the ORF region of the target mRNA and preferably should be at least 75 bp after the initiation ATG and at least 75 bp before the stop codon. Based on these criteria, or on other known criteria (e.g., Reynolds et al. (2004) Nature Biotechnol. 22:326-30), siRNA molecules can be designed by one of ordinary skill in the art.


III. Genomically Guided Therapeutics


Another embodiment of the present invention is a method for developing a genomically guided AN1792 (a genomically guided therapeutic product) comprising determining gene expression patterns for AD subjects who are not likely to develop encephalitis after administration of AN1792 and/or who are likely to develop an. IgG response after administration of AN1792. The method of the present invention is useful in making genomically guided AN1792 which comprises AN1792 and a label comprising an indication of a target population genomically defined to be not likely to develop encephalitis after administration of AN1792 and/or likely to develop an IgG response after administration of AN1792. As used herein a label comprising an indication of a target population genomically defined to be not likely to develop encephalitis and/or likely to develop an IgG response, is any type of medium that may be provided together with AN1792, such as a leaflet, a package insert, a list of instructions, an instruction manual, a computer readable medium, a label on a bottle, or any other type of medium which conveys to the pharmacist, physician, or any other healthcare provider, and/or the patient the desired target population.


The genomically guided AN1792 includes AN1792 having an improved therapeutic response profile for an individual or a group of individuals belonging to a genomically defined population selected from a nongenomically defined population having AD, wherein the genomically defined population is preidentified as having (or not having) a particular gene expression pattern and wherein the particular gene expression pattern is associated with an improved response to AN1792. The compositions of the present invention are administered to at least one individual of the genomically defined population and are capable of treating AD in the genomically defined population more effectively or safely than treating a nongenomically defined population of individuals having AD. As noted, the genomically defined population would typically be identified as part of the indication by information printed on the label or packaging of, or otherwise provided with, genomically guided AN1792.


In addition, the present invention is directed to a defined population of cells originating from and residing in diverse mammalian individuals, preferably human, wherein said population is formed by determining the presence of a gene expression pattern associated with a characteristic response to AN1792 and wherein the population of cells is exposed to a therapeutically effective amount of AN1792. The present invention is also directed to a defined and isolated population of cells originating from diverse mammalian individuals, preferably human, wherein said population comprises a gene expression pattern associated with a characteristic response to AN1792 and wherein the population of cells is exposed to a therapeutically effective amount of AN1792. Such cells may be cultured in vitro and may be useful for the study of AN1792 in vitro.


Another aspect of the invention relates to a method comprising the steps of providing at least one peripheral blood sample of an AD patient; and comparing an expression profile of one or more genes in the at least one peripheral blood sample to at least one reference expression profile from an AD patient treated with AN1792 of said one or more genes. Each of the genes is differentially expressed in peripheral blood mononuclear cells (PBMCs) of AD patients who developed encephalitis, or did not develop an IgG response, or both, in response to AN1792 treatment as compared to AD patients who did not develop encephalitis, or did develop an IgG response, or both, respectively, in response to AN1792 treatment.


Diagnostic or screening methods based on differentially expressed gene products are well known in the art. In accordance with one aspect of the present invention, the differential expression patterns of an AD patient likely to develop encephalitis and/or not develop an IgG response in response to AN1792 treatment can be determined by measuring the level of RNA transcripts of these genes in peripheral blood samples. Suitable methods for this purpose include, but are not limited to, RT-PCR, Northern Blot, in situ hybridization, Southern Blot, slot-blotting, nuclease protection assays and polynucleotide arrays. The peripheral blood samples can be either whole blood, or samples containing enriched PBMCs. In other embodiments of the invention, the source of genes can be a bodily fluids or a tissue other than blood.


In general, RNA isolated from peripheral blood samples can be amplified to cDNA or cRNA before detection and/or quantification. The isolated RNA can be either total RNA or mRNA. Suitable amplification methods include, but are not limited to, RT-PCR, isothermal amplification, ligase chain reaction, and Qbeta replicase. The amplified nucleic acid products can be detected and/or quantified through hybridization to labeled probes. Amplification primers or hybridization probes can be prepared from the gene sequence of differentially expressed genes using methods well known in the art.


The differential expression patterns of genes associated with the likelihood of developing encephalitis and/or of not developing an IgG response can also be determined by measuring the levels of polypeptides encoded by these genes in peripheral blood. 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.


Suitable antibodies include, but are not limited to, polyclonal antibodies, monoclonal antibodies, chimeric antibodies, humanized antibodies, single chain antibodies, Fab fragments and fragments produced by Fab expression libraries. Such antibodies can be prepared by methods well known in the art. Available antibodies may also be used.


In a further aspect of the invention, there is provided a system comprising a computer readable memory that stores at least one reference expression profile of one or more genes in peripheral blood samples of a reference AD patient, wherein each of said one or more genes is differentially expressed in PBMCs of AD patients who are likely to develop encephalitis, or not likely to develop an IgG response, or both, respectively, in response to AN1792 treatment as compared to AD patients who are not likely to develop encephalitis, or are likely to develop an IgG response, or both, respectively, in response to AN1792 treatment. A program capable of comparing an expression profile of interest to the reference expression profile, and a processor capable of executing the program, is also provided in the system.


For the method of treatment for AD of the present invention, AN1792 is administered in a therapeutically effective amount. AN1792 may be administered orally, topically, parenterally, by inhalation or spray (e.g., nasally), or rectally in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants and vehicles. The term parenteral as used herein includes percutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, or intrathecal injection or infusion techniques and the like. Preferably, the AN1792 is administered as a pharmaceutical formulation comprising AN1792 and a pharmaceutically acceptable carrier. AN1792 may be present in association with one or more nontoxic pharmaceutically acceptable carriers and/or diluents and/or adjuvants, and, if desired, other active ingredients. The pharmaceutical compositions containing AN1792 may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.


Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preservative agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain AN1792 in admixture with nontoxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques. In some cases such coatings may be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed.


Formulations for oral use may also be presented as hard gelatin capsules wherein the AN1792 is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.


Aqueous suspensions contain AN1792 in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.


Oily suspensions may be formulated by suspending AN1792 in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents and flavoring agents may be added to provide palatable oral preparations. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.


Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide AN1792 in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents or suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present.


Pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil or a mineral oil or mixtures of these. Suitable emulsifying agents may be naturally occurring gums, for example gum acacia or gum tragacanth, naturally occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents.


Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents. The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono-or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.


AN1792 may also be administered in the form of suppositories, e.g., for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable nonirritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter and polyethylene glycols.


AN1792 may be administered parenterally in a sterile medium. AN1792, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle. Advantageously, adjuvants, local anesthetics, preservatives and buffering agents can be dissolved in the vehicle.


In one embodiment, the AN1792 peptide antigen is provided as a sterile liquid suspension, which appears as a hazy, colorless liquid suspension and which includes 0.5 mg/mL, in 10 mM glycine, 10 mM sodium citrate, 0.4% polysorbate 80, 5% sucrose, at a pH of 6.0. The AN1792 is administered together with QS-21 adjuvant, which is provided as a sterile, clear solution, and includes 1.0 mg/mL, in phosphate buffered saline with 0.4% polysorbate 80 at a pH of 6.5.


QS-21 (Stimulon™; Antigenics, Inc., Framingham, Mass.; U.S. Pat. No. 5,057,540) is a naturally occurring saponin molecule purified from the South American tree Quillaja saponaria Molina. Numerous studies in laboratory animals have demonstrated the adjuvant activity of QS-21 and have established its safety profile. Rabbit toxicity studies with single or multiple injections of various doses of QS-21 alone or combined with various antigens have documented a pattern of mild to moderate inflammation (hemorrhage, necrosis and edema) at the injection site and no significant organ toxicity. Slight alterations in white blood cell counts (leukocytosis and leukopenia) and creatinine kinase are common. Pharmacokinetic data collected after a single IM injection of tritium-labeled QS-21 in rabbits show QS-21 highly concentrated in the lymph nodes draining the injection area. Excretion occurs primarily through the kidneys, and both QS-21 and its metabolites are found in the urine. Studies in mice, rabbits and monkeys with QS-21 adjuvanted immunotherapeutics show improvement in B and T cell effector function, especially an increase in achieved antibody titers, induction of antigen-specific cytotoxic T lymphocytes, immunoglobulin class switching, affinity maturation and broadening of antigen-primed B cell repertoire.


In another embodiment, polysorbate 80 is a component of the formulated drug product AN1792 and the adjuvant, QS-21. It is a nonionic surfactant used widely as an emulsifying agent in the preparation of stable oil-in-water pharmaceutical emulsions. It is also used as a solubilization agent or as a wetting agent in the formulation of oral and parenteral suspensions. There have been occasional reports of rare contact hypersensitivity to polysorbates following their topical use and reports of tuberculin type hypersensitivity following intramuscular injection in combination with vitamin A. Polysorbates have also been associated with serious adverse events, including some deaths in low-birth weight infants following intravenous administration of a vitamin E preparation containing a mixture of polysorbate 20 and 80.


The AN1792 and QS-21 are preferably administered by intramuscular injection into deltoid muscle. If multiple administrations are desired, sides may be alternated for each injection session. Several administrations may be necessary to achieve the best results; in one embodiment, administrations are given as follows: a first injection is given at day 1; one month later, a second injection is given; 2 months after injection 2, a third injection is given; 3 months after injection 3, a fourth injection is given; 3 months after injection 4, a fifth injection is given; and 3 months after injection 5, a sixth injection is given, for a total of six injections in one year.


At present, the anti-AN1792 titer necessary to achieve a beneficial therapeutic effect in human AD is unknown. Whereas the PDAPP (platelet-derived growth factor-driven amyloid precursor protein) transgenic mouse develops several AD-like neuropathologies, the progression of pathology in this model may very well take a more aggressive course than in human AD, as the changes occur in months and the expression levels APP/Aβ are several fold higher than in nontransgenic species. The lowest titers in PDAPP efficacy studies that have resulted in lessening of neuropathological progression have been in the range of 1-2,000. In addition, a fragment of Aβ(1-5) attached to a carrier protein and combined with complete Freund's adjuvant/incomplete Freund's adjuvant was effective in preventing neuropathology despite raising a peak geometric titer of only 2,400.


It will be understood, however, that the specific dose level and administration dosing schedule for any particular patient will depend upon a variety of factors including the activity of the AN1792 employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy, as well as the antibody titer that is desired.


The following examples are intended to illustrate the invention and should not be construed as limiting the invention in any way


EXAMPLES

An exploratory search for predictors of clinical responses to AN1792 immunization in the preimmunization gene expression patterns in PBMCs of patients with mild to moderate AD was undertaken. Accordingly, pharmacogenomic analyses have been performed with the intention of determining associations between gene expression patterns and clinical response parameters.


Predictors of response were sought because the incidence of antibody responsiveness in the Phase I study was relatively low (48%), an incidence that would have more than doubled the number of patients required in a Phase II evaluation of efficacy (as measured by cognitive function) associated with anti-AN1792 antibody response. Therefore, a wide and unbiased pharmacogenomic-based search for genes whose expression levels prior to immunization were significantly associated with postimmunization positive antibody titer was designed. Consequently, blood samples were obtained from 123 treated U.S. patients (five of which developed meningoencephalitis) and 30 patients in the placebo group. Simultaneous analysis of the expression levels of approximately 22,000 sequences in each preimmune blood sample obtained from all consenting subjects was performed using the Affymetrix U133A GeneChip®. In the Phase Ia trials of AN1792, by the time encephalitis was recognized as a severe adverse event, preimmune blood samples from five of the six U.S. encephalitis patients had been collected for pharmacogenomic studies. (The sixth U.S. encephalitis patient had not consented to the pharmacogenomic portion of the study, and therefore no blood sample was available from this patient for the pharmacogenomics study).


In summary, as developed below, associations between preimmunization gene expression patterns in peripheral blood mononuclear cells of AD patients, that were either placed under in vitro culture conditions (Example 1) or unstimulated (Example 2), and postimmunization clinical responses have been found. Corroboration of these findings may be of interest and may be made by showing the same associations in a second (independent) sample set (e.g., samples from the European clinical trial patients).


Example 1
Association Between Gene Expression Patterns of in Vitro Stimulated (Cultured) Samples and Adverse Clinical Responses
Example 1.1
Materials and Methods—Sample Preparation

Consent to the pharmacogenomic study was optional and obtained after approval by local institutional review boards in the U.S. (E.U. patients were not included in the pharmacogenomic study). Blood was collected from patients in the U.S. at the screening visit and was shipped overnight at room temperature to the Pharmacogenomic Laboratory in Andover, Mass. For each sample, the peripheral blood mononuclear cell (PBMC) fraction was purified by CPT fractionation, as described below, and 2×106 of these cells (the baseline sample, i.e., the first daughter sample for baseline measurements) were snap frozen; these represent cells that were not subject to in vitro culture (see. Example 1.1.3.1). The remaining cells were divided into four equal aliquots and cultured in vitro overnight in conditions described below. Cells were then harvested and snap frozen. The culturing step was performed because it was reasoned that preimmunization gene expression profiles in PBMCs associated with a postimmunization clinical response to AN1792 might most likely be revealed by exposing PBMCs to AN1792 as an antigen in culture. The hypothesis behind this reasoning was that immunotherapeutic responsiveness may reflect a state of “preexisting readiness” to respond to AN1792, and this state may be reflected in the gene expression profile of PBMCs prior to immunotherapy. Accordingly, both AN1792-stimulated and control cultures were set up for each sample. Total RNA was purified from each sample, and RNA expression levels of each of 22,000 sequences were assayed, as described below. Statistical analyses were performed to identify genes whose expression patterns showed a statistically significant association with antibody responsiveness, development of encephalitis or the presence of ApoE4 alleles. FIG. 1 shows a summary of the design of this Example 1.


Example 1.1.1
Purification of PBMCs by CPT Fractionation

Fractionation of PBMCs by CPT (cell preparation tube) fractionation was performed using a single screening visit blood sample drawn into a CPT Cell Preparation Vacutainer Tube (BD Vacutainer Systems, Franklin Lakes, N.J.). The target volume was 8 ml, but in some cases this target was not reached. Samples that were not received at Pharmacogenomics Laboratory within a day of collection were excluded from the study. Upon receipt, differential cell counts were performed. The PBMC fraction was then purified according to the CPT protocol (BD Vacutainer Systems) and differential cell count performed on the purified PBMC fraction. CPT purification resulted in greater than 99% reduction in RBC representation in all 141 study samples. CPT purification did not alter by more than 15% the percentage of monocytes relative to PBMCs. The efficiency of removal of neutrophils by CPT fractionation is shown in FIG. 2. For the samples of FIG. 2, CPT tubes were inverted gently eight times, 300 μl was removed in a counting vial for the Pentra 60 C+ analyzer (ABX Diagnostics; Montpellier, France) and differential counts performed. PBMC purification on the remaining sample was performed by centrifugation in a horizontal swinging rotor bucket at 1500×g for 20 minutes. The PBMC fraction was removed and washed by adding 5 ml phosphate buffered saline (PBS), gently inverting eight times, and transferring into a 15 ml conical tube. This procedure was repeated using 3 ml PBS. The PBMC fraction was then pelleted at 450×g for 5 minutes. The supernatant (PBS) was discarded, cells were resuspended in 3 ml PBS, and 300 μl of this was removed for cell differential counts using a Pentra 60 C+ analyzer. Closed symbols represent the percentage neutrophils before CPT fractionation; open symbols represent the percentage neutrophils after CPT fractionation.


Post-CPT fractionation, the percentage of neutrophils averaged 11% of the neutrophil percentage before fractionation, with a standard deviation of 11. As seen in FIG. 2, in eight cases (patients 17, 23, 36, 44, 271, 288, 311, and 756) CPT fractionation failed to reduce the percentage of neutrophils to less than 20%. (As shown in Table 2 (see also Table 6), one of these eight patients, patient 311, was removed from analysis due to an operator error identified during QC review.) It has been reported (Schmielau and Finn (2001) Cancer Res. 61:4756-60) that changes in neutrophils upon activation cause them to sediment aberrantly and copurify with PBMCs, suggesting that density change is a marker of their activation. Therefore it is likely that the seven samples included in data analysis that have a relatively high number of neutrophils in the post-CPT PBMC fraction came from patients with a higher than normal percentage of activated neutrophils. Since this parameter (activated neutrophils) could potentially impact gene expression profiles, upon unblinding of the samples, the characteristics of these seven samples among the patient groups were analyzed to determine whether there was an over- or under-representation of samples with high neutrophil content in any of the patient groups. Table 2 lists characteristics of samples with post-CPT fractionation neutrophil content >20%, and shows that patients with high neutrophil content are represented in both the antibody responding and nonresponding groups. None of the five patients who developed encephalitis are among the patients with high post-CPT fractionation neutrophil content.


Of the seven patients with high postfractionation neutrophil content, one received placebo, four are IgM nonresponders and three are IgM responders. As mentioned above, data from patient 311 was removed from analysis due to an operator error identified during QC review.


Example 1.1.2
Overnight Culture Conditions

All in vitro culture was done in upright tissue culture flasks (Falcon, catalog number 353108; Fischer Scientific, Pittsburgh, Pa.) in complete culture media consisting of RPMI 1640, 10% heat inactivated fetal calf serum (0.9 EU/ml), 100 u/ml penicillin and 100 μg/ml streptomycin (GIBCO/BRL; Gaithersburg, Md.), 2 mM glutamine (GIBCO/BRL), 5×10−5 M 2-mercaptoethanol. Cultures were incubated at 37° C. with 5% CO2 overnight. In cases where at least 1×107 cells were available, 2.5×106 cells were added to 5 ml of treatment group stimulation media for each of four culture groups. (Stimulation media for each of the four groups is described below.) In cases where cell number was <1×107, 25% of the available cells were added to 5 ml of treatment group stimulation media for each of the four treatment groups.


Example 1.1.3
Generation of Five Daughter Samples from Each Patient Sample

Five daughter samples were generated from each patient sample received. FIG. 3 provides a summary of the samples generated and the samples selected for analysis. As detailed below, five daughter samples were generated from each available purified PBMC sample. One of these daughter samples was not placed in culture (first daughter sample). The other four daughter samples were cultured overnight as described above (second through fifth daughter samples).


Example 1.1.3.1
Baseline (First Daughter) Samples—(Unstimulated)

An aliquot consisting of 2×106 cells was removed from the purified PBMC fraction, pelleted by centrifugation, resuspended in 300 μl RLT Buffer (Qiagen, Valencia, Calif.) containing 2-mercaptoethanol (the starting buffer for RNA purification), snap frozen, and stored at −80° C. Initially, gene expression analysis was performed on a small subset (22) of the baseline samples. The remaining samples were retained pending the results derived from the in vitro-stimulated samples. Analysis of the entire set of baseline (unstimulated) samples (independent of the analysis provided in this Example 1) is addressed in Example 2.


Example 1.1.3.2
AN1792-Stimulated (Second Daughter) Samples

Cells cultured in media supplemented with AN1792 (10 μg/ml) and a cocktail of immune stimulatory adjuvants consisting of 10 U/ml rhIL-12 (Wyeth, Cambridge, Mass.), 1.5 ng/ml rhIL-2 (R&D Systems, Minneapolis, Minn.), 1.5 ng/ml rhIL-6 (R&D Systems), 10 ng/ml rhIL-7 (R&D), and 10 μg/ml hB7.2 IgG1 (Wyeth). Gene expression analysis was performed on all available samples from this culture condition.


Example 1.1.3.3
Control for AN1792-Stimulated (Third Daughter) Samples—(AN1792 Vehicle-Stimulated)

Cells were cultured under conditions identical to those for the AN1792-stimulated samples except that, as a placebo control, the buffer for AN1792 (10 mM glycine, 10 mM citrate, 5% sucrose, 0.4% PS-80, pH 6.0) was added at the same concentration as in the AN1792-stimulated samples. Gene expression analysis was performed on all available samples from this culture condition.


Example 1.1.3.4
PHA-Stimulated (Fourth Daughter) Samples

Cells were cultured in complete media with 1:150 dilution of Bacto PHA (Phytohemagglutinin P, DIFCO, Becton, Dickinson and Company, BD Biosciences, San Jose, Calif.: 1% solution in 0.85% saline). Gene expression analysis was performed on a small subset (22) of the samples from this culture condition.


Example 1.1.3.5
Control for PHA-Stimulated (Fifth Daughter) Samples—(PHA Vehicle-Stimulated)

Cells were cultured under conditions identical to those for the PHA-stimulated samples except that no PHA was added to the culture. Gene expression analysis was performed on a small subset (22) of the samples from this culture condition.


Example 1.1.4
Cell Harvest and RNA Purification

Nonadherent cells were harvested and pelleted. RLT buffer and 2-mercaptoethanol (350 μl) were added to the flask to allow for the harvest of adherent cells. This suspension was then added to the spun pellet of nonadherent cells. These suspensions were then snap frozen on dry ice and stored at −80° C. RNA purification was performed using QIAshredders and Qiagen RNeasy mini-kits.


Example 1.1.5
RNA Amplification and Generation of GeneChip Hybridization Probe

A probe for hybridization, i.e., biotinylated cRNA, was made from each sample by a two-cycle IVT amplification protocol (with biotinylated nucleotides incorporated during the second cycle). Due to the small amount of sample available, the two-cycle protocol was necessary for generation of sufficient biotinylated cRNA (10 μg of biotinylated cRNA from 50 ng of total RNA) for hybridization. The published Affymetrix two-cycle protocol was followed. Any sample for which the total RNA yield was <50 ng, or which yielded <10 μg of biotinylated cRNA after the IVT amplification reactions was excluded from further processing. Ten μg of biotinylated cRNA from each sample was fragmented to form a hybridization mixture. An eleven member standard curve, comprising gene fragments derived from cloned bacterial and bacteriophage sequences, was also included (spiked) in each hybridization mixture at concentrations ranging from 0.5 pM to 150 pM, representing RNA frequencies of approximately 3.3 to 1000 ppm (see Hill et al. (2001) Genome Biology 2 (12):research0055.1-0055.13). The biotinylated standard curve fragments were synthesized by T7-polymerase-driven IVT reactions from plasmid-based templates. The spiked biotinylated RNA fragments serve both as an internal standard to assess chip sensitivity and as a standard curve to convert measured fluorescent difference averages from individual genes into RNA frequencies in ppm. A reaction mixture (containing biotinylated cRNA and the 11 member standard curve) for each sample was hybridized for 16 hr at 45° C. to the Affymetrix HG-U133A oligonucleotide GeneChip, which interrogates the RNA levels of over 22,000 sequences.


Example 1.2
Materials and Methods—Determination of Expression Patterns
Example 1.2.1
Determination of Gene Expression Frequencies

The hybridization mixtures were removed and stored, and the arrays were washed and stained with streptavidin R-phycoerythrin (Molecular Probes, Inc., Eugene, Oreg.) using GeneChip Fluidics Station 400 (Affymetrix, Inc.) and scanned with a Hewlett Packard GeneArray Scanner (Hewlett Packard, Palo Alto, Calif.) following the manufacturer's instructions. Array images were processed using the Affymetrix MicroArray Suite 5.0 software (MAS 5.0; Affymetrix, Inc.) 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 MAS 5.0. Using the Gene Expression Data System (GEDS) as a graphical user interface, a sample description was provided to the Expression Profiling Information and Knowledge System (EPIKS) Oracle database, and the correct cel file was associated with the description. The database processes then invoked the MAS 5.0 software to create probeset summary values: probe intensities were summarized for each message using the Affymetrix Signal algorithm, and the Affymetrix Absolute Detection metric (Absent, Present, or Marginal, as defined by the MAS 5.0 software) for each probeset. MAS 5.0 was also used for the first pass normalization by scaling the trimmed mean to a value of 100. The database processes also calculated a series of chip QC (quality control) metrics and stored all the raw data and QC calculations back to the database.


Example 1.2.2
Inclusion Criteria for GeneChip Results

The EPIKS database contained all GeneChip results including those that must be excluded from the analysis. Excluded data consist of GeneChip results for: a) samples other than those stimulated in culture with AN1792 or its control, and b) replicate chips. Replicate GeneChip results were generated both when samples were rerun due to QC failure and when replicates were run to assess between-chip variability. To ensure equal weight per sample, only one chip (the last chip run for any given sample) per culture condition per patient sample was used in the analyses. All samples whose chips failed QC specifications were rerun and passed. Therefore no samples were lost to analysis due to GeneChip QC failure. Table 3 lists chip QC inclusion specifications used in this analysis (although other means of quality control for GeneChips or other DNA microarray chips may be used).


Example 1.2.3
Normalization and Filtering of Gene Expression Data

Frequency values for chips meeting inclusion criteria were normalized to control for chip-to-chip differences. The scaled frequency method of Hill et al. ((2001) Genome Biology 2 (12):research0055.1-0055.13) was used. Genes that do not have any relevant information were filtered from the dataset. This occurred in two stages: 1) any gene that was called Absent on all GeneChips (as determined by the Affymetrix Absolute Detection metric in MAS 5.0) was removed from the dataset; and 2) any gene that was expressed at a normalized frequency of <10 ppm on all GeneChips was removed from the dataset to ensure that any gene kept in the analysis set was detected at a frequency of at least 10 ppm at least once. (In previous studies, high variability had been observed in frequency measurements below 10.) The total number of genes in the analysis after these filtering steps were performed was 10,168.


Example 1.2.4
Identification and Reporting of Outlier Samples

To identify outlier samples, we computed the square of the pairwise Pearson correlation coefficients (r2) among all pairs of samples using Splus (Version 5.1) (ITC Computer Systems, University of Virginia). Specifically, we started from the G×S matrix of expression values, where G is the total number of genes and S is the total number of samples. We calculated r2 between all pairs of columns (samples) in this matrix. The result was a symmetric S×S matrix of r2 values (see Weinstein et al. (1997) “An information-intensive approach to the molecular pharmacology of cancer,” Science 275:343-49). This matrix measures the similarity between each sample and all other samples in the analysis. Since all of these samples come from (relatively) elderly human PBMCs treated according to common protocols, the expectation is that the correlation coefficients reveal a high degree of similarity in general (i.e., the expression levels of the majority of the 10,168 transcripts are similar in all samples analyzed). To summarize the similarity of samples, for each sample the average of the r2 values between that sample and the other samples studied in this Example 1 was calculated (Table 4).


The closer the value of average r2 is to 1, the more alike the sample is to the other samples within the analysis. Low average r2 values indicate that the gene expression profile of the sample is an “outlier” in terms of overall gene expression patterns. Outlier status can indicate either that the sample has a gene expression profile that deviates significantly from the other samples within the analysis, or that the technical quality of the sample was inferior. Therefore, the pharmacogenomic supplemental statistical analysis plan of this study stipulated the step of identifying any outliers (average r2 value <0.75) and conducting an analysis of the individual gene expression profile of each outlier. There are a total of seven samples (listed in Table 5) that meet this criterion.


The r2 outlier samples identified in Table 5 include one particularly critical sample: the AN1792-stimulated sample from patient 33. Patient 33 is one of five encephalitis patients. The gene expression profiles of the seven r2 outlier samples were examined, and it was determined that they all contain sequences that are expressed throughout the linear range of the standard curve. None of the samples shows gene expression frequencies either uniformly lower or higher than average. Therefore, it is highly unlikely that the r2 status of these outliers is due to a technical failure of the in vitro transcription (IVT) reactions or other factors related to sample quality.


Example 1.2.5
Merging of Clinical and Gene Expression Data

Relevant clinical data received from StatProbe, Inc. (Ann Arbor, Mich.) (pertaining to treatment group, maximum IgG titer for all visits, maximum IgM titer for all visits, ApoE4 type, and encephalitis status), along with demographic data and treatment group, were merged with the gene expression data by donor identification number (the randomization number that was assigned to each patient in the study).


Example 1.2.6
Samples Analyzed for Gene Expression Levels
Example 1.2.6.1
Sample Inclusion Criteria

Inclusion in the study required 1) that samples arrive at the Pharmacogenomics Laboratory within one day of collection, 2) that culture conditions were within specifications, 3) an RNA yield >50 ng, and 4) an IVT yield >10 μg. Table 6 accounts for all samples received for this Example 1, and identifies the number of patients in this study. Of the 172 enrolled U.S. patients, 167 consented to inclusion in the pharmacogenomic portion of the study. Of the 167 samples, six did not meet shipping specifications, and an additional 12 did not meet culture and storage specifications. Eight samples yielded insufficient product for chip hybridization, and an additional eight samples were removed due to an operator error identified during QC review. Therefore, the total number of AN1792-stimulated samples analyzed in this Example 1 is 133.


Example 1.2.6.2
Demographics of Patients

Sixty-four (64) of the patients in this Example 1 were female and 69 were male. Ages ranged from 53 to 87 years. Patient demographics are shown in Table 7.


The vast majority of patients (86%) were Caucasian. Hispanic (9%), Black (3%), Asian (1%), and unknown (2%) comprised the remainder. Gender representation was balanced within these groups and is shown in Table 8. All five encephalitis patients are Caucasian females born between August 1918 and December 1929.


The pharmacogenomic supplemental statistical analysis plan of this Example 1 defines IgG responders as having a maximum titer≧2200 at any time point. The maximum titer of partial IgG responders was >200<2200, and of nonresponders was ≦200. Patients with an IgM titer>100 at any time point are defined as IgM responders. Table 9 gives a breakdown of study samples by gender, response category, and ApoE type.


Example 1.2.6.3
Overview of Approach to Statistical Analyses (Pharmacogenomic Supplemental Statistical Analysis Plan)

Two approaches, analysis of variance (ANOVA) and signal-to-noise metrics (described below), were used in this Example 1 to identify significant associations between preimmunization gene expression patterns of in vitro stimulated samples and patient antibody response, development of encephalitis, and ApoE4 type. These two approaches were designed to find different types of associations in complex sets of data, and therefore different relationships can be identified by the two methods.


Two types of gene expression metrics were used: the logarithm of the gene frequency of the AN1792-stimulated culture, and the logarithm of the ratio of the gene frequency of the AN1792-stimulated culture to the gene frequency of the control culture for each patient sample. This latter metric is equivalent to the difference between the logarithms of the gene frequencies for the two culture conditions.


Example 1.2.6.3.1
ANOVA

For each gene in the final data analysis set, ANOVA was used to determine whether there is a significant association between the gene frequency metric and 1) antibody response (IgM), 2) antibody response (IgG), 3) ApoE4 type, and 4) development of encephalitis. In the ANOVA analysis, raw p values were adjusted for multiplicity according to the false discovery rate (FDR) procedure of Benjamini and Hochberg ((1995) J. Royal Stat. Soc. B57:289-300), as well as the stepdown bootstrap procedure of Westfall and Young ((1993) Resampling-Based Multiple Testing: Examples and Methods for p-Value Adjustment. John Wiley and Sons, Inc., New York; p. 67). Genes with an FDR of <0.05 are reported. At this threshold, 5% of findings are likely to be false positives. The tables presenting the statistical data also provide the raw (unadjusted) p value for each of these genes. Because it has been reported (Xiao et al. (2002) BMC Genomics 3:28) that the genes identified through the FDR procedure are more likely to be of biological relevance than those identified by the stepdown bootstrap procedure of Westfall and Young, and because the analyses of these data support the same conclusion, the FDR procedure is the focus of the analysis.


Example 1.2.6.3.2
GeneCluster

The GeneCluster application chooses marker genes by a signal-to-noise metric and evaluates them for their association with a given response parameter using a weighted voting algorithm (Golub et al. (1999) Science 286:531-37). Genes are assigned a score, and the 95th percentile scores in randomly permuted data are provided for comparison. Genes with a score greater than that reported in the 95th percentile column for randomly permuted data are reported as showing a significant association with a patient group. The probability of seeing a gene that scores this high by chance is less than 0.05. In cases where the number of genes showing a significant association is greater than 100, only the first 100 genes are reported.


In analyses where no gene shows significance at the 0.05 level by GeneCluster, but ANOVA did identify genes at the 0.05 significance level, the top 50 genes in GeneCluster showing significance at the 0.1 level are reported for the purposes of discussion and comparison with the genesets identified through ANOVA analysis.


Example 1.3
Materials and Methods—Data Analysis
Example 1.3.1
Metrics of Data Submitted for Analysis

For each of the four clinical parameters (IgG response, IgM response, ApoE4 type, and encephalitis outcome), two distinct sets of analyses were done for the cultured samples: analysis using the gene frequency in AN1792-stimulated samples, and analysis using the ratio (fold change of frequency) of the AN1792-stimulated sample and its control-stimulated sample. Two distinct sets of genes were submitted for these two types of analyses: 1) only those genes where the ratio of the maximum gene frequency to the minimum gene frequency is >2 (the metric used for this analysis is the frequency of samples stimulated in culture with AN1792); and 2) all genes that passed the filtering criteria (called Present, and with at least one frequency >10 ppm). The metric for this set of genes is the ratio of the frequency of the AN1792 cultures sample to the frequency for the diluent control sample.


Example 1.3.2
Definitions of Groups Compared
Example 1.3.2.1
Analysis of Association Between Gene Expression Metric and Development of Encephalitis

The five female encephalitis patients were compared to the 44 treated female nonencephalitis patients.


Example 1.3.2.2
Analysis of Association Between Gene Expression Metric and IgG Titer

The 22 treated patients with a maximum IgG titer≧2200 (responders) were compared to the 60 treated patients with a maximum IgG titer≦200 (nonresponders). (Data from patients with maximum titers between 200 and 2200 were not used in the identification of statistically significant associations, but were analyzed once the statistical programs had identified genes of interest.)


Example 1.3.2.3
Analysis of Association Between Gene Expression Metric and IgM Titer

The 81 treated patients with a maximum IgM titer>100 (responders) were compared to the 27 treated patients with a maximum IgM titer<100 (nonresponders).


Example 1.3.2.4
Analysis of Association Between Gene Expression Metric and ApoE4

All patients (treated and placebo) for which ApoE4 typing is known (104 patients) were included in this analysis. The 70 ApoE4 positive patients (homozygous and heterozygous) were compared to the 34 ApoE4 negative patients.


Example 1.4
Results—Gene Expression Association with Encephalitis
Example 1.4.1
Gene Expression Levels Showing Association with Encephalitis Using the Metric of Gene Frequency in AN1792-Stimulated Cultures

The logarithm of the gene frequency of the AN1792-stimulated culture was calculated for each gene for each of the five female encephalitis patients and each of the 44 female nonencephalitis patients receiving immunotherapy. ANOVA and GeneCluster analyses were conducted comparing these two groups.


Example 1.4.1.1
ANOVA

In the ANOVA analysis of the frequencies of genes in the AN1792-stimulated samples, 118 probesets had an association with encephalitis with a false discovery rate (FDR)<0.05. The unadjusted p values for these genes with FDR<0.05 ranged from 0.000001 to ≦0.0006. These 118 probesets represent 96 genes of known function and 17 sequences whose functions are not yet known. The balance (five probesets) represents genes tiled more than once on the U133A chip, and thus identified more than once by ANOVA. The 113 genes associated with encephalitis by ANOVA with FDR<0.05 are listed in alphabetical order in Table 10.


Example 1.4.1.2
GeneCluster Analysis

Using GeneCluster, genes with elevated expressions most closely associated with encephalitis were identified, and 162 of these genes had a permutation-based p value <0.05. None had a permutation-based p value <0.01. The narrow range of permutation-based p values for the 162 genes identified (>0.01, <0.05) reflects the small sample size of the encephalitis group and the similarity in expression patterns of a large number of the genes identified (discussed in more detail below). The 100 genes with the top scores in GeneCluster for association between increased expression and encephalitis (out of the aforementioned 162 genes) are shown in Table 11.


Using GeneCluster, no gene whose decreased expression was closely associated with encephalitis had a permutation-based p value <0.05, although there were a large number of genes that just missed this cutoff. However, the results indicate that there are genes associated with decreased expression levels in encephalitis both by ANOVA (FDR<0.05) and by GeneCluster (if the GeneCluster permutation-based p value criterion is relaxed to <0.1). For the purposes of discussion and for comparison with ANOVA, therefore, the list of genes selected by GeneCluster as associated with a decreased level of expression in encephalitis patients (permutation-based p value <0.1) were compiled and analyzed. The 50 genes most closely associated with decreased levels of expression in encephalitis patients (all of which met the permutation-based p value <0.1 criterion) are shown in Table 12.


Example 1.4.1.3
Comparison of Genes Identified through ANOVA and GeneCluster Analyses

To assess the overlap in the list of genes identified by ANOVA and GeneCluster, the list of 113 genes identified by ANOVA with FDR<0.05 (Table 10) was compared to the lists of genes associated with encephalitis by GeneCluster analyses. Of the 200 genes identified in GeneCluster as most closely associated with elevated levels of expression in encephalitis patients, 59 overlapped with the 68 genes identified by ANOVA as having elevated levels of expression in encephalitis patients and FDR<0.05. Of the 200 genes identified in GeneCluster as most closely associated with decreased levels of expression in encephalitis patients, 44 overlapped with the 45 genes identified by ANOVA as having decreased levels of expression in encephalitis patients and FDR<0.05. By this method of assessing overlap, therefore, 91% (103 out of 113) of the most significant genes identified by ANOVA analysis were also selected by the GeneCluster application.


Example 1.4.1.4
Expression Patterns of Genes Associated with Encephalitis by ANOVA and GeneCluster

A detailed examination of the expression patterns of the genes listed in Tables 10, 11, and 12 reveals relevant information that is not apparent through mere survey of the p values. First, the gene expression profiles of the five encephalitis patients appear to fall into two fairly distinct patterns. The expression profiles of encephalitis patients 19, 33, and 503 are more similar to each other than they are to the profiles of encephalitis patients 299 and 301. In addition, the profiles of patients 19, 33 and 503 deviate from normal more often than those of patients 299 and 301. For approximately 73% of the genes shown in Table 10, at least three encephalitis patients (usually patients 19, 33, and 503) express at levels associated with encephalitis. Examples of this expression pattern are shown in FIGS. 4-9. (For many of the remaining 27% of genes listed in Table 10, abnormal gene expression levels were observed in only one or two of the encephalitis patients. These genes are not addressed further.)



FIG. 4 shows the expression pattern of TPR in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described in materials and methods (Example 1.1.3). TPR, translocated promoter region, also called tumor-potentiating region, has been implicated in oncogenesis involving the met oncogene. For this figure, as for subsequent figures (FIGS. 5-13), the following description applies. Frequency values are reported as ppm. The horizontal line represents the geometric mean frequency for that group. The vertical lines separate patient groups. The seven patients groups are: 1) female encephalitis patients, 2) immunized IgG titer negative (i.e., maximum titer≦200) females, 3) immunized female patients with maximum IgG titer>200<2200, 4) immunized female patients with maximum IgG titer≧2200, 5) immunized IgG titer negative (i.e., maximum titer≦200) males, 6) immunized male patients with maximum IgG titer>200<2200, and 7) immunized male patients with maximum IgG titer≧2200. The open circles represent absent calls; the closed circles represent present calls. Note the high probability of false absent calls; an increased number of false negative calls (transcripts called absent when actually present) results from the extreme 3′ bias introduced by the two-round IVT protocol. Due to the small amounts of sample available, the two-round IVT protocol was necessary.



FIG. 5 shows the expression pattern of NKTR in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described above in Example 1.1.3. NKTR, natural killer tumor recognition sequence, also known as natural killer triggering receptor, is involved in the activation of the innate immune system.



FIG. 6 shows the expression pattern of XTP2 in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described above in Example 1.1.3. XTP2, HbxAg transactivating protein 2, is thought to be implicated in cell activation events associated with hepatitis B virus infection.



FIG. 7 shows the expression pattern of SRPK2 in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described above in Example 1.1.3. SRPK2, SFRS protein kinase 2 (protein kinase, arginine/serine splicing factor 2), has been implicated in posttranscriptional regulation of gene expression.



FIG. 8 shows the expression pattern of THOC2 in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described in Example 1.1.3. THOC2, THO complex 2, has been implicated in the control of gene transcription.



FIG. 9 shows the expression pattern of PSME3 in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described in Example 1.1.3. PSME3, proteasome activator subunit 3, is a subunit of the protease responsible for the generation of peptides loaded onto MHC class I molecules.


Four of the encephalitis patients (usually patients 19, 33, 299 and 503) express 23% of the genes listed in Table 10 at levels associated with encephalitis. Patient 301 is much less clearly distinguishable from nonencephalitis patients by gene-expression profile. A total of 14 (12%) of the genes listed in Table 10 are expressed by all five patients at levels associated with encephalitis. However, the expression levels associated with encephalitis for these 14 genes are less distinct between the encephalitis and nonencephalitis groups than for genes that capture only three or four of the encephalitis patients. These 14 genes are listed in Table 13. Examples of the expression patterns for four of these genes are shown in FIGS. 10 through 13.



FIG. 10 shows the expression pattern of DAB2 in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described in Example 1.1.3. DAB2, disabled homologue 2, mitogen-responsive phosphoprotein, competes with SOS for binding to GRB2 and thus is implicated in control of growth rate.



FIG. 11 shows the expression pattern of SCAP2 in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described in Example 1.1.3. SCAP2, src family-associated phosphoprotein 2, is an adaptor protein thought to play an essential role in the src-signaling pathway.



FIG. 12 shows the expression pattern of furin in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described in Example 1.1.3. Furin is a processing enzyme involved in activation of TGF1, an anti-inflammatory cytokine.



FIG. 13 shows the expression pattern of CD54 (ICAM1) in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described in Example 1.1.3. CD54, intracellular adhesion molecule 1 (ICAM1), is a ligand for lymphocyte function-associated antigens and is involved in response to antigen.


As encephalitis patient 301 expresses only 12% of the genes listed in Table 10 at levels associated with encephalitis, the expression profile of this patient can be considered more “normal” than the profiles of the other encephalitis patients. Of the five encephalitis patients, patient 33 expressed the most genes (105 of 113) listed in Table 10 at levels associated with encephalitis. The ranking of encephalitis patients in terms of most genes expressed at levels associated with encephalitis is: 33, 19, 503, 299 and 301.


A second trend in gene expression profiles that is not apparent through survey of the statistical associations emerges from the examination of the expression levels of genes associated with encephalitis in individual AN1792 nonencephalitis patients. Data from males were not used to identify genes associated with encephalitis, because all the encephalitis patients in the study were female and the comparator group used was the 44 female AN1792 nonencephalitis patients. Although all the encephalitis patients were female in this study, it is not believed at this time that gender plays a role in predicting whether a patient will develop encephalitis, because in the European Phase IIA clinical trials several males developed encephalitis. Examination of the profiles in males, therefore, offers an opportunity to assess whether samples that were not used to identify associations with encephalitis have profiles consistent with those identified through analysis of the female samples. Table 14 depicts the level of agreement in terms of gene expression profile and clinical diagnosis of encephalitis when the data are analyzed with the inclusion of male nonencephalitis patients (Table 14 is discussed further below in Example 1.8.1).


Using the genes that capture the three most severe encephalitis patients (19, 33, and 503), the false positives are restricted to a few (three or four) patients, and it is often the same three or four patients captured. IgG nonresponding male patients 252 and 752, and partial responding female patient 8 (maximum IgG titer 208) express many of the genes most closely associated with encephalitis at or close to the levels associated with encephalitis. As seen in Table 14 and discussed above, genes that capture all five encephalitis patients also capture an increased number of nonmeningoencephalitic patients, and IgG responders are among the nonencephalitis patients captured. (For example, patients 5, 12, 32, 508, and 755 are IgG responding nonencephalitis patients who express some genes at levels associated with encephalitis.) Another set of genes is the set consisting of the three genes that correctly classify 60% of the encephalitis developer patients and incorrectly classify 4% of the encephalitis nondeveloper patients (i.e., SRPK2, TPR, and NKTR). Another set of genes is the set consisting of the three genes that correctly classify 100% of the encephalitis developer patients, and incorrectly classify 25% of the encephalitis nondeveloper patients (i.e., SCAP2, PACE (furin), and DAB2). Another set of genes is the set consisting of SRPK2, TPR, NKTR, SCAP2, PACE (furin), and DAB2.


Example 1.4.1.5
Comparison of Gene Expression Patterns in AN1792-Stimulated and Control Cultures

The identification of gene expression patterns associated with encephalitis in cultures stimulated with AN1792 raised the question of whether in vitro stimulation with AN1792 was required for detection of encephalitis-associated gene expression patterns. To answer this question, the expression patterns in control cultures of the genes associated with encephalitis by the metric of gene frequency in AN1792-stimulated cultures were analyzed. Table 15 shows the association between encephalitis and the metric of frequency in control cultures for 23 of the genes most closely associated with encephalitis by the metric of frequency in the control cultures (for genes that are also shown in Table 10).


This result indicates that detection of statistically significant associations between preimmunization gene expression and postimmunization development of encephalitis may not require in vitro stimulation with AN1792. Of the 113 genes associated with encephalitis using the metric of gene frequency in AN1792-stimulated cultures, 64 genes also show an association using the metric of gene frequency in control cultures (setting the cutoff at raw p<0.005 (FDR<0.18)). The detection of the association with encephalitis in both the AN1792-stimulated and control cultures is evidence both that the associations can be detected without in vitro exposure to AN1792 and that, since the associations have been detected in two sets of samples, the associations have sound technical and statistical support.


The analysis of the control cultures also reveal genes that, whereas associated with encephalitis using the AN1792-stimulated culture frequency metric, show absolutely no association using the metric of frequency in control cultures. The 12 most extreme examples of this gene expression pattern are shown in Table 16. Note that two of the genes in Table 16, PSMF1 and TAP2, are functionally related to antigen processing.


Example 1.4.2
Using the Metric of Ratio of the Frequency in AN1792-Stimulated Samples to the Frequency in Control Culture Samples to Identify Gene Expression Levels with Association to Encephalitis

The logarithm of the ratio of the gene frequency of the AN1792-stimulated culture to the gene frequency of the control was calculated for each gene for the five female encephalitis patients and the 44 treated nonencephalitis female patients. This is equivalent to the difference between the logarithms of the gene frequencies for the two culture conditions.


Example 1.4.2.1
ANOVA

By this ratio metric, ANOVA found no association with encephalitis with FDR<0.05. The lowest (best association) was FDR=0.104, and there were five genes at this FDR value. This result indicates that the association found by ANOVA did not reach the level of statistical significance (0.05) stipulated in the pharmacogenomic supplemental statistical analysis plan of this study. This finding is consistent with the result (noted above) indicating the detection of strong associations between encephalitis and gene expression levels in control (i.e., without AN1792) stimulated cultures.


Example 1.4.2.2
GeneCluster Analysis

By the ratio metric, GeneCluster identified 13 genes that were associated with encephalitis with a permutation-based p value <0.05. The permutation-based p value was >0.01 for all 13 genes listed. These 13 genes, along with their associated raw (unadjusted) p and FDR values by ANOVA, are shown in Table 17. For all genes listed, AN1792 stimulation resulted in a decrease in gene expression frequency. Note that the associations with encephalitis detected using the ratio metric are much weaker (both by ANOVA and GeneCluster) than the associations detected using the frequency metric, again indicating that exposure to antigen (AN1792) in vitro may play a minor role in revealing the associations between gene expression and postimmunization development of encephalitis.


Example 1.5
Results—Gene Expression Association with IgG Responsiveness
Example 1.5.1
Gene Expression Levels Showing Association with IgG Responsiveness Using the Metric of Gene Frequency in AN1792-Stimulated Cultures

The goal of the search for correlates with antibody response was to identify markers that would allow the preimmunization identification of likely nonresponders in what was, at the onset of this study, a planned Phase III study. If the incidence of nonresponders could be lowered through a prescreening test, the power of the clinical trial could be increased.


Example 1.5.1.1
ANOVA

ANOVA was performed by comparing data from the 60 nonresponders (maximum titer≦200) to the 22 IgG responders (maximum titer≧2200) and the 60 nonresponders to the 26 IgG partial (or low) responders (maximum IgG titer>200 and <2200). ANOVA identified 375 genes associated with IgG responsiveness with FDR<0.05 (raw p<0.000919). These data indicate numerous statistically significant differences between IgG responders and nonresponders in the preimmunization PBMC gene expression profiles. However, this number of genes far exceeds the number required to reach the goal of identifying a small geneset associated with likely nonresponsiveness; thus, Table 18 lists only the 15 genes associated with IgG responsiveness by ANOVA with FDR<0.011. The adjusted p values (by Westfall and Young stepdown bootstrap procedure for multiplicity adjustment) for these 15 genes are also shown in Table 18. Note that 11 of the genes listed show an association with IgG response with adjusted p≦0.05.


Example 1.5.1.2
GeneCluster Analysis

By GeneCluster analysis, more than 500 genes showed an association between gene expression level and IgG response at the 0.01 level of significance. For a more focused analysis, genes associated with a permutation-based p value <0.00005 were selected. (This significance level indicates that the GeneCluster score for the gene is higher than observed in the top 0.005 percentile of randomly permuted data.) At this extremely stringent level of significance, four genes showed association with IgG response. These were granulin, FC fragment of IgG receptor transporter alpha (FCGRT), isoleucine-tRNA synthetase (IARS), and minichromosome maintenance, S. cerevisiae homolog 3 (MCM3). These four genes were also among the 11 most significant associations identified through ANOVA (see Table 18). The gene expression frequencies of the four genes significant at the 0.00005 level by GeneCluster analysis are shown in FIGS. 14-17 for each of the patients in the analysis.



FIG. 14 shows the gene expression levels of IARS, isoleucine-tRNA synthetase, (in individual patients by response group) in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described in Example 1.1.3. For this figure, as for subsequent figures (FIGS. 15-17), the following description applies. Frequency values are reported as ppm. The horizontal line represents the geometric mean frequency for that group. IgG nonresponders: maximum titer≦200; partial IgG responders: maximum titer>200<2200; IgG responders: maximum titer≧2200.



FIG. 15 shows the gene expression levels of FCGRT, Fc fragment of IgG receptor transporter alpha, in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described in Example 1.1.3.



FIG. 16 shows the gene expression levels of granulin in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described in Example 1.1.3.



FIG. 17 shows the gene expression levels of MCM3 (thought to be involved in the DNA replication process) in samples stimulated overnight in culture with AN1792 and the cocktail of immune adjuvants described in Example 1.1.3.



FIGS. 14 through 17 show that, for the set of samples in this study at least, nonresponsiveness is associated with high expression levels of granulin and FCGRT and low expression levels of IARS and MCM3. Expression levels of partial responders (maximum IgG titer>200<2200) are intermediate between nonresponders and responders.


Increasing the permutation-based p value from 0.00005 (four genes identified) to 0.00007 in GeneCluster results in an increase of 226 in the number of genes identified. The large number of genes identified at the 0.00007 level of significance (also an extremely stringent criterion) reflects numerous differences in gene expression between the IgG responder and nonresponder groups. Of the 230 genes identified in GeneCluster at the 0.0007 significance level, 217 were also identified as associated by ANOVA, indicating a high concordance between the genes identified by the two applications. This level of concordance is similar to that observed for the associations identified between gene expression profiles and encephalitis.


Example 1.5.1.3
Correlation Between Expression Levels and IgG Response Group

The data in Table 18 and FIGS. 14-17 suggest that preimmunization gene expression profiling has the potential to identify a fraction of the population least likely to respond. Therefore, using the four genes identified by GeneCluster analysis, the correlation between expression levels and IgG response groups was assessed. Table 19 shows the correlation between expression pattern and IgG responsiveness of the individual genes for the four genes identified by GeneCluster analysis.


Example 1.5.2
Using the Metric of Ratio of the Frequency in AN1792-Stimulated Samples to the Frequency in Control Culture Samples to Identify Gene Expression Levels With Association to IgG Responsiveness

ANOVA and GeneCluster analyses were run using the metric of the ratio of gene frequency values in AN1792-stimulated cultures to gene frequency values in control cultures. Neither analysis revealed association that met the 0.05 significance level cutoff. These data indicate that the associations found using the gene frequency metric were not dependent on in vitro stimulation with AN1792.


Example 1.6
Results—Lack of Association Between Gene Expression Pattern and IgM Responsiveness

ANOVA and GeneCluster analyses were performed comparing treated IgM responders and nonresponders. Both the metric of gene frequency in AN1792-stimulated samples and the metric of the ratio of the gene frequency of the AN1792-stimulated culture to the gene frequency of the control were used in these analyses. No association was found in which FDR<0.05 (ANOVA) or permutation-based p value <0.05 (GeneCluster).


Example 1.7
Results—Lack of Association Between Gene Expression Pattern and the Presence of the ApoE4 Allele

ANOVA was performed comparing gene expression patterns of ApoE4 homozygous, ApoE4 heterozygous, and ApoE4 negative patients. Data from both treated and placebo patients were included in this analysis. GeneCluster analysis was performed comparing ApoE4 negative patients to ApoE4 positive patients. Both the metric of gene frequency in AN1792-stimulated samples and the metric of the ratio of the gene frequency of the AN1792-stimulated culture to the gene frequency of the control were used in these analyses. No association was found that met the 0.05 level of significance. In fact, the two top scoring genes detected by GeneCluster were gender-specific genes encoded by the Y chromosome. The identification of Y chromosome-encoded genes reflects the fact that there are 12 more males than females in the ApoE4 negative group (see Table 9). Therefore, no significant correlation between gene expression pattern in PBMCs and ApoE4 type was detected by this study.


Example 1.8
Discussion
Example 1.8.1
Gene Expression Patterns Associated with Encephalitis

Based on the evidence showing strong associations between preimmunization gene expression patterns and postimmunization development of encephalitis, there is evidence to suggest that certain genes may be associated with development of encephalitis. These results can be viewed as providing a basis for the formulation of hypotheses that may help explain why some patients were susceptible to the development of encephalitis. The data are consistent with the hypothesis that the patients who developed encephalitis were predisposed to do so because some pathways related to immune function were in a state of increased activation. In assessing this information on gene expression profiles associated with encephalitis, it should be noted that the sample set of five encephalitis patients is extremely small and contains considerable diversity. Also, as treatment was halted after two or three immunizations, it is not known whether other patients would have developed encephalitis had immunizations continued. For example, speculation on the data in Table 14 could either favor the interpretation that gene SCAP2 incorrectly groups 11 of 103 treated nonencephalitis patients with the encephalitis group, or that these 11 additional patients may be at increased risk of developing encephalitis. This study does not provide sufficient data to distinguish between these possibilities. It is also possible that increased risk of encephalitis is correlated with a gene expression profile that requires some combination of genes to be expressed at levels associated with encephalitis. However, as noted above, regardless of the interpretation, the analysis would result in the prediction that certain nonencephalitis-prone patients would likely develop encephalitis, rather than the prediction that encephalitis-prone patients would not get encephalitis. Because the goal of the present invention is to ensure that patients at risk of encephalitis be identified in order to avoid an adverse reaction to immunotherapy and to provide a targeted therapeutic for AN1792, excluding a small percentage of patients that would otherwise be good candidates is within the goal of the present invention.


The results disclosed here do suggest that certain gene expression patterns may be useful in preimmunization assessment of the relative risk of encephalitis. The number of genes associated with FDR<0.05 is large (113 genes), and there is variation among these genes with respect to both the number of encephalitis patients that express at levels associated with encephalitis, and the number of nonencephalitis patients that express at levels associated with encephalitis. Therefore, as an illustrative example, or exercise, regarding the potential for using these data to classify patients, three criteria for inclusion on a selected list of six encephalitis-association genes useful in classification were set; inclusion on the list required meeting either the first and third criteria or the second and third criteria. The first criterion was belonging to the group of genes that capture three of the five encephalitis patients (see, e.g., FIGS. 4-8). The second criterion was belonging to the group of genes that capture all five encephalitis patients (see, e.g., FIGS. 10, 11 and 13). The third criterion was belonging to the group of genes for which statistically significant associations with encephalitis have been observed both in AN1792-stimulated and control cultures (see Table 15). This last criterion increases the likelihood that genes with true associations are selected by requiring that both sets of data pass a rigorous statistical filter.


Using these criteria for inclusion on the list of genes with potential as “risk assessment genes,” the genes TPR, NKTR, XTP-2, and SRPK2 are examples of genes that were included because they met the first and third criteria. DAB2 and SCAP2 are examples that were included because they met the second and third criteria. A list of genes containing these six genes only results in the accurate classification of five out of five encephalitis patients, and incorrectly classifies about 25-30% (depending on the cutoff) of nonencephalitis patients (see also Table 14).


ASRGL1 is the gene most closely associated with encephalitis by ANOVA (see Table 10), and also shows an extremely strong association by GeneCluster analysis (see Table 11). Inclusion of this single gene on the list of potential “risk assessment genes” would raise the misclassification rate among nonencephalitis patients to about 40%. However, as noted in the footnotes to Table 14, the preponderance of the misclassified patients are male. (With a cutoff of F>20, 100% of the patients misclassified by this gene are male. With a cut-off of F>12, 64% of the misclassified patients are male.) To a great extent, two facts explain the high false positive rate when ASRGL1 is included in the set of genes used for risk assessment: (1) that data from female patients only was used to calculate the strength of the association with encephalitis, and (2) that high levels of expression in nonencephalitis patients are strongly associated with being male. These issues call into question the true strength of the association between ASRGL1 and encephalitis. Three possibilities regarding why high levels of ASRGL1 are extremely strongly associated with encephalitis in females but not in males are: (1) the data reflect a true gender difference, (2) identification of ASRGL1 is a false positive (noting that the FDR<0.05 cutoff allows for the false identification of about six genes), and (3) the association exists but is much less strong than when calculated excluding males.


The findings by GeneCluster are consistent with the findings by ANOVA in that both show numerous differences in gene expression between the meningoencephalitic and nonmeningoencephalitic groups. Genes selected by ANOVA are not expected to be identical to genes selected by GeneCluster due to the differences in algorithms used to select the genes and the nonequivalent methods of calculating p values. However, it is of interest to compare the lists of genes identified by ANOVA and GeneCluster because the level of overlap between the gene lists gives both an indication of the robustness of the methods and an understanding of differing weights given to pattern recognition by each of the approaches. GeneCluster places greater weight than ANOVA on the requirement that all five encephalitis patients group together with respect to the expression frequency of the identified gene. ANOVA places greater weight than GeneCluster on outliers (compared to nonencephalitis patients) even if only one or two of the encephalitis patients express at levels deviant from normal. Therefore, as a result of the different algorithms used by the two applications, both applications identify as associated with encephalitis genes where three of the five encephalitis patients express at levels outside the normal range, but ANOVA will tend to identify the encephalitis association more strongly than will GeneCluster. GeneCluster, on the other hand, will rank more highly genes that are expressed at similar levels by all five encephalitis patients, even if the average expression level in encephalitis patients falls at the outer limits of the range within normal patients.


Many of the statistically significant associations between gene expression patterns that were observed in the gene frequencies in cultures stimulated in vitro with AN1792 were also observed in control cultures that were not exposed to AN1792. This result indicates that detection of many aspects of the gene expression profile associated with a predisposition to the development of encephalitis does not require in vitro exposure to AN1792. This conclusion is also consistent with the results using the ratio metric (fold change in frequency in AN1792-stimulated cultures as compared with control cultures). The ratio metric revealed no association meeting the FDR<0.05 level by ANOVA, and the associations revealed by GeneCluster were much less robust than those identified using the frequency metric.


Example 1.8.2
Biological Pathways Associated with Encephalitis

Caution must be exercised in drawing conclusions on biological mechanisms based solely on gene expression profiles. The gene expression profiles of the encephalitis patients indicate that these patients may be prone to process and react differently to antigen. Examination of the expression levels of ICAM1 (FIG. 13), PSME3 (FIG. 9) and XTP2 (FIG. 6) illustrate this point. There may also be differences in the innate immunity pathway (see FIG. 5 for the NKTR expression profile).


Many of the genes showing the most significant association with encephalitis are functionally related to the control of transcription. The identified differences in gene expression patterns could therefore be the result of activation (or deactivation) of genes under common transcriptional control. This interpretation fits with the observation that certain genesets show a consistent pattern in certain patients (for example patients 8, 19, 33, 252, 503, and 752), hinting that these genesets are behaving as a correlated set in a small number of patients. This type of correlation is well recognized in gene expression analysis, and is factored in the algorithms used by GeneCluster.


There is also some suggestion within the data that patients that express a significant number of genes at levels associated with encephalitis may be at reduced risk if they do not develop a significant IgG titer (≧2200). Patients 8, 252 and 752 fall into this category. This hypothesis fits with the clinical information that, whereas IgG responders most often do not develop encephalitis, those patients who do develop encephalitis are likely to have significant IgG titers.


The genes identified as associated with encephalitis by the ratio metric of frequency in AN1792-stimulated cultures to frequency in control cultures are functionally related to immune function including response to cytokines, control of apoptosis and chemotaxis, signal transduction and control of proliferation. These data are consistent with a difference between nonencephalitis and encephalitis patients in terms of immune system response to exposure to AN1792, but the associations found are relatively weak.


Example 1.8.3
Gene Expression Profile Associations with IgG Nonresponsiveness

Both GeneCluster and ANOVA indicate that there are numerous statistically significant differences between the preimmunization gene expression profiles of IgG responders and nonresponders. These numerous differences may be a reflection of a few different biological pathways being activated in the two groups. This kind of difference can result in activation and deactivation of genes that are under common transcriptional control and consequently behave as correlated sets. This type of correlation is well recognized in gene expression analysis, and is factored in the algorithms used by GeneCluster. Many of the genes showing the most significant association with IgG nonresponsiveness are functionally related to the control of transcription.


The association between high levels of FCGRT with IgG nonresponsiveness is an intriguing finding. This gene is believed to function in the transport of IgG in some forms of immunity. The association of low levels of IARS with nonresponsiveness is another fascinating and unexpected finding. The autoimmune diseases polymyositis and dermatomyositis are a consequence of autoantibodies directed against one or more of the aminoacyl-tRNA synthetases with subsequent lymphocytic destruction of myocytes. Six of 20 human aminoacyl-tRNA synthetases have been identified as targets in these autoimmune diseases. In light of this information, the association identified in this study between low levels of IARS and IgG nonresponsiveness suggests that high levels of IARS may be associated with hyperresponsiveness, and the destruction observed in autoimmune disease might be an adaptive response aimed at controlling high activity of this gene. The MCM3 gene is thought to be involved in DNA replication. Thus it is possible that the gene may function in the replication of lymphocytes known to be necessary for T and B cell responses. Low levels of this gene are associated with nonresponsiveness, a finding consistent with the hypothesis that this gene functions in the proliferative phase of the in vivo immune response.


No gene associated with IgG responsiveness was identified by the ratio metric of frequency in AN1792-stimulated cultures to frequency in control cultures. This finding indicates that the gene expression patterns associated with IgG responsiveness are intrinsic characteristics of the patients that do not depend for detection on in vitro exposure to AN1792. It is possible, therefore, that the gene expression profiles associated with IgG responsiveness in this study are general surrogate markers for the ability to respond to immunotherapy. Such markers have not been identified, and these findings, if validated, could help in understanding the incidence of immunotherapeutic nonresponsiveness in general, and especially in the elderly.


No statistically significant association was found between gene expression profiles and IgM response, although the same trend that is statistically significant in the IgG analysis is detectable in the IgM analysis (but does not reach statistical significance). For example, the four highest expressors of FCGRT are IgM nonresponders and IgG nonresponders. The same is true for the four highest expressors of granulin and the five highest expressors of CST3.


Example 1.9
Conclusions

By ANOVA and GeneCluster analyses, statistically significant associations have been detected between the gene expression profiles of PBMCs of patients prior to immunization with AN1792 and the postimmunization development of encephalitis. In addition, statistically significant associations were found between the preimmunization gene expression profile in PBMCs and postimmunization development of IgG response.


No statistically significant associations were found between gene expression profiles and either IgM response or ApoE4 type. For many of the genes associated with IgG responsiveness, however, a similar trend is present in the comparison of IgM responders and nonresponders, but the trend does not reach statistical significance for a single gene.


Example 2
Association of Gene Expression Profiles of Unstimulated Samples with Either Favorable or Adverse Clinical Responses
Example 2.1
Materials and Methods—Sample Collection and Preparation
Example 2.1.1
Sample Collection

Consent to the pharmacogenomic portion of the study was optional and obtained after approval by local institutional review boards in the U.S. (E.U. patients were not included in the pharmacogenomic study). All gene expression analyses were conducted on RNA purified from peripheral blood mononuclear cells (PBMCs) collected prior to immunization. Blood samples were collected from consenting subjects at the screening visit (between 9 and 54 days prior to the first immunization) and were shipped overnight at room temperature to the Clinical Pharmacogenomic Laboratory at Wyeth Research in Andover, Mass., and PBMCs were purified as described in Examples 1.1 and 1.1.1 above (see also Burczynski et al. (2005) Clin. Cancer Res. 11:1181-89). CPT purification resulted in greater than 99% reduction in RBC representation in all 153 study samples, and CPT purification did not alter by more than 15% the percentage of monocytes relative to PBMCs. The efficiency of removal of neutrophils by CPT fractionation is shown in FIG. 2 and discussed in Example 1.1.1 (see also Table 2; see generally Example 1.1.3.1). A fraction of the PBMCs (2×106 cells) was pelleted and frozen on dry ice for the isolation of RNA samples. The remaining PBMCs were consigned to in vitro studies (described in Example 1).


Example 2.1.2
Sample Preparation: RNA Purification

The purified PBMC fraction was pelleted by centrifugation, resuspended in 300 μl RLT Buffer (Qiagen, Valencia, Calif.) containing 2-mercaptoethanol (the starting buffer for RNA purification), snap frozen and stored at −80° C. prior to gene expression analysis. RNA was purified using QIA shredders and Qiagen RNeasy® mini-kits. In particular, labeled targets for oligonucleotide arrays were prepared using 50 ng of total RNA. Biotinylation of cRNA (generated using two-cycle IVT amplification), hybridization to the HG-U133A Affymetrix GeneChip Array®, and conversion of signal values to normalized parts per million (Hill et al. (2001) Genome Biol. 2:research0055.1-0055.13) are described below. Data for 9,678 probesets that were called ‘present’ and with frequency ≧10 parts per million in at least one of the samples were subjected to the statistical analyses described below, while probesets that did not meet these criteria were excluded. SAS was used for all analyses unless otherwise noted.


Example 2.1.3
Sample Preparation: Microarray Targets Labeling

Labeled cRNA for hybridization to microarrays was prepared using a two-round in vitro transcription (IVT) amplification procedure. The two-round procedure was necessary because the RNA yield (from 2×106 starting PBMCs) was less than 1 μg in some cases. Total RNA was converted to 1st strand cDNA by priming with 40 pmol of T7-(dT)24 primer (Genset Corp). Primer and total RNA were incubated at 70° C. for 10 minutes and then held at 50° C. until the addition of first-strand buffer [250 mM Tris-HCl (pH 8.3), 375 mM KCl, 15 mM MgCl2], 10 mM DTT, 500 μM each of dNTP mix, and 40 U RNAseOUT (all from Invitrogen). Samples were then incubated at 50° C. for 2 minutes followed by the addition of the 200 U of SuperScript™ II Reverse Transcriptase (Invitrogen) and incubation at 50° C. for 1 hour.


Double-stranded cDNA was synthesized by incubating the 1st strand cDNA at 16° C. for 2 hours with second-strand buffer plus, 200 μM of each dNTP, 10 U of E. coli DNA ligase, 40 U of E. coli DNA Polymerase I, 2 U of E. coli Rnase H, (all from Invitrogen), and DEPC-treated water (Ambion) to a final volume of 150 μl. Six units of T4 DNA Polymerase (BioLabs) were then added and samples were incubated for 5 minutes at 16° C. The reaction was stopped by the addition of 20 mM EDTA (Ambion), and samples were placed on ice.


Using paramagnetic beads (Polysciences, Inc.) and a 3-in-1 magnetic particle separator (CPG, Inc), cDNA was purified by solid-phase reversible immobilization (DeAngelis et al. (1995) Nucleic Acids Res. 23:4742-43). Purified cDNA (10 μl) was transcribed into nonlabeled cRNA in an IVT reaction in 0.8×IVT buffer (Ambion), 2.9 mM each of rNTP mix (Amersham), 40 U of RNase Inhibitor (Ambion), 4.3 mM DTT (Invitrogen), 450 U T7 Polymerase (Epicentre) and DEPC-treated water (Ambion) to a final volume of 35 μl and incubation at 37° C. for at least 16 hours.


The nonlabeled cRNA was purified using the Qiagen RNeasy® Mini Kit and RNA cleanup protocol (according to manufacturer's protocol). For the second round of amplification, samples were lyophilized to 10 μl. cRNA was then reverse-transcribed into cDNA using 150 ng of random hexamer (Wyeth) at 70° C. for 10 minutes, and then held at 50° C.


First strand cDNA synthesis for the second IVT procedure was performed in first strand buffer [250 mM Tris-HCl (pH 8.3), 375 mM KCl, 15 mM MgCl2], 10 mM DTT, 500 μM of each dNTP mix, and 40 U RNAseOUT (all from Invitrogen) with incubation at 37° C. for 2 minutes followed by addition of 200 U SuperScript™ II Reverse Transcriptase (Invitrogen) to a final volume of 20 μl. Synthesis was completed at 37° C. for 1 hour. Two units of E. coli RNase H (Invitrogen) were added and the mixture was incubated at 37° C. for 20 minutes and 95° C. for 2 minutes, and then chilled on ice. Samples were then primed with 20 pmol of T7-(dT)24 Primer (Genset Corp.) at 70° C. for 10 minutes and chilled on ice.


Second strand cDNA synthesis for the second IVT procedure was initiated using second-strand buffer plus, 200 μM each of dNTP, 40 U of E. coli Polymerase I, 2 U of E. coli RNase H, (all from Invitrogen) and DEPC-treated water (Ambion) to a final volume of 150 μl, and incubated at 16° C. for 2 hours. Six units of T4 DNA polymerase (BioLabs) were added and sample was incubated for 5 minutes at 16° C. The reaction was stopped by addition of 20 mM EDTA (Ambion) and samples were placed on ice. cDNA was purified by binding paramagnetic beads as described above. Second-round purified cDNA (10 μl) was transcribed into biotin-labeled cRNA by IVT using 1×IVT buffer (Ambion), rNTP mix containing 3 mM of GTP, 1.5 mM of ATP and 1.2 mM each of CTP and UTP (Amersham), 0.4 mM each of Bio-11 CTP and Bio-11 UTP (Perkin Elmer), 40 U of RNase Inhibitor (Ambion), 10 mM DTT (Invitrogen), 2,500 U T7 Polymerase (Epicentre) and water (Ambion) in a final volume of 60 μl followed by incubation at 37° C. for at least 16 hours. The biotin-labeled cRNA was purified using the Qiagen Rneasy® Mini-kit and RNA cleanup protocol according to manufacturer's instructions. Quantification of cRNA yield was performed using UV absorbance 280/260. Ten μg of labeled cRNA was fragmented in 40 mM Tris-acetate pH 8.0, 100 mM KOAc, 30 mM MgOAc for 33 minutes at 94° C. in a final volume of 40 μl. This labeled target was hybridized with MES buffer, 30 μg herring sperm DNA, 150 μg acetylated BSA, 50 pM Bio 948, and RNase free water to a final volume of 300 μl, then incubated at 99° C. for 10 minutes, and then held at 45° C. for 5 minutes.


Example 2.1.4
Sample Preparation: Hybridization of Labeled cRNA to Microarray

Biotinylated cRNA was hybridized to the Affymetrix HG-U133A GeneChip array as described in the Affymetrix Technical Manual.


Example 2.2
Materials and Methods—Determination of Gene Expression Patterns
Example 2.2.1
Determination of Gene Expression Frequencies

Gene expression frequencies of unstimulated patient samples procured from patients who were IgG and/or IgM (antibody) responders (titer≧2200), partial antibody responders (200≦titer<2,200), antibody nonresponders (titer<200), encephalitis developers and/or encephalitis nondevelopers in response to AN1792 were determined as described above (Example 1.2.1) according to certain inclusion criteria for GeneChip Results, also described above (Example 1.2.2 and Table 3). Briefly, MAS 5.0 software was used to compute signal values (i.e., probe intensities) and absent/present calls for each probeset on each array (marginal calls were counted as absent calls due the filter criteria). MAS 5.0 was also used for the first pass normalization by scaling the trimmed mean to a value of 100. The database processes also calculated a series of chip QC (quality control) metrics and stored all the raw data and QC calculations back to the database. QC metrics were stored with the raw data in the database, e.g., as in Example 1.2.2. The signal values for each probeset were converted to frequency values representative of the number of transcripts present in 106 transcripts (ppm) by reference to a standard curve (see, e.g., Example 1.2.3). Data for 9,678 probesets that were called ‘present’ and with frequency ≧10 ppm in at least one of the samples were included in the study. GeneChip data that passed all quality control criteria, as described in Example 1.2.2, were generated from 123 treated and 30 placebo groups (see Table 3 for GeneChip quality control criteria for study inclusion). SAS was used for all analyses unless otherwise noted.


Example 2.2.2
Merging of Clinical and Gene Expression Data

Relevant clinical data pertaining to treatment group, maximum IgG titer for all visits, maximum IgM titer for all visits, encephalitis status, and demographic data were received from StatProbe, Inc. (Ann Arbor, Mich.). The clinical data were merged with the gene expression data by donor identification number. (See also, generally, Example 1.2.5.)


Example 2.2.3
Sample Inclusion Criteria and Patient Demographics
Example 2.2.3.1
Sample Inclusion Criteria

Inclusion for study in this Example 2 required 1) that samples arrive at the Pharmacogenomics Laboratory within one day of collection, 2) an RNA yield >50 ng, and 3) an IVT yield >10 μg. Table 20 accounts for all samples received, and identifies the number of patients in this study (see also FIG. 18). Of the 172 enrolled U.S. patients, 167 consented to inclusion in the pharmacogenomic portion of the study. Of the 167 samples, six did not meet shipping specifications and eight samples yielded insufficient product for chip hybridization. Of the 153 samples remaining, 123 samples were procured from patients treated with AN1792 and 30 samples were procured from placebo patients; note that the 30 samples from the placebo patients were irrelevant to the analysis presented herein for this Example 2.


Example 2.2.3.2
Demographics of Patients

Seventy-five (75) of the patients in the study of this Example 2 were female and 78 were male. The average age was 73 years. Patient demographics for the 123 treated patients are shown in Table 21.


Subjects were assigned to response groups based on postimmunization maximum titer during follow-up. For both IgM and IgG the response groups were: 1) nonresponders, (titer<200); 2) partial responders (200≦titer<2,200); and 3) responders (titer≧2200). Table 22 gives a breakdown of study samples by gender and response category.


Example 2.2.4
Materials and Methods—Pharmacogenomic Statistical Analysis Plan
Example 2.2.4.1
Identification and Removal of Genes Significantly Associated with Covariates

Analyses were conducted to identify factors that might have confounding effects on associations between gene expression levels and response groups. Preimmunization differential blood cell counts and gender were two such factors investigated, and both were identified as significant covariates. For each gene, analysis of covariance (ANCOVA) was used to test for associations of expression level with these two covariates (i.e., with gender; monocyte:lymphocyte ratio). Log-transformed expression was modeled as a function of sex and the monocyte:lymphocyte ratio. To avoid potential confounding with IgG response or the development of encephalitis, these ANCOVAs were run using data only from IgG nonresponders (n=70). Genes were considered significantly associated with either sex or the monocyte:lymphocyte ratio if the unadjusted F-test p value for the respective effect was <0.01. Because all five encephalitis patients for these analyses were female, genes significantly associated with gender were not included in further analyses. Genes identified as having a significant linear association between expression levels and the CPT monocyte:lymphocyte ratio were also removed from further analyses. It is recognized that genes removed from analysis for these reasons may have been associated both with the identified covariable and the response class. Therefore genes associated with response class could be under-reported. Removal of these genes resulted in 8,239 remaining probesets to be further analyzed.


Example 2.2.4.2
Criteria for Selection of Genes Associated with Antibody Responsiveness

Subjects were assigned prior to unblinding to response groups based on postimmunization maximum titer during follow-up. As described above, for both IgM and IgG the response groups were: 1) nonresponders, (titer<200); 2) partial responders (200≦titer<2,200); and 3) responders (titer≧2200). The numbers of patients in each of these groups are shown in Table 22. The proportional odds logistic regression model was used to determine if significant associations existed between preimmunization gene expression levels and postimmunization response groups. The analyses were run using both all immunized subjects in the study (n=123), and with the exclusion of the five encephalitis patients (n=118). It should be noted that all patients from the U.S. that developed meningoencephalitis were IgG responders and all patients but one from the E.U. that developed meningoencephalitis were IgG responders, and distinction was sought between genes related to risk of encephalitis and those associated with IgG responsiveness. Raw p values were adjusted for multiplicity according to the false discovery rate (FDR) procedure of Benjamini and Hochberg ((1995) J. Roy. Stat. Soc. B. 57:289-300; see also, Xiao et al. (2002) BMC Genomics 3:28). Genes were selected as significantly associated with response if: a) the FDR for association with response was <0.1, a criterion that allows for an estimated 10% false positive identifications; b) the odds ratio between responders and others (nonresponders plus partial responders) was >3 fold; c) the FDR from the analysis excluding meningoencephalitis patients was at least twice as significant as the FDR for association with meningoencephalitis; and d) the FDR for association with encephalitis was >0.1. These selection steps identified genes with an odds ratio of at least 3 between responders and others, where the chance of a false positive association was at most 10%, with genes most significantly associated with encephalitis excluded. No genes were found to be significantly associated with the IgM response groups.


Example 2.2.4.3
Identification of Genes Associated with Risk of Encephalitis

The binary logistic regression model was used to determine if significant associations existed between preimmunization gene expression levels and postimmunization development of meningoencephalitis. Treated patients who developed meningoencephalitis (n=5) were compared to those who did not (n=118). The small number of meningoencephalitic subjects resulted in large odds ratios (>10) with some exceedingly wide confidence intervals (2 to 3 orders of magnitude). Because all encephalitis subjects were also IgG antibody responders, genes associated with antibody response (with the encephalitis patients excluded) were filtered from the list of encephalitis-associated genes. Genes were selected as significantly associated with encephalitis if: a) the odds ratio between meningoencephalitics and nonmeningoencephalitics was >3 fold; b) the FDR was <0.1; c) the odds ratio for association with meningoencephalitis was at least two times greater than that for association with IgG response; d) the FDR for association with IgG response was >0.1; and e) the odds ratio for IgG response was less than 2 fold. Due to the observation that some genes with IgG odds ratios between 2 and 4 fold had meningoencephalitis odds ratios up to hundreds fold higher, exceptions were made to filtering rule (e) when the odds ratio for association with meningoencephalitis was at least five-fold greater than the odds ratio for association with IgG response. These selection steps identified genes associated with an odds ratio of at least 3 between the meningoencephalitics and nonmeningoencephalitics, where the chance of a false positive association was at most 10%, with genes most significantly associated with an IgG response excluded.


Example 2.2.4.4
Use of GeneCluster to Select Best Gene Subset

GeneCluster (see www.broad.mit.edu/cancer/software/genecluster2/gc2.html) (Golub et al. (1999) Science 286:531-37) was used both as a method of demonstrating associations between the expression levels of the 8,239 probesets remaining (see Example 2.2.4.1) and response group using ANOVA-based methods, and to select gene expression patterns that most accurately assigned samples to the correct response class (i.e., correct response group). Gene selection was based on weighted voting. Statistical significance was assessed by a permutation-based p value. For the analysis of antibody response groups, partial responders were excluded from this analysis. Classifiers for encephalitis were chosen using data from all immunized subjects.


Example 2.2.4.5
Selection of Two-Gene Combinations that Most Accurately Segregate Meningoencephalitics from Nonmeningoencephalitics

The ability of two-gene models to discriminate between meningoencephalitics and nonmeningoencephalitics was evaluated by logistic regression models using as covariates all 287,661 pairwise combinations of genes meeting the criteria for association with meningoencephalitis. For each model, the sum of the absolute values of the log-odds for all subjects was used as a ranking measure to indicate the strength of the discrimination. To estimate the FDRs for this large set of logistic regression models, the full analysis was rerun 200 times with random permutation of the class labels to compute resampling-based FDRs (Reiner et al. (2003) Bioinformatics 19:368-75). These analyses were carried out using R statistics package 1.9.1, which can be found at www.R-project.org (R Development Core Team (2004) R Foundation for Statistical Computing).


Example 2.2.4.6
Pathway Analysis

These data were generated through the use of Ingenuity Pathways Analysis (Summer 04 Release V1), a web-delivered application that explores networks such as gene expression array data sets (see www.ingenuity.com). Biological functions were assigned to the overall analysis by using findings that have been extracted from the scientific literature and stored in the Ingenuity Pathways Knowledge Base. The biological functions assigned to the analysis are ranked according to the significance of that biological function to the analysis. A Fischer's exact test is used to calculate a p value determining the probability that the biological function assigned to the analysis is explained by chance alone.


Example 2.3
Results

A total of 372 patients, 172 from the U.S. and 200 from the E.U., were enrolled in the clinical trial. Participation in the pharmacogenomic portion of the study was optional and offered to U.S. patients only, and 97% agreed to participate. Consent was obtained after approval by local institutional review boards. FIG. 18 shows the disposition of patients with respect to the pharmacogenomic portion of the study. GeneChips that passed quality control inclusion criteria (detailed in Table 3) were generated from 123 treated and 30 placebo patients. The search for gene expression levels associated with response to immunization was conducted by comparing preimmunization expression levels between subjects grouped according to postimmunization response (as measured by maximum anti-AN1792 titer or the development of meningoencephalitis). Of the 6 U.S. patients who ultimately developed meningoencephalitis, 5 had consented to pharmacogenomics; there were 12 E.U. patients who developed meningoencephalitis.


Example 2.3.1
Identification and Removal from Analysis of Genes Associated with Monocyte Proportion and Gender Covariables

A statistically significant correlation (p=0.012) was detected between monocyte-to-lymphocyte ratio and IgG responsiveness, with a high proportion of monocytes associated with nonresponsiveness. The top 16 samples for this metric fell within the nonresponder group (see FIG. 19). The association between monocyte proportion and IgM response groups was not statistically significant, and trended in the opposite direction from the association with IgG responsiveness. Despite the statistically significant association between the IgG response and proportion of monocytes, however, the monocyte-to-lymphocyte ratio was not itself a useful biomarker of likely nonresponsiveness because the majority (77%) of nonresponders fell within the range of responders (see FIG. 18). The significance of the association did, nevertheless, point to the need to account for monocyte proportion covariate in analyses of associations between IgG responsiveness and gene expression. Another concern was that, although there were males among the E.U. patients who developed meningoencephalitis, all five U.S. encephalitis patients in the pharmacogenomic study were female, precipitating the need to account for sex-related differences in analyses of associations between encephalitis and gene expression. Sequences significantly associated with monocyte proportion and/or sex were identified by ANCOVA and removed from further analysis; they are listed in alphabetical order in Table 23. It is recognized that genes removed from analyses for these reasons may have been associated both with the identified covariate and the response class, and therefore, genes associated with response class could be under-reported. It should be noted that although the genes listed in Table 23 are excluded from Tables 24-37 (i.e., in Example 2), some genes listed in Table 23 may be included in Tables 10-12 and 18 (i.e., some of the genes listed in Tables 10-12 and 18 (see Example 1) are included in Table 23 as associated with covariates). After removal of these genes significantly associated with monocyte-to-lymphocyte ratio and/or sex, 8,239 probesets remained for further analysis.


Example 2.3.2
Identification of Predictive Biomarkers of IgG Response

The search for gene expression levels associated with antibody response was conducted by comparing preimmunization expression levels between subjects grouped according to postimmunization maximum IgM and IgG titer. No genes met the criteria for significant association of preimmunization gene expression levels and postimmunization IgM titer. In contrast, there were 366 sequences (from 318 genes and 17 unmapped sequences) that met the selection criteria for association with IgG response. MRPS31 (mitochondrial ribosomal protein 31) had the smallest (most significant) false discovery rate (FDR=0.0003, with a p value unadjusted for multiplicity of 1.07E−7 and odds ratio encephalitis=5.5). The highest observed odds ratio was 10.3 (for PTMA, prothymosin, alpha), indicating that elevated expression of this gene was strongly associated with IgG response. The lowest odds ratio (calculated with encephalitics) was 0.098 (GLUD1, glutamate dehydrogenase 1), indicating that decreased expression of this gene was strongly associated with IgG response. The FDRs and odds ratios for genes identified as associated with IgG response are shown in Table 24.


Example 2.3.3
Biological Pathways Associated with IgG Response

Pathway analyses indicate that, prior to immunization, the ability to mount an IgG response is highly correlated with expression patterns of genes directly involved in the protein synthesis machinery. Ingenuity Global Analysis reports highly significant (p value=9.53E−12 to 1.29E−3) associations with the protein synthesis categories (a measure of the likelihood that genes that participate in protein synthesis are biomarkers associated with IgG responsiveness). In addition to the genes identified by Ingenuity, 22 additional genes were identified that directly participate in translational events. All of the IgG response-associated genes directly involved in the protein synthetic machinery were expressed at higher levels in IgG responders. The most significant of these genes are shown in Table 25. In contrast, 42% of the IgG response-associated genes involved in other functions were expressed at lower levels in IgG responders. Functions significantly represented among these genes were transcription, cell cycle, cell growth and proliferation, protein trafficking, DNA repair and recombination, and protein synthesis regulation. A selection of these genes is shown in Table 26. The annotation of IgG response-associated genes is shown in Table 27.


Example 2.3.4
Selection of Genes that Accurately Classify IgG Responders

Using the weighted voting algorithm as implemented in GeneCluster, a set of 24 sequences (from the 7,479 sequences remaining after removal from 9,678 probesets of genes significantly associated with monocyte-to-lymphocyte ratio and/or sex (see Example 2.3.1) and of genes significantly associated with encephalitis (see Example 2.3.5)) were identified as the most accurate classifier. All 24 sequences had a permutation-based p value <0.01, and all but one (RAB3-GAP150) had a permutation-based p value <0.001. Table 28 lists the descriptions of the 24 genes, and respective odds ratios and FDRs for IgG and encephalitis, that are best at accurate classification of the IgG responders (the 24 genes identify 76 patients correctly and 19 patients incorrectly; of the incorrectly identified patients, 6 are IgG responders). Table 29 lists the classification of each patient (i.e., patients that were IgG responders or IgG nonresponders) and the confidence score using these 24 classifier genes. Table 30 is a list of the 6 best classifiers of an IgG response (a subset of the 24 genes in Table 28); this set correctly identifies 75 patients but incorrectly identifies 20 patients. Table 31 lists the classification of each patient and the confidence score using these 6 classifier genes.


Example 2.3.5
Identification of Predictive Biomarkers for Development of Encephalitis

There were 760 sequences (from 689 genes and 8 unmapped sequences) that met the selection criteria for association with encephalitis. These associations were identified by comparing the gene expression levels of the 5 patients who developed meningoencephalitis to the gene expression levels of the 118 treated patients who did not. The gene most significantly associated (unadjusted p=5.07E−7, FDR=0.004, odds ratio=230) with encephalitis was STAT1, a critical gene in a proinflammatory signal transduction pathway. The highest odds ratio observed was 3,136 (for NHP2L1, with increased expression associated with encephalitis). The lowest odds ratio was 1.0E−4 (for HEAB, with decreased expression associated with encephalitis). For 364 sequences (48%) of the 760 meningoencephalitis-associated sequences, the odds ratios were greater than 10 fold (greater than 10 or less than 0.1), but the confidence limits were often very broad due to the small size of the encephalitis group and the heterogeneity within it. The development of encephalitis was associated with the decreased expression of 41% of the sequences. The FDRs and odds ratios for the meningoencephalitis-associated sequences are shown in Table 32.


Example 2.3.6
Genes and Biological Pathways Associated with Development of Encephalitis

Of the 760 sequences associated with encephalitis, 63 were replicate identifications (i.e., multiple probesets mapping to the same gene). The majority of these sequences were mapped by Ingenuity; among the unmapped sequences, five subsequently were mapped to known genes by homology search. Ingenuity Global Analysis assigns 56% of encephalitis-associated genes to “High Level Functions” and “Global Canonical Pathways.” Significantly represented were genes related to the control of apoptosis and proinflammatory immune response, or to the downstream functions of control of cell cycle, cell proliferation, protein synthesis and protein trafficking (see Table 33 for annotation of genes associated with meningoencephalitis). Ingenuity Pathway Analysis reports p values for the significance of the link between encephalitis-associated genes and cell death categories as ranging from 7.46E−7 to 4.65E−2, and for the link between associated genes and cell cycle functions as ranging from 4.35E−9 to 4.65E−2. Genes related to TNF/Fas, TGFβ and p53 pathways were highly represented among genes related to the control of cell death (see Table 34). A selection of these genes and their association with meningoencephalitis is shown in Table 35. While the encephalitis-associated genes in Table 35 were selected on the basis of known involvement in TNF and/or Fas pathways and other immune response-related cell death and cell activation pathways, the list does not encompass all such genes.


Example 2.3.7
Selection of Genes that Accurately Classify Patients Who Develop Encephalitis

Using the frequency data from all immunized subjects, eight genes (selected from the 760 encephalitis-associated sequences, and shown in Table 36) that accurately assigned 4 of 5 encephalitis patients and 111 (94%) of nonencephalitis patients were identified using weighted voting and leave-one-out cross-validation in GeneCluster. The confidence scores for the classification of the five encephalitis patients and a representative selection of nonencephalitis patients are shown in FIG. 20. The one encephalitis patient who was assigned to the incorrect group was assigned with the highest possible confidence score. Therefore, additional analyses were conducted to determine whether a model weighted toward the capture of all five encephalitis patients would correctly classify this patient as among those who developed encephalitis.


Selection of optimal classifiers by the pairwise combination logistic regression approach was designed to find the two-gene combinations that best distinguished the meningoencephalitics from nonmeningoencephalitics. No functional annotation is available on nuclear protein ukP68 (NpukP68), which was one of the two genes in the top ranked logistic regression-based classifier pair. STAT1 appeared in the third-highest ranked two-gene classifier, with an odds ratio for association with encephalitis of 230.4. Remarkably, for 18 of the top 20 two-gene combinations (listed in Table 37), one of the genes in the two-gene combination was either STAT1 or NpukP68, indicating a very strong association between high expression of either of these two genes and the development of encephalitis. FIG. 21 shows expression level plots of the top ranked and third ranked gene combinations (pairs). FIG. 22 shows the expression level plots for the remaining 18 top-ranked gene pairs. Both FIG. 21 and FIG. 22 display the association of expression profiles for the pairs of genes listed in Table 37 with either the clinical response of encephalitis development or encephalitis nondevelopment.


Example 2.4
Discussion

This invention identified 318 genes whose expression levels prior to immunization with AN1792 are significantly associated with IgG responsiveness to AN1792 immunization (i.e., can be also be used to assess IgG nonresponsiveness). No such risk factors were identified for IgM nonresponsiveness. Expression levels of genes associated with IgG response in partial responders (200≦titer<2,200) were consistently intermediate between nonresponders (titer<200) and responders (titer≧2200), a trend that provides additional evidence of the relationship between preimmunization gene expression pattern and IgG response.


The vast majority of genes associated with IgG response are related to biological functions (protein synthesis and trafficking, RNA processing, cellular assembly and organization, and cell cycle control) that are not specific to the immune system. The incidence of responsiveness in this study was relatively low (53 of 123 with titer>200), and the patients were elderly (mean age 74 years). Since responsiveness to immunization is known to decline with age (Westmoreland et al. (1990) Epidemiol. Infect. 104:499-509; Looney et al. (2001) J. Clin. Immunol. 21:30-36; Rey (1997) Bull. Soc. Pathol. Exot. 90 (4):245-52; Arreaza et al (1993) Clin. Exp. Immunol. 92:169-73; Salvador et al. (2003) Immunol. Allergy Clin. North Am. 23 (1):133-48), age may influence the expression levels of genes directly involved in protein synthesis and the other functions identified by this invention as associated with IgG response.


The invention identified 689 genes whose expression levels prior to immunization with AN1792 are significantly associated with development of encephalitis following immunization. These risk factors were identified by comparing the gene expression levels of the five patients who developed encephalitis to the levels of the 118 treated patients who did not develop encephalitis. In contrast to the IgG associated genes, functional annotation of genes associated with encephalitis indicated a preponderance of genes of particular importance in pathways related to the control of the immune system and inflammation. Those who developed encephalitis had, prior to immunization, detectable perturbations in pathways controlling the TNF and other proinflammatory and apoptotic cascades. Perturbations favoring both anti-apoptotic and pro-apoptotic activities were detected, possibly suggesting compensatory activation to counteract deleterious effects of perturbation in apoptosis. This is also supported by perturbations in a large number of cell cycle, growth, and proliferation genes. The STAT gene family plays a central role in proinflammatory cytokine activation and in apoptotic cascades. Perturbation in the expression levels of STAT1, STAT3 (3′ untranslated region), and STAT5 were found to be highly significant risk factors for encephalitis. High expression of a variety of other genes involved in proinflammatory cascades, such as IL-9, IL-19, IL-25, IL-27R, and CD80, were also associated with encephalitis. Elevated expression of the coding region and decreased expression of the 3′ untranslated region of STAT5B were associated with development of meningoencephalitis, suggesting that variants of STAT5B mRNA make different contributions to the “meningoencephalitis-prone” gene expression pattern.


All five encephalitis patients for whom gene expression data were available were IgG responders. It is therefore notable that IgG responders who developed encephalitis expressed some protein synthesis and trafficking genes at levels significantly lower than nonmeningoencephalitic IgG responders. Remarkably, for a number of genes (RPS7, RPLP1, RPS24, and RPL9), lower expression levels were associated with development of encephalitis, while higher expression levels were associated with IgG response. Another distinction between the IgG response associated genes and the meningoencephalitis-associated genes is that, although protein synthesis is identified as a significant category among both sets, the preponderance (˜80%) of IgG response-associated genes in this category are directly involved in the protein synthetic machinery, and that all of these were expressed at higher levels in IgG responders. In contrast, the majority of meningoencephalitis-associated genes categorized as involved in protein synthesis regulate protein expression, with only approximately half expressed at higher levels in the meningoencephalitis group. These data provide an additional line of evidence that preimmunization gene expression patterns associated with risk of encephalitis are distinguishable from those associated with IgG response.


Logistic regression using pairwise combinations of genes was applied to identify the most accurate two-gene combination classifier of patients at risk of developing meningoencephalitis. This analytical approach identified the combination of expression levels of NPukP68 and AKAP13 (PRKA anchor protein 13 anchor) as the top biomarkers for separating all 5 meningoencephalitics from nonmeningoencephalitics. No functional annotation is available on NPukP68, but elevated expression was associated with an odds ratio of 651. Either NPukP68 or STAT1 (odds ratio of 230.4) appears as one of the genes listed in eighteen of the 20 top ranked pairwise combinations.


Of the five meningoencephalitis patients, encephalitis, one expressed the vast majority of 760 meningoencephalitis associated sequences at levels associated with the nonmeningoencephalitis group. However, this patient expressed numerous genes at levels associated with encephalitis following 24-hour in vitro stimulation with a stimulatory cytokine cocktail and the AN1792 antigen (i.e., the protocol in Example 1; see patient 33, e.g., in FIGS. 4-13). These observations together suggest that a small number of critical genes may profoundly influence the consequences of both in vivo and in vitro immune stimulation.


The inventors have identified highly significant associations between PBMC preimmunization gene expression patterns and postimmunization anti-AN1792 IgG responses and postimmunization development of meningoencephalitis. These results may be of use in identifying patients at risk of developing a severe adverse event in active immunotherapy for Alzheimer's disease, and in identifying those patients that are likely to respond to immunotherapy.


All references cited in this application are incorporated by reference in their entireties as if fully set forth herein.

TABLE 1Stringency examplesStringencyPolynucleotideHybridHybridization TemperatureWash TemperatureConditionHybridLength (bp)1and Buffer2and Buffer2ADNA:DNA>5065° C.; 1X SSC -or-65° C.; 0.3X SSC42° C.; 1X SSC, 50%formamideBDNA:DNA<50TB*; 1X SSCTB*; 1X SSCCDNA:RNA>5067° C.; 1X SSC -or-67° C.; 0.3X SSC45° C.; 1X SSC, 50%formamideDDNA:RNA<50TD*; 1X SSCTD*; 1X SSCERNA:RNA>5070° C.; 1X SSC -or-70° C.; 0.3X SSC50° C.; 1X SSC, 50%formamideFRNA:RNA<50TF*; 1X SSCTF*; 1X SSCGDNA:DNA>5065° C.; 4X SSC -or-65° C.; 1X SSC42° C.; 4X SSC, 50%formamideHDNA:DNA<50TH*; 4X SSCTH*; 4X SSCIDNA:RNA>5067° C.; 4X SSC -or-67° C.; 1X SSC45° C.; 4X SSC, 50%formamideJDNA:RNA<50TJ*; 4X SSCTJ*; 4X SSCKRNA:RNA>5070° C.; 4X SSC -or-67° C.; 1X SSC50° C.; 4X SSC, 50%formamideLRNA:RNA<50TL*; 2X SSCTL*; 2X SSCMDNA:DNA>5050° C.; 4X SSC -or-50° C.; 2X SSC40° C.; 6X SSC, 50%formamideNDNA:DNA<50TN*; 6X SSCTN*; 6X SSCODNA:RNA>5055° C.; 4X SSC -or-55° C.; 2X SSC42° C.; 6X SSC, 50%formamidePDNA:RNA<50Tp*; 6X SSCTp*; 6X SSCQRNA:RNA>5060° C.; 4X SSC -or-60° C.; 2X SSC45° C.; 6X SSC, 50%formamideRRNA:RNA<50TR*; 4X SSCTR*; 4X SSC
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.
2SSPE (1xSSPE is 0.15 M NaCl, 10 mM NaH2PO4, and 1.25 mM EDTA, pH 7.4) can be substituted for SSC (1xSSC is 0.15 M NaCl and 15 mM sodium citrate) in the hybridization and wash buffers; washes are performed for 15 minutes after hybridization is complete.

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(log10Na+) + 0.41(% G + C) − (600/N), where N is the number of bases in the hybrid, and
Na+ is the concentration of sodium ions in the hybridization buffer (Na+ for 1X SSC = 0.165 M). Additional examples of stringency conditions for polynucleotide hybridization are provided in Sambrook et al., Molecular
# Cloning: A Laboratory Manual, Chs. 9 & 11, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY (1989), and Ausubel et al., eds., Current Protocols in Molecular Biology, Sects. 2.10 & 6.3-6.4, John Wiley & Sons, Inc. (1995), herein incorporated by reference.









TABLE 2










Characteristics of samples with % neutrophils >20%


post CPT fractionation
















Maxi-
Maxi-




Post-CPT

mum
mum



Pre-CPT
% Neutro-
Treatment
IgG
IgM


Patient
% Neutrophils
phils
Group
Titer
Titer















17
63
38
Immunotherapy
50
25


23
64
47
Immunotherapy
50
25


36
62
29
Immunotherapy
4111
13275


44
53
25
Immunotherapy
126
957


271
79
37
Immunotherapy
172
28554


288
59
25
Placebo
50
25


756
67
40
Immunotherapy
50
25
















TABLE 3








Criteria for chip inclusion in the final dataset


















Chip sensitivity
<6.1



Raw Q
<7



Scale factor
<4 and >1/4



Cell saturation ratio
<0.00005



QC P probability frequency
<20



QC P probability average difference
<250



Number of outliers across the array
<1600



Defect on visual inspection
Absent

















TABLE 4










r2 values for all study samples












AN1792-

Control



Patient
stimulated
S.D.
culture
S.D.


number
sample
AN1792
sample
control














2
0.90
0.05
0.84
0.07


3
0.92
0.05
0.88
0.06


5
0.89
0.05
0.87
0.06


6
0.90
0.06
0.88
0.06


7
0.89
0.04
0.81
0.06


8
0.88
0.05
0.79
0.06


9
0.90
0.05

N.A.


12
0.92
0.04
0.89
0.05


14
0.86
0.05
0.86
0.06


15
0.90
0.05
0.85
0.06


17
0.90
0.05
0.82
0.06


18
0.87
0.06
0.87
0.05


19
0.84
0.06
0.84
0.07


20
0.91
0.05
0.88
0.06


22
0.90
0.05
0.72
0.08


23
0.88
0.05
0.88
0.04


24
0.88
0.05
0.87
0.07


25
0.86
0.06
0.83
0.06


26
0.88
0.06
0.86
0.06


27
0.81
0.08
0.82
0.07


29
0.91
0.05
0.89
0.06


30
0.87
0.05
0.85
0.05


31
0.88
0.04
0.88
0.05


32
0.88
0.05
0.87
0.06


33
0.72
0.06
0.89
0.06


34
0.91
0.04
0.89
0.05


36
0.88
0.05
0.87
0.06


37
0.91
0.04

N.A.


40
0.88
0.06
0.86
0.07


41
0.89
0.06
0.88
0.06


43
0.92
0.04
0.88
0.05


44
0.90
0.05
0.89
0.05


45
0.90
0.05
0.88
0.05


46
0.91
0.04
0.88
0.06


49
0.80
0.05
0.71
0.07


50
0.88
0.06
0.84
0.07


52
0.90
0.05
0.87
0.06


53
0.90
0.05
0.89
0.05


54
0.89
0.05
0.87
0.07


55
0.91
0.04

N.A.


56
0.86
0.05
0.88
0.06


60
0.90
0.05
0.78
0.07


61
0.89
0.04
0.88
0.05


62
0.88
0.05
0.86
0.07


64
0.88
0.04
0.88
0.06


65
0.88
0.05

N.A.


66
0.89
0.04
0.85
0.07


67
0.89
0.04
0.86
0.07


68
0.85
0.06
0.87
0.06


69
0.92
0.04

N.A.


71
0.89
0.05
0.87
0.07


73
0.85
0.04
0.88
0.06


251
0.92
0.04
0.89
0.05


252
0.89
0.04
0.86
0.06


254
0.90
0.05
0.88
0.05


255
0.91
0.04
0.83
0.06


256
0.88
0.06
0.87
0.06


257
0.90
0.04
0.83
0.06


258
0.90
0.05
0.88
0.06


259
0.90
0.05
0.90
0.05


260
0.89
0.06
0.89
0.05


262
0.89
0.04
0.88
0.05


264
0.90
0.06
0.89
0.05


266
0.91
0.05
0.88
0.06


267
0.90
0.05
0.89
0.05


268
0.89
0.05
0.69
0.06


269
0.88
0.06
0.87
0.06


270
0.87
0.05
0.88
0.06


271
0.88
0.05
0.85
0.06


272
0.90
0.06
0.88
0.06


273
0.91
0.04
0.87
0.05


274
0.92
0.04
0.88
0.05


275
0.89
0.05
0.88
0.06


277
0.89
0.05
0.85
0.06


279
0.88
0.05
0.89
0.05


280
0.90
0.05
0.89
0.06


281
0.91
0.04
0.86
0.06


282
0.90
0.05
0.88
0.05


283
0.84
0.06
0.73
0.06


284
0.91
0.05
0.86
0.06


285
0.87
0.06
0.87
0.06


286
0.89
0.05
0.88
0.05


287
0.91
0.04
0.89
0.06


288
0.91
0.04
0.87
0.06


289
0.90
0.05
0.89
0.06


290
0.90
0.04
0.82
0.06


291
0.91
0.04
0.84
0.07


292
0.90
0.05
0.87
0.06


293
0.89
0.05
0.86
0.07


294
0.90
0.05
0.86
0.06


295
0.91
0.04
0.90
0.05


296
0.91
0.04
0.89
0.05


297
0.89
0.06
0.86
0.07


299
0.87
0.06
0.89
0.06


300
0.91
0.04
0.89
0.06


301
0.88
0.05
0.83
0.06


303
0.84
0.06
0.86
0.07


304
0.90
0.05
0.87
0.06


306
0.89
0.04
0.85
0.07


307
0.87
0.05

N.A.


308
0.79
0.07

N.A.


309
0.90
0.05
0.88
0.06


310
0.90
0.04
0.84
0.06


312
0.91
0.04
0.86
0.06


313
0.89
0.05

N.A.


314
0.88
0.05
0.89
0.06


315
0.90
0.04
0.88
0.05


316
0.88
0.04
0.70
0.06


317
0.89
0.05
0.88
0.06


318
0.89
0.04
0.88
0.06


319
0.88
0.06

N.A.


502
0.91
0.05
0.88
0.05


503
0.84
0.06
0.82
0.07


504
0.89
0.05
0.88
0.05


505
0.89
0.06
0.84
0.05


508
0.89
0.05
0.86
0.06


510
0.91
0.04
0.89
0.05


511
0.89
0.05
0.86
0.06


513
0.90
0.05
0.87
0.06


514
0.90
0.05
0.87
0.06


515
0.85
0.06
0.84
0.06


752
0.90
0.05
0.88
0.06


753
0.91
0.05
0.88
0.06


755
0.90
0.05
0.90
0.06


756
0.89
0.04
0.86
0.06


758
0.90
0.05
0.87
0.06


759
0.91
0.05
0.87
0.05


760
0.90
0.05
0.89
0.05


761
0.90
0.05
0.86
0.06


762
0.90
0.04
0.89
0.05


763
0.87
0.06
0.78
0.09


764
0.90
0.05
0.86
0.07


765
0.88
0.04
0.74
0.06
















TABLE 5










Responder status of r2 value outlier samples












Patient

IgG



Patient
treatment

response
IgM response


number
group
Culture condition
group
group














33
AN1792
AN1792-
IgG responder
IgM responder




stimulated
and





meningoen-





cephalitic


22
AN1792
Diluent control-
IgG Responder
IgM responder




stimulated


49
Placebo
Diluent control-
Not applicable
Not applicable




stimulated


268
Placebo
Diluent control-
Not applicable
Not applicable




stimulated


283
Placebo
Diluent control-
Not applicable
Not applicable




stimulated


316
AN1792
Diluent control-
Nonresponder
Nonresponder




stimulated


765
AN1792
Diluent control-
Nonresponder
Nonresponder




stimulated
















TABLE 6










Samples received by pharmacogenomic laboratory











Number














Enrolled U.S. patients
172



Enrolled patients who consented to
167



pharmacogenomic portion of study



Samples within shipping specifications
161



AN1792-stimulated samples within
149



culture and storage specifications



AN1792-stimulated samples with >50 ng RNA
141



AN1792-stimulated samples with >10 μg IVT
141



AN1792-stimulated samples removed
8



due to operator error identified



during QC review



Total number of AN1792-stimulated
133



samples in study



Patients represented by paired
124



(antigen-stimulated and



diluent control) samples



Diluent control samples unavailable
9



due to insufficient



yield of mRNA or IVT

















TABLE 7










Patients in study, by year of birth












Number of
Number of


Year of Birth
Number of Patients
Female Patients
Male Patients













1915-1920
27
17
10


1921-1925
30
19
11


1926-1930
30
9
21


1931-1935
20
6
14


1936-1940
19
7
12


1941-1945
2
1
1


1946-1950
4
4
0


Unknown
1
1
0


Cumulative total
133
64
69
















TABLE 8










Gender of patients in study, by race













Caucasian
Hispanic
Black
Asian
Unknown





Females
56
6
2
0
0


Males
58
6
2
1
2
















TABLE 9










Samples in pharmacogenomic study













Male
Female
Total
















Placebo
11
14
25



Treated
59
49
108



Typed ApoE4 negative
23
11
34



Typed ApoE4 positive
34
36
70



Treated IgG responders
10
12
22



Treated IgG partial responders
12
14
26



Treated IgG nonresponders
37
23
60



Treated IgM responders
40
41
81



Treated IgM nonresponders
19
8
27



Meningoencephalitis patients
0
5
5

















TABLE 10










GENES ASSOCIATED WITH MENINGOENCEPHALITIS BY ANOVA


(sorted alphabetically by gene name)

















Average







expression in







meningoencephalitis







patients relative to







average in


Accession


Unadjusted

nonencephalitis


number
Gene description
Gene name
p value
FDR
patients





NM_005736
ARP1 actin-related protein
ACTR1A
0.000088
0.0203
lower



1 homolog A, centractin



alpha (yeast)


NM_015999
Adiponectin receptor 1
ADIPOR1
0.000158
0.0271
lower


AK021586
Agrin
AGRN
0.000042
0.0169
lower


M90360
A kinase (PRKA) anchor
AKAP13
0.000037
0.0163
higher



protein 13


NM_014481
APEX nuclease
APEX2
0.000221
0.0330
lower



(apurinic/apyrimidinic



endonuclease) 2


NM_001655
archain 1
ARCN1
0.000016
0.0092
higher


BC005851
Rho GDP dissociation
ARHGDIA
0.000344
0.0390
lower



inhibitor (GDI) alpha


NM_012099
CD3-epsilon-associated
ASE-1
0.000314
0.0389
lower



protein; antisense to



ERCC-1


NM_025080
asparaginase like 1
ASRGL1
0.000001
0.0087
higher


U26455
ataxia telangiectasia
ATM
0.000232
0.0338
higher



mutated (includes



complementation groups



A, C and D)


NM_001687
ATP synthase, H+
ATP5D
0.000476
0.0448
lower



transporting, mitochondrial



F1 complex, delta subunit


M62762
ATPase, H+ transporting,
ATP6V0C
0.000526
0.0468
lower



lysosomal 16 kDa, V0



subunit c


NM_001693
ATPase, H+ transporting,
ATP6V1B2
0.000085
0.0203
lower



lysosomal 56/58 kDa, V1



subunit B, isoform 2


NM_016311
ATPase inhibitory factor 1
ATPIF1
0.000414
0.0413
higher


NM_017450
BAI1-associated protein 2
BAIAP2
0.000160
0.0271
lower


NM_004640
HLA-B associated
BAT1
0.000323
0.0389
lower



transcript 1


AA102574
bromodomain adjacent to
BAZ1A
0.000010
0.0087
higher



zinc finger domain, 1A


NM_001707
B-cell CLL/lymphoma 7B
BCL7B
0.000069
0.0184
lower


NM_004634
bromodomain and PHD
BRPF1
0.000149
0.0266
higher



finger containing, 1


NM_018944
chromosome 21 open
C21ORF45
0.000253
0.0352
higher



reading frame 45


AL545982
chaperonin containing
CCT2
0.000231
0.0338
lower



TCP1, subunit 2 (beta)


AF098641
CD44 antigen (homing
CD44
0.000152
0.0266
lower



function and Indian blood



group system)


NM_001783
CD79A antigen
CD79A
0.000484
0.0449
lower



(immunoglobulin-



associated alpha)


AB017493
core promoter element
COPEB
0.000063
0.0184
higher



binding protein


U69546
CUG triplet repeat, RNA
CUGBP2
0.000393
0.0412
higher



binding protein 2


BE046443
cylindromatosis (turban
CYLD
0.000384
0.0408
higher



tumor syndrome)


NM_001343
disabled homolog 2,
DAB2
0.000065
0.0184
higher



mitogen-responsive



phosphoprotein



(Drosophila)


BG530850
DEAD (Asp-Glu-Ala-Asp)
DDX18
0.000189
0.0305
higher



box polypeptide 18


BE963238
DEAD (Asp-Glu-Ala-Asp)
DDX52
0.000404
0.0413
higher



box polypeptide 52


AW081113
SR rich protein
DKFZP564B0769
0.000008
0.0087
higher


NM_001961
eukaryotic translation
EEF2
0.000106
0.0225
lower



elongation factor 2


BG481972
eukaryotic translation
EIF5
0.000006
0.0087
higher



initiation factor 5


BF445047
epithelial membrane
EMP1
0.000461
0.0442
lower



protein 1


NM_004459
fetal Alzheimer antigen
FALZ
0.000059
0.0184
higher


NM_012179
F-box only protein 7
FBXO7
0.000025
0.0123
lower


NM_018115
hypothetical protein
FLJ10498
0.000183
0.0300
lower



FLJ10498


NM_024845
hypothetical protein
FLJ14154
0.000049
0.0184
lower



FLJ14154


NM_017736
hypothetical protein
FLJ20274
0.000080
0.0203
higher



FLJ20274


NM_017775
hypothetical protein
FLJ20343
0.000315
0.0389
higher



FLJ20343


AU145053
formin binding protein 1
FNBP1
0.000409
0.0413
higher


NM_002030
formyl peptide receptor-
FPRL2
0.000501
0.0459
lower



like 2


NM_002569
furin (paired basic amino
FURIN
0.000264
0.0363
lower



acid cleaving, enzyme)


BE439987
growth arrest-specific 7
GAS7
0.000240
0.0344
higher


BE646414
golgi associated, gamma
GGA2
0.000271
0.0364
higher



adaptin ear containing,



ARF binding protein 2


BG420237
heat shock 90 kDa protein
HSPCA
0.000207
0.0318
higher



1, alpha


AA284705
intercellular adhesion
ICAM1
0.000010
0.0087
lower



molecule 1 (CD54), human



rhinovirus receptor


BG261322
translation initiation factor
IF2
0.000041
0.0169
higher



IF2


NM_016281
STE20-like kinase
JIK
0.000250
0.0352
higher


BF382924
joined to JAZF1
JJAZ1
0.000327
0.0389
higher


NM_003772
jerky homolog-like
JRKL
0.000425
0.0420
higher



(mouse)


D26488
KIAA0007 protein
KIAA0007
0.000070
0.0184
higher


AI673812
KIAA0553 protein
KIAA0553
0.000014
0.0087
higher


AU153525
KIAA0652 gene product
KIAA0652
0.000558
0.0485
lower


AI629033
KIAA0872 protein
KIAA0872
0.000063
0.0184
lower


BF223224
kinesin family member 5B
KIF5B
0.000063
0.0184
higher


BF673699
v-Ki-ras2 Kirsten rat
KRAS2
0.000127
0.0252
higher



sarcoma 2 viral oncogene



homolog


AK001105
LAG 1 longevity assurance
LASS2
0.000062
0.0184
lower



homolog 2 (S. cerevisiae)


NM_017526
leptin receptor
LEPR
0.000346
0.0390
higher


U82276
leukocyte
LILRA2
0.000131
0.0252
lower



immunoglobulin-like



receptor, subfamily A



(with TM domain),



member 2


BF965566
leucine rich repeat (in
LRRFIP1
0.000011
0.0087
higher



FLII) interacting protein 1


AI972475
LYRIC/3D3
LYRIC
0.000129
0.0252
higher


AI566096
likely ortholog of mouse
M96
0.000411
0.0413
higher



metal response element



binding transcription factor 2


AF067173
mago-nashi homolog,
MAGOH
0.000472
0.0448
higher



proliferation-associated



(Drosophila)


AI471665
MYC-associated zinc
MAZ
0.000451
0.0436
lower



finger protein (purine-



binding transcription factor


AL556619
methyl-CpG binding
MBD4
0.000333
0.0389
higher



domain protein 4


NM_014763
mitochondrial ribosomal
MRPL19
0.000320
0.0389
higher



protein L19


BC001165
N-ethylmaleimide-
NAPA
0.000564
0.0486
lower



sensitive factor attachment



protein, alpha


AI361805
natural killer-tumor
NKTR
0.000006
0.0087
higher



recognition sequence


BC004952
likely ortholog of mouse
NSPC1
0.000008
0.0087
lower



nervous system polycomb 1


NM_022731
nuclear ubiquitous casein
NUCKS
0.000198
0.0310
higher



kinase and cyclin-



dependent kinase substrate


NM_005022
profilin 1
PFN1
0.000148
0.0266
lower


NM_024165
PHD finger protein 1
PHF1
0.000486
0.0449
lower


NM_004279
peptidase (mitochondrial
PMPCB
0.000180
0.0300
higher



processing) beta


NM_004774
PPAR binding protein
PPARBP
0.000296
0.0386
higher


J03223
proteoglycan 1, secretory
PRG1
0.000216
0.0328
lower



granule


BC001423
proteasome (prosome,
PSME3
0.000021
0.0108
lower



macropain) activator



subunit 3 (PA28 gamma;



Ki)


BG029917
proteasome (prosome,
PSMF1
0.000543
0.0476
lower



macropain) inhibitor



subunit 1 (PI31)


AF348514
prothymosin, alpha (gene
PTMA
0.000083
0.0203
higher



sequence 28)


NM_002872
ras-related C3 botulinum
RAC2
0.000088
0.0203
lower



toxin substrate 2 (rho



family, small GTP binding



protein Rac2)


NM_021039
S100 calcium binding
S100A11
0.000329
0.0389
lower



protein A11 (calgizzarin)


NM_014845
Sac domain-containing
SAC3
0.000342
0.0390
higher



inositol phosphatase 3


NM_003930
src family associated
SCAP2
0.000095
0.0213
higher



phosphoprotein 2


NM_012430
SEC22 vesicle trafficking
SEC22L2
0.000146
0.0266
lower



protein-like 2 (S. cerevisiae)


AV702810
SET translocation
SET
0.000070
0.0184
higher



(myeloid leukemia-



associated)


NM_031286
SH3 domain binding
SH3BGRL3
0.000150
0.0266
lower



glutamic acid-rich protein



like 3


NM_020239
small protein effector 1 of
SPEC1
0.000071
0.0184
higher



Cdc42


AW149364
SFRS protein kinase 2
SRPK2
0.000004
0.0087
higher


M25077
Sjogren syndrome antigen
SSA2
0.000328
0.0389
higher



A2 (60 kDa,



ribonucleoprotein



autoantigen SS-A/Ro)


NM_004760
serine/threonine kinase 17a
STK17A
0.000096
0.0213
lower



(apoptosis-inducing)


NM_016930
syntaxin 18
STX18
0.000105
0.0225
higher


NM_000544
transporter 2, ATP-binding
TAP2
0.000378
0.0408
lower



cassette, sub-family B



(MDR/TAP)


NM_006521
transcription factor binding
TFE3
0.000385
0.0408
lower



to IGHM enhancer 3


AL031651
tranglutaminase 2
TGM2
0.000018
0.0099
lower


BG403671
THO complex 2
THOC2
0.000015
0.0088
higher


NM_003807
tumor necrosis factor
TNFSF14
0.000195
0.0310
higher



(ligand) superfamily,



member 14


BF110993
translocated promoter
TPR
0.000003
0.0087
higher



region (to activated MET



oncogene)


U84404
ubiquitin protein ligase
UBE3A
0.000066
0.0184
higher



E3A (human papilloma



virus E6-associated



protein, Angelman



syndrome)


AI557312
Unknown
Unknown
0.000011
0.0087
higher


AW301861
Unknown
Unknown
0.000014
0.0087
higher


AV726646
Unknown
Unknown
0.000027
0.0123
higher


BE737027
Unknown
Unknown
0.000047
0.0182
higher


AA910371
Unknown
Unknown
0.000057
0.0184
higher


BF680255
Unknown
Unknown
0.000116
0.0237
higher


BE857772
Unknown
Unknown
0.000272
0.0364
higher


AI345238
Unknown
Unknown
0.000292
0.0385
higher


BF984434
Unknown
Unknown
0.000349
0.0390
higher


AA292281
Unknown
Unknown
0.000377
0.0408
higher


BF796940
Unknown
Unknown
0.000408
0.0413
higher


U82278
Unknown
Unknown
0.000450
0.0436
lower


AV753392
Unknown
Unknown
0.000529
0.0468
higher


U79458
WW domain binding
WBP2
0.000013
0.0087
higher



protein 2


BE729523
HbxAg transactivated
XTP2
0.000012
0.0087
higher



protein 2


BC002323
Zyxin
ZYX
0.000309
0.0389
lower
















TABLE 11










TOP 100 GENES IDENTIFIED BY GENECLUSTER AS ASSOCIATED WITH INCREASED EXPRESSION LEVELS


IN MENINGOENCEPHALITIS PATIENTS
















5%



Accession
Gene name (sorted


Permuted



number
by ANOVA FDR)
Gene description
Score
Score
ANOVA FDR















NM_025080
ASRGL1
asparaginase like 1
1.47
1.40
0.009


NM_013448
BAZ1A
bromodomain adjacent to zinc finger domain, 1A
0.77
0.76
0.009


NM_001969
EIF5
eukaryotic translation initiation factor 5
0.91
0.89
0.009


AK025600
KIAA0553
KIAA0553 protein
0.72
0.70
0.009


NM_004735
LRRFIP1
leucine rich repeat (in FLII) interacting protein 1
0.74
0.72
0.009


NM_003138
SRPK2
SFRS protein kinase 2
0.82
0.81
0.009


XM_211847
Unknown
Unknown
0.92
0.90
0.009


NM_001862
Unknown
Unknown
0.77
0.76
0.009


XM_047325
THOC2
THO complex 2
0.74
0.72
0.009


BQ772224
Unknown
Unknown
0.86
0.85
0.012


NM_006738
AKAP13
A kinase (PRKA) anchor protein 13
0.75
0.74
0.016


NM_020239
SPEC1
Small protein effector 1 of Cdc42
0.88
0.86
0.018


NM_130839
UBE3A
ubiquitin protein ligase E3A (human papilloma virus E6-associated protein,
1.35
1.31
0.018




Angelman syndrome)


NM_002823
PTMA
prothymosin, alpha (gene sequence 28)
0.89
0.88
0.020


NM_003930
SCAP2
src family associated phosphoprotein 2
1.65
1.46
0.021


NM_016930
STX18
syntaxin 18
1.31
1.20
0.022


NM_001015
Unknown
Unknown
0.77
0.76
0.024


NM_033360
KRAS2
v-Ki-ras2 Kirsten rat sarcoma 2 viral oncogene homolog
0.73
0.71
0.025


NM_004634
BRPF1
bromodomain and PHD finger containing, 1
0.92
0.90
0.027


NM_004279
PMPCB
Peptidase (mitochondrial processing) beta
0.83
0.82
0.030


NM_005348
HSPCA
heat shock 90 kDa protein 1, alpha
0.73
0.71
0.032


NM_005890
GAS7
growth arrest-specific 7
0.72
0.70
0.034


NM_004774
PPARBP
PPAR binding protein
1.37
1.34
0.039


NM_015355
JJAZ1
joined to JAZF1
0.92
0.90
0.039


NM_014763
MRPL19
mitochondrial ribosomal protein L19
0.89
0.88
0.039


NM_004600
SSA2
Sjogren syndrome antigen A2 (60 kDa, ribonucleoprotein autoantigen SS-
0.9
0.89
0.039




A/Ro)


NM_014845
SAC3
Sac domain-containing inositol phosphatase 3
0.76
0.74
0.039


NM_001328
Unknown
Unknown
0.75
0.74
0.039


NM_016311
ATPIF1
ATPase inhibitory factor 1
0.73
0.71
0.041


AK022200
DDX52
DEAD (Asp-Glu-Ala-Asp) box polypeptide 52
0.84
0.83
0.041


NM_007358
M96
likely ortholog of mouse metal response element binding transcription factor 2
0.92
0.92
0.041


NM_002370
MAGOH
mago-nashi homolog, proliferation-associated (Drosophila)
0.75
0.73
0.045


NM_012201
GLG1
Golgi apparatus protein 1
0.76
0.75
0.057


NM_004539
NARS
asparaginyl-tRNA synthetase
0.83
0.83
0.062


NM_012385
P8
p8 protein (candidate of metastasis 1)
0.9
0.88
0.062


NM_000988
Unknown
Unknown
0.82
0.82
0.062


NM_006649
SDCCAG16
serologically defined colon cancer antigen 16
0.74
0.72
0.063


NM_006024
TAX1BP1
Tax1 (human T-cell leukemia virus type I) binding protein 1
0.73
0.71
0.067


NM_003328
TXK
TXK tyrosine kinase
0.78
0.78
0.068


CA313371
MBP
myelin basic protein
0.77
0.76
0.069


NM_000376
VDR
vitamin D (1,25-dihydroxyvitamin D3) receptor
0.91
0.89
0.070


NM_004623
TTC4
tetratricopeptide repeat domain 4
0.93
0.92
0.074


NM_019071
ING3
inhibitor of growth family, member 3
0.88
0.87
0.076


NM_002823
PTMA
prothymosin, alpha (gene sequence 28)
0.85
0.85
0.076


NM_014170
HSPC135
HSPC135 protein
0.74
0.73
0.079


NM_000181
GUSB
glucuronidase, beta
0.77
0.77
0.082


NM_004871
GOSR1
golgi SNAP receptor complex member 1
0.76
0.75
0.084


NM_001004
Unknown
Unknown
0.73
0.70
0.084


BC010161
ALU
ALU Sequence
0.78
0.77
0.084


AI478300
ALU
ALU Sequence
0.78
0.77
0.088


NM_014810
CAP350
centrosome-associated protein 350
0.81
0.81
0.088


CB530067
CTSB
cathepsin B
0.76
0.76
0.088


NM_004442
EPHB2
EphB2
0.75
0.73
0.088


NM_005102
FEZ2
fasciculation and elongation protein zeta 2 (zygin II)
0.77
0.76
0.088


NM_014171
CRIPT
postsynaptic protein CRIPT
0.74
0.72
0.090


NM_001412
EIF1A
eukaryotic translation initiation factor 1A
0.92
0.90
0.094


AB095946
IPO9
importin 9
0.78
0.78
0.094


NM_152227
SNX5
sorting nexin 5
0.73
0.71
0.094


NM_144498
OSBPL2
oxysterol binding protein-like 2
0.76
0.76
0.096


NM_015523
DKFZP566E144
small fragment nuclease
0.76
0.76
0.096


BC036583
PRKAR2A
protein kinase, cAMP-dependent, regulatory, type II, alpha
0.84
0.83
0.097


AK021482
ALAD
aminolevulinate, delta-, dehydratase
0.78
0.78
0.097


NM_001637
AOAH
acyloxyacyl hydrolase (neutrophil)
0.82
0.81
>0.1


NM_005104
BRD2
bromodomain containing 2
0.75
0.74
>0.1


NM_003796
C19ORF2
chromosome 19 open reading frame 2
0.88
0.87
>0.1


NM_006807
CBX1
Chromobox homolog 1 (HP1 beta homolog Drosophila)
0.82
0.81
>0.1


NM_016052
CGI-115
CGI-115 protein
0.77
0.76
>0.1


NM_022802
CTBP2
C-terminal binding protein 2
0.82
0.82
>0.1


NM_006565
CTCF
CCCTC-binding factor (zinc finger protein)
0.75
0.74
>0.1


AW984453
CUGBP1
CUG triplet repeat, RNA binding protein 1
0.72
0.70
>0.1


NM_001363
DKC1
dyskeratosis congenita 1, dyskerin
0.73
0.71
>0.1


NM_015497
DKFZP564G2022
DKFZP564G2022 protein
0.83
0.82
>0.1


NM_173801
FLJ12178
hypothetical protein FLJ12178
0.88
0.87
>0.1


AF271783
FLJ21174
hypothetical protein FLJ21174
0.92
0.91
>0.1


NM_002027
FNTA
farnesyltransferase, CAAX box, alpha
0.76
0.75
>0.1


NM_177442
FTSJ2
FtsJ homolog 2 (E. coli)
0.73
0.72
>0.1


NM_024629
KLIP1
KSHV latent nuclear antigen interacting protein 1
0.8
0.80
>0.1


XM_300615
KNS2
kinesin 2 60/70 kDa
0.74
0.72
>0.1


NM_018479
LOC55862
uncharacterized hypothalamus protein HCDASE
0.73
0.71
>0.1


NM_014462
LSM1
LSM1 homolog, U6 small nuclear RNA associated (S. cerevisiae)
0.74
0.73
>0.1


NM_016019
LUC7L2
LUC7-like 2 (S. cerevisiae)
0.74
0.73
>0.1


NM_018848
MKKS
McKusick-Kaufman syndrome
0.82
0.81
>0.1


NM_018657
MYNN
Myoneurin
0.74
0.73
>0.1


NM_017852
NALP2
NACHT, LRR and PYD containing protein 2
0.75
0.73
>0.1


NM_002484
NUBP1
nucleotide binding protein 1 (MinD homolog, E. coli)
0.76
0.75
>0.1


NM_002552
ORC4L
Origin recognition complex, subunit 4-like (yeast)
0.81
0.81
>0.1


NM_002815
PSMD11
proteasome (prosome, macropain) 26S subunit, non-ATPase, 11
0.77
0.76
>0.1


NM_014676
PUM1
pumilio homolog 1 (Drosophila)
0.73
0.71
>0.1


AK024423
RNASEH1
ribonuclease H1
0.83
0.82
>0.1


NM_006414
RPP38
ribonuclease P (38 kD)
0.76
0.75
>0.1


NM_013306
SNX15
sorting nexin 15
0.76
0.74
>0.1


NM_003563
SPOP
speckle-type POZ protein
0.76
0.75
>0.1


NM_003765
STX10
syntaxin 10
0.84
0.84
>0.1


NM_006351
TIMM44
Translocase of inner mitochondrial membrane 44 homolog (yeast)
0.83
0.82
>0.1


NM_003313
TSTA3
Tissue specific transplantation antigen P35B
0.81
0.81
>0.1


NM_003314
TTC1
tetratricopeptide repeat domain 1
0.75
0.73
>0.1


NM_180699
U1SNRNPBP
U1-snRNP binding protein homolog
0.72
0.70
>0.1


AK092175
Unknown
Unknown
0.73
0.70
>0.1


NM_017528
WBSCR22
Williams Beuren syndrome chromosome region 22
0.73
0.71
>0.1


NM_018253
YAP
YY1 associated protein
0.77
0.77
>0.1
















TABLE 12










TOP 50 GENES IDENTIFIED BY GENECLUSTER AS


ASSOCIATED WITH LOWER EXPRESSION LEVELS


IN MENINGOENCEPHALITIS PATIENTS












Gene name





Accession
(sorted by


ANOVA


number
ANOVA FDR)
Gene description
Score
FDR














NM_032673
NSPC1
likely ortholog of mouse nervous
1.01
0.0087




system polycomb 1


NM_012478
WBP2
WW domain binding protein 2
0.89
0.0087


NM_000201
ICAM1
intercellular adhesion molecule 1
0.87
0.0087




(CD54), human rhinovirus receptor


NM_001655
ARCN1
archain 1
0.88
0.0092


BQ581290
TGM2
transglutaminase 2
0.91
0.0099


XM_300774
AGRN
agrin
0.91
0.0169


NM_024845
FLJ14154
hypothetical protein FLJ14154
0.88
0.0184


NM_001707
BCL7B
B-cell CLL/lymphoma 7B
0.86
0.0184


NM_014940
KIAA0872
KIAA0872 protein
0.86
0.0184


NM_001693
ATP6V1B2
ATPase, H+ transporting, lysosomal
0.9
0.0203




56/58 kDa, V1 subunit B, isoform 2


NM_017450
BAIAP2
BAI1-associated protein 2
1.09
0.0271


NM_018115
FLJ10498
hypothetical protein FLJ10498
0.89
0.0300


NM_002727
PRG1
proteoglycan 1, secretory granule
0.92
0.0328


NM_014481
APEX2
APEX nuclease
1.05
0.0330




(apurinic/apyrimidinic endonuclease) 2


NM_002569
FURIN
furin (paired basic amino acid
1.16
0.0363




cleaving enzyme)


NM_000544
TAP2
transporter 2, ATP-binding cassette,
1.09
0.0408




sub-family B (MDR/TAP)


NM_006521
TFE3
transcription factor binding to IGHM
0.88
0.0408




enhancer 3


NM_005620
S100A11
S100 calcium binding protein A11
0.87
0.0408




(calgizzarin)


BM930256
EMP1
epithelial membrane protein 1
0.89
0.0442


NM_024165
PHF1
PHD finger protein 1
0.94
0.0449


NM_014741
KIAA0652
KIAA0652 gene product
0.84
0.0485


CD644158
RAB5B
RAB5B, member RAS oncogene
0.88
0.0596




family


NM_004309
ARHGDIA
Rho GDP dissociation inhibitor
0.83
0.0619




(GDI) alpha


AI911044
PDI2
peptidyl arginine deiminase, type II
0.84
0.0651


XM_114002
NY-REN-24
NY-REN-24 antigen
0.89
0.0659


NM_002355
M6PR
mannose-6-phosphate receptor
0.86
0.0705




(cation dependent)


CD643922
STK17B
serine/threonine kinase 17b
0.87
0.0711




(apoptosis-inducing)


NM_012075
C16ORF35
chromosome 16 open reading frame
0.91
0.0742




35


AB002368
XPO6
exportin 6
0.92
0.0793


NM_148175
PPIL2
peptidylprolyl isomerase
0.86
0.0836




(cyclophilin)-like 2


NM_022060
ABHD4
abhydrolase domain containing 4
0.96
0.0884


NM_014734
KIAA0247
KIAA0247 gene product
0.93
0.0884


NM_078467
CDKN1A
cyclin-dependent kinase inhibitor 1A
0.84
0.0884




(p21, Cip1)


NM_002530
NTRK3
neurotrophic tyrosine kinase,
1.07
0.0945




receptor, type 3


AK023816
UNK_AK023816

Homo sapiens cDNA FLJ13754 fis,

0.86
0.0979




clone PLACE3000362.


NM_000201
ICAM1
intercellular adhesion molecule 1
1.02
>0.1




(CD54), human rhinovirus receptor


BU627400
CTSB
cathepsin B
0.97
>0.1


BX117520
FLJ13910
hypothetical protein FLJ13910
0.94
>0.1


AW969709
F2RL1
coagulation factor II (thrombin)
0.94
>0.1




receptor-like 1


XM_114002
NY-REN-24
NY-REN-24 antigen
0.91
>0.1


AB040972
FLJ11560
hypothetical protein FLJ11560
0.91
>0.1


NM_006865
LILRA3
leukocyte immunoglobulin-like
0.9
>0.1




receptor, subfamily A (without TM




domain), member 3


BI020084
MKRN1
makorin, ring finger protein, 1
0.9
>0.1


NM_170665
ATP2A2
ATPase, Ca++ transporting, cardiac
0.89
>0.1




muscle, slow twitch 2


NM_016525
UBAP1
ubiquitin associated protein 1
0.87
>0.1


NM_000167
GK
glycerol kinase
0.85
>0.1


NM_015444
RIS1
Ras-induced senescence 1
0.84
>0.1


NM_000270
NP
nucleoside phosphorylase
0.84
>0.1


NM_023079
FLJ13855
hypothetical protein FLJ13855
0.84
>0.1
















TABLE 13










GENES THAT CAPTURE FIVE-OUT-OF-FIVE


MENINGOENCEPHALITIS PATIENTS















Cutoff level for




Accession


association with
Unadjusted


number
Gene name
Gene description
meningoencephalitis
p value
FDR















AA284705
ICAM1
intercellular adhesion
F < 5
0.000010
0.0087




molecule 1 (CD54),




human rhinovirus receptor


NM_001343
DAB2
disabled homolog 2,
F > 11
0.000065
0.0184




mitogen-responsive




phosphoprotein




(Drosophila)


U84404
UBE3A
ubiquitin protein ligase
F > 16
0.000066
0.0184




E3A (human papilloma




virus E6-associated




protein, Angelman




syndrome)


AF348514
PTMA
prothymosin, alpha (gene
F > 80
0.000083
0.0203




sequence 28)


NM_003930
SCAP2
src family associated
F > 75
0.000095
0.0213




phosphoprotein 2


NM_016930
STX18
syntaxin 18
F > 25
0.000105
0.0225


AI972475
LYRIC
LYRIC/3D3
F > 20
0.000129
0.0252


U26455
ATM
ataxia telangiectasia
F > 24
0.000232
0.0338




mutated (includes




complementation groups




A, C and D)


NM_016281
JIK
STE20-like kinase
F > 48
0.000250
0.0352


NM_002569
FURIN
furin (paired basic amino
F < 10
0.000264
0.0363




acid cleaving enzyme;




PACE)


BE646414
GGA2
golgi associated, gamma
F > 256
0.000271
0.0364




adaptin ear containing,




ARF binding protein 2


NM_004774
PPARBP
PPAR binding protein
F > 8
0.000296
0.0386


NM_017775
FLJ20343
hypothetical protein
F > 39
0.000315
0.0389




FLJ20343


NM_000544
TAP2
transporter 2, ATP-
F < 5
0.000378
0.0408




binding cassette,




subfamily B (MDR/TAP)
















TABLE 14










Selected examples of accuracy of classification


using expression patterns associated


with meningoencephalitis













Meningoen-






cephalitis
Nonencephalitis





patients
patients
Identification




correctly
incorrectly
numbers




identifieda/
identified/
of patients


Gene
Metric
total (%)
total (%)
incorrectly


name
applied
(5 in group)
(103 in group)
classified





SRPK2
F > 5
3/(60%)
 3/(3%)
8, 252, 752


NKTR
F > 5
3/(60%)
 3/(3%)
8, 252, 752


TPR
present
3/(60%)
 4/(4%)
8, 14, 252, 752


ASRGL1
F > 20
4/(80%)
 5/(5%)
43, 53, 273,






297, 316*


ASRGL1
F > 12
5/(100%)
22/(21%)
2, 6, 8, 15, 22,






23, 24, 25, 36,






40, 43, 53, 60,






254, 270, 271,






273, 295, 297,






312, 316,






753**


SCAP2
F > 75
5/(100%)
11/(11%)
5, 7, 12, 14, 24,






32, 258, 271,






753, 755, 758


DAB2
F > 11
5/(100%)
13/(13%)
6, 8, 14, 15, 32,






50, 254, 271,






281, 300,






316, 753, 755








aFrequency cutoffs were selected as capturing the number of meningoencephalitis patients indicated. The total number of nonencephalitis patients incorrectly classified due to expressing at least one gene



# above the cutoff is 36 (out of 103). If the requirement to capture encephalitis patient 301 is dropped, the total number of nonencephalitis patients misclassified due to expressing at least one gene above cutoff is 9.




*All of these patients are male. Therefore, data for these patients were not considered in calculating the statistical significance of the association of this gene with meningoencephalitis.





**Of these patients, 14 are male. Data from males were not used in calculating the association between expression level and meningoencephalitis.














TABLE 15










Examples of genes that show an association with meningoencephalitis


in both AN1792-stimulated and control cultures














p value metric:
FDR metric:






gene
gene




frequency in
frequency in
p value metric:
FDR metric:



Gene name
antigen-
antigen-
gene frequency in
gene frequency in


Accession
(sorted
positive
positive
antigen-negative
antigen-negative


number
alphabetically)
cultures
cultures
cultures
cultures





M90360
AKAP13
0.000037
0.016
0.000035
0.017


NM_025080
ASRGL1
0.000001
0.009
0.000076
0.024


AA102574
BAZ1A
0.000010
0.009
0.000021
0.013


NM_001343
DAB2
0.000065
0.018
0.000286
0.055


BG530850
DDX18
0.000189
0.030
0.000372
0.066


AW081113
DKFZP564B0769
0.000008
0.009
0.000001
0.002


BG481972
EIF5
0.000006
0.009
0.000019
0.013


NM_004459
FALZ
0.000059
0.018
0.000054
0.020


NM_017736
FLJ20274
0.000080
0.020
0.000027
0.014


AU145053
FNBP1
0.000409
0.041
0.000067
0.024


BE646414
GGA2
0.000271
0.036
0.000809
0.092


BG420237
HSPCA
0.000207
0.032
0.000081
0.024


NM_000201
ICAM1
0.000010
0.009
0.001032
0.100


AI673812
KIAA0553
0.000014
0.009
0.000002
0.003


BF673699
KRAS2
0.000127
0.025
0.000083
0.024


BF965566
LRRFIP1
0.000011
0.009
0.000056
0.020


AI972475
LYRIC
0.000129
0.025
0.000007
0.006


AI566096
M96
0.000411
0.041
0.000205
0.045


AI361805
NKTR
0.000006
0.009
0.000017
0.013


AW149364
SRPK2
0.000004
0.009
0.000025
0.013


BG403671
THOC2
0.000015
0.009
0.001291
0.108


BF110993
TPR
0.000003
0.009
0.000000
0.002


BE729523
XTP2
0.000012
0.009
0.000001
0.002
















TABLE 16










Examples of genes that show an association with meningo-


encephalitis in AN1792-stimulated cultures and no


association in control cultures














p value
FDR:
p value
FDR




metric:
metric:
metric:
metric:




gene
gene
gene
gene



Gene
frequency
frequency
frequency
frequency



name
in
in
in
in



(sorted
antigen-
antigen-
antigen-
antigen-


Accession
alpha-
positive
positive
negative
negative


number
betically)
cultures
cultures
cultures
cultures















NM_014481
APEX2
0.000221
0.033002
0.863468
0.959865


NM_017450
BAIAP2
0.00016
0.027081
0.853139
0.957957


NM_001707
BCL7B
0.000069
0.018403
0.540282
0.827374


AF098641
CD44
0.000152
0.026582
0.705258
0.903721


NM_018115
FLJ10498
0.000183
0.029962
0.919492
0.979341
















TABLE 17










Genes associated with meningoencephalitis by GeneCluster analysis using the ratio metric












Gene name






(sorted


Accession
alpha-

Unadjusted
ANOVA


number
betically)
Gene description
p value
FDR














NM_014576
ACF
apobec-1 complementation factor
0.000329
0.223274


U48705
DDR1
discoidin domain receptor family,
0.00004
0.10434




member 1


NM_000173
GP1BA
glycoprotein Ib (platelet), alpha
0.000694
0.301068




polypeptide


AK024651
GPR107
G protein-coupled receptor 107
0.000131
0.111287


BC002842
HIST1H2BD
histone 1, H2bd
0.000051
0.10434


BE888744
IFIT2
interferon-induced protein with




tetratricopeptide repeats 2
0.000504
0.269983


AU150943
LOC221061
hypothetical protein LOC221061
0.000062
0.105681


NM_005446
P2RXL1
purinergic receptor P2X-like 1,
0.001729
0.342444




orphan receptor


AL512687
PM5
pM5 protein
0.001386
0.319676


M57399
PTN
pleiotrophin (heparin binding growth
0.00206
0.342444




factor 8, neurite growth-promoting




factor 1)


NM_021908
ST7
suppression of tumorigenicity 7
0.000025
0.10434


AK023816
UNK_AK023816

Homo sapiens cDNA FLJ13754 fis, clone

0.000098
0.110884




PLACE3000362.


X93006
Unknown
Unknown
0.001095
0.301068
















TABLE 18










Genes associated with IgG responsiveness by ANOVA using gene frequency metric


(FDR < 0.011)



















Average








expression in








IgG








nonresponders



Gene name




relative to


Accession
(sorted by

Unadjusted
ANOVA
Adjusted
average in IgG


number
p value)
Gene description
p value
FDR
p value
responders





NM_013417
IARS
isoleucine-tRNA
6.389E−07
0.004997
0.01
lower




synthetase


NM_004279
PMPCB
peptidase (mitochondrial
9.394E−07
0.004997
0.01
lower




processing) beta


NM_173638
Unknown
Unknown
0.000001
0.004997
0.01
lower


NM_005736
ACTR1A
ARP1 actin-related protein
0.000002
0.004997
0.02
higher




1 homolog A, centractin




alpha (yeast)


NM_000099
CST3
cystatin C (amyloid
0.000002
0.004997
0.02
higher




angiopathy and cerebral




hemorrhage)


NM_004107
FCGRT
Fc fragment of IgG,
0.000002
0.004997
0.02
higher




receptor, transporter, alpha


XM_086398
Unknown
Unknown
0.000002
0.004997
0.02
lower


NM_003328
TXK
TXK tyrosine kinase
0.000003
0.007541
0.04
lower


NM_000754
COMT
catechol-O-
0.000004
0.007702
0.05
higher




methyltransferase


NM_002087
GRN
Granulin
0.000004
0.007541
0.04
higher


NM_002388
MCM3
MCM3 minichromosome
0.000005
0.007702
0.05
lower




maintenance deficient 3




(S. cerevisiae)


NM_024835
LZK1
C3HC4-type zinc finger
0.000006
0.009032
0.07
lower




protein


NM_005216
DDOST
dolichyl-
0.000009
0.010942
0.09
higher




diphosphooligosaccharide-




protein glycosyltransferase


NM_005022
PFN1
profilin 1
0.000009
0.010942
0.09
higher


XM_295598
SF3A1
splicing factor 3a, subunit
0.000009
0.010942
0.09
lower




1, 120 kDa
















TABLE 19










Response groups as segregated by the four genes most strongly


associated with IgG responsiveness by GeneCluster









Patients in 70th percentile of gene frequency level



associated with IgG responsiveness












Average






expression



in IgG



nonresponders
IgG non-
IgG
IgG partial



relative
responders/
responders/
responders/



to average
(%) (60
(%) (22
(%) (26


Gene
in IgG
patients
patients
patients


name
responders
in group)
in group)
in group)





Granulin
Higher
25/(42%)
0/(0%)
7/(27%)


FCGRT
Higher
26/(43%)
0/(0%)
6/(23%)


IARS
Lower
25/(42%)
1/(5%)
6/(23%)


MCM3
Lower
23/(38%)
1/(5%)
9/(34%)
















TABLE 20










Samples Received By Pharmacogenomic Laboratory









Number












Enrolled U.S. patients
172


Enrolled patients who consented to pharmacogenomic
167


portion of study


Samples within shipping specifications
161


Samples that generated data from chips that
153


met all QC criteria
















TABLE 21










Patients in Study by Year of Birth










Age (years)
Number of Patients














≧80
29



70-80
61



60-70
29



50-60
4



Cumulative total
123

















TABLE 22










Samples in Pharmacogenomic Study











Male
Female
Total














Placebo
12
18
30


Treated
66
57
123


Treated IgG responders
12
13
25


Treated IgG partial responders
12
16
28


Treated IgG nonresponders
42
28
70


Treated IgM responders
20
20
40


Treated IgM partial responders
18
21
39


Treated IgM nonresponders
28
16
44


Meningoencephalitis patients
0
5
5



















TABLE 23











Affymetrix




Qualifier
Gene Name









213266_at
76P



209993_at
ABCB1



202804_at
ABCC1



213485_s_at
ABCC10



214033_at
ABCC6



201873_s_at
ABCE1



200965_s_at
ABLIM1



49452_at
ACACB



222011_s_at
ACAT2



221641_s_at
ACATE2



201630_s_at
ACP1



204393_s_at
ACPP



207275_s_at
ACSL1



202422_s_at
ACSL4



200720_s_at
ACTR1A



219623_at
ACTR5



204639_at
ADA



202604_x_at
ADAM10



202381_at
ADAM9



202912_at
ADM



204183_s_at
ADRBK2



211071_s_at
AF1Q



203566_s_at
AGL



201491_at
AHSA1



212980_at
AHSA2



215051_x_at
AIF1



209901_x_at
AIF1



213095_x_at
AIF1



212543_at
AIM1



202587_s_at
AK1



201675_at
AKAP1



203156_at
AKAP11



210517_s_at
AKAP12



201425_at
ALDH2



214221_at
ALMS1



214366_s_at
ALOX5



204446_s_at
ALOX5



202125_s_at
ALS2CR3



204294_at
AMT



218575_at
ANAPC1



206385_s_at
ANK3



212289_at
ANKRD12



213005_s_at
ANKRD15



213035_at
ANKRD28



202888_s_at
ANPEP



201012_at
ANXA1



201590_x_at
ANXA2



210427_x_at
ANXA2



208816_x_at
ANXA2P2



201302_at
ANXA4



200782_at
ANXA5



205639_at
AOAH



221937_at
AP1GBP1



64418_at
AP1GBP1



203300_x_at
AP1S2



211047_x_at
AP2S1



203410_at
AP3M2



202442_at
AP3S1



209870_s_at
APBA2



209871_s_at
APBA2



221492_s_at
APG3L



201687_s_at
API5



211404_s_at
APLP2



208702_x_at
APLP2



214875_x_at
APLP2



208703_s_at
APLP2



221087_s_at
APOL3



202268_s_at
APPBP1



202630_at
APPBP2



39248_at
AQP3



205568_at
AQP9



201526_at
ARF5



202211_at
ARFGAP3



57082_at
ARH



221790_s_at
ARH



38149_at
ARHGAP25



37117_at
ARHGAP8



213039_at
ARHGEF18



208736_at
ARPC3



211963_s_at
ARPC5



210980_s_at
ASAH1



213902_at
ASAH1



212818_s_at
ASB1



206743_s_at
ASGR1



206130_s_at
ASGR2



204244_s_at
ASK



205047_s_at
ASNS



218987_at
ATF7IP



208758_at
ATIC



212672_at
ATM



203454_s_at
ATOX1



207522_s_at
ATP2A3



211755_s_at
ATP5F1



207809_s_at
ATP6AP1



200078_s_at
ATP6V0B



212041_at
ATP6V0D1



200096_s_at
ATP6V0E



201972_at
ATP6V1A



201089_at
ATP6V1B2



201527_at
ATP6V1F



209903_s_at
ATR



208002_s_at
BACH



221234_s_at
BACH2



217911_s_at
BAG3



219667_s_at
BANK1



202121_s_at
BC-2



203053_at
BCAS2



214390_s_at
BCAT1



202030_at
BCKDK



219528_s_at
BCL11B



203685_at
BCL2



205681_at
BCL2A1



203140_at
BCL6



206465_at
BG1



204493_at
BID



210201_x_at
BIN1



210538_s_at
BIRC3



202592_at
BLOC1S1



206126_at
BLR1



211729_x_at
BLVRA



203773_x_at
BLVRA



215460_x_at
BRD1



205715_at
BST1



204901_at
BTRC



202096_s_at
BZRP



218889_at
C10ORF117



220147_s_at
C12ORF14



219099_at
C12ORF5



218422_s_at
C13ORF10



218852_at
C14ORF10



218139_s_at
C14ORF108



212460_at
C14ORF147



219526_at
C14ORF169



219316_s_at
C14ORF58



221940_at
C18B11



222099_s_at
C19ORF13



214173_x_at
C19ORF2



213390_at
C19ORF7



218456_at
C1QDC1



202878_s_at
C1QR1



217835_x_at
C20ORF24



212996_s_at
C21ORF108



203996_s_at
C21ORF2



221984_s_at
C2ORF17



213615_at
C3F



210054_at
C4ORF15



214661_s_at
C4ORF9



220751_s_at
C5ORF4



220088_at
C5R1



218195_at
C6ORF211



219006_at
C6ORF66



218877_s_at
C6ORF75



218116_at
C9ORF78



219147_s_at
C9ORF95



221631_at
CACNA1I



211984_at
CALM1



210349_at
CAMK4



218309_at
CAMKIINALPHA



212252_at
CAMKK2



201850_at
CAPG



201238_s_at
CAPZA2



201949_x_at
CAPZB



37012_at
CAPZB



218929_at
CARF



211208_s_at
CASK



206011_at
CASP1



211367_s_at
CASP1



209970_x_at
CASP1



207467_x_at
CAST



207625_s_at
CBFA2T2



209682_at
CBLB



212914_at
CBX7



204655_at
CCL5



1405_i_at
CCL5



200953_s_at
CCND2



208796_s_at
CCNG1



213743_at
CCNT2



221511_x_at
CCPG1



205098_at
CCR1



206337_at
CCR7



201947_s_at
CCT2



206587_at
CCT6B



201743_at
CD14



203645_s_at
CD163



215049_x_at
CD163



208653_s_at
CD164



205789_at
CD1D



205831_at
CD2



206545_at
CD28



209555_s_at
CD36



206488_s_at
CD36



213539_at
CD3D



206804_at
CD3G



210031_at
CD3Z



216942_s_at
CD58



211744_s_at
CD58



205173_x_at
CD58



213958_at
CD6



200663_at
CD63



203507_at
CD68



209795_at
CD69



214049_x_at
CD7



210895_s_at
CD86



205758_at
CD8A



206761_at
CD96



205627_at
CDA



213151_s_at
CDC10



221556_at
CDC14B



209658_at
CDC16



201853_s_at
CDC25B



207318_s_at
CDC2L5



209288_s_at
CDC42EP3



209286_at
CDC42EP3



218157_x_at
CDC42SE1



204995_at
CDK5R1



218315_s_at
CDK5RAP1



209501_at
CDR2



204029_at
CELSR2



204066_s_at
CENTG2



205642_at
CEP1



202195_s_at
CGI-100



218102_at
CGI-26



219590_x_at
CGI-30



214426_x_at
CHAF1A



219049_at
CHGN



214665_s_at
CHP



204065_at
CHST10



221059_s_at
CHST6



221058_s_at
CKLF



219161_s_at
CKLF



212752_at
CLASP1



219947_at
CLECSF6



208659_at
CLIC1



221042_s_at
CLMN



200743_s_at
CLN2



204050_s_at
CLTA



207270_x_at
CMRF35



203291_at
CNOT4



203642_s_at
COBLL1



203073_at
COG2



208818_s_at
COMT



221676_s_at
CORO1C



203663_s_at
COX5A



211025_x_at
COX5B



201943_s_at
CPD



201940_at
CPD



210069_at
CPT1B



208146_s_at
CPVL



201200_at
CREG



210766_s_at
CSE1L



203104_at
CSF1R



203591_s_at
CSF3R



202332_at
CSNK1E



204619_s_at
CSPG2



215646_s_at
CSPG2



211571_s_at
CSPG2



204620_s_at
CSPG2



221731_x_at
CSPG2



201201_at
CSTB



212905_at
CSTF2T



203947_at
CSTF3



218924_s_at
CTBS



200765_x_at
CTNNA1



210844_x_at
CTNNA1



200839_s_at
CTSB



200838_at
CTSB



201487_at
CTSC



200766_at
CTSD



203657_s_at
CTSF



202295_s_at
CTSH



202087_s_at
CTSL



202902_s_at
CTSS



209665_at
CYB561D2



203922_s_at
CYBB



203923_s_at
CYBB



201066_at
CYC1



208923_at
CYFIP1



215785_s_at
CYFIP2



221903_s_at
CYLD



213295_at
CYLD



201926_s_at
DAF



201678_s_at
DC12



203799_at
DCL-1



204246_s_at
DCTN3



218013_x_at
DCTN4



214909_s_at
DDAH2



202262_x_at
DDAH2



203409_at
DDB2



212690_at
DDHD2



201241_at
DDX1



204977_at
DDX10



208149_x_at
DDX11



208159_x_at
DDX11



208896_at
DDX18



200694_s_at
DDX24



218819_at
DDX26



215693_x_at
DDX27



219108_x_at
DDX27



221780_s_at
DDX27



205000_at
DDX3Y



220890_s_at
DDX47



202447_at
DECR1



215158_s_at
DEDD



205382_s_at
DF



203385_at
DGKA



217989_at
DHRS8



212674_s_at
DHX30



205726_at
DIAPH2



219374_s_at
DIBD1



201479_at
DKC1



221541_at
DKFZP434B044



202560_s_at
DKFZP547E1010



213657_s_at
DKFZP547K1113



37590_g_at
DKFZP547K1113



212333_at
DKFZP564F0522



221265_s_at
DKFZP564O1664



210006_at
DKFZP564O243



208092_s_at
DKFZP566A1524



221970_s_at
DKFZP586L0724



213199_at
DKFZP586P0123



36552_at
DKFZP586P0123



214247_s_at
DKK3



212727_at
DLG3



201681_s_at
DLG5



218794_s_at
DLP



212730_at
DMN



203301_s_at
DMTF1



205963_s_at
DNAJA3



200666_s_at
DNAJB1



202867_s_at
DNAJB12



202500_at
DNAJB2



213088_s_at
DNAJC9



212538_at
DOCK9



208872_s_at
DP1



203717_at
DPP4



204646_at
DPYD



200762_at
DPYSL2



217868_s_at
DREV1



204751_x_at
DSC2



203635_at
DSCR3



208892_s_at
DUSP6



208891_at
DUSP6



208893_s_at
DUSP6



57532_at
DVL2



202968_s_at
DYRK2



218660_at
DYSF



218482_at
E(Y)2



219551_at
EAF2



204858_s_at
ECGF1



220048_at
EDAR



204642_at
EDG1



212830_at
EGFL5



222221_x_at
EHD1



218935_at
EHD3



201018_at
EIF1AX



201017_at
EIF1AX



201016_at
EIF1AX



201019_s_at
EIF1AX



204409_s_at
EIF1AY



209429_x_at
EIF2B4



212351_at
EIF2B5



201142_at
EIF2S1



201530_x_at
EIF4A1



31845_at
ELF4



220386_s_at
EML4



201324_at
EMP1



201325_s_at
EMP1



207610_s_at
EMR2



201313_at
ENO2



209473_at
ENTPD1



207691_x_at
ENTPD1



204076_at
ENTPD4



212375_at
EP400



204505_s_at
EPB49



200843_s_at
EPRS



202176_at
ERCC3



202414_at
ERCC5



201328_at
ETS2



201329_s_at
ETS2



204328_at
EVER1



217838_s_at
EVL



204714_s_at
F5



209271_at
FALZ



203974_at
FAM16AX



203184_at
FBN2



209696_at
FBP1



213145_at
FBXL14



209004_s_at
FBXL5



212231_at
FBXO21



218432_at
FBXO3



204232_at
FCER1G



214511_x_at
FCGR1A



216950_s_at
FCGR1A



203561_at
FCGR2A



218831_s_at
FCGRT



205237_at
FCN1



201798_s_at
FER1L3



205418_at
FES



219069_at
FGIF



204834_at
FGL2



206492_at
FHIT



201540_at
FHL1



219117_s_at
FKBP11



200709_at
FKBP1A



58780_s_at
FLJ10357



218274_s_at
FLJ10415



218993_at
FLJ10581



221806_s_at
FLJ10707



217884_at
FLJ10774



222132_s_at
FLJ10842



218125_s_at
FLJ10853



218347_at
FLJ10900



218552_at
FLJ10948



209688_s_at
FLJ10996



218307_at
FLJ11164



213694_at
FLJ11220



218633_x_at
FLJ11342



39650_s_at
FLJ11383



219361_s_at
FLJ12484



219765_at
FLJ12586



218312_s_at
FLJ12895



218370_s_at
FLJ12903



218532_s_at
FLJ20152



219734_at
FLJ20174



219646_at
FLJ20186



219809_at
FLJ20195



220306_at
FLJ20202



218652_s_at
FLJ20265



218710_at
FLJ20272



219460_s_at
FLJ20507



219258_at
FLJ20516



217961_at
FLJ20551



221229_s_at
FLJ20628



218932_at
FLJ20729



217895_at
FLJ20758



218366_x_at
FLJ20859



219315_s_at
FLJ20898



218483_s_at
FLJ21827



65635_at
FLJ21865



212918_at
FLJ22028



219435_at
FLJ22170



222143_s_at
FLJ22405



221081_s_at
FLJ22457



219359_at
FLJ22635



218454_at
FLJ22662



218754_at
FLJ23323



218776_s_at
FLJ23375



208903_at
FLJ46061



210607_at
FLT3LG



212232_at
FNBP4



200090_at
FNTA



204829_s_at
FOLR2



206015_s_at
FOXJ3



203064_s_at
FOXK2



214148_at
FOXM1



202945_at
FPGS



205119_s_at
FPR1



210773_s_at
FPRL1



210772_at
FPRL1



209702_at
FTO



205324_s_at
FTSJ1



213594_x_at
FUSIP1



209893_s_at
FUT4



209892_at
FUT4



217897_at
FXYD6



210105_s_at
FYN



202812_at
GAA



200645_at
GABARAP



219013_at
GALNT11



218885_s_at
GALNT12



213049_at
GARNL1



211067_s_at
GAS7



204793_at
GASP



209603_at
GATA3



209602_s_at
GATA3



209604_s_at
GATA3



203765_at
GCA



218912_at
GCC1



212139_at
GCN1L1



202182_at
GCN5L2



206589_at
GFI1



202722_s_at
GFPT1



208914_at
GGA2



209249_s_at
GHITM



204222_s_at
GLIPR1



204221_x_at
GLIPR1



209276_s_at
GLRX



217807_s_at
GLTSCR2



35820_at
GM2A



205349_at
GNA15



214157_at
GNAS



204000_at
GNB5



201921_at
GNG10



207157_s_at
GNG5



212335_at
GNS



212334_at
GNS



208798_x_at
GOLGIN-67



210425_x_at
GOLGIN-67



210279_at
GPR18



200736_s_at
GPX1



220864_s_at
GRIM19



204396_s_at
GRK5



211284_s_at
GRN



216041_x_at
GRN



200678_x_at
GRN



200696_s_at
GSN



201912_s_at
GSPT1



205541_s_at
GSPT2



205770_at
GSR



201470_at
GSTO1



205930_at
GTF2E1



202605_at
GUSB



214501_s_at
H2AFY



209818_s_at
HABP4



202282_at
HADH2



211699_x_at
HBA1



202300_at
HBXIP



218345_at
HCA112



219484_at
HCFC2



218450_at
HEBP1



218603_at
HECA



212815_at
HELIC1



218306_s_at
HERC1



201944_at
HEXB



203020_at
HHL



38340_at
HIP1R



209558_s_at
HIP1R



204512_at
HIVEP1



205936_s_at
HK3



205671_s_at
HLA-DOB



214438_at
HLX1



206074_s_at
HMGA1



203665_at
HMOX1



204112_s_at
HNMT



211732_x_at
HNMT



209068_at
HNRPDL



204647_at
HOMER3



208470_s_at
HPR



202854_at
HPRT1



219403_s_at
HPSE



218092_s_at
HRB



203202_at
HRB2



209971_x_at
HRI



218508_at
HSA275986



213598_at
HSA9761



204405_x_at
HSA9761



200941_at
HSBP1



209657_s_at
HSF2



221771_s_at
HSMPP8



221597_s_at
HSPC171



212493_s_at
HYPB



218805_at
IAN4L1



204744_s_at
IARS



210439_at
ICOS



203596_s_at
IFIT5



204785_x_at
IFNAR2



202727_s_at
IFNGR1



201642_at
IFNGR2



201393_s_at
IGF2R



210095_s_at
IGFBP3



212827_at
IGHM



206420_at
IGSF6



202491_s_at
IKBKAP



209575_at
IL10RB



204773_at
IL11RA



201888_s_at
IL13RA1



201887_at
IL13RA1



203679_at
IL1RL1LG



212657_s_at
IL1RN



221658_s_at
IL21R



220054_at
IL23A



205291_at
IL2RB



217804_s_at
ILF3



208594_x_at
ILT8



203126_at
IMPA2



205376_at
INPP4B



203006_at
INPP5A



204706_at
INPP5E



213792_s_at
INSR



200995_at
IPO7



200993_at
IPO7



205995_x_at
IQCB1



220034_at
IRAK3



33304_at
ISG20



201656_at
ITGA6



205055_at
ITGAE



210213_s_at
ITGB4BP



211339_s_at
ITK



202747_s_at
ITM2A



202746_at
ITM2A



203723_at
ITPKB



201189_s_at
ITPR3



206700_s_at
JARID1D



212496_s_at
JMJD2B



202138_x_at
JTV1



201464_x_at
JUN



212192_at
KCTD12



200700_s_at
KDELR2



203712_at
KIAA0020



212789_at
KIAA0056



213483_at
KIAA0073



212510_at
KIAA0089



203492_x_at
KIAA0092



203493_s_at
KIAA0092



213006_at
KIAA0146



212844_at
KIAA0179



212733_at
KIAA0226



212735_at
KIAA0226



212053_at
KIAA0251



212621_at
KIAA0286



40016_g_at
KIAA0303



212356_at
KIAA0323



203288_at
KIAA0355



203049_s_at
KIAA0372



202713_s_at
KIAA0391



203959_s_at
KIAA0478



36545_s_at
KIAA0542



212946_at
KIAA0564



212675_s_at
KIAA0582



212579_at
KIAA0650



212663_at
KIAA0674



212311_at
KIAA0746



212314_at
KIAA0746



212546_s_at
KIAA0826



212548_s_at
KIAA0826



212570_at
KIAA0830



36888_at
KIAA0841



212402_at
KIAA0853



209760_at
KIAA0922



213407_at
KIAA0931



209654_at
KIAA0947



216996_s_at
KIAA0971



213092_x_at
KIAA0974



201270_x_at
KIAA1068



213271_s_at
KIAA1117



209379_s_at
KIAA1128



209378_s_at
KIAA1128



212453_at
KIAA1279



203086_at
KIF2



203087_s_at
KIF2



221219_s_at
KLHDC4



221221_s_at
KLHL3



206785_s_at
KLRC2



211954_s_at
KPNB3



211955_at
KPNB3



201003_x_at
KUA-UEV



204385_at
KYNU



210663_s_at
KYNU



217388_s_at
KYNU



203041_s_at
LAMP2



203042_at
LAMP2



202020_s_at
LANCL1



217933_s_at
LAP3



200673_at
LAPTM4A



200618_at
LASP1



211005_at
LAT



209881_s_at
LAT



207734_at
LAX



221011_s_at
LBH



204891_s_at
LCK



204012_s_at
LCMT2



201030_x_at
LDHB



221558_s_at
LEF1



202594_at
LEPROTL1



202595_s_at
LEPROTL1



201105_at
LGALS1



208949_s_at
LGALS3



208934_s_at
LGALS8



202726_at
LIG1



210660_at
LILRA1



211100_x_at
LILRA2



207857_at
LILRA2



211101_x_at
LILRA2



210146_x_at
LILRB2



207697_x_at
LILRB2



210225_x_at
LILRB3



211133_x_at
LILRB3



211135_x_at
LILRB3



220036_s_at
LIMR



206440_at
LIN7A



201847_at
LIPA



212697_at
LOC162427



214838_at
LOC375035



221249_s_at
LOC81558



214791_at
LOC93349



47560_at
LPHN1



212276_at
LPIN1



202460_s_at
LPIN2



220532_s_at
LR8



211596_s_at
LRIG1



200785_s_at
LRP1



209841_s_at
LRRN3



202245_at
LSS



214574_x_at
LST1



211582_x_at
LST1



210629_x_at
LST1



207339_s_at
LTB



203005_at
LTBR



217842_at
LUC7L2



205859_at
LY86



215967_s_at
LY9



206584_at
LY96



202625_at
LYN



218437_s_at
LZTFL1



203362_s_at
MAD2L1



206363_at
MAF



209014_at
MAGED1



218176_at
MAGEF1



218573_at
MAGEH1



210092_at
MAGOH



204777_s_at
MAL



210017_at
MALT1



214180_at
MAN1C1



209166_s_at
MAN2B1



204089_x_at
MAP3K4



214339_s_at
MAP4K1



206296_x_at
MAP4K1



210449_x_at
MAPK14



202788_at
MAPKAPK3



201669_s_at
MARCKS



205819_at
MARCO



214363_s_at
MATR3



209332_s_at
MAX



218440_at
MCCC1



35147_at
MCF2L



212246_at
MCFD2



201930_at
MCM6



219952_s_at
MCOLN1



219066_at
MDS018



219698_s_at
METTL4



201126_s_at
MGAT1



219797_at
MGAT4A



222120_at
MGC13138



214696_at
MGC14376



221756_at
MGC17330



221904_at
MGC21688



222064_s_at
MGC2744



221255_s_at
MGC2963



212313_at
MGC29816



204699_s_at
MGC29875



204700_x_at
MGC29875



202365_at
MGC5139



221580_s_at
MGC5306



218750_at
MGC5306



200899_s_at
MGEA5



204168_at
MGST2



204917_s_at
MLLT3



200644_at
MLP



204959_at
MNDA



209583_s_at
MOX2



212885_at
MPHOSPH10



215731_s_at
MPHOSPH9



212197_x_at
M-RIP



214771_x_at
M-RIP



218027_at
MRPL15



208787_at
MRPL3



201717_at
MRPL49



209609_s_at
MRPL9



211594_s_at
MRPL9



218259_at
MRTF-B



210356_x_at
MS4A1



217418_x_at
MS4A1



219607_s_at
MS4A4A



219666_at
MS4A6A



41220_at
MSF



202911_at
MSH6



218773_s_at
MSRB



213511_s_at
MTMR1



216095_x_at
MTMR1



218716_x_at
MTO1



203774_at
MTR



210386_s_at
MTX1



207727_s_at
MUTYH



202431_s_at
MYC



201960_s_at
MYCBP2



209124_at
MYD88



212082_s_at
MYL6



213733_at
MYO1F



202423_at
MYST3



212462_at
MYST4



48612_at
N4BP1



221867_at
N4BP1



212653_s_at
NACSIN



202944_at
NAGA



218231_at
NAGK



218189_s_at
NANS



204749_at
NAP1L3



201414_s_at
NAP1L4



37005_at
NBL1



219079_at
NCB5OR



209949_at
NCF2



207677_s_at
NCF4



205147_x_at
NCF4



203315_at
NCK2



219231_at
NCOA6IP



214181_x_at
NCR3



211583_x_at
NCR3



208759_at
NCSTN



210817_s_at
NDP52



214867_at
NDST2



203621_at
NDUFB5



211752_s_at
NDUES7



203413_at
NELL2



217722_s_at
NEUGRIN



211105_s_at
NFATC1



202584_at
NFX1



202215_s_at
NFYC



217963_s_at
NGFRAP1



218240_at
NKIRAS2



200902_at
NM_004261.1



205006_s_at
NMT2



200875_s_at
NOL5A



217962_at
NOLA3



211951_at
NOLC1



217950_at
NOSIP



213775_x_at
NP220



209798_at
NPAT



200701_at
NPC2



200063_s_at
NPM1



203814_s_at
NQO2



204791_at
NR2C1



204651_at
NRF1



217850_at
NS



210023_s_at
NSPC1



213061_s_at
NTAN1



217802_s_at
NUCKS



207545_s_at
NUMB



209073_s_at
NUMB



218768_at
NUP107



212247_at
NUP205



213945_s_at
NUP210



202900_s_at
NUP88



214945_at
NY-REN-7



201599_at
OAT



201364_s_at
OAZ2



201365_at
OAZ2



200790_at
ODC1



203569_s_at
OFD1



206323_x_at
OPHN1



202074_s_at
OPTN



210028_s_at
ORC3L



204957_at
ORC5L



218556_at
ORMDL2



209627_s_at
OSBPL3



209626_s_at
OSBPL3



202780_at
OXCT1



210401_at
P2RX1



210448_s_at
P2RX5



218589_at
P2RY5



208051_s_at
PAIP1



202759_s_at
PALM2



218771_at
PANK4



213534_s_at
PASK



216945_x_at
PASK



212825_at
PAXIP1L



205353_s_at
PBP



214177_s_at
PBXIP1



214512_s_at
PC4



209361_s_at
PCBP4



214937_x_at
PCM1



210156_s_at
PCMT1



218014_at
PCNT1



212422_at
PDCD11



212593_s_at
PDCD4



202731_at
PDCD4



222317_at
PDE3B



204735_at
PDE4A



204491_at
PDE4D



212390_at
PDE4DIP



214099_s_at
PDE4DIP



214129_at
PDE4DIP



208690_s_at
PDLIM1



202671_s_at
PDXK



219132_at
PELI2



218472_s_at
PELO



218590_at
PEO1



55616_at
PERLD1



204992_s_at
PFN2



200886_s_at
PGAM1



208454_s_at
PGCP



200737_at
PGK1



200738_s_at
PGK1



219394_at
PGS1



222125_s_at
PH-4



212660_at
PHF15



218517_at
PHF17



203691_at
PI3



212506_at
PICALM



205452_at
PIGB



212120_at
PIGF



212240_s_at
PIK3R1



219788_at
PILRA



222218_s_at
PILRA



220954_s_at
PILRB



204269_at
PIM2



201192_s_at
PITPN



218667_at
PJA1



204612_at
PKIA



202732_at
PKIG



201251_at
PKM2



60528_at
PLA2G4B



206214_at
PLA2G7



205372_at
PLAG1



207002_s_at
PLAGL1



203471_s_at
PLEK



201136_at
PLP2



202430_s_at
PLSCR1



202446_s_at
PLSCR1



214081_at
PLXDC1



219700_at
PLXDC1



208890_s_at
PLXNB2



213241_at
PLXNC1



213677_s_at
PMS1



218224_at
PNMA1



203366_at
POLG



218016_s_at
POLR3E



203782_s_at
POLRMT



32502_at
PP1665



212199_at
PP784



200661_at
PPGB



203063_at
PPM1F



216347_s_at
PPP1R13B



41577_at
PPP1R16B



212750_at
PPP1R16B



201877_s_at
PPP2R5C



32541_at
PPP3CC



207000_s_at
PPP3CC



206174_s_at
PPP6C



200975_at
PPT1



201494_at
PRCP



203057_s_at
PRDM2



218329_at
PRDM4



201619_at
PRDX3



201858_s_at
PRG1



202741_at
PRKACB



213093_at
PRKCA



209048_s_at
PRKCBP1



209049_s_at
PRKCBP1



210038_at
PRKCQ



210039_s_at
PRKCQ



38269_at
PRKD2



204061_at
PRKX



206279_at
PRKY



204447_at
PROSAPIP1



209440_at
PRPS1



221036_s_at
PSFL



209337_at
PSIP1



208805_at
PSMA6



200039_s_at
PSMB2



201400_at
PSMB3



202353_s_at
PSMD12



218371_s_at
PSPC1



211178_s_at
PSTPIP1



219938_s_at
PSTPIP2



206278_at
PTAFR



206631_at
PTGER2



205171_at
PTPN4



206687_s_at
PTPN6



202897_at
PTPNS1



204960_at
PTPRCAP



203554_x_at
PTTG1



204020_at
PURA



201608_s_at
PWP1



201607_at
PWP1



221666_s_at
PYCARD



202990_at
PYGL



205174_s_at
QPCT



201482_at
QSCN6



219622_at
RAB20



209514_s_at
RAB27A



210951_x_at
RAB27A



217763_s_at
RAB31



217764_s_at
RAB31



204214_s_at
RAB32



207405_s_at
RAD17



212646_at
RAFTLIN



218337_at
RAI16



202100_at
RALB



201711_x_at
RANBP2



210676_x_at
RANBP2L1



212842_x_at
RANBP2L1



212127_at
RANGAP1



209284_s_at
RAP140



209285_s_at
RAP140



204070_at
RARRES3



205590_at
RASGRP1



203185_at
RASSF2



201092_at
RBBP7



212331_at
RBL2



203250_at
RBM16



218593_at
RBM28



213852_at
RBM8A



204098_at
RBMX2



212820_at
RC3



213878_at
RECQL



202296_s_at
RER1



220570_at
RETN



204023_at
RFC4



216834_at
RGS1



202988_s_at
RGS1



209324_s_at
RGS16



201453_x_at
RHEB



204951_at
RHOH



214700_x_at
RIF1



209684_at
RIN2



218598_at
RINT-1



209941_at
RIPK1



213338_at
RIS1



209882_at
RIT1



218269_at
RNASE3L



213397_x_at
RNASE4



213566_at
RNASE6



207735_at
RNF125



215031_x_at
RNF126



217865_at
RNF130



219104_at
RNF141



204040_at
RNF144



219035_s_at
RNF34



212696_s_at
RNF4



218286_s_at
RNF7



203160_s_at
RNF8



202683_s_at
RNMT



208270_s_at
RNPEP



210479_s_at
RORA



210426_x_at
RORA



217559_at
RPL10L



200809_x_at
RPL12



221726_at
RPL22



213084_x_at
RPL23A



212039_x_at
RPL3



211073_x_at
RPL3



213689_x_at
RPL5



200908_s_at
RPLP2



201011_at
RPN1



205562_at
RPP38



214001_x_at
RPS10



200949_x_at
RPS20



201909_at
RPS4Y1



212928_at
RPS5P1



204171_at
RPS6KB1



218909_at
RPS6KC1



221524_s_at
RRAGD



212589_at
RRAS2



212590_at
RRAS2



201477_s_at
RRM1



219549_s_at
RTN3



211509_s_at
RTN4



36129_at
RUTBC1



200660_at
S100A11



205863_at
S100A12



203186_s_at
S100A4



202917_s_at
S100A8



203535_at
S100A9



213262_at
SACS



32099_at
SAFB2



204900_x_at
SAP30



218854_at
SART2



200069_at
SART3



213988_s_at
SAT



203408_s_at
SATB1



39835_at
SBF1



209146_at
SC4MOL



211423_s_at
SC5DL



205790_at
SCAP1



218217_at
SCPEP1



202541_at
SCYE1



202071_at
SDC4



212607_at
SDCCAG8



202228_s_at
SDFR1



219349_s_at
SEC5L1



218265_at
SECISBP2



219351_at
SEDL



204563_at
SELL



210124_x_at
SEMA4F



208939_at
SEPHS1



212414_s_at
SEPT6



213666_at
SEPT6



212413_at
SEPT6



214298_x_at
SEPT6



212415_at
SEPT6



217977_at
SEPX1



202833_s_at
SERPINA1



212268_at
SERPINB1



213572_s_at
SERPINB1



206034_at
SERPINB8



218346_s_at
SESN1



200687_s_at
SF3B3



213370_s_at
SFMBT1



212001_at
SFRS14



201129_at
SFRS7



200044_at
SFRS9



201698_s_at
SFRS9



220642_x_at
SH120



201312_s_at
SH3BGRL



201311_s_at
SH3BGRL



204019_s_at
SH3YL1



221519_at
SHFM3



221833_at
SIAH1



201998_at
SIAT1



52940_at
SIGIRR



211761_s_at
SIP



201381_x_at
SIP



220485_s_at
SIRPB2



218878_s_at
SIRT1



205484_at
SIT



206181_at
SLAMF1



210422_x_at
SLC11A1



206600_s_at
SLC16A5



209003_at
SLC25A11



202433_at
SLC35B1



218826_at
SLC35F2



218237_s_at
SLC38A1



212110_at
SLC39A14



211030_s_at
SLC6A6



201195_s_at
SLC7A5



203579_s_at
SLC7A6



204588_s_at
SLC7A7



202983_at
SMARCA3



210357_s_at
SMOX



205596_s_at
SMURF2



218788_s_at
SMYD3



213447_at
SNRPN



201522_x_at
SNRPN



206042_x_at
SNURF



218404_at
SNX10



210648_x_at
SNX3



216841_s_at
SOD2



212807_s_at
SORT1



212780_at
SOS1



207777_s_at
SP140



216274_s_at
SPC18



217827_s_at
SPG21



202524_s_at
SPOCK2



202523_s_at
SPOCK2



204011_at
SPRY2



214925_s_at
SPTAN1



215235_at
SPTAN1



203127_s_at
SPTLC2



217995_at
SQRDL



210959_s_at
SRD5A1



211056_s_at
SRD5A1



214789_x_at
SRP46



207040_s_at
ST13



204150_at
STAB1



208992_s_at
STAT3



206118_at
STAT4



202693_s_at
STK17A



202695_s_at
STK17A



202786_at
STK39



211106_at
SUPT3H



201483_s_at
SUPT4H1



212894_at
SUPV3L1



209447_at
SYNE1



202761_s_at
SYNE2



205691_at
SYNGR3



212828_at
SYNJ2



205804_s_at
T3JAM



216925_s_at
TAL1



201463_s_at
TALDO1



212978_at
TA-LRRP



204770_at
TAP2



202813_at
TARBP1



37278_at
TAZ



203386_at
TBC1D4



213400_s_at
TBL1X



208130_s_at
TBXAS1



202396_at
TCERG1



209153_s_at
TCF3



205255_x_at
TCF7



212764_at
TCF8



217909_s_at
TCFL4



203303_at
TCTE1L



200803_s_at
TEGT



219131_at
TERE1



203611_at
TERF2



218104_at
TEX10



206715_at
TFEC



210215_at
TFR2



208249_s_at
TGDS



201506_at
TGFBI



204731_at
TGFBR3



212910_at
THAP11



218492_s_at
THAP7



204064_at
THOC1



202393_s_at
TIEG



201666_at
TIMP1



203167_at
TIMP2



208838_at
TIP120A



208700_s_at
TKT



208699_x_at
TKT



206472_s_at
TLE3



212769_at
TLE3



204924_at
TLR2



209150_s_at
TM9SF1



212194_s_at
TM9SF4



218113_at
TMEM2



202644_s_at
TNFAIP3



203508_at
TNFRSF1B



219423_x_at
TNFRSF25



210847_x_at
TNFRSF25



211841_s_at
TNFRSF25



206150_at
TNFRSF7



210314_x_at
TNFSF13



209499_x_at
TNFSF13



211495_x_at
TNFSF13



209500_x_at
TNFSF13



207892_at
TNFSF5



212261_at
TNRC15



201870_at
TOMM34



201519_at
TOMM70A



221601_s_at
TOSO



221602_s_at
TOSO



204529_s_at
TOX



201690_s_at
TPD52



201379_s_at
TPD52L2



200822_x_at
TPI1



201731_s_at
TPR



210972_x_at
TRA@



209671_x_at
TRA@



205599_at
TRAF1



204352_at
TRAF5



201391_at
TRAP1



219434_at
TREM1



218425_at
TRIAD3



202478_at
TRIB2



218145_at
TRIB3



217147_s_at
TRIM



213009_s_at
TRIM37



209390_at
TSC1



221493_at
TSPYL1



210645_s_at
TTC3



208073_x_at
TTC3



208195_at
TTN



214983_at
TTTY15



218184_at
TULP4



203246_s_at
TUSC4



208864_s_at
TXN



208959_s_at
TXNDC4



207668_x_at
TXNDC7



204122_at
TYROBP



213876_x_at
U2AF1L2



200058_s_at
U5-200KD



219192_at
UBAP2



221839_s_at
UBAP2



211764_s_at
UBE2D1



203109_at
UBE2M



218011_at
UBL5



202706_s_at
UMPS



220998_s_at
UNC93B1



213274_s_at
UNK_AA020826



212993_at
UNK_AA114166



221728_x_at
UNK_AA628440



214686_at
UNK_AA868898



211563_s_at
UNK_AB006572



222108_at
UNK_AC004010



211796_s_at
UNK_AF043179



211429_s_at
UNK_AF119873



217473_x_at
UNK_AF229163



222001_x_at
UNK_AI160126



202969_at
UNK_AI216690



50376_at
UNK_AI278629



213152_s_at
UNK_AI343248



64064_at
UNK_AI435089



217526_at
UNK_AI478300



213161_at
UNK_AI583393



212239_at
UNK_AI680192



215399_s_at
UNK_AI683900



221918_at
UNK_AI742210



204860_s_at
UNK_AI817801



221850_x_at
UNK_AI826075



221973_at
UNK_AI983904



217028_at
UNK_AJ224869



216044_x_at
UNK_AK027146



40446_at
UNK_AL021366



202789_at
UNK_AL022394



212642_s_at
UNK_AL023584



213540_at
UNK_AL031228



203608_at
UNK_AL031230



212636_at
UNK_AL031781



209733_at
UNK_AL034399



212234_at
UNK_AL034550



213213_at
UNK_AL035669



212400_at
UNK_AL043266



213817_at
UNK_AL049435



214948_s_at
UNK_AL050136



216199_s_at
UNK_AL109942



212430_at
UNK_AL109955



212098_at
UNK_AL134724



212737_at
UNK_AL513583



212606_at
UNK_AL536319



213193_x_at
UNK_AL559122



212501_at
UNK_AL564683



212222_at
UNK_AU143855



221876_at
UNK_AU151157



214218_s_at
UNK_AV699347



202124_s_at
UNK_AV705253



212274_at
UNK_AV705559



215633_x_at
UNK_AV713720



202073_at
UNK_AV757675



213839_at
UNK_AW028110



214735_at
UNK_AW166711



212429_s_at
UNK_AW194657



210926_at
UNK_AY014272



211474_s_at
UNK_BC004948



211725_s_at
UNK_BC005884



213564_x_at
UNK_BE042354



213281_at
UNK_BE327172



203640_at
UNK_BE328496



212693_at
UNK_BE670928



221971_x_at
UNK_BE672818



208988_at
UNK_BE675843



208785_s_at
UNK_BE893893



204276_at
UNK_BE895437



215438_x_at
UNK_BE906054



213503_x_at
UNK_BE908217



213189_at
UNK_BE966695



212114_at
UNK_BE967207



212071_s_at
UNK_BE968833



221842_s_at
UNK_BE972394



213011_s_at
UNK_BF116254



212638_s_at
UNK_BF131791



212624_s_at
UNK_BF339445



213567_at
UNK_BF431965



202405_at
UNK_BF432532



212037_at
UNK_BF508848



212509_s_at
UNK_BF968134



209815_at
UNK_BG054916



202515_at
UNK_BG251175



214658_at
UNK_BG286537



222280_at
UNK_BG491393



205038_at
UNK_BG540504



211902_x_at
UNK_L34703



209670_at
UNK_M12959



210915_x_at
UNK_M15564



212237_at
UNK_N64780



203580_s_at
UNK_NM_003983



203130_s_at
UNK_NM_004522



205961_s_at
UNK_NM_004682



204474_at
UNK_NM_005081



203501_at
UNK_NM_006102



202475_at
UNK_NM_006326



203062_s_at
UNK_NM_014641



206003_at
UNK_NM_014645



205340_at
UNK_NM_014797



205953_at
UNK_NM_014813



203674_at
UNK_NM_014877



204568_at
UNK_NM_014924



206053_at
UNK_NM_014930



203956_at
UNK_NM_014941



204411_at
UNK_NM_017596



220486_x_at
UNK_NM_017698



218873_at
UNK_NM_017710



218829_s_at
UNK_NM_017780



218331_s_at
UNK_NM_017782



205510_s_at
UNK_NM_017976



218594_at
UNK_NM_018072



220452_x_at
UNK_NM_021031



208540_x_at
UNK_NM_021039



201963_at
UNK_NM_021122



218764_at
UNK_NM_024064



219253_at
UNK_NM_024121



219431_at
UNK_NM_024605



218505_at
UNK_NM_024673



220251_at
UNK_NM_024998



220999_s_at
UNK_NM_030778



214328_s_at
UNK_R01140



58308_at
UNK_R71157



211612_s_at
UNK_U62858



49485_at
UNK_W22625



203519_s_at
UPF2



203234_at
UPP1



201903_at
UQCRC1



210053_at
USMG5



208723_at
USP11



203965_at
USP20



220419_s_at
USP25



201498_at
USP7



221513_s_at
UTP14A



203992_s_at
UTX



208067_x_at
UTY



219675_s_at
UXS1



201337_s_at
VAMP3



211749_s_at
VAMP3



202550_s_at
VAPB



204254_s_at
VDR



208623_s_at
VIL2



208622_s_at
VIL2



217949_s_at
VKORC1



220990_s_at
VMP1



205922_at
VNN2



212323_s_at
VPS13D



203856_at
VRK1



213773_x_at
WBSCR20A



213670_x_at
WBSCR20C



214100_x_at
WBSCR20C



213460_x_at
WBSCR20C



221581_s_at
WBSCR5



218882_s_at
WDR3



212533_at
WEE1



34225_at
WHSC2



213836_s_at
WIPI49



203827_at
WIPI49



205667_at
WRN



201760_s_at
WSB2



209375_at
XPC



218767_at
XPMC2H



211946_s_at
XTP2



213077_at
YTHDC2



204787_at
Z39IG



214032_at
ZAP70



203026_at
ZBTB5



213051_at
ZC3HAV1



220104_at
ZC3HAV1



212704_at
ZCCHC11



213853_at
ZCSL3



218078_s_at
ZDHHC3



202978_s_at
ZF



201368_at
ZFP36L2



203556_at
ZHX2



202136_at
ZMYND11



219854_at
ZNF14



200050_at
ZNF146



204327_s_at
ZNF202



218005_at
ZNF22



213934_s_at
ZNF23



204937_s_at
ZNF274



209494_s_at
ZNF278



209431_s_at
ZNF278



219228_at
ZNF331



214760_at
ZNF337



40569_at
ZNF42



219848_s_at
ZNF432



215359_x_at
ZNF44



214482_at
ZNF46



218735_s_at
ZNF544



221645_s_at
ZNF83



206572_x_at
ZNF85



200808_s_at
ZYX



212893_at
ZZZ3


























TABLE 24
















In Global





Unadjusted p
Odds Ratio
Odds Ratio



Analysis




value for IgG
for IgG
for IgG



(functional
In Key




association,
association,
association,



categories
High




calculated
calculated
calculated
Affymetrix


and
Level



FDR IgG
with
with
without
probeset

Multiple/
canonical
Functional


Gene
association
encephalitics
encephalitics
encephalitics
qualifier
Description
Single
pathways)
categories
























PTBP1
0.00203
3.57E-05
0.112
0.096
211270_x_at
polypyrimidine
multiple
Yes
Yes








tract binding








protein 1


GLUD1
0.0151
1.26E-03
0.098
0.110
200946_x_at
glutamate
single
Yes
No








dehydrogenase








1


MKNK1
0.0192
1.85E-03
0.147
0.121
209467_s_at
MAP kinase
single
Yes
Yes








interacting








serine/threonine








kinase 1


SLC12A9
0.000732
1.67E-06
0.117
0.125
220371_s_at
solute carrier
single
No
No








family 12








(potassium/








chloride








transporters),








member 9


HDGF
0.00241
5.05E-05
0.137
0.155
216484_x_at
hepatoma-
single
Yes
No








derived








growth factor








(high-mobility








group protein








1-like)


ACTR1A
0.00671
3.03E-04
0.171
0.156
200721_s_at
ARP1 actin-
single
Yes
No








related protein








1 homolog A,








centractin








alpha (yeast)


GORASP2
0.00703
3.40E-04
0.219
0.176
207812_s_at
golgi
single
No
No








reassembly








stacking








protein 2,








55kDa


BLCAP
0.00456
1.56E-04
0.175
0.179
201032_at
bladder cancer
single
No
No








associated








protein


DKFZP564J157
0.0018
2.85E-05
0.158
0.187
217794_at
DKFZp564J157
single
No
No








protein


FLJ10315
0.00139
1.16E-05
0.174
0.191
218770_s_at
hypothetical
single
No
No








protein








FLJ10315


CRKL
0.0192
1.86E-03
0.180
0.192
212180_at
v-crk sarcoma
single
Yes
No








virus CT10








oncogene








homolog








(avian)-like


EXT2
0.0261
3.14E-03
0.198
0.192
202012_s_at
exostoses
single
Yes
No








(multiple) 2


CDC40
0.00559
2.21E-04
0.177
0.203
203376_at
cell division
single
Yes
No








cycle 40








homolog








(yeast)


FLJ11560
0.00151
1.90E-05
0.189
0.203
211433_x_at
KIAA1539
multiple
No
No


OAZIN
0.0208
2.12E-03
0.210
0.204
212461_at
ornithine
single
Yes
No








decarboxylase








antizyme








inhibitor


COPS7A
0.0333
4.47E-03
0.258
0.207
209029_at
COP9
single
No
No








constitutive








photomorpho-








genic homolog








subunit 7A








(Arabidopsis)


STOM
0.0322
4.29E-03
0.235
0.208
201060_x_at
stomatin
single
No
No


NPEPPS
0.0187
1.77E-03
0.266
0.209
201454_s_at
aminopeptidase
single
Yes
No








puromycin








sensitive


SGPL1
0.027
3.31E-03
0.191
0.209
212321_at
sphingosine-1-
multiple
Yes
No








phosphate








lyase 1


MAP2K3
0.00282
6.47E-05
0.195
0.214
215499_at
mitogen-
single
Yes
Yes








activated








protein kinase








kinase 3


SEC31L1
0.0134
1.02E-03
0.190
0.215
210616_s_at
SEC31-like 1
single
Yes
No








(S.cerevisiae)


ATP6V0A1
0.00387
1.15E-04
0.175
0.217
212383_at
ATPase, H+
single
No
No








transporting,








lysosomal V0








subunit a








isoform 1


CBARA1
0.0114
7.57E-04
0.192
0.217
216903_s_at
calcium
single
No
No








binding atopy-








related








autoantigen 1


TXNRD1
0.0573
1.06E-02
0.285
0.218
201266_at
thioredoxin
single
Yes
Yes








reductase 1


TM9SF2
0.0206
2.08E-03
0.194
0.222
201078_at
transmembrane 9
single
Yes
No








superfamily








member 2


SH3BP2
0.00404
1.24E-04
0.207
0.223
209370_s_at
SH3-domain
single
Yes
No








binding








protein 2


VCP
0.00283
6.67E-05
0.172
0.226
208648_at
valosin-
single
Yes
No








containing








protein


KIAA0676
0.0326
4.34E-03
0.224
0.227
215994_x_at
KIAA0676
single
No
No








protein


FLJ10307
0.00537
2.01E-04
0.209
0.228
218753_at
hypothetical
single
No
No








protein








FLJ10307


PAFAH1B1
0.00451
1.53E-04
0.212
0.228
200815_s_at
platelet-
single
Yes
Yes








activating








factor








acetylhydrolase,








isoform Ib,








alpha subunit








45kDa


EIF4A1
0.0299
3.79E-03
0.221
0.231
211787_s_at
eukaryotic
single
Yes
Yes








translation








initiation








factor 4A,








isoform 1


MFN2
0.00115
6.12E-06
0.205
0.236
201155_s_at
mitofusin 2
single
Yes
No


ACTR1B
0.0317
4.14E-03
0.220
0.239
202135_s_at
ARP1 actin-
single
Yes
No








related protein








1 homolog B,








centractin beta








(yeast)


MGC10433
0.00363
1.01E-04
0.192
0.239
205740_s_at
hypothetical
single
No
No








protein








MGC10433


CANX
0.0265
3.23E-03
0.226
0.242
200068_s_at
calnexin
single
Yes
No


LOC285148
0.0072
3.50E-04
0.204
0.244
213532_at
hypothetical
single
No
No








protein








LOC285148


ATP6V0C
0.00358
9.87E-05
0.215
0.245
36994_at
ATPase, H+
single
Yes
No








transporting,








lysosomal








16kDa, V0








subunit c


DAG1
0.014
1.10E-03
0.256
0.246
205417_s_at
dystroglycan 1
single
Yes
Yes








(dystrophin-








associated








glycoprotein








1)


K-ALPHA-1
0.0116
7.87E-04
0.225
0.246
211058_x_at
tubulin, alpha,
multiple
No
No








ubiquitous


PLOD
0.00146
1.68E-05
0.245
0.246
200827_at
procollagen-
single
Yes
No








lysine, 2-








oxoglutarate








5-dioxygenase








(lysine








hydroxylase,








Ehlers-Danlos








syndrome type








VI)


PLOD3
0.0236
2.65E-03
0.277
0.246
202185_at
procollagen-
single
Yes
No








lysine, 2-








oxoglutarate








5-dioxygenase








3


COBRA1
0.00336
8.60E-05
0.257
0.249
202757_at
cofactor of
single
Yes
No








BRCA1


NPL4
0.0243
2.81E-03
0.284
0.249
217796_s_at
nuclear protein
single
Yes
No








localization 4


SDF2
0.0965
2.31E-02
0.227
0.250
203090_at
stromal cell-
single
Yes
No








derived factor








2


PPP2R4
0.00261
5.83E-05
0.237
0.251
208874_x_at
protein
single
Yes
No








phosphatase








2A, regulatory








subunit B′(PR








53)


DNASE1L1
0.00182
2.98E-05
0.231
0.255
203912_s_at
deoxyribonuc1
single
Yes
No








ease I-like 1


LASS2
0.00355
9.71E-05
0.259
0.255
222212_s_at
LAG1
single
No
No








longevity








assurance








homolog 2 (S.








cerevisiae)


XPO7
0.00287
6.85E-05
0.298
0.256
212166_at
exportin 7
single
Yes
Yes


GBA
0.0116
7.92E-04
0.264
0.260
209093_s_at
glucosidase,
single
No
No








beta; acid








(includes








glucosyl-








ceramidase)


PLAGL2
0.0343
4.72E-03
0.269
0.260
202924_s_at
pleiomorphic
single
Yes
Yes








adenoma








gene-like 2


MGC16824
0.0163
1.40E-03
0.226
0.262
203173_s_at
esophageal
single
No
No








cancer








associated








protein


MGC13024
0.0238
2.71E-03
0.273
0.266
221864_at
hypothetical
single
No
No








protein








MGC13024


KIAA0494
0.0111
7.20E-04
0.228
0.269
201776_s_at
K1AA0494
single
No
No








gene product


SMP1
0.0833
1.87E-02
0.278
0.272
217766_s_at
small
single
No
No








membrane








protein 1


HK1
0.0468
7.87E-03
0.320
0.273
200697_at
hexokinase 1
single
Yes
No


KIAA1193
0.0115
7.74E-04
0.251
0.275
44822_s_at
KIAA1193
single
No
No


FLJ13910
0.0297
3.77E-03
0.257
0.276
212482_at
hypothetical
single
No
No








protein








FLJ13910


NUP214
0.0176
1.61E-03
0.264
0.278
202155_s_at
nucleoporin
single
Yes
Yes








214kDa


SH3GLB1
0.0764
1.65E-02
0.279
0.279
209090_s_at
SH3-domain
single
Yes
No








GRB2-like








endophilin B1


TM6SF1
0.0138
1.07E-03
0.271
0.279
219892_at
transmembrane 6
single
No
No








superfamily








member 1


XPO6
0.0161
1.39E-03
0.237
0.279
211982_x_at
exportin 6
single
No
No


C21ORF97
0.0686
1.40E-02
0.286
0.280
218019_s_at

single
No
No


SMARCD2
0.0211
2.20E-03
0.268
0.283
201827_at
SWI/SNF
single
Yes
No








related, matrix








associated,








actin








dependent








regulator of








chromatin,








subfamily d,








member 2


ETHE1
0.00865
4.76E-04
0.289
0.285
204034_at
ethylmalonic
single
No
No








encephalopath








y1


DCTN1
0.0065
2.82E-04
0.300
0.287
211780_x_at
dynactin 1
single
Yes
No








(p150, glued








homolog,








Drosophila)



0.0439
7.18E-03
0.280
0.289
213184_at


N/A
N/A


TUBA6
0.0104
6.38E-04
0.301
0.290
211750_x_at
tubulin alpha 6
multiple
No
No


FURIN
0.0106
6.68E-04
0.290
0.291
201945_at
furin (paired
single
Yes
No








basic amino








acid cleaving








enzyme)


RAB2L
0.0477
8.06E-03
0.249
0.292
209110_s_at
ral guanine
single
Yes
No








nucleotide








dissociation








stimulator-








like 2


UBE2G1
0.0373
5.41E-03
0.274
0.294
209141_at
ubiquitin-
single
No
No








conjugating








enzyme E2G 1








(UBC7








homolog, C.








elegans)


CENTA1
0.00887
4.95E-04
0.255
0.295
90265_at
centaurin,
single
Yes
No








alpha 1


DR1
0.0412
6.43E-03
0.252
0.297
207654_x_at
down-
single
Yes
No








regulator of








transcription








1, TBP-








binding








(negative








cofactor 2)


MAPK7
0.00208
3.76E-05
0.255
0.297
35617_at
mitogen-
single
Yes
No








activated








protein








kinase 7


MPST
0.00964
5.58E-04
0.259
0.297
203524_s_at
mercapto-
single
No
No








pyruvate








sulfur-








transferase


MXD4
0.00144
1.27E-05
0.254
0.297
212346_s_at
MAX
single
Yes
No








dimerization








protein 4


PEMT
0.00144
1.39E-05
0.259
0.298
207621_s_at
phosphatidylet
single
Yes
No








hanolamine N-








methyl-








transferase


DULLARd
0.00939
5.35E-04
0.263
0.299
200035_at
dullard
single
No
No








homolog








(Xenopus









laevis)



GDI1
0.00287
6.90E-05
0.283
0.302
201864_at
GDP
single
Yes
No








dissociation








inhibitor 1


ARPC1B
0.00202
3.49E-05
0.276
0.304
201954_at
actin related
single
Yes
No








protein ⅔








complex,








subunit 1B,








41kDa


IMPDH1
0.00256
5.63E-05
0.274
0.304
204169_at
IMP (inosine
single
Yes
No








monophosphate)








dehydrogenase








1


PABPC4
0.00456
1.56E-04
0.256
0.304
201064_s_at
poly(A)
single
Yes
Yes








binding








protein,








cytoplasmic 4








(inducible








form)


KDELR2
0.0192
1.85E-03
0.297
0.305
200698_at
KDEL (Lys-
single
Yes
Yes








Asp-Glu-Leu)








endoplasmic








reticulum








protein








retention








receptor 2


BAT3
0.0147
1.20E-03
0.326
0.306
201255_x_at
HLA-B
single
No
No








associated








transcript 3


JWA
0.067
1.34E-02
0.320
0.306
200760_s_at
cytoskeleton
single
Yes
No








related








vitamin








A responsive








protein


MGC5508
0.0558
1.02E-02
0.323
0.306
201361_at
hypothetical
single
No
No








protein








MGC5508


PEPD
0.00541
2.06E-04
0.273
0.308
202108_at
peptidase D
single
No
No


CLIC4
0.00564
2.25E-04
0.318
0.312
201560_at
chloride
single
Yes
No








intracellular








channel 4


GRINA
0.00167
2.47E-05
0.281
0.313
212090_at
glutamate
single
No
No








receptor,








ionotropic, N-








methyl D-








asparate-








associated








protein 1








(glutamate








binding)


GTF2A2
0.0277
3.44E-03
0.277
0.314
202678_at
general
single
No
No








transcription








factor IIA, 2,








12kDa


ELK1
0.0999
2.44E-02
0.324
0.315
203617_x_at
ELK1,
single
Yes
No








member of








ETS oncogene








family


RELA
0.00702
3.33E-04
0.260
0.315
201783_s_at
v-rel
single
Yes
Yes








reticuloendoth








eliosis viral








oncogene








homolog A,








nuclear factor








of kappa light








polypeptide








gene enhancer








in B-cells 3,








p65 (avian)


AMFR
0.0543
9.67E-03
0.313
0.319
202204_s_at
autocrine
single
Yes
No








motility factor








receptor


SLC9A8
0.0217
2.32E-03
0.315
0.321
212947_at
solute carrier
single
No
No








family 9








(sodium/hydro








gen exchanger),








isoform 8


APG4B
0.0383
5.72E-03
0.326
0.322
212280_x_at
APG4
single
Yes
No








autophagy 4








homolog B (S.








cerevisiae)


POLR2L
0.00358
9.92E-05
0.296
0.322
211730_s_at
polymerase
single
Yes
No








(RNA) II








(DNA








directed)








polypeptide L,








7.6kDa


SNX27
0.0131
9.63E-04
0.314
0.325
221498_at
sorting nexin
single
No
No








family








member 27


ADRM1
0.00547
2.14E-04
0.284
0.326
201281_at
adhesion
single
No
No








regulating








molecule 1


FLJ10521
0.079
1.73E-02
0.319
0.326
221656_s_at
hypothetical
single
No
No








protein








FLJ10521


KIAA0121
0.0265
3.22E-03
0.327
0.328
212399_s_at
vestigial like 4
single
No
No








(Drosophila)


MYO9B
0.0442
7.28E-03
0.284
0.328
214780_s_at
myosin IXB
single
Yes
No


LENG4
0.0101
6.00E-04
0.302
0.329
205634_x_at
leukocyte
single
Yes
No








receptor








cluster (LRC)








member 4


SGSH
0.0139
1.08E-03
0.311
0.331
35626_at
N-sulfoglu-
single
Yes
No








cosamine








sulfohydrolase








(sulfamidase)


CORO1B
0.00144
1.48E-05
0.303
0.333
64486_at
coronin, actin
single
No
No








binding








protein,1B


RRAGD
0.00512
1.87E-04
0.276
0.334
221523_s_at
Ras-related
single
No
No








GTP binding








D


XBP1
0.0599
1.13E-02
0.292
0.334
200670_at
X-box binding
single
Yes
No








protein 1


CKAP4
0.0365
5.21E-03
0.302
0.335
200998_s_at
cytoskeleton-
single
No
No








associated


PP9099
0.0122
8.61E-04
0.319
0.335
204436_at
PH domain-
single
No
No








containing








protein


ARF3
0.00478
1.68E-04
0.320
0.338
200011_s_at
ADP-
single
Yes
Yes








ribosylation








factor 3


LOC51257
0.024
2.74E-03
0.332
0.338
210075_at
hypothetical
single
No
No








protein








LOC51257


PXN
0.0174
1.57E-03
0.296
0.342
201087_at
paxillin
single
Yes
No


SNN
0.00635
2.74E-04
0.300
0.344
218033_s_at
stannin
single
Yes
No


CAMTA2
0.0276
3.41E-03
0.301
0.350
212948_at
calmodulin
single
No
No








binding








transcription








activator 2


C20ORF35
0.0209
2.16E-03
0.306
0.351
218094_s_at
chromosome
single
No
No








20 open








reading frame








35


FLJ13725
0.0586
1.10E-02
0.322
0.354
45749_at
hypothetical
single
No
No








protein








FLJ13725


GRK6
0.0194
1.89E-03
0.311
0.355
210981_s_at
G protein-
single
Yes
No








coupled








receptor








kinase 6


FLJ12287
0.0101
6.18E-04
0.332
0.357
219259_at
sema domain,
single
No
No








immunoglobulin








domain (Ig),








transmembrane








domain (TM)








and short








cytoplasmic








domain,








(semaphorin)








4A


CDKN1A
0.00396
1.20E-04
0.325
0.365
202284_s_at
cyclin-
single
Yes
Yes








dependent








kinase








inhibitor 1A








(p21, Cip1)


KPNA6
0.0117
8.02E-04
0.320
0.365
212101_at
karyopherin
single
Yes
Yes








alpha 6








(importin








alpha 7)


CAB45
0.00387
1.16E-04
0.328
0.367
217855_x_at
calcium
single
No
No








binding








protein Cab45








precursor


GALNACT-2
0.0196
1.92E-03
0.315
0.367
222235_s_at
chondroitin
single
Yes
No








sulfate








GalNAcT-2


WDR13
0.0114
7.58E-04
0.326
0.371
222138_s_at
WD repeat
single
No
No








domain 13


OS-9
0.0184
1.73E-03
0.329
0.374
200714_x_at
amplified in
single
No
No








osteosarcoma


SRF
0.0116
7.97E-04
0.3 19
0.374
202401_s_at
serum
single
Yes
Yes








response








factor (c-fos








serum








response








element-








binding








transcnption








factor)


CHK
0.0114
7.68E-04
0.327
0.376
204266_s_at
choline kinase
single
Yes
No








alpha


TM9SF4
0.0399
6.06E-03
0.332
0.376
212198_s_at
transmembrane
single
No
No








9 superfamily








protein








member 4


CLN2
0.00611
2.55E-04
0.331
0.379
200742_s_at
ceroid-
single
Yes
No








lipofuscinosis,








neuronal 2,








late infantile








(Jansky-








Bielschowsky








disease)


LOC92482
0.037
5.31E-03
3.028
2.691
213220_at
hypothetical
single
No
No








protein








LOC92482


SS18L2
0.0277
3.43E-03
3.151
2.704
218283_at
synovial
single
No
No








sarcoma








translocation








gene on








chromosome








18-like 2


AKR1C1
0.0586
1.10E-02
3.098
2.712
216594_x_at
aldo-keto
single
Yes
No








reductase








family 1,








member C1








(dihydrodiol








dehydrogenase








1; 20-alpha








3-alpha)








hydroxysteroid








dehydrogenase)


CALM1
0.0214
2.26E-03
3.136
2.722
209563_x_at
calmodulin 1
single
Yes
No








(phosphorylase








kinase, delta)


DHX15
0.0435
7.09E-03
3.049
2.726
201385_at
DEAH (Asp-
single
No
No








Glu-Ala-His)








box








polypeptide 15


KIAA0252
0.0407
6.31E-03
3.099
2.786
212302_at
K1AA0252
single
No
No


NDUFA4
0.0501
8.60E-03
3.034
2.790
217773_s_at
NADH
single
Yes
No








dehydrogenase








(ubiquinone) 1








alpha








subcomplex,








4, 9kDa


FLJ10460
0.00426
1.38E-04
3.085
2.791
220071_x_at
hypothetical
single
No
No








protein








FLJ10460


LSM5
0.00482
1.71E-04
3.126
2.815
211747_s_at
LSM5
single
No
No








homolog, U6








small nuclear








RNA associated








(S. cerevisiae)


ALMS1
0.00371
1.08E-04
3.045
2.829
214707_x_at
Alstrom
single
Yes
No








syndrome 1



0.00211
3.97E-05
3.099
2.874
216006_at


N/A
N/A



0.00283
6.67E-05
3.291
2.879
217713_x_at


N/A
N/A


LAMR1
0.00169
2.59E-05
3.016
2.903
216806_at
laminin
single
Yes
No








receptor 1








(ribosomal








protein SA,








67kDa)


GTF2H2
0.00283
6.56E-05
3.033
2.926
221540_x_at
general
single
No
No








transcription








factor IIH,








polypeptide 2,








44kDa


TLE1
0.0399
6.07E-03
3.140
2.932
203221_at
transducin-like
single
Yes
No








enhancer of








split 1 (E(sp1)








homolog,








Drosophila)


XRCC2
0.00165
2.33E-05
3.033
2.934
207598_x_at
X-ray repair
single
Yes
Yes








complementing








defective








repair in








Chinese








hamster cells 2


DSPP
0.0227
2.51E-03
3.374
2.943
221681_s_at
dentin
single
Yes
No








sialophospho








protein


EIF3S1
0.0395
5.97E-03
3.184
2.948
208264_s_at
eukaryotic
single
Yes
Yes








translation








initiation








factor 3,








subunit 1








alpha, 35kDa


SLC30A5
0.0274
3.38E-03
3.230
2.967
218989_x_at
solute carrier
single
Yes
No








family 30








(zinc








transporter),








member 5


ZNF261
0.0458
7.66E-03
3.022
2.995
207559_s_at
zinc finger
single
Yes
No








protein 261


NAB1
0.0838
1.89E-02
3.184
2.996
211139_s_at
NGFI-A
single
Yes
No








binding








protein 1








(EGR1








binding








protein 1)


RIOK3
0.00115
5.92E-06
3.312
3.004
215588_x_at
RIO kinase
single
Yes
No








3 (yeast)


ZNF505
0.0193
1.87E-03
3.026
3.006
215758_x_at
zinc finger
single
No
No








protein 505


SNRPB2
0.00676
3.08E-04
3.596
3.015
202505_at
small nuclear
single
Yes
No








ribonucleo-








protein








polypeptide B″


UBL1
0.04
6.13E-03
3.159
3.033
211069_s_at
SMT3
single
Yes
Yes








suppressor of








mif two 3








homolog 1








(yeast)


TBCA
0.0071
3.44E-04
3.344
3.041
203667_at
tubulin-
single
No
No








specific








chaperone a


POLR1B
0.0011
5.46E-06
3.590
3.049
220113_x_at
polymerase
single
Yes
No








(RNA) I








polypeptide B,








128kDa


TCEAL1
0.0241
2.77E-03
3.163
3.062
204045_at
transcription
single
Yes
No








elongation








factor A (SII)-








like 1


MGC48332
0.0165
1.43E-03
3.326
3.063
213256_at
hypothetical
single
No
No








protein








MGC48332


SCD4
0.00773
3.96E-04
3.441
3.071
214036_at


N/A
N/A


FLJ22256
0.0113
7.39E-04
3.503
3.081
220856_x_at


N/A
N/A


TAX1BP1
0.0433
7.03E-03
3.335
3.085
200977_s_at
Tax1 (human
single
Yes
No








T-cell








leukemia virus








type I) binding








protein 1


FLJ10287
0.0177
1.64E-03
3.262
3.091
219130_at
hypothetical
single
No
No








protein








FLJ10287


CYCS
0.0281
3.51E-03
3.257
3.092
208905_at
cytochrome c,
single
Yes
No








somatic


CGI-12
0.00755
3.79E-04
3.378
3.117
219363_s_at
CGI-12
single
No
No








protein


RBBP6
0.00544
2.10E-04
3.065
3.128
212781_at
retinoblastoma
single
Yes
No








binding








protein 6


GTSE1
0.0219
2.35E-03
3.070
3.133
211040_x_at
G-2 and S-
single
No
No








phase








expressed 1


RPL26
0.00121
7.25E-06
3.342
3.133
222229_x_at
ribosomal
single
Yes
Yes








protein L26


PIGL
0.042
6.64E-03
3.247
3.142
205873_at
phosphatidylin
single
Yes
No








ositol glycan,








class L


NOL5A
0.0284
3.56E-03
3.760
3.146
200874_s_at
nucleolar
single
No
No








protein 5A








(56kDa with








KKE/D








repeat)


C1GALT1
0.0132
9.79E-04
3.612
3.150
219439_at
core 1 UDP-
single
No
No








galactose:N-








acetylgalactos








amine-alpha-R








beta 1,3-








galactosyl-








transferase


NIF3L1BP1
0.00355
9.67E-05
3.506
3.150
218334_at
Ngg1
single
No
No








interacting








factor 3 like








1 binding








protein 1


P38IP
0.0361
5.14E-03
3.621
3.160
220408_x_at
transcription
single
No
No








factor (p38








interacting








protein)


FTLL1
0.0128
9.29E-04
3.273
3.170
217703_x_at


N/A
N/A


NIP30
0.0382
5.68E-03
3.641
3.175
217896_s_at
NEFA-
single
No
No








interacting








nuclear protein








NIP30


LONP
0.00209
3.81E-05
3.226
3.197
221834_at
peroxisomal
single
No
No








Ion protease


DNAJC8
0.0997
2.43E-02
3.323
3.199
212491_s_at
DnaJ (Hsp40)
single
No
No








homolog,








subfamily C,








member 8


TCEB1
0.0777
1.70E-02
3.198
3.200
202824_s_at
transcription
single
Yes
No








elongation








factor B (SIII),








polypeptide I








(15kDa,








elongin C)


OIP2
0.00874
4.81E-04
3.544
3.206
215136_s_at
exosome
single
No
No








component 8


C14ORF123
0.00368
1.05E-04
3.718
3.209
218571_s_at
chromosome
single
No
No








14 open








reading frame








123


MCAM
0.0733
1.54E-02
3.745
3.222
211042_x_at
melanoma cell
single
Yes
No








adhesion








molecule


MPP2
0.0223
2.42E-03
3.682
3.243
207984_s_at
membrane
single
Yes
No








protein,








palmitoylated








2 (MAGUK








p55 subfamily








member 2)


LOC57149
0.0526
9.29E-03
3.251
3.244
203897_at
hypothetical
single
No
No








protein A-








211C6.1


FLJ23233
0.0505
8.72E-03
3.601
3.245
58367_s_at
hypothetical
single
No
No








protein








FLJ23233


P29
0.00698
3.28E-04
3.659
3.247
202553_s_at
GCIP-
single
No
No








interacting








protein p29


DNAH3
0.018
1.67E-03
3.282
3.250
209751_s_at
Mitogen

N/A
N/A








Activated








Protein








Kinase








Kinase


SON
0.0373
5.41E-03
3.843
3.258
214988_s_at
SON DNA
single
Yes
No








binding








protein


NONO
0.0513
8.98E-03
3.745
3.275
210470_x_at
non-POU
single
No
No








domain








containing,








octamer-








binding


PGF
0.000656
7.16E-07
3.555
3.285
215179_x_at
placental
single
Yes
No








growth factor,








vascular








endothelial








growth factor-








related protein


MCM3AP
0.00923
5.22E-04
3.627
3.293
215582_x_at
MCM3
single
Yes
Yes








mini-








chromosome








maintenance








deficient 3








(S. cerevisiae)








associated








protein


WBSCR5
0.00211
4.04E-05
3.921
3.317
211768_at
Williams-
single
Yes
No








Beuren








syndrome








chromosome








region 5


TMPO
0.00976
5.74E-04
3.992
3.321
209753_s_at
thymopoietin
single
Yes
Yes


NTRK3
0.0148
1.21E-03
3.298
3.323
217033_x_at
neurotrophic
single
Yes
Yes








tyrosine








kinase,








receptor,








type 3


SOD1
0.0301
3.83E-03
3.742
3.337
200642_at
superoxide
single
Yes
Yes








dismutase 1,








soluble








(amyotrophic








lateral








sclerosis 1








(adult))


TCTEL1
0.0433
7.05E-03
3.245
3.359
201999_s_at
t-complex-
single
Yes
No








associated-








testis-








expressed








1-like 1


GSTM3
0.00354
9.54E-05
3.641
3.363
202554_s_at
glutathione S-
single
Yes
No








transferase








M3 (brain)


C60RF62
0.0672
1.35E-02
3.245
3.390
208809_s_at
chromosome 6
single
No
No








open reading








frame 62


ZNF263
0.0237
2.68E-03
3.509
3.390
203707_at
zinc finger
single
Yes
No








protein 263


NEDD5
0.075
1.60E-02
3.861
3.393
200015_s_at
neural
single
No
No








precursor cell








expressed,








developmentally








down-regulated 5


CPA2
0.0157
1.33E-03
3.506
3.416
206212_at
carboxy-
single
Yes
No








peptidase A2








(pancreatic)


PEX16
0.0559
1.02E-02
3.690
3.422
49878_at
peroxisomal
single
Yes
No








biogenesis








factor 16


RPL35
0.0158
1.35E-03
3.439
3.434
200002_at
ribosomal
multiple
No
No








protein L35


FACL6
0.0188
1.79E-03
3.228
3.436
211207_s_at
acyl-CoA
single
Yes
No








synthetase








long-chain








family








member 6


FOXO1A
0.0328
4.40E-03
3.167
3.436
202724_s_at
forkhead box
single
Yes
Yes








O1A








(rhabdomyo-








sarcoma)


TGFB3
0.0237
2.67E-03
3.511
3.440
209747_at
transforming
single
Yes
Yes








growth factor,








beta 3


RPL24
0.0682
1.38E-02
3.055
3.445.
214143_x_at
ribosomal
single
No
No








protein L24


HSPC128
0.00512
1.88E-04
3.394
3.457
218936_s_at
HSPC128
single
No
No








protein


PSKH1
0.0137
1.05E-03
3.788
3.459
213141_at
protein serine
single
No
No








kinase H1


RANBP9
0.0805
1.78E-02
3.008
3.471
202582_s_at
RAN binding
single
Yes
No








protein 9


SNAP25
0.0337
4.59E-03
3.119
3.476
202507_s_at
synaptosomal-
single
Yes
No








associated








protein, 25kDa


FLJ23476
0.0126
9.08E-04
3.680
3.501
218647_s_at
ischemia/reper
single
No
No








fusion








inducible








protein


PHF2
0.0669
1.33E-02
3.458
3.502
212726_at
PHD finger
single
No
No








protein 2


FLJ20331
0.00256
5.62E-05
3.855
3.510
215063_x_at
hypothetical
single
No
No








protein








FLJ20331


SMARCA5
0.0278
3.47E-03
3.504
3.519
213251_at
SWI/SNF
single
Yes
No








related, matrix








associated,








actin








dependent








regulator of








chromatin,








subfamily a,








member 5


UQCRB
0.00582
2.36E-04
3.910
3.523
209065_at
ubiquinol-
multiple
Yes
No








cytochrome c








reductase








binding








protein


DKFZp566N034
0.00301
7.46E-05
4.150
3.525
208238_x_at


N/A
N/A


TRAPCC2
0.0224
2.46E-03
3.842
3.530
206853_s_at


N/A
N/A


SLC35E1
0.0152
1.28E-03
3.961
3.538
79005_at
solute carrier
single
No
No








family 35,








member E1


DT1P1A10
0.0318
4.17E-03
4.334
3.551
213079_at
hypothetical
single
No
No








protein








DT1P1A10


PRKDC
0.00631
2.70E-04
3.401
3.553
208694_at
protein kinase,
single
Yes
Yes








DNA-








activated,








catalytic








polypeptide


TTC13
0.037
5.30E-03
3.397
3.554
219481_at
tetratrico-
single
No
No








peptide repeat








domain 13


NFRKB
0.0586
1.10E-02
3.162
3.570
213028_at
nuclear factor
single
No
No








related to








kappa B








binding








protein


B2M
0.0662
1.32E-02
3.074
3.582
201891_s_at
beta-2-
single
Yes
No








microglobulin


VAMP4
0.0241
2.77E-03
3.512
3.591
213480_at
vesicle-
single
Yes
No








associated








membrane








protein 4


HSPA8
0.0337
4.58E-03
3.651
3.602
221891_x_at
heat shock
single
Yes
No








70kDa protein








8


Unknown
0.00665
2.95E-04
4.084
3.610
215557_at


N/A
N/A


MAP3K7
0.032
4.23E-03
3.601
3.616
215476_at


N/A
N/A


0
0.0397
6.03E-03
3.602
3.628
212436_at


N/A
N/A


RARG-1
0.00541
2.05E-04
4.107
3.631
202882_x_at
nucleolar
multiple
No
No








protein 7,








27kDa


SSB
0.00795
4.21E-04
3.664
3.642
201139_s_at
Sjogren
single
Yes
Yes








syndrome








antigen B








(autoantigen








La)


HNRPH1
0.0111
7.23E-04
3.327
3.643
213619_at
heterogeneous
single
Yes
No








nuclear








ribonucleo-








protein H1 (H)


HCDI
0.0906
2.11E-02
3.791
3.649
213398_s_at
chromosome
single
No
No








14 open








reading frame








124


COX7A3
0.00631
2.71E-04
4.532
3.654
217249_x_at
cytochrome c
single
Yes
No








oxidase








subunit VIIa








polypeptide 3








(liver)


CDK2
0.00404
1.25E-04
4.295
3.677
204252_at
cyclin-
single
Yes
Yes








dependent








kinase 2


ZNF-U69274
0.00547
2.14E-04
4.158
3.688
204847_at
zinc finger and
single
No
No








BTB domain








containing 11


ZFP95
0.018
1.67E-03
3.562
3.694
203730_s_at
zinc finger
single
No
No








protein 95








homolog








(mouse)


Unannotated
0.00249
5.25E-05
4.290
3.722
215628_x_at


N/A
N/A


PITPNC1
0.0956
2.28E-02
3.059
3.752
219155_at
phosphatidylin
single
Yes
No








ositol transfer








protein,








cytoplasmic 1


ATP5I
0.0144
1.15E-03
3.511
3.755
209492_x_at
ATP synthase,
single
No
No








H+








transporting,








mitochondrial








F0 complex,








subunit e


ING1L
0.0297
3.75E-03
3.537
3.766
205981_s_at
inhibitor of
single
Yes
No








growth family,








member 1-like


FLJ34588
0.0206
2.07E-03
3.860
3.767
212410_at
Smhs2
single
No
No








homolog (rat)


FLJ117l2
0.0427
6.87E-03
3.380
3.779
219056_at
hypothetical
single
No
No








protein








FLJ11712


PTD004
0.0353
4.94E-03
3.883
3.798
219293_s_at
hypothetical
single
No
No








protein








PTD004


MCFD2
0.0687
1.40E-02
3.299
3.808
212245_at
multiple
single
No
No








coagulation








factor








deficiency 2


HSPA4
0.0212
2.23E-03
4.047
3.833
208815_x_at
heat shock
single
Yes
No








70kDa protein








4


RPL19
0.037
5.32E-03
3.543
3.836
200029_at
ribosomal
single
Yes
Yes








protein L19


NDUFA6
0.00489
1.76E-04
3.979
3.843
202001_s_at
NADH
single
Yes
No








dehydrogenase








(ubiquinone) 1








alpha








subcomplex,








6, 14kDa



0.00126
7.93E-06
4.337
3.850
217446_x_at


N/A
N/A


HNRPD
0.0375
5.51E-03
4.357
3.860
200073_s_at
heterogeneous
multiple
Yes
No








nuclear








ribonucleo-








protein D (AU-








rich element








RNA binding








protein 1,








37kDa)


GCSH
0.00755
3.80E-04
4.230
3.885
213129_s_at
glycine
single
Yes
No








cleavage








system protein








H








(aminomethyl








carrier)


BUB3
0.0419
6.59E-03
4.233
3.890
201457_x_at
BUB3
multiple
Yes
Yes








budding








uninhibited by








benzimidazoles








3 homolog








(yeast)


RPL26L1
0.00676
3.07E-04
4.211
3.893
218830_at
ribosomal
single
No
No








protein L26-








like 1


GPRC5D
0.0171
1.52E-03
4.305
3.928
221297_at
G protein-
single
No
No








coupled








receptor,








family C,








group 5,








member D


NDUFS5
0.0348
4.83E-03
3.213
3.929
201757_at
NADH
single
Yes
No








dehydrogenase








(ubiquinone)








Fe-S protein 5,








15kDa








(NADH-








coenzyme Q








reductase)


ESRRBL1
0.00189
3.19E-05
4.429
3.996
218100_s_at
estrogen-
single
Yes
No








related








receptor beta








like 1


LOC144983
0.0735
1.55E-02
3.185
3.999
216559_x_at
hypothetical
single
No
No








protein








LOC144983


NDUFB8
0.00671
3.04E-04
4.431
4.011
201227_s_at
NADH
single
Yes
No








dehydrogenase








(ubiquinone) 1








beta








subcomplex,








8, 19kDa


FLJ10996
0.0773
1.68E-02
3.150
4.015
219774_at
hypothetical
single
No
No








protein








FLJ10996


RPS15
0.0867
1.98E-02
3.348
4.049
200819_s_at
ribosomal
single
Yes
Yes








protein S15


USP9X
0.00537
2.00E-04
4.430
4.050
201100_s_at
ubiquitin
single
Yes
No








specific








protease 9, X-








linked (fat








facets-like,








Drosophila)


MFN1
0.00994
5.90E-04
4.588
4.073
207098_s_at
mitofusin 1
single
Yes
No


HNRPDL
0.0635
1.23E-02
4.040
4.074
209067_s_at
heterogeneous
single
Yes
No








nuclear








ribonucleo-








protein D-like


ABCE1
0.0118
8.21E-04
3.786
4.103
201872_s_at
ATP-binding
single
No
No








cassette, sub-








family E








(OABP),








member 1


KIAA0036
0.00959
5.49E-04
4.375
4.135
211707_s_at
IQ
single
No
No








calmodulin-








binding motif








containing 1


UBA2
0.0215
2.28E-03
4.168
4.190
201177_s_at
SUMO-1
single
No
No








activating








enzyme








subunit 2


FKSG17
0.000732
2.00E-06
4.514
4.213
211445_x_at
FKSG17
single
No
No


LEREPO4
0.0172
1.54E-03
4.607
4.214
201595_s_at
likely ortholog
single
Yes
No








of mouse








immediate








early response,








erythropoietin








4


RPL35A
0.0502
8.64E-03
3.493
4.215
213687_s_at
ribosomal
single
No
No








protein L35a


TRIM44
0.0382
5.64E-03
4.971
4.232
217760_at
tripartite
single
No
No








motif-








containing 44


RAD21
0.0133
9.91E-04
4.274
4.257
200608_s_at
RAD21
single
Yes
Yes








homolog (S.








pombe)


NDUFB4
0.0171
1.53E-03
5.235
4.272
218226_s_at
NADH
single
Yes
No








dehydrogenase








(ubiquinone) 1








beta








subcomplex,








4, 15kDa


EEF1A1
0.071
1.47E-02
3.476
4.296
213477_x_at
eukaryotic
single
Yes
Yes








translation








elongation








factor 1 alpha








1


TTC17
0.0246
2.86E-03
3.633
4.346
218972_at
tetratrico-
single
No
No








peptide repeat








domain 17


PLEKHH1
0.0168
1.48E-03
3.955
4.370
64942_at


N/A
N/A


PSMB1
0.0549
9.90E-03
5.083
4.385
214288_s_at
proteasome
single
No
No








(prosome,








macropain)








subunit, beta








type, 1


RWDD1
0.0133
9.94E-04
3.926
4.411
219598_s_at
RWD domain
single
No
No








containing 1


TAF7
0.0635
1.24E-02
4.126
4.421
201023_at
TAF7 RNA
single
Yes
No








polymerase II,








TATA box








binding








protein (TBP)-








associated








factor, 55kDa


RPL27
0.0555
1.00E-02
3.661
4.435
200025_s_at
ribosomal
single
No
No








protein L27


RPL5
0.0114
7.59E-04
4.900
4.532
200937_s_at
ribosomal
multiple
No
No








protein L5


LOC153561
0.00092
4.23E-06
5.268
4.610
213089_at
hypothetical
single
No
No








protein








LOC153561


RPL38
0.0856
1.95E-02
3.262
4.643
202029_x_at
ribosomal
single
No
No








protein L38


RPL36AL
0.0114
7.47E-04
4.472
4.645
201406_at
ribosomal
multiple
No
No








protein L36a-








like


CCNH
0.0988
2.39E-02
3.661
4.670
204093_at
cyclin H
single
Yes
No


TMSB10
0.0425
6.74E-03
4.215
4.699
217733_s_at
thymosin,
single
Yes
No








beta 10


RPS25
0.0306
3.93E-03
3.331
4.701
200091_s_at
ribosomal
single
No
No








protein S25


SP100
0.0125
8.95E-04
3.849
4.739
202863_at
nuclear
single
Yes
No








antigen Sp100


VDAC3
0.00384
1.14E-04
5.809
4.751
208845_at
voltage-
single
Yes
No








dependent








anion channel








3


H2AFY
0.00139
1.18E-05
5.427
4.763
220375_s_at
H2A histone
single
No
No








family,








member Y


FLJ14668
0.011
7.10E-04
4.890
4.774
215947_s_at
hypothetical
single
No
No








protein








FLJ14668


RRN3
0.0196
1.92E-03
3.931
4.808
216908_x_at
RNA
single
Yes
No








polymerase I








transcription








factor RRN3


MAN1C1
0.000899
3.93E-06
5.500
4.877
218918_at
mannosidase,
single
Yes
No








alpha, class








1C, member 1


HARS
0.00651
2.85E-04
5.305
4.881
202042_at
histidyl-tRNA
single
Yes
No








synthetase


CDC16
0.00384
1.14E-04
5.179
4.904
209659_s_at
CDC16 cell
single
Yes
No








division cycle








16 homolog








(S. cerevisiae)


MRCL3
0.00482
1.70E-04
4.825
5.009
201319_at
myosin
single
Yes
No








regulatory








light chain








MRCL3


SEC63
0.00817
4.40E-04
6.364
5.076
201916_s_at
SEC63-like
single
Yes
No








(S. cerevisiae)


RPL31
0.03
3.81E-03
4.290
5.084
200963_x_at
ribosomal
single
Yes
Yes








protein L31


NIFU
0.0508
8.80E-03
3.867
5.110
209075_s_at
iron-sulfur
single
Yes
No








cluster








assembly








enzyme


MTMR9
0.0186
1.76E-03
6.285
5.141
204837_at
myotubularin
single
No
No








related protein








9


RPL36A
0.00129
9.75E-06
5.312
5.196
217256_x_at
ribosomal
single
No
No








protein L36a



0.0454
7.55E-03
3.958
5.219
200012_x_at


N/A
N/A


CHCHD7
0.042
6.64E-03
3.531
5.286
218642_s_at
coiled-coil-
single
No
No








helix-coiled-








coil-helix








domain








containing 7


RPS17
0.00354
9.59E-05
5.070
5.351
212578_x_at
ribosomal
single
No
No








protein S17


PCM1
0.00216
4.26E-05
6.574
5.370
214118_x_at
pericentriolar
single
No
No








material 1


RPL34
0.0427
6.86E-03
4.804
5.509
200026_at
ribosomal
single
No
No








protein L34


CBX3
0.0211
2.20E-03
6.761
5.622
200037_s_at
chromobox
single
No
No








homolog 3








(HP1 gamma








homolog,








Drosophila)


MRPS31
0.000293
1.07E-07
5.502
5.742
212603_at
mitochondrial
single
No
No








ribosomal








protein S31


RPS3A
0.0419
6.59E-03
4.500
5.768
212391_x_at
ribosomal
multiple
Yes
Yes








protein S3A


RPS19
0.00703
3.39E-04
5.278
5.773
202649_x_at
ribosomal
single
Yes
No








protein S19


RPS15A
0.0543
9.70E-03
3.647
5.841
200781_s_at
ribosomal
single
No
No








protein S15a


NTAN1
0.00489
1.75E-04
6.521
5.993
213062_at
N-terminal
single
Yes
No








asparagine








amidase


GTF3A
0.02
1.98E-03
5.494
6.029
201338_x_at
general
single
Yes
Yes








transcription








factor IIIA


FLJ13213
0.0265
3.22E-03
4.494
6.102
217828_at
hypothetical
single
No
No








protein








FLJ13213


RPS7
0.0363
5.17E-03
4.105
6.111
213941_x_at
ribosomal
single
Yes
Yes








protein S7


NAP1L1
0.0791
1.74E-02
5.196
6.233
212967_x_at
nucleosome
multiple
Yes
No








assembly








protein 1-like








1


RPS6
0.0227
2.51E-03
5.313
6.356
209134_s_at
ribosomal
multiple
Yes
Yes








protein S6


RPS27
0.00161
2.12E-05
5.908
6.361
200741_s_at
ribosomal
single
No
No








protein S27








(metallopansti








mulin 1)


DDX5
0.0895
2.07E-02
4.224
6.423
200033_at
DEAD (Asp-
single
No
No








Glu-Ala-Asp)








box








polypeptide 5


RPLP1
0.061
1.17E-02
4.328
6.622
200763_s_at
ribosomal
single
Yes
Yes








protein, large,








P1


RPS24
0.0176
1.60E-03
4.942
6.800
200061_s_at
ribosomal
single
Yes
Yes








protein S24


PTPN2
0.00752
3.77E-04
6.736
7.030
213136_at
protein
multiple
Yes
Yes








tyrosine








phosphatase,








non-receptor








type 2


RPL4
0.00294
7.14E-05
6.268
7.111
200089_s at
ribosomal
multiple
Yes
Yes








protein L4


RPL22
0.0111
7.15E-04
5.862
7.320
220960_x_at
ribosomal
multiple
Yes
No








protein L22


MRPS22
0.0116
7.92E-04
7.774
7.370
219220_x_at
mitochondrial
single
No
No








ribosomal








protein S22


GPR153
0.0174
1.57E-03
5.836
7.470
220725_x_at


N/A
N/A


MAPRE1
0.00298
7.30E-05
8.747
7.718
200712_s_at
microtubule-
single
Yes
No








associated








protein,








RP/EB family,








member 1


HMGN2
0.0735
1.56E-02
5.160
7.778
208668_x_at
high-mobility
single
Yes
No








group








nucleosomal








binding








domain 2


RPL17
0.00283
6.65E-05
7.454
8.038
200038_s_at
ribosomal
multiple
No
No








protein L17


RPL9
0.022
2.36E-03
5.775
8.273
200032_s_at
ribosomal
single
Yes
Yes








protein L9


FLJ20003
0.000336
2.04E-07
8.503
8.278
219067_s_at
chromosome
single
No
No








10 open








reading frame








86


RPL14
0.00211
3.94E-05
7.439
8.807
200074_s_at
ribosomal
multiple
Yes
Yes








protein L14


RPL6
0.00546
2.13E-04
8.309
8.928
200034_s_at
ribosomal
single
Yes
Yes








protein L6


RPL7
0.0159
1.37E-03
6.500
9.048
200717_x_at
ribosomal
multiple
Yes
Yes








protein L7


FLJ11021
0.0189
1.79E-03
6.351
9.554
202302_s_at
similar to
single
No
No








splicing factor,








arginine/serine-








rich 4


EIF4G2
0.0549
9.90E-03
8.478
10.152
200004_at
eukaryotic
single
Yes
Yes








translation








initiation








factor 4








gamma, 2


BTF3
0.00687
3.17E-04
10.155
10.364
211939_x_at
basic
multiple
No
No








transcription








factor 3


SFRS14
0.00144
1.58E-05
8.161
10.381
213505_s_at
splicing factor,
single
No
No








arginine/serine-








rich 14


SRP14
0.0176
1.60E-03
8.130
11.379
200007_at
signal
single
Yes
Yes








recognition








particle 14kDa








(homologous








Alu RNA








binding








protein)


PTMA
0.00593
2.44E-04
10.333
11.508
200773_x_at
prothymosin,
single
Yes
No








alpha (gene








sequence 28)


RLPS4X
0.00483
1.72E-04
9.330
13.462
216342_x_at
ribosomal
multiple
No
No








protein S4,








X-linked
















TABLE 25










Association Between Preimmunization Expression Levels of


Genes Involved in Protein Synthesis Machinery and Post-


immunization IgG Response











FDR
Odds Ratio




association
association


Gene
with IgG
with IgG


Name
response
response
Gene Description













MRPS31
0.000293
5.502
mitochondrial ribosomal





protein S31


RPL7
0.000732
5.846
ribosomal protein L7


RPL26
0.00121
3.342
ribosomal protein L26


RPL36A
0.00129
5.312
ribosomal protein L36a


RPS27
0.00161
5.908
ribosomal rotein S27





(metallopanstimulin 1)


RPL14
0.00211
7.439
ribosomal protein L14


RPL17
0.00283
7.454
ribosomal protein L17


RPL4
0.00294
6.268
ribosomal protein L4


RPL7
0.00336
6.044
ribosomal protein L7


RPS17
0.00354
5.070
ribosomal protein S17


RPL14
0.00373
5.310
ribosomal protein L14


RPL17
0.00435
6.380
ribosomal protein L17


RPS4X
0.00483
9.330
ribosomal protein S4,





X-linked


RPL6
0.00546
8.309
ribosomal protein L6


RPS3A
0.00631
3.179
ribosomal protein S3A


RPL17
0.00645
5.874
ribosomal protein L17


RPL26L1
0.00676
4.211
ribosomal protein L26-like 1


RPS19
0.00703
5.278
ribosomal protein S19


RPL35
0.00735
3.013
ribosomal protein L35


SSB
0.00795
3.664
Sjogren syndrome antigen B





(autoantigen La)


RPL22
0.0111
5.862
ribosomal protein L22


RPL36AL
0.0114
4.472
ribosomal protein L36a-like


RPL5
0.0114
4.900
ribosomal protein L5


MRPS22
0.0116
7.774
mitochondrial ribosomal





protein S22


RPL35
0.0158
3.439
ribosomal protein L35


RPL7
0.0159
6.500
ribosomal protein L7


RPL22
0.0174
5.543
ribosomal protein L22


RPS24
0.0176
4.942
ribosomal protein S24


RPL36AL
0.0179
4.078
ribosomal protein L36a-like


RPL9
0.022
5.775
ribosomal protein L9


RPS4X
0.0222
5.131
ribosomal protein S4,





X-linked


RPS6
0.0227
5.313
ribosomal protein S6


RPL4
0.0229
4.112
ribosomal protein L4


RPL22
0.0269
5.178
ribosomal protein L22


RPL31
0.03
4.290
ribosomal protein L31


RPS25
0.0306
3.331
ribosomal protein S25


FOXO1A
0.0328
3.167
forkhead box O1A





(rhabdomyosarcoma)


RPL5
0.0354
3.356
ribosomal protein L5


RPS7
0.0363
4.105
ribosomal protein S7


RPL19
0.037
3.543
ribosomal protein L19


EIF3S1
0.0395
3.184
eukaryotic translation





initiation factor 3,





subunit 1a


RPS3A
0.0419
4.500
ribosomal protein S3A


RPL34
0.0427
4.804
ribosomal protein L34


RPL4
0.0431
3.436
ribosomal protein L4


RPL21
0.0454
3.958
ribosomal protein L21





(gene or pseudogene)


RPL35A
0.0502
3.493
ribosomal protein L35a


RPS15A
0.0543
3.647
ribosomal protein S15a


EIF4G2
0.0549
8.478
eukaryotic translation





initiation factor 4 gamma,2
















TABLE 26










Selection of Genes Associated with IgG Responsiveness











Ingenuity category
Gene
FDR
OR
Description














Protein trafficking
SRP14
0.018
8.13
signal recognition particle 14kDa


Protein trafficking
MCM3AP
0.009
3.63
MCM3 minichromosome maintenance






deficient 3 associated protein


Protein trafficking
UBL1
0.04
3.16
SMT3 suppressor of mif two 3 homolog 1






(yeast)


Protein trafficking
KPNA6
0.012
0.32
karyopherin alpha 6 (importin alpha 7)


Protein trafficking
ARF3
0.005
0.32
ADP-ribosylation factor 3


Protein trafficking
XPO7
0.003
0.29
exportin 7


Protein trafficking
KDELR2
0.019
0.29
KDEL endoplasmic reticulum protein






retention receptor 2


Protein trafficking
NUP214
0.018
0.26
nucleoporin 214kDa


Protein synthesis
EIF4G2
0.055
8.48
eukaryotic translation initiation factor 4






gamma, 2


Protein synthesis
RPL6
0.005
8.31
ribosomal protein L6


Protein synthesis
RPL4
0.003
6.27
ribosomal protein L4


Protein synthesis
RPL7
0.003
6.04
ribosomal protein L7


Protein synthesis
RPL9
0.022
5.77
ribosomal protein L9


Protein synthesis
RPS6
0.0227
5.31
ribosomal protein S6


Protein synthesis
RPL14
0.004
5.31
ribosomal protein L14


Protein synthesis
RPS24
0.018
4.94
ribosomal protein S24


Protein synthesis
RPLP1
0.061
4.33
ribosomal protein, large, P1


Protein synthesis
RPL31
0.03
4.29
ribosomal protein L31


Protein synthesis
RPS7
0.036
4.11
ribosomal protein S7


Protein synthesis
SSB
0.008
3.66
Sjogren syndrome antigen B


Protein synthesis
RPL19
0.037
3.54
ribosomal protein L19


Protein synthesis
EEF1A1
0.071
3.48
eukaryotic translation elongation factor 1






alpha 1


Protein synthesis
RPS15
0.087
3.35
ribosomal protein S15


Protein synthesis
RPL26
0.001
3.34
ribosomal protein L26


Protein synthesis
EIF3S1
0.039
3.18
eukaryotic translation initiation factor 3,






subunit 1 alpha, 35kDa


Protein synthesis
FOXO1A
0.033
3.17
forkhead box O1A (rhabdomyosarcoma)


Protein synthesis
TXNRD1
0.057
0.28
thioredoxin reductase 1


Protein synthesis
RELA
0.007
0.26
v-rel reticuloendotheliosis viral oncogene






homolog A, nuclear factor of kappa light






polypeptide gene enhancer in B-cells 3,






p65 (avian)


Protein synthesis
PABPC4
0.004
0.26
poly(A) binding protein, cytoplasmic 4






(inducible form)


Protein synthesis
EIF4A1
0.099
0.22
eukaryotic translation initiation factor 4A,






isoform 1


Protein synthesis
MAP2K3
0.0028
0.19
mitogen-activated protein kinase kinase 3


Protein synthesis
MIKNK1
0.019
0.15
MAP kinase interacting serine/threonine






kinase 1


Protein synthesis
PTBP1
0.002
0.11
polypyrimidine tract binding protein


DNA repair,
CDK2
0.004
4.29
cyclin-dependent kinase 2


replication,


recombination


DNA repair,
TMPO
0.009
3.99
thymopoietin


replication,


recombination


DNA repair,
PRKDC
0.006
3.40
protein kinase, DNA-activated, catalytic


replication,



polypeptide, role in VDJ recombination


recombination


DNA repair,
BUB3
0.021
3.12
BUB3 budding uninhibited by


replication,



benzimidazoles 3 homolog (yeast)


recombination


DNA repair,
XRCC2
0.002
3.03
X-ray repair complementing defective


replication,



repair, role in VDJ recombination.


recombination


DNA repair,
CDKN1A
0.004
0.32
cyclin-dependent kinase inhibitor 1A


replication,



(p21, Cip1)


recombination


DNA repair,
PAFAH1B1
0.0045
0.21
platelet-activating factor acetylhydrolase


replication,



beta subunit (PAF-AH beta)


recombination







FDR = false discovery rate; OR = odds ratio














TABLE 27










Annotation of IgG-associated Genes.













Gene assigned by Ingenuity to one of


+HL,34




the following functions: cell-cycle



(includes DNA synthesis, cell growth



and proliferation), cell death, cell



signaling and interaction (includes



cell signaling and cell-to-cell



signaling and interaction), immune



functions (includes immune and


Identified



lymphatic system development and


by more
Affymetrix



function and immune response),
FDR IgG
Odds
than one
probeset


Gene Name
protein synthesis and trafficking
association
Ratio
probeset
identifier















MRPS31
Yes
0.0003
5.502
No
212603_at


FLJ20003
Not assigned to function by Ingenuity
0.0003
8.503
No
219067_s_at


PGF
No
0.0007
3.555
No
215179_x_a


SLC12A9
Not assigned to function by Ingenuity
0.0007
0.117
No
220371_s_at


FKSG17
Not assigned to function by Ingenuity
0.0007
4.514
No
211445_x_a


MAN1C1
No
0.0009
5.5
No
218918_at


LOC153561
Not assigned to function by Ingenuity
0.0009
5.268
No
213089_at


POLR1B
No
0.0011
3.59
No
220113_x_a


MFN2
No
0.0012
0.205
No
201155_s_at


RIOK3
No
0.0012
3.312
No
215588_x_a


RPL26
Yes
0.0012
3.342
No
222229_x_a


RPL36A
Yes
0.0013
5.312
No
217256_x_a


FLJ10315
Not assigned to function by Ingenuity
0.0014
0.174
No
218770_s_at


H2AFY
Not assigned to function by Ingenuity
0.0014
5.427
No
220375_s_at


MXD4
No
0.0014
0.254
No
212346_s_at


EMT
No
0.0014
0.259
No
207621_s_at


CORO1B
Not assigned to function by Ingenuity
0.0014
0.303
No
64486_at


SFRS14
Not assigned to function by Ingenuity
0.0014
8.161
No
213505_s_at


PLOD
No
0.0015
0.245
No
200827_at


FLJ11560
Not assigned to function by Ingenuity
0.0015
0.189
Yes
211433_x_a


RPS27
Yes
0.0016
5.908
No
200741_s_at


XRCC2
Yes
0.0017
3.033
No
207598_x_a


GRINA
Not assigned to function by Ingenuity
0.0017
0.281
No
212090_at


LAMR1
No
0.0017
3.016
No
216806_at


DKFZPS64J157
Not assigned to function by Ingenuity
0.0018
0.158
No
217794_at


DNASE1L1
No
0.0018
0.231
No
203912_s_at


ESRRBL1
No
0.0019
4.429
No
218100_s_at


ARPC1B
No
0.0020
0.276
No
201954_at


PTBP1
Yes
0.0020
0.112
Yes
211270_x_a


MAPK7
No
0.0021
0.255
No
35617_at


LONP
Not assigned to function by Ingenuity
0.0021
3.226
No
221834_at


WBSCR5
No
0.0021
3.921
No
211768_at


unannotated
Not assigned to function by Ingenuity
0.0021
3.099
No
216006_at


RPL14
Yes
0.0021
7.439
Yes
200074_s_at


PCM1
Not assigned to function by Ingenuity
0.0022
6.574
No
214118_x_a


HDGF
No
0.0024
0.137
No
216484_x_a


unannotated
Not assigned to function by Ingenuity
0.0025
4.29
No
215628_x_a


IMPDH1
No
0.0026
0.274
No
204169_at


FLJ20331
Not assigned to function by Ingenuity
0.0026
3.855
No
215063_x_a


PPP2R4
No
0.0026
0.237
No
208874_x_a


MAP2K3
Yes
0.0028
0.195
No
215499_at


VCP
No
0.0028
0.172
No
208648_at


GTF2H2
Not assigned to function by Ingenuity
0.0028
3.033
No
221540_x_a


PPP2CA
Not assigned to function by Ingenuity
0.0028
3.291
No
217713_x_a


RPL17
Yes
0.0028
7.454
Yes
200038_s_at


GDI1
No
0.0029
0.283
No
201864_at


XPO7
Yes
0.0029
0.298
No
212166_at


RPL4
Yes
0.0029
6.268
Yes
200089_s_at


MAPRE1
No
0.0030
8.747
No
200712_s_at


DKFZp566N034
Not assigned to function by Ingenuity
0.0030
4.15
No
208238_x_a


COBRA1
No
0.0034
0.257
No
202757_at


GSTM3
No
0.0035
3.641
No
202554_s_at


RPS17
Yes
0.0035
5.07
No
212578_x_a


LASS2
Not assigned to function by Ingenuity
0.0036
0.259
No
222212_s_at


NIF3L1BP1
Not assigned to function by Ingenuity
0.0036
3.506
No
218334_at


ATP6V0C
No
0.0036
0.215
No
36994_at


POLR2L
No
0.0036
0.296
No
211730_s_at


MGC10433
Not assigned to function by Ingenuity
0.0036
0.192
No
205740_s_at


C14ORF123
Not assigned to function by Ingenuity
0.0037
3.718
No
218571_s_at


ALMS1
No
0.0037
3.045
No
214707_x_a


CDC16
No
0.0038
5.179
No
209659_s_at


VDAC3
No
0.0038
5.809
No
208845_at


ATP6V0A1
Not assigned to function by Ingenuity
0.0039
0.175
No
212383_at


CAB45
Not assigned to function by Ingenuity
0.0039
0.328
No
217855_x_a


CDKN1A
Yes
0.0040
0.325
No
202284_s_at


SH3BP2
No
0.0040
0.207
No
209370_s_at


CDK2
Yes
0.0040
4.295
No
204252_at


FLJ10460
Not assigned to function by Ingenuity
0.0043
3.085
No
220071_x_a


PAFAH1B1
Yes
0.0045
0.212
No
200815_s_at


BLCAP
Not assigned to function by Ingenuity
0.0046
0.175
No
201032_at


PABPC4
Yes
0.0046
0.256
No
201064_s_at


ARF3
Yes
0.0048
0.32
No
200011_s_at


MRCL3
No
0.0048
4.825
No
201319_at


LSM5
Not assigned to function by Ingenuity
0.0048
3.126
No
211747_s_at


RPS4X
Yes
0.0048
9.33
Yes
216342_x_a


NDUFA6
No
0.0049
3.979
No
202001_s_at


NTAN1
No
0.0049
6.521
No
213062_at


RRAGD
Not assigned to function by Ingenuity
0.0051
0.276
No
221523_s_at


HSPC128
Not assigned to function by Ingenuity
0.0051
3.394
No
218936_s_at


USP9X
No
0.0054
4.43
No
201100_s_at


FLJ10307
Not assigned to function by Ingenuity
0.0054
0.209
No
218753_at


RARG-1
Not assigned to function by Ingenuity
0.0054
4.107
Yes
202882_x_a


PEPD
Not assigned to function by Ingenuity
0.0054
0.273
No
202108_at


RBBP6
No
0.0054
3.065
No
212781_at


RPL6
Yes
0.0055
8.309
No
200034_s_at


ADRM1
Not assigned to function by Ingenuity
0.0055
0.284
No
201281_at


ZNF-U69274
Not assigned to function by Ingenuity
0.0055
4.158
No
204847_at


CDC40
No
0.0056
0.177
No
203376_at


CLIC4
No
0.0056
0.318
No
201560_at


UQCRB
No
0.0058
3.91
Yes
209065_at


PTMA
No
0.0059
10.333
No
200773_x_a


CLN2
No
0.0061
0.331
No
200742_s_at


COX7A3
No
0.0063
4.532
No
217249_x_a


PRKDC
Yes
0.0063
3.401
No
208694_at


SNN
No
0.0064
0.3
No
218033_s_at


DCTN1
No
0.0065
0.3
No
211780_x_a


HARS
No
0.0065
5.305
No
202042_at


Unknown
Not assigned to function by Ingenuity
0.0067
4.084
No
215557_at


ACTR1A
No
0.0067
0.171
No
200721_s_at


NDUFB8
No
0.0067
4.431
No
201227_s_at


SNRPB2
No
0.0068
3.596
No
202505_at


RPL26L1
Yes
0.0068
4.211
No
218830_at


BTF3
Not assigned to function by Ingenuity
0.0069
10.155
Yes
211939_x_a


P29
Not assigned to function by Ingenuity
0.0070
3.659
No
202553_s_at


RELA
Yes
0.0070
0.26
No
201783_s_at


GORASP2
Not assigned to function by Ingenuity
0.0070
0.219
No
207812_s_at


RPS19
Yes
0.0070
5.278
No
202649_x_a


TBCA
Not assigned to function by Ingenuity
0.0071
3.344
No
203667_at


LOC285148
Not assigned to function by Ingenuity
0.0072
0.204
No
213532_at


PTPN2
Yes
0.0075
6.736
Yes
213136_at


GCSH
No
0.0076
4.23
No
213129_s_at


CGI-12
Not assigned to function by Ingenuity
0.0076
3.378
No
219363_s_at


SCD4
Not assigned to function by Ingenuity
0.0077
3.441
No
214036_at


SSB
yes
0.0080
3.664
No
201139_s_at


SEC63
No
0.0082
6.364
No
201916_s_at


ETHE1
Not assigned to function by Ingenuity
0.0087
0.289
No
204034_at


OIP2
Not assigned to function by Ingenuity
0.0087
3.544
No
215136_s_at


CENTA1
No
0.0089
0.255
No
90265_at


MCM3AP
Yes
0.0092
3.627
No
215582_x_a


DULLARD
Not assigned to function by Ingenuity
0.0094
0.263
No
200035_at


KIAA0036
Not assigned to function by Ingenuity
0.0096
4.375
No
211707_s_at


MPST
Not assigned to function by Ingenuity
0.0096
0.259
No
203524_s_at


TMPO
Yes
0.0098
3.992
No
209753_s_at


MFN1
No
0.0099
4.588
No
207098_s_at


LENG4
No
0.0101
0.302
No
205634_x_a


FLJ12287
Not assigned to function by Ingenuity
0.0101
0.332
No
219259_at


TUBA6
Not assigned to function by Ingenuity
0.0104
0.301
Yes
211750_x_a


FURIN
No
0.0106
0.29
No
201945_at


FLJ14668
Not assigned to function by Ingenuity
0.0110
4.89
No
215947_s_at


HNRPH1
No
0.0111
3.327
No
213619_at


KIAA0494
Not assigned to function by Ingenuity
0.0111
0.228
No
201776_s_at


RPL22
Yes
0.0111
5.862
Yes
220960_x_a


FLJ22256
Not assigned to function by Ingenuity
0.0113
3.503
No
220856_x_a


CHK
No
0.0114
0.327
No
204266_s_at


CBARA1
Not assigned to function by Ingenuity
0.0114
0.192
No
216903_s_at


WDR13
Not assigned to function by Ingenuity
0.0114
0.326
No
222138_s_at


RPL36AL
yes
0.0114
4.472
Yes
201406_at


RPL5
yes
0.0114
4.9
Yes
200937_s_at


KIAA1193
Not assigned to function by Ingenuity
0.0115
0.251
No
4822_s_at


K-ALPHA-1
Not assigned to function by Ingenuity
0.0116
0.225
Yes
211058_x_a


GBA
Not assigned to function by Ingenuity
0.0116
0.264
No
209093_s_at


SRF
Yes
0.0116
0.319
No
202401_s_at


MRPS22
yes
0.0116
7.774
No
219220_x_a


KPNA6
Yes
0.0117
0.32
No
212101_at


ABCE1
Not assigned to function by Ingenuity
0.0118
3.786
No
201872_s_at


PP9099
Not assigned to function by Ingenuity
0.0122
0.319
No
204436_at


SP100
No
0.0125
3.849
No
202863_at


FLJ23476
Not assigned to function by Ingenuity
0.0126
3.68
No
218647_s_at


FTLL1
Not assigned to function by Ingenuity
0.0128
3.273
No
217703_x_a


SNX27
Not assigned to function by Ingenuity
0.0131
0.314
No
221498_at


C1GALT1
Not assigned to function by Ingenuity
0.0132
3.612
No
219439_at


RWDD1
Not assigned to function by Ingenuity
0.0133
3.926
No
219598_s_at


RAD21
Yes
0.0133
4.274
No
200608_s_at


SEC31L1
No
0.0134
0.19
No
210616_s_at


PSKH1
Not assigned to function by Ingenuity
0.0137
3.788
No
213141_at


TM6SF1
Not assigned to function by Ingenuity
0.0138
0.271
No
219892_at


SGSH
No
0.0139
0.311
No
35626_at


DAG1
Yes
0.0140
0.256
No
205417_s_at


ATP5I
Not assigned to function by Ingenuity
0.0144
3.511
No
209492_x_a


BAT3
Not assigned to function by Ingenuity
0.0147
0.326
No
201255_x_a


NTRK3
Yes
0.0148
3.298
No
217033_x_a


GLUD1
No
0.0151
0.098
No
200946_x_a


SLC35E1
Not assigned to function by Ingenuity
0.0152
3.961
No
79005_at


CPA2
No
0.0157
3.506
No
206212_at


RPL35
yes
0.0158
3.439
Yes
200002_at


RPL7
Yes
0.0159
6.5
Yes
200717_x_a


XPO6
Not assigned to function by Ingenuity
0.0161
0.237
No
211982_x_a


MGC16824
Not assigned to function by Ingenuity
0.0163
0.226
No
203173_s_at


MGC48332
Not assigned to function by Ingenuity
0.0165
3.326
No
213256_at


PLEKHH1
Not assigned to function by Ingenuity
0.0168
3.955
No
64942_at


NDUFB4
No
0.0171
5.235
No
218226_s_at


GPRC5D
Not assigned to function by Ingenuity
0.0171
4.305
No
221297_at


LEREPO4
No
0.0172
4.607
No
201595_s_at


PXN
No
0.0174
0.296
No
201087_at


GPR153
Not assigned to function by Ingenuity
0.0174
5.836
No
220725_x_a


NUP214
Yes
0.0176
0.264
No
202155_s_at


RPS24
Yes
0.0176
4.942
No
200061_s_at


SRP14
Yes
0.0176
8.13
No
200007_at


FLJ10287
Not assigned to function by Ingenuity
0.0177
3.262
No
219130_at


DNAH3
Not assigned to function by Ingenuity
0.0180
3.282
No
209751_s_at


ZFP95
Not assigned to function by Ingenuity
0.0180
3.562
No
203730_s_at


OS-9
Not assigned to function by Ingenuity
0.0184
0.329
No
200714_x_a


MTMR9
Not assigned to function by Ingenuity
0.0186
6.285
No
204837_at


NPEPPS
No
0.0187
0.266
No
201454_s_at


FACL6
No
0.0188
3.228
No
211207_s_at


FLJ11021
Not assigned to function by Ingenuity
0.0189
6.351
No
202302_s_at


CRKL
No
0.0192
0.18
No
212180_at


MKNK1
Yes
0.0192
0.147
No
209467_s_at


KDELR2
Yes
0.0192
0.297
No
200698_at


ZNF505
Not assigned to function by Ingenuity
0.0193
3.026
No
215758_x_a


GRK6
No
0.0194
0.311
No
210981_s_at


GALNACT-2
No
0.0196
0.315
No
222235_s_at


RRN3
No
0.0196
3.931
No
216908_x_a


GTF3A
Yes
0.0200
5.494
No
201338_x_a


TM9SF2
No
0.0206
0.194
No
201078_at


FLJ34588
Not assigned to function by Ingenuity
0.0206
3.86
No
212410_at


OAZIN
No
0.0208
0.21
No
212461_at


C20ORF35
Not assigned to function by Ingenuity
0.0209
0.306
No
218094_s_at


SMARCD2
No
0.0211
0.268
No
201827_at


CBX3
Not assigned to function by Ingenuity
0.0211
6.761
No
200037_s_at


HSPA4
No
0.0212
4.047
No
208815_x_a


CALM1
No
0.0214
3.136
No
209563_x_a


UBA2
Not assigned to function by Ingenuity
0.0215
4.168
No
201177_s_at


SLC9A8
Not assigned to function by Ingenuity
0.0217
0.315
No
212947_at


GTSE1
Not assigned to function by Ingenuity
0.0219
3.07
No
211040_x_a


RPL9
Yes
0.0220
5.775
No
200032_s_at


MPP2
No
0.0223
3.682
No
207984_s_at


TRAPCC2
Not assigned to function by Ingenuity
0.0224
3.842
No
206853_s_at


DSPP
No
0.0227
3.374
No
221681_s_at


RPS6
Yes
0.0227
5.313
Yes
209134_s_at


PLOD3
No
0.0236
0.277
No
202185_at


ZNF263
No
0.0237
3.509
No
203707_at


TGFB3
Yes
0.0237
3.511
No
209747_at


MGC13024
Not assigned to function by Ingenuity
0.0238
0.273
No
221864_at


LOCS1257
Not assigned to function by Ingenuity
0.0240
0.332
No
210075_at


TCEAL1
No
0.0241
3.163
No
204045_at


VAMP4
No
0.0241
3.512
No
213480_at


NPL4
No
0.0243
0.284
No
217796_s_at


TTC17
Not assigned to function by Ingenuity
0.0246
3.633
No
218972_at


EXT2
No
0.0261
0.198
No
202012_s_at


CANX
No
0.0265
0.226
No
200068_s_at


KIAA0121
Not assigned to function by Ingenuity
0.0265
0.327
No
212399_s_at


FLJ13213
Not assigned to function by Ingenuity
0.0265
4.494
No
217828_at


SGPL1
No
0.0270
0.191
Yes
212321_at


SLC30A5
No
0.0274
3.23
No
218989_x_a


CAMTA2
Not assigned to function by Ingenuity
0.0276
0.301
No
212948_at


GTF2A2
Not assigned to function by Ingenuity
0.0277
0.277
No
202678_at


SS18L2
Not assigned to function by Ingenuity
0.0277
3.151
No
218283_at


SMARCA5
No
0.0278
3.504
No
213251_at


CYCS
No
0.0281
3.257
No
208905_at


NOL5A
Not assigned to function by Ingenuity
0.0284
3.76
No
200874_s_at


ING1L
No
0.0297
3.537
No
205981_s_at


FLJ13910
Not assigned to function by Ingenuity
0.0297
0.257
No
212482_at


EIF4A1
Yes
0.0299
0.221
No
211787_s_at


RPL31
Yes
0.0300
4.29
No
200963_x_a


SOD1
Yes
0.0301
3.742
No
200642_at


RPS25
Yes
0.0306
3.331
No
200091_s_at


ACTR1B
No
0.0317
0.22
No
202135_s_at


DT1P1A10
Not assigned to function by Ingenuity
0.0318
4.334
No
213079_at


MAP3K7
Not assigned to function by Ingenuity
0.0320
3.601
No
215476_at


STOM
Not assigned to function by Ingenuity
0.0322
0.235
No
201060_x_a


K1AA0676
Not assigned to function by Ingenuity
0.0326
0.224
No
215994_x_a


FOXO1A
Yes
0.0328
3.167
No
202724_s_at


COPS7A
Not assigned to function by Ingenuity
0.0333
0.258
No
209029_at


SNAP25
No
0.0337
3.119
No
202507_s_at


HSPA8
No
0.0337
3.651
No
221891_x_a


PLAGL2
Yes
0.0343
0.269
No
202924_s_at


NDUFS5
No
0.0348
3.213
No
201757_at


PTD004
Not assigned to function by Ingenuity
0.0353
3.883
No
219293_s_at


P38IP
Not assigned to function by Ingenuity
0.0361
3.621
No
220408_x_a


RPS7
Yes
0.0363
4.105
No
213941_x_a


CKAP4
Not assigned to function by Ingenuity
0.0365
0.302
No
200998_s_at


LOC92482
Not assigned to function by Ingenuity
0.0370
3.028
No
213220_at


TTC13
Not assigned to function by Ingenuity
0.0370
3.397
No
219481_at


RPL19
Yes
0.0370
3.543
No
200029_at


SON
No
0.0373
3.843
No
214988_s_at


UBE2G1
Not assigned to function by Ingenuity
0.0373
0.274
No
209141_at


HNRPD
No
0.0375
4.357
Yes
200073_s_at


NIP30
Not assigned to function by Ingenuity
0.0382
3.641
No
217896_s_at


TRIM44
Not assigned to function by Ingenuity
0.0382
4.971
No
217760_at


APG4B
No
0.0383
0.326
No
212280_x_a


EIF3S1
Yes
0.0395
3.184
No
208264_s_at


0
Not assigned to function by Ingenuity
0.0397
3.602
No
212436_at


TLE1
No
0.0399
3.14
No
203221_at


TM9SF4
Not assigned to function by Ingenuity
0.0399
0.332
No
212198_s_at


UBL1
Yes
0.0400
3.159
No
211069_s_at


K1AA0252
Not assigned to function by Ingenuity
0.0407
3.099
No
212302_at


DR1
No
0.0412
0.252
No
207654_x_a


BUB3
Yes
0.0419
4.233
Yes
201457_x_a


RPS3A
Yes
0.0419
4.5
Yes
212391_x_a


PIGL
No
0.0420
3.247
No
205873_at


CHCHD7
Not assigned to function by Ingenuity
0.0420
3.531
No
218642_s_at


TMSB10
No
0.0425
4.215
No
217733_s_at


FLJ11712
Not assigned to function by Ingenuity
0.0427
3.38
No
219056_at


RPL34
Not assigned to function by Ingenuity
0.0427
4.804
No
200026_at


TCTEL1
No
0.0433
3.245
No
201999_s_at


TAX1BP1
No
0.0433
3.335
No
200977_s_at


DHX15
Not assigned to function by Ingenuity
0.0435
3.049
No
201385_at


TRIM33
Not assigned to function by Ingenuity
0.0439
0.28
No
213184_at


MYO9B
No
0.0442
0.284
No
214780_s_at


RPL21 (or
yes
0.0454
3.958
No
200012_x_a


RPL21


Pseudogene)


ZNF261
No
0.0458
3.022
No
207559_s_at


HK1
No
0.0468
0.32
No
200697_at


RAB2L
No
0.0477
0.249
No
209110_s_at


NDUFA4
No
0.0501
3.034
No
217773_s_at


RPL35A
yes
0.0502
3.493
No
213687_s_at


FLJ23233
Not assigned to function by Ingenuity
0.0505
3.601
No
58367_s_at


NIFU
No
0.0508
3.867
No
209075_s_at


NONO
Not assigned to function by Ingenuity
0.0513
3.745
No
210470_x_a


LOC57149
Not assigned to function by Ingenuity
0.0526
3.251
No
203897_at


AMFR
No
0.0543
0.313
No
202204_s_at


RPS15A
yes
0.0543
3.647
No
200781_s_at


PSMB1
Not assigned to function by Ingenuity
0.0549
5.083
No
214288_s_at


EIF4G2
Yes
0.0549
8.478
No
200004_at


RPL27
yes
0.0555
3.661
No
200025_s_at


MGC5508
Not assigned to function by Ingenuity
0.0558
0.323
No
201361_at


PEX16
No
0.0559
3.69
No
49878_at


TXNRD1
Yes
0.0573
0.285
No
201266_at


AKR1C1
No
0.0586
3.098
No
216594_x_a


FLJ13725
Not assigned to function by Ingenuity
0.0586
0.322
No
45749_at


NFRKB
Not assigned to function by Ingenuity
0.0586
3.162
No
213028_at


XBP1
No
0.0599
0.292
No
200670_at


RPLP1
Yes
0.0610
4.328
No
200763_s_at


HNRPDL
No
0.0635
4.04
No
209067_s_at


TAF7
No
0.0635
4.126
No
201023_at


B2M
No
0.0662
3.074
No
201891_s_at


PHF2
Not assigned to function by Ingenuity
0.0669
3.458
No
212726_at


JWA
No
0.0670
0.32
No
200760_s_at


C6ORF62
Not assigned to function by Ingenuity
0.0672
3.245
No
208809_s_at


RPL24
Not assigned to function by Ingenuity
0.0682
3.055
No
214143_x_a


C21ORF97
Not assigned to function by Ingenuity
0.0686
0.286
No
218019_s_at


MCFD2
Not assigned to function by Ingenuity
0.0687
3.299
No
212245_at


EEF1A1
Yes
0.0710
3.476
No
213477_x_a


MCAM
No
0.0733
3.745
No
211042_x_a


HMGN2
No
0.0735
5.16
No
208668_x_a


LOC144983
Not assigned to function by Ingenuity
0.0735
3.185
No
216559_x_a


NEDD5
Not assigned to function by Ingenuity
0.0750
3.861
No
200015_s_at


SH3GLB1
No
0.0764
0.279
No
209090_s_at


FLJ10996
Not assigned to function by Ingenuity
0.0773
3.15
No
219774_at


TCEB1
No
0.0777
3.198
No
202824_s_at


FLJ10521
Not assigned to function by Ingenuity
0.0790
0.319
No
221656_s_at


NAP1L1
No
0.0791
5.196
Yes
212967_x_a


RANBP9
No
0.0805
3.008
No
202582_s_at


SMP1
Not assigned to function by Ingenuity
0.0833
0.278
No
217766_s_at


NAB1
No
0.0838
3.184
No
211139_s_at


RPL38
yes
0.0856
3.262
No
202029_x_a


RPS15
Yes
0.0867
3.348
No
200819_s_at


DDX5
Not assigned to function by Ingenuity
0.0895
4.224
No
200033_at


HCDI
Not assigned to function by Ingenuity
0.0906
3.791
No
213398_s_at


PITPNC1
No
0.0956
3.059
No
219155_at


SDF2
No
0.0965
0.227
No
203090_at


CCNH
No
0.0988
3.661
No
204093_at


DNAJC8
Not assigned to function by Ingenuity
0.0997
3.323
No
212491_s_at


ELK1
No
0.0999
0.324
No
203617_x_a
























TABLE 28









Affymetrix



IgG






probeset

Level in IgG
Signal-to-
Odds
FDR
Encephalitis
FDR


Rank
identifier
Gene Description
Responders
Noise Score
Ratio
IgG
Odds Ratio
Encephalitis























1
202344_at
HSF1 - heat shock
Decreased
0.82
0.383
0.00129
0.185
0.047996




transcription factor 1


2
205875_s_at
TREX1 - three prime
Decreased
0.8
0.411
0.00159
0.318
0.06771




repair exonuclease 1


3
212907_at
UNK_AI972416-
Decreased
0.78
0.434
0.0229
0.594
0.536404




Human hbc647 mRNA




sequence.


4
201574_at
ETF1 - eukaryotic
Decreased
0.78
0.194
0.000715
0.075
0.041668




translation termination




factor 1


5
218037_at
MGC3035 - hypothetical
Decreased
0.76
0.208
0.0011
0.065
0.038226




protein MGC3035


6
209215_at
TETRAN - tetracycline
Decreased
0.76
0.399
0.00134
0.421
0.205058




transporter-like protein


7
201360_at
CST3 - cystatin C
Decreased
0.74
0.481
0.00311
0.395
0.073239




(amyloid angiopathy and




cerebral hemorr


8
215706_x_at
ZYX-zyxin
Decreased
0.74
0.547
0.00256
0.471
0.108013


9
201954_at
ARPC1B - actin related
Decreased
0.73
0.276
0.00202
0.263
0.242364




protein ⅔ complex,




subunit 1B, 41k


10
221725_at
WASF2 - WAS protein
Decreased
0.72
0.108
0.00144
0.026
0.076275




family, member 2


11
201720_s_at
LAPTM5-Lysosomal-
Decreased
0.69
0.212
0.00455
0.076
0.067654




associated multispanning




membrane protei


12
217811_at
SELT - selenoprotein T
Decreased
0.68
0.454
0.0032
0.245
0.073377


13
202373_s_at
RAB3-GAP150 - rab3
Increased
0.76
5.468
0.00659
70.713
0.051166




GTPase-activating




protein, non-catalytic




subu


14
216806_at
UNK_AL136306 -
Increased
0.73
3.016
0.00169
2.453
0.369437




Consensus includes




gb:AL136306




/DEF = Human DNA sequ


15
213509_x_at
CES2 - carboxylesterase
Increased
0.68
2.777
0.00129
11.698
0.01765




2 (intestine, liver)


16
216508_x_at
UNK_AC007277 -
Increased
0.67
2.476
0.048
7.724
0.183538




Consensus includes




gb:AC007277




/DEF = Homo sapiens B


17
218918_at
MAN1C1 - mannosidase,
Increased
0.66
5.5
0.000899
9.161
0.129904




alpha, class 1C, member




1


18
212637_s_at
WWP 1 - WW domain-
Increased
0.66
2.226
0.0496
3.469
0.275866




containing protein 1


19
215221_at
UNK_AK025064-
Increased
0.64
2.717
0.00828
2.156
0.477634




Homo sapiens cDNA:




FLJ21411 fis, clone




COL03986.


20
202909_at
EPM2AIP1 - EPM2A
Increased
0.63
3.836
0.00167
8.301
0.067315




(laforin) interacting




protein 1


21
204528_s_at
NAP1L1 - nucleosome
Increased
0.62
4.079
0.000732
2.709
0.357819




assembly protein 1-like 1


22
203371_s_at
NDUFB3 - NADH
Increased
0.61
4.865
0.00985
52.955
0.053723




dehydrogenase




(ubiquinone) 1 beta




subcomplex,


23
208845_at
UNK_BC002456-
Increased
0.61
5.809
0.00384
11.618
0.159569




gb:BC002456.1/




DEF = Homo sapiens,




voltage-dependent


24
200685_at
SFRS11 - splicing factor,
Increased
0.6
1.998
0.0264
1.19
0.877872




arginine/serine-rich 11




















TABLE 29








Patient
Confidence
True

Correct


ID
Score
Class
Classification
Classification



















2
0.643
IGG-RESP
IGG-RESP
Yes


4
0.715
IGG-RESP
IGG-RESP
Yes


5
0.817
IGG-RESP
IGG-RESP
Yes


7
0.889
IGG-NON
IGG-NON
Yes


10
0.805
IGG-NON
IGG-NON
Yes


12
0.853
IGG-NON
IGG-RESP
No


14
0.744
IGG-NON
IGG-NON
Yes


15
0.047
IGG-RESP
IGG-RESP
Yes


16
0.601
IGG-RESP
IGG-RESP
Yes


17
0.916
IGG-NON
IGG-NON
Yes


18
1.000
IGG-NON
IGG-NON
Yes


19
0.744
IGG-RESP
IGG-RESP
Yes


22
0.520
IGG-RESP
IGG-RESP
Yes


23
0.477
IGG-NON
IGG-NON
Yes


25
1.000
IGG-NON
IGG-NON
Yes


28
0.018
IGG-RESP
IGG-NON
No


29
0.162
IGG-RESP
IGG-RESP
Yes


31
0.379
IGG-RESP
IGG-NON
No


32
1.000
IGG-RESP
IGG-RESP
Yes


33
0.334
IGG-RESP
IGG-RESP
Yes


34
0.844
IGG-NON
IGG-NON
Yes


36
0.556
IGG-RESP
IGG-RESP
Yes


40
0.563
IGG-RESP
IGG-NON
No


41
0.310
IGG-RESP
IGG-NON
No


43
0.602
IGG-NON
IGG-NON
Yes


44
0.835
IGG-NON
IGG-NON
Yes


48
0.756
IGG-NON
IGG-NON
Yes


52
0.911
IGG-NON
IGG-NON
Yes


53
0.002
IGG-RESP
IGG-RESP
Yes


54
0.958
IGG-NON
IGG-NON
Yes


55
0.935
IGG-NON
IGG-RESP
No


57
0.817
IGG-NON
IGG-NON
Yes


64
0.403
IGG-RESP
IGG-NON
No


66
0.062
IGG-NON
IGG-NON
Yes


68
0.822
IGG-NON
IGG-NON
Yes


69
1.000
IGG-NON
IGG-NON
Yes


70
0.621
IGG-NON
IGG-NON
Yes


71
0.868
IGG-NON
IGG-NON
Yes


252
0.703
IGG-RESP
IGG-NON
No


254
0.947
IGG-NON
IGG-NON
Yes


255
0.446
IGG-RESP
IGG-RESP
Yes


258
0.967
IGG-NON
IGG-NON
Yes


259
0.565
IGG-RESP
IGG-NON
No


260
0.172
IGG-NON
IGG-NON
Yes


262
1.000
IGG-NON
IGG-NON
Yes


263
0.747
IGG-NON
IGG-NON
Yes


266
0.739
IGG-NON
IGG-NON
Yes


269
0.971
IGG-NON
IGG-NON
Yes


271
0.038
IGG-NON
IGG-NON
Yes


274
0.479
IGG-NON
IGG-NON
Yes


277
0.885
IGG-RESP
IGG-RESP
Yes


279
1.000
IGG-NON
IGG-NON
Yes


280
0.846
IGG-NON
IGG-NON
Yes


281
0.642
IGG-NON
IGG-NON
Yes


285
1.000
IGG-NON
IGG-NON
Yes


286
0.573
IGG-NON
IGG-RESP
No


287
0.108
IGG-RESP
IGG-NON
No


289
0.833
IGG-NON
IGG-NON
Yes


290
0.553
IGG-NON
IGG-NON
Yes


291
1.000
IGG-NON
IGG-NON
Yes


293
0.713
IGG-RESP
IGG-RESP
Yes


294
0.811
IGG-NON
IGG-NON
Yes


295
0.429
IGG-NON
IGG-NON
Yes


296
0.088
IGG-RESP
IGG-NON
No


299
0.704
IGG-RESP
IGG-RESP
Yes


300
0.041
IGG-NON
IGG-NON
Yes


301
0.634
IGG-RESP
IGG-RESP
Yes


302
0.811
IGG-NON
IGG-NON
Yes


303
0.748
IGG-NON
IGG-NON
Yes


304
0.986
IGG-NON
IGG-NON
Yes


306
0.905
IGG-RESP
IGG-NON
No


307
0.980
IGG-NON
IGG-NON
Yes


308
0.492
IGG-RESP
IGG-NON
No


314
1.000
IGG-NON
IGG-NON
Yes


315
1.000
IGG-NON
IGG-NON
Yes


316
0.942
IGG-NON
IGG-NON
Yes


319
0.721
IGG-NON
IGG-NON
Yes


503
0.907
IGG-RESP
IGG-RESP
Yes


506
0.170
IGG-NON
IGG-NON
Yes


507
0.341
IGG-NON
IGG-NON
Yes


508
0.798
IGG-RESP
IGG-RESP
Yes


509
0.201
IGG-RESP
IGG-NON
No


514
0.987
IGG-NON
IGG-NON
Yes


515
0.667
IGG-NON
IGG-NON
Yes


516
0.942
IGG-NON
IGG-RESP
No


752
0.990
IGG-RESP
IGG-NON
No


753
0.400
IGG-NON
IGG-NON
Yes


755
0.115
IGG-NON
IGG-RESP
No


756
0.892
IGG-NON
IGG-NON
Yes


757
0.613
IGG-NON
IGG-NON
Yes


758
0.552
IGG-NON
IGG-NON
Yes


760
0.712
IGG-NON
IGG-NON
Yes


762
0.574
IGG-NON
IGG-RESP
No


763
0.352
IGG-NON
IGG-NON
Yes


765
0.995
IGG-NON
IGG-NON
Yes























TABLE 30












IgG






Affymetrix

Signal-to-
Odds
FDR
Encephalitis
FDR


Rank
identifier
Gene Description
Noise Score
Ratio
IgG
Odds Ratio
Encephalitis






















1
202344_at
HSF1 - heat shock
0.82
0.383
0.00129
0.185
0.047996




transcription factor 1


2
205875_s_at
TREX 1 - three prime
0.8
0.411
0.00159
0.318
0.06771




repair exonuclease 1


3
212907_at
UNK_A1972416-
0.78
0.434
0.0229
0.594
0.536404




Human hbc647 mRNA




sequence.


4
202373_s_at
RAB3-GAP150 - rab3
0.76
5.468
0.00659
0.713
0.051166




GTPase-activating




protein, non-catalytic




subu


5
216806_at
UNK_AL136306-
0.73
3.016
0.00169
2.453
0.369437




Consensus includes




gb:AL136306/




DEF = Human DNA




sequ


6
213509_x_at
CES2 -
0.68
2.777
0.00129
11.698
0.01765




carboxylesterase 2




(intestine, liver)



















TABLE 31








Patient
Confidence
True



ID
Score
Class
Classification


















2
0.694
IGG-RESP
IGG-RESP


4
0.975
IGG-RESP
IGG-RESP


5
1.000
IGG-RESP
IGG-RESP


7
0.577
IGG-NON
IGG-NON


10
1.000
IGG-NON
IGG-NON


12
1.000
IGG-NON
IGG-RESP


14
0.186
IGG-NON
IGG-NON


15
1.000
IGG-RESP
IGG-RESP


16
0.118
IGG-RESP
IGG-RESP


17
0.867
IGG-NON
IGG-NON


18
1.000
IGG-NON
IGG-NON


19
1.000
IGG-RESP
IGG-RESP


22
0.516
IGG-RESP
IGG-RESP


23
1.000
IGG-NON
IGG-NON


25
1.000
IGG-NON
IGG-NON


28
1.000
IGG-RESP
IGG-NON


29
0.045
IGG-NON
IGG-RESP


31
0.257
IGG-NON
IGG-NON


32
1.000
IGG-RESP
IGG-RESP


33
0.297
IGG-RESP
IGG-RESP


34
0.229
IGG-NON
IGG-NON


36
0.858
IGG-RESP
IGG-RESP


40
0.569
IGG-RESP
IGG-NON


41
0.869
IGG-RESP
IGG-NON


43
0.119
IGG-RESP
IGG-NON


44
0.686
IGG-NON
IGG-NON


48
0.241
IGG-NON
IGG-NON


52
0.592
IGG-NON
IGG-NON


53
0.234
IGG-RESP
IGG-RESP


54
1.000
IGG-NON
IGG-NON


55
0.965
IGG-NON
IGG-RESP


57
1.000
IGG-NON
IGG-NON


64
0.176
IGG-RESP
IGG-NON


66
1.000
IGG-RESP
IGG-NON


68
1.000
IGG-NON
IGG-NON


69
1.000
IGG-NON
IGG-NON


70
0.450
IGG-NON
IGG-NON


71
0.593
IGG-NON
IGG-NON


252
0.681
IGG-RESP
IGG-NON


254
1.000
IGG-NON
IGG-NON


255
0.676
IGG-RESP
IGG-RESP


258
1.000
IGG-NON
IGG-NON


259
1.000
IGG-NON
IGG-NON


260
0.272
IGG-NON
IGG-NON


262
1.000
IGG-NON
IGG-NON


263
0.623
IGG-NON
IGG-NON


266
0.973
IGG-NON
IGG-NON


269
1.000
IGG-NON
IGG-NON


271
0.577
IGG-NON
IGG-NON


274
0.665
IGG-RESP
IGG-NON


277
1.000
IGG-RESP
IGG-RESP


279
1.000
IGG-NON
IGG-NON


280
1.000
IGG-NON
IGG-NON


281
0.338
IGG-NON
IGG-NON


285
1.000
IGG-NON
IGG-NON


286
0.036
IGG-RESP
IGG-RESP


287
0.382
IGG-NON
IGG-NON


289
0.156
IGG-NON
IGG-NON


290
0.902
IGG-NON
IGG-NON


291
1.000
IGG-NON
IGG-NON


293
0.042
IGG-RESP
IGG-RESP


294
1.000
IGG-NON
IGG-NON


295
0.437
IGG-NON
IGG-NON


296
0.034
IGG-NON
IGG-NON


299
1.000
IGG-RESP
IGG-RESP


300
0.574
IGG-RESP
IGG-NON


301
0.966
IGG-RESP
IGG-RESP


302
1.000
IGG-NON
IGG-NON


303
0.200
IGG-NON
IGG-NON


304
1.000
IGG-NON
IGG-NON


306
1.000
IGG-RESP
IGG-NON


307
1.000
IGG-NON
IGG-NON


308
1.000
IGG-RESP
IGG-NON


314
1.000
IGG-NON
IGG-NON


315
1.000
IGG-NON
IGG-NON


316
0.996
IGG-NON
IGG-NON


319
0.937
IGG-NON
IGG-NON


503
1.000
IGG-RESP
IGG-RESP


506
0.861
IGG-RESP
IGG-NON


507
0.639
IGG-NON
IGG-NON


508
1.000
IGG-RESP
IGG-RESP


509
0.601
IGG-RESP
IGG-NON


514
1.000
IGG-NON
IGG-NON


515
0.225
IGG-NON
IGG-NON


516
0.766
IGG-NON
IGG-RESP


752
0.908
IGG-RESP
IGG-NON


753
0.244
IGG-NON
IGG-NON


755
0.020
IGG-NON
IGG-RESP


756
1.000
IGG-NON
IGG-NON


757
1.000
IGG-NON
IGG-NON


758
1.000
IGG-NON
IGG-NON


760
0.990
IGG-NON
IGG-NON


762
1.000
IGG-NON
IGG-RESP


763
0.402
IGG-NON
IGG-NON


765
1.000
IGG-NON
IGG-NON
















TABLE 32










Genes Associated with Meningoencephalitis














Odds







Ratio for




association



Meningo-
with



encephalitis
meningo-
Unadjusted

Affymetrix


Gene
FDR
encephalitis
p values
Description
identifier















STAT1
0.004
230.416
5.10E-07
signal transducer and
209969_s_at






activator of transcription






1, 91kDa


NHP2L1
0.010
3136.203
2.17E-05
NHP2 non-histone
201076_at






chromosome protein 2-






like 1 (S. cerevisiae)


C10ORF7
0.010
673.31
7.19E-06
chromosome 10 open
201725_at






reading frame 7


FLJ11806
0.010
651.763
1.99E-05
nuclear protein UKp68
213064_at


ZW10
0.010
470.958
3.04E-05
ZW10 homolog,
204812_at






centromere/kinetochore






protein (Drosophila)


C12ORF22
0.010
459.155
1.83E-05
chromosome 12 open
221260_s_at






reading frame 22


ICMT
0.010
417.532
1.62E-05
isoprenylcysteine
201609_x_at






carboxyl






methyltransferase


RABGAP1
0.010
303.809
1.13E-05
RAB GTPase activating
204028_s_at






protein 1


TRAP240
0.010
68.675
9.52E-06
thyroid hormone receptor
201986_at






associated protein 1


SEC24C
0.010
66.791
3.08E-05
SEC24 related gene
202361_at






family, member C (S.






cerevisiae)


BRD2
0.010
56.318
1.06E-05
bromodomain containing
208686_s_at






2


KPNB1
0.010
32.282
3.83E-05
karyopherin (importin)
208975_s_at






beta 1


GZMB
0.010
31.809
3.68E-05
granzyme B (granzyme
210164_at






cytotoxic T-






lymphocyte-associated






serine esterase 1)


FNBP3
0.010
13.972
3.19E-05
formin binding protein 3
213729_at


KLF2
0.010
0.038
3.72E-05
Kruppel-like factor 2
219371_s_at






(lung)


STK17B
0.010
0.025
1.38E-05
serine/threonine kinase
205214_at






17b (apoptosis-inducing)


JARID1B
0.010
0.006
3.93E-05
Jumonji, AT rich
211202_s_at






interactive domain 1B






(RBP2-like)


MGC21416
0.011
8.373
5.98E-05
hypothetical protein
212341_at






MGC21416


STAT3
0.011
6.606
5.96E-05
signal transducer and
208991_at






activator of transcription






3 (acute-phase response






factor)


OSBPL8
0.011
4.201
5.85E-05
oxysterol binding
212582_at






protein-like 8


BTG2
0.011
0.033
5.26E-05
BTG family, member 2
201236_s_at


UBE2D3
0.011
0.002
5.40E-05
ubiquitin-conjugating
200669_s_at






enzyme E2D 3 (UBC4/5






homolog, yeast)


HEAB
0.011
0.001
5.41E-05
ATP/GTP-binding
204370_at






protein


ATP6V1D
0.011
172.543
6.85E-05
ATPase, H+ transporting,
208899_x_at






lysosomal 34kDa, V1






subunit D


KIF5B
0.011
3.731
7.12E-05
kesin family member
201991_s_at






5B


DC8
0.012
69.508
8.01E-05
DKFZP566O1646
209484_s_at






protein


CD84
0.013
23.97
1.01E-04
CD84 antigen (leukocyte
205988_at






antigen)


Unknown
0.013
0.015
9.59E-05
no sequence similarity to
211444_at






any genes or proteins


GLTSCR1
0.013
0.013
9.14E-05
glioma tumor suppressor
219445_at






candidate region gene 1


UGCG
0.013
14.445
1.10E-04
UDP-glucose ceramide
204881_s_at






glucosyltransferase


SFRS21P
0.014
57.281
1.14E-04
splicing factor,
206989_s_at






arginine/serine-rich 2,






interacting protein


MMP24
0.014
0.022
1.16E-04
matrix metalloproteinase
78047_s_at






24 (membrane-inserted)


GCDH
0.014
50.321
1.33E-04
glutaryl-Coenzyme A
203500_at






dehydrogenase


TNPO3
0.014
21.713
1.32E-04
transportin 3
212318_at


MBD4
0.014
8.79
1.28E-04
methyl-CpG binding
209579_s_at






domain protein 4


PABPC1
0.014
0.006
1.29E-04
poly(A) binding protein,
215823_x_at






cytoplasmic 1


VDR
0.014
7.092
1.50E-04
vitamin D (1,25-
204255_s_at






dihydroxyvitamin D3)






receptor


H2AFY
0.015
0.016
1.62E-04
H2A histone family,
207168_s_at






member Y


CBX6
0.016
34.482
1.79E-04
chromobox homolog 6
202047_s_at


IL2RA
0.016
11.266
1.77E-04
interleukin 2 receptor,
211269_s_at






alpha


TTC3
0.016
5.376
1.78E-04
tetratricopeptide repeat
208662_s_at






domain 3


STAT5B
0.016
0.029
1.81E-04
signal transducer and
212549_at






activator of transcription






5B


TRIP13
0.016
17.331
1.88E-04
thyroid hormone receptor
204033_at






interactor 13


FLJ23441
0.016
17.419
1.95E-04
hypothetical protein
219217_at






FLJ23441


STXBP2
0.016
0.095
1.94E-04
syntaxin binding protein
209367_at






2


LRRFIP1
0.016
18.564
1.99E-04
leucine rich repeat (in
201862_s_at






FLII) interacting protein






1


PADI2
0.016
0.145
2.08E-04
peptidyl arginine
209791_at






deiminase, type II


HNRPC
0.016
324.673
2.15E-04
heterogeneous nuclear
214737_x_at






ribonucleoprotein C






(C1/C2)


PTPRC
0.017
4.891
2.29E-04
protein tyrosine
212587_s_at






phosphatase, receptor






type, C


PTDSR
0.018
0.018
2.46E-04
phosphatidylserine
212723_at






receptor


HUMGT198A
0.018
8.097
2.57E-04
GT198, complete ORF
205956_x_at


TPR
0.018
4.823
2.56E-04
translocated promoter
201730_s_at






region (to activated MET






oncogene)


DUT
0.018
40.207
2.74E-04
dUTP pyrophosphatase
208955_at


RAB1A
0.018
0.003
2.71E-04
RAB1A, member RAS
208724_s_at






oncogene family


HMG2L1
0.019
5.679
2.87E-04
high-mobility group
212596_s_at






protein 2-like 1


RIN3
0.019
0.105
2.92E-04
Ras and Rab interactor 3
60471_at


PDCD8
0.019
119.631
3.15E-04
programmed cell death 8
205512_s_at






(apoptosis-inducing






factor)


GLS
0.019
60.862
3.19E-04
glutaminase
203159_at


CSE1L
0.019
38.753
3.13E-04
CSE1 chromosome
201112_s_at






segregation 1-like (yeast)


RNMT
0.019
0.050
3.15E-04
RNA (guanine-7-)
202684_s_at






methyltransferase


TFE3
0.019
0.041
3.18E-04
transcription factor
206649_s_at






binding to IGHM






enhancer 3


FLJ12788
0.020
167.936
3.23E-04
hypothetical protein
218838_s_at






FLJ12788


MGAT2
0.020
20.774
3.29E-04
mannosyl (alpha-1,6-)-
203102_s_at






glycoprotein beta-1,2-N-






acetylglucosaminyl-






transferase


CGI-37
0.021
10.964
3.67E-04
comparative gene
219031_s_at






identification transcript






37


LUC7A
0.021
7.673
3.58E-04
cisplatin resistance-
208835_s_at






associated overexpressed






protein


FBXW7
0.021
5.619
3.66E-04
F-box and WD-40
218751_s_at






domain protein 7






(archipelago homolog,






Drosophila)


DICER1
0.021
0.073
3.62E-04
Dicer1, Dcr-1 homolog
216260_at






(Drosophila)


UBCE7IP5
0.021
0.036
3.52E-04
likely ortholog of mouse
204598_at






ubiquitin conjugating






enzyme 7 interacting






protein 5


C21ORF80
0.021
0.032
3.62E-04
protein O-
209578_s_at






fucosyltransferase 2


TXNL2
0.021
152.265
3.83E-04
thioredoxin-like 2
209080_x_at


PRKRA
0.022
0.027
3.98E-04
protein kinase,
209139_s_at






interferon-inducible






double stranded RNA






dependent activator


BARD1
0.022
11.776
4.11E-04
BRCA1 associated RING
205345_at






domain 1


SH3BP5
0.022
11.205
4.16E-04
SH3-domain binding
201810_s_at






protein 5 (BTK-






associated)


OBRGRP
0.022
4.025
4.13E-04
leptin receptor gene-
202378_s_at






related protein


C1ORF33
0.023
12.564
4.43E-04
chromosome 1 open
220688_s_at






reading frame 33


M96
0.023
9.28
4.39E-04
likely ortholog of mouse
203346_s_at






metal response element






binding transcription






factor 2


Unknown
0.023
0.109
4.44E-04
Unknown containing a
214801_at






LAP1C protein domain


IPO4
0.023
29.56
4.62E-04
importin 4
218305_at


DNCL1
0.023
6.81
4.56E-04
dynein, cytoplasmic,
200703_at






light polypeptide 1


BAZ1A
0.023
6.808
4.63E-04
bromodomain adjacent to
217985_s_at






zinc finger domain, 1A


NALP1
0.023
0.133
4.53E-04
NACHT, leucine rich
218380_at






repeat and PYD






containing 1


GNAS
0.023
0.071
4.59E-04
GNAS complex locus
200780_x_at


TH1L
0.024
13.185
4.76E-04
TH1-like (Drosophila)
220607_x_at


IRS2
0.024
0.060
4.80E-04
insulin receptor substrate
209185_s_at






2


LTF
0.025
0.325
5.08E-04
lactotransferrin
202018_s_at


MIRAB13
0.026
0.109
5.40E-04
molecule interacting with
221779_at






Rab13


BATF
0.026
9.718
5.45E-04
basic leucine zipper
205965_at






transcription factor,






ATF-like


FLN29
0.026
176.965
5.51E-04
FLN29 gene product
35254_at


HAX1
0.026
34.12
5.59E-04
HS1 binding protein
201145_at


MYO1B
0.026
18.41
5.61E-04
myosin IB
212365_at


SLC5A3
0.026
4.832
5.56E-04
solute carrier family 5
213164_at






(inositol transporters),






member 3


PADI4
0.026
0.108
5.62E-04
peptidyl arginine
220001_at






deiminase, type IV


STK10
0.026
0.052
5.72E-04
serine/threonine kinase
40420_at






10


RAB2
0.027
0.002
5.96E-04
RAB2, member RAS
208734_x_at






oncogene family


BPI
0.027
0.219
6.23E-04
bactericidal/permeability-
205557_at






increasing protein


DEFA4
0.027
0.196
6.31E-04
defensin, alpha 4,
207269_at






corticostatin


KPNA6
0.028
34.224
6.49E-04
karyopherin alpha 6
212103_at






(importin alpha 7)


C19ORF10
0.028
45.058
6.57E-04
chromosome 19 open
221739_at






reading frame 10


DKFZPS64G2022
0.028
11.966
6.66E-04
DKFZP564G2022
212202_s_at






protein


SNRK
0.028
0.043
6.63E-04
SNF-1 related kinase
209481_at


GBP1
0.028
5.53
6.70E-04
guanylate binding protein
202269_x_at






1, interferon-inducible,






67kDa


ZFP36
0.029
0.108
7.02E-04
zinc finger protein 36,
201531_at






C3H type, homolog






(mouse)


ZNF238
0.029
0.120
7.15E-04
zinc finger protein 238
212774_at


SIPA1
0.029
0.053
7.17E-04
signal-induced
204164_at






proliferation-associated






gene 1


CXCL10
0.029
7.825
7.34E-04
chemokine (C-X-C
204533_at






motif) ligand 10


RRM2
0.029
5.394
7.24E-04
ribonucleotide reductase
209773_s_at






M2 polypeptide


RAB31
0.029
3.04
7.52E-04
RAB31, member RAS
217762_s_at






oncogene family


USP36
0.029
0.071
7.53E-04
ubiquitin specific
220370_s_at






protease 36


PTP4A1
0.029
0.034
7.54E-04
protein tyrosine
200732_s_at






phosphatase type IVA,






member 1


DPCK
0.029
156.071
7.58E-04
Coenzyme A synthase
201913_s_at


ALDOC
0.029
11.591
7.75E-04
aldolase C, fructose-
202022_at






bisphosphate


PXMP3
0.030
39.115
8.14E-04
peroxisomal membrane
210296_s_at






protein 3, 35kDa






(Zellweger syndrome)


ZFP36L1
0.030
0.036
8.11E-04
zinc finger protein 36,
211962_s_at






C3H type-like 1


CYLN2
0.030
0.060
8.26E-04
cytoplasmic linker 2
211031_s_at


STAU
0.031
0.078
8.49E-04
staufen, RNA binding
213037_x_at






protein (Drosophila)


PHF1
0.031
0.130
8.60E-04
PHD finger protein 1
202928_s_at


HN1
0.031
18.055
8.74E-04
hematological and
217755_at






neurological expressed 1


STOML2
0.031
6.512
8.78E-04
stomatin (EPB72)-like 2
215416_s_at


ARID3B
0.031
0.149
8.77E-04
AT rich interactive
218964_at






domain 3B (BRIGHT-






like)


IL19
0.031
8.869
8.93E-04
interleukin 19
220745_at


WSX1
0.032
46.587
9.17E-04
interleukin 27 receptor,
205926_at






alpha


NFE2L1
0.032
33.502
9.06E-04
nuclear factor (erythroid-
200759_x_at






derived 2)-like 1


TDE1
0.032
17.535
9.38E-04
tumor differentially
211769_x_at






expressed 1


NALP2
0.032
16.21
9.48E-04
NACHT, leucine rich
221690_s_at






repeat and PYD






containing 2


POLA
0.032
14.919
8.99E-04
polymerase (DNA
204835_at






directed), alpha


CKLFSF6
0.032
13.746
9.50E-04
chemokine-like factor
217947_at






super family 6


SSH1
0.032
11.182
9.13E-04
slingshot homolog 1
221753_at






(Drosophila)


MINK
0.032
0.145
9.49E-04
misshapen/NIK-related
214246_x_at






kinase


DKFZP434H132
0.032
0.143
9.22E-04
DKFZP434H132 protein
215087_at


JM5
0.032
0.114
9.56E-04
WD repeat domain, X-
209216_at






linked 1


FLJ13479
0.032
0.010
9.37E-04
hypothetical protein
219047_s_at






FLJ13479


MKI67
0.032
69.144
1.01E-03
antigen identified by
212021_s_at






monoclonal antibody Ki-






67


RBX1
0.032
27.734
1.01E-03
ring-box 1
218117—at


TIMM13
0.032
22.616
1.00E-03
translocase of inner
218188_s_at






mitochondrial membrane






13 homolog (yeast)


ECHDC1
0.032
16.161
1.01E-03
enoyl Coenzyme A
219974_x_at






hydratase domain






containing 1


KIAA0930
0.032
14.228
1.01E-03
chromosome 22 open
212421_at






reading frame 9


HEG
0.032
6.044
1.02E-03
HEG homolog
212822_at


MASK
0.032
5.562
1.01E-03
ankyrin repeat and KH
208772_at






domain containing 1


JUNB
0.032
0.108
1.02E-03
jun B proto-oncogene
201473_at


C9ORF28
0.032
0.037
1.01E-03
chromosome 9 open
52975_at






reading frame 28


RLF
0.032
0.028
1.01E-03
rearranged L-myc fusion
204243_at






sequence


AB026190
0.033
12.367
1.06E-03
Kelch motif containing
204177_s_at






protein


GTF2H5
0.033
8.729
1.09E-03
GTF2H5, general
213357_at






transcription factor IIH,






polypeptide 5


RBMS1
0.033
5.153
1.09E-03
RNA binding motif,
209868_s_at






single stranded






interacting protein 1


ENIGMA
0.033
0.081
1.09E-03
PDZ and LIM domain 7
203370_s_at






(enigma)


MIR
0.033
0.128
1.10E-03
c-mir, cellular modulator
221824_s_at






of immune recognition


SRRM2
0.033
5.461
1.11E-03
serine/arginine repetitive
208610_s_at






matrix 2


SRR
0.033
15.068
1.12E-03
serine racemase
219205_at


MCL1
0.033
0.058
1.12E-03
myeloid cell leukemia
200797_s_at






sequence 1 (BCL2-






related)


FACL5
0.034
89.075
1.17E-03
acyl-CoA synthetase
218322_s_at






long-chain family






member 5


CPSF1
0.034
0.209
1.16E-03
cleavage and
33132_at






polyadenylation specific






factor 1, 160kDa


AK2
0.034
17.668
1.19E-03
adenylate kinase 2
212175_s_at


PTTG11P
0.034
0.004
1.19E-03
pituitary tumor-
200677_at






transforming 1






interacting protein


GTPBP1
0.034
0.032
1.19E-03
GTP binding protein 1
219357_at


UNG
0.035
10.732
1.24E-03
uracil-DNA glycosylase
202330_s_at


RPS28
0.035
0.215
1.23E-03
ribosomal protein S28
216380_x_at


PAX5
0.035
8.402
1.24E-03
paired box gene 5 (B-cell
221969_at






lineage specific






activator)


PSMD8
0.035
11.013
1.29E-03
proteasome (prosome,
200820_at






macropain) 26S subunit,






non-ATPase, 8


NUDT1
0.035
10.67
1.29E-03
nudix (nucleoside
204766_s_at






diphosphate linked






moiety X)-type motif 1


SLC25A12
0.035
52.625
1.30E-03
solute carrier family 25
203339_at






(mitochondrial carrier,






Aralar), member 12


C1ORF24
0.036
12.539
1.31E-03
chromosome 1 open
217966_s_at






reading frame 24


HTATIP2
0.036
15.356
1.32E-03
HIV-1 Tat interactive
207180_s_at






protein 2, 30kDa


SRPK2
0.036
3.184
1.34E-03
SFRS protein kinase 2
203181_x_at


PRKAR1A
0.036
16.407
1.34E-03
protein kinase, cAMP-
200604_s_at






dependent, regulatory,






type I, alpha (tissue






specific extinguisher 1)


CD80
0.036
26.52
1.36E-03
CD80 antigen (CD28
207176_s_at






antigen ligand 1, B7-1






antigen)


MGC3248
0.036
20.329
1.37E-03
dynactin 4
209231_s_at


UBXD2
0.036
6.211
1.39E-03
UBX domain containing
212007_at






2


GALNT1
0.036
36.544
1.40E-03
UDP-N-acetyl-alpha-D-
201723_s_at






galactosamine:polypeptide






N-acetylgalactosaminyl-






transferase 1 (GalNAc-T1)


STX18
0.036
23.897
1.43E-03
syntaxin 18
218763_at


PDCD11
0.036
15.892
1.41E-03
programmed cell death
212424_at






11


ISGF3G
0.036
7.836
1.42E-03
interferon-stimulated
203882_at






transcription factor 3,






gamma 48kDa


RAB7
0.036
0.083
1.42E-03
RAB7, member RAS
211960_s_at






oncogene family


CDC42
0.036
0.051
1.42E-03
cell division cycle 42
210232_at






(GTP binding protein,






25kDa)


NFATC1
0.036
80.225
1.55E-03
nuclear factor of
210162_s_at






activated T-cells,






cytoplasmic, calcineurin-






dependent 1


PSMD1
0.036
19.918
1.51E-03
proteasome (prosome,
201198_s_at






macropain) 26S subunit,






non-ATPase, 1


COL4A3BP
0.036
17.703
1.55E-03
collagen, type IV, alpha
219625_s_at






3 (Goodpasture antigen)






binding protein


NR3C1
0.036
11.05
1.55E-03
nuclear receptor
201865_x_at






subfamily 3, group C,






member 1






(glucocorticoid receptor)


SEC63
0.036
7.604
1.54E-03
SEC63-like (S.
201914_s_at






cerevisiae)


PSMD11
0.036
5.523
1.46E-03
proteasome (prosome,
208777_s_at






macropain) 26S subunit,






non-ATPase, 11


H2AV
0.036
0.268
1.57E-03
H2A histone family,
212206_s_at






member V


CABIN1
0.036
0.162
1.55E-03
calcineurin binding
37652_at






protein 1


NET1
0.036
0.146
1.53E-03
neuroepithelial cell
201830_s_at






transforming gene 1


NFIL3
0.036
0.116
1.44E-03
nuclear factor,
203574_at






interleukin 3 regulated


MOAP1
0.036
0.115
1.47E-03
modulator of apoptosis 1
212508_at


SKP1A
0.036
0.113
1.47E-03
S-phase kinase-
200719_at






associated protein 1A






(p19A)


FLJ11127
0.036
0.069
1.53E-03
hypothetical protein
219694_at






FLJ11127


G1P3
0.036
0.069
1.58E-03
interferon, alpha-
204415_at






inducible protein (clone






IFI-6-16)


BNIP3L
0.036
0.044
1.55E-03
BCL2/adenovirus E1B
221478_at






19kDa interacting protein






3-like


C6ORF82
0.036
0.041
1.50E-03
chromosome 6 open
221488_s_at






reading frame 82


XTP2
0.037
4.327
1.62E-03
HBxAg transactivated
214055_x_at






protein 2


MBNL3
0.037
0.058
1.62E-03
muscleblind-like 3
219814_at






(Drosophila)


PDHB
0.037
33.983
1.63E-03
pyruvate dehydrogenase
208911_s_at






(lipoamide) beta


CKS1B
0.038
16.085
1.71E-03
CDC28 protein kinase
201897_s_at






regulatory subunit 1B


GALNS
0.038
0.227
1.71E-03
galactosamine (N-
206335_at






acetyl)-6-sulfate






sulfatase (Morquio






syndrome,






mucopolysaccharidosis






type IVA)


USP12
0.038
48.047
1.72E-03
USP12, ubiquitin
213327_s_at






specific protease 12


EIF5
0.038
8.566
1.73E-03
eukaryotic translation
208290_s_at






initiation factor 5


KIAA0650
0.038
0.146
1.73E-03
KIAA0650 protein
212577_at


UQCRFS1
0.038
0.060
1.74E-03
ubiquinol-cytochrome c
208909_at






reductase, Rieske iron-






sulfur polypeptide 1


ACO1
0.038
49.485
1.78E-03
aconitase 1, soluble
207071_s_at


MRPL13
0.038
9.48
1.77E-03
mitochondrial ribosomal
218049_s_at






protein L13


SCGF
0.038
0.120
1.75E-03
stem cell growth factor;
211709_s_at






lymphocyte secreted C-






type lectin


CHC1L
0.038
0.084
1.79E-03
chromosome
204759_at






condensation 1-like


TRIAD3
0.039
29.384
1.80E-03
TRIAD3 protein
218426_s_at


RFP
0.039
35.742
1.87E-03
ret finger protein
212116_at


PSMD13
0.039
16.384
1.84E-03
proteasome (prosome,
201233_at






macropain) 26S subunit,






non-ATPase, 13


ACOX1
0.039
15.909
1.86E-03
acyl-Coenzyme A
209600_s_at






oxidase 1, palmitoyl


ITGAV
0.039
12.837
1.82E-03
integrin, alpha V
202351_at






(vitronectin receptor,






alpha polypeptide,






antigen CD51)


SEC23B
0.039
11.687
1.83E-03
Sec23 homolog B (S.
201583_s_at






cerevisiae)


RPA3
0.039
10.718
1.84E-03
replication protein A3,
209507_at






14kDa


KLF7
0.039
7.918
1.83E-03
Kruppel-like factor 7
204334_at






(ubiquitous)


AGTPBP1
0.039
0.099
1.87E-03
ATP/GTP binding
204500_s_at






protein 1


CGI-127
0.039
0.039
1.86E-03
yippee protein
217783_s_at


KIAA0892
0.039
0.071
1.90E-03
KIAA0892
212505_s_at


APLP2
0.039
0.155
1.92E-03
amyloid beta (A4)
208248_x_at






precursor-like protein 2


IL7R
0.039
3.182
1.94E-03
interleukin 7 receptor
205798_at


SR140
0.039
0.144
1.95E-03
U2-associated SR140
212058_at






protein


HMGCL
0.040
11.109
1.99E-03
3-hydroxymethyl-3-
202772_at






methylglutaryl-






Coenzyme A lyase






(hydroxymethylglutarica






ciduria)


TDP1
0.040
10.611
1.98E-03
tyrosyl-DNA
219715_s_at






phosphodiesterase 1


VDAC3
0.040
7.789
1.97E-03
voltage-dependent anion
208846_s_at






channel 3


HIPK1
0.040
0.025
2.01E-03
homeodomain interacting
212291_at






protein kinase 1


FLJ14639
0.040
38.716
2.03E-03
nuclear factor of
212809_at






activated T-cells,






cytoplasmic, calcineurin-






dependent 2 interacting






protein


CGI-01
0.040
9.62
2.04E-03
CGI-01 protein
212405_s_at


FLJ11078
0.040
0.094
2.02E-03
hypothetical protein
219354—at






FLJ11078


CGI-128
0.041
54.238
2.10E-03
CGI-128 protein
218074_at


IL9
0.041
9.187
2.12E-03
interleukin 9
208193_at


NUP43
0.041
6.165
2.13E-03
nucleoporin 43kDa
219007_at


CCNL1
0.041
0.153
2.12E-03
cyclin L1
220046_s_at


GORASP2
0.041
0.100
2.13E-03
golgi reassembly
208843_s_at






stacking protein 2,






5kDa


AP162
0.041
0.069
2.15E-03
pleckstrin homology
212717_at






domain containing,






family M (with RUN






domain) member 1


PLSCR3
0.041
0.029
2.16E-03
phospholipid scramblase
56197_at






3


MGLL
0.042
14.896
2.21E-03
monoglyceride lipase
211026_s_at


NCOA3
0.042
13.528
2.23E-03
nuclear receptor
207700_s_at






coactivator 3


RNUT1
0.042
11.552
2.22E-03
RNA, U transporter 1
207438_s_at


ALEX3
0.042
5.508
2.21E-03
armadillo repeat
217858_s_at






containing, X-linked 3


TNFSF10
0.042
4.806
2.24E-03
tumor necrosis factor
202688_at






(ligand) superfamily,






member 10


PPP6C
0.042
0.045
2.24E-03
protein phosphatase 6,
203529_at






catalytic subunit


CENPC1
0.042
0.106
2.25E-03
centromere protein C 1
204739_at


NR1D1
0.042
0.197
2.25E-03
nuclear receptor
204760_s_at






subfamily 1, group D,






member 1


MTMR2
0.042
11.89
2.28E-03
myotubularin related
203211_s_at






protein 2


FDPS
0.042
11.053
2.27E-03
farnesyl diphosphate
201275_at






synthase (farnesyl






pyrophosphate






synthetase,






dimethylallyltrans-






transferase,






geranyltranstransferase)


FLJ12439
0.042
6.764
2.27E-03
hypothetical protein
219420_s_at






FLJ12439


TFEB
0.042
0.138
2.27E-03
transcription factor EB
50221_at


Unknown
0.042
0.315
2.31E-03
no sequence similanty to
222315_at






other genes or proteins


KIAA1332
0.042
0.061
2.31E-03
F-box protein 42
221813_at


C14ORF159
0.042
0.132
2.32E-03
chromosome 14 open
218298_s_at






reading frame 159


PSME2
0.042
10.064
2.34E-03
proteasome (prosome,
201762_s_at






macropain) activator






subunit 2 (PA28 beta)


MPHOSPH6
0.043
10.656
2.38E-03
M-phase phosphoprotein
203740_at






6


YWHAB
0.043
10.596
2.40E-03
tyrosine 3-
217717_s_at






monooxygenase/tryptophan






5-monooxygenase






activation protein, beta






polypeptide


MCM7
0.043
7.75
2.40E-03
MCM7 minichromosome
208795_s_at






maintenance deficient 7






(S. cerevisiae)


PSMD2
0.043
334.893
2.43E-03
proteasome (prosome,
200830_at






macropain) 26S subunit,






non-ATPase, 2


AMPD2
0.043
0.122
2.45E-03
adenosine
212360_at






monophosphate






deaminase 2 (isoform L)


CCNE1
0.044
6.88
2.48E-03
cyclin E1
213523_at


MMP7
0.044
6.512
2.48E-03
matrix metalloproteinase
204259_at






7 (matrilysin, uterine)


GTF2H1
0.044
12.954
2.51E-03
general transcription
202453_s_at






factor IIH, polypeptide 1,






62kDa


FNBP1
0.044
5.151
2.52E-03
formin binding protein 1
213940_s_at


UBD
0.044
7.847
2.54E-03
ubiquitin D
205890_s_at


FLJ38984
0.045
19.598
2.57E-03
hypothetical protein
212791_at






FLJ38984


TLE4
0.045
0.108
2.58E-03
transducin-like enhancer
204872_at






of split 4 (E(sp1)






homolog, Drosophila)


ITM2B
0.045
0.032
2.60E-03
integral membrane
217732_s_at






protein 2B


HSD17B7
0.045
14.99
2.62E-03
hydroxysteroid (17-beta)
220081_x_at






dehydrogenase 7


KIAA1115
0.047
33.455
2.74E-03
KIAA1115
209229_s_at


COAS1
0.047
3.854
2.74E-03
chomosome one
214693_x_at






amplified sequence 1






cyclophilin


XRCC5
0.047
17.167
2.77E-03
X-ray repair
208643_s_at






complementing defective






repair in Chinese hamster






cells 5 (double-strand-






break rejoining; Ku






autoantigen, 80kDa)


STMN1
0.047
11.125
2.76E-03
stathmin 1/oncoprotein
200783_s_at






18


CTLA4
0.047
8.016
2.77E-03
cytotoxic T-lymphocyte-
221331_x_at






associated protein 4


STAG2
0.047
6.595
2.78E-03
stromal antigen 2
207983_s_at


KIAA0404
0.047
0.144
2.78E-03
KIAA0404 protein
213300_at


SF3B4
0.047
0.180
2.80E-03
splicing factor 3b,
209044_x_at






subunit 4, 49kDa


CXCL9
0.047
6.108
2.81E-03
chemokine (C-X-C
203915_at






motif) ligand 9


ITGAX
0.047
0.032
2.85E-03
integrin, alpha X (antigen
210184_at






CD11C (p150), alpha






polypeptide)


FLJ14888
0.048
25.043
2.86E-03
hypothetical protein
213031_s_at






FLJ14888


FLJ10803
0.048
31.56
2.90E-03
hypothetical protein
209445_x_at






FLJ10803


OSBPL9
0.048
288.036
2.91E-03
oxysterol binding
218047_at






protein-like 9


PTEN
0.048
0.107
2.91E-03
phosphatase and tensin
204054_at






homolog (mutated in






multiple advanced






cancers 1)


EFHD2
0.048
0.128
2.93E-03
EF hand domain
217992_s_at






containing 2


PPIH
0.048
29.937
2.99E-03
peptidyl prolyl isomerase
204228_at






H (cyclophilin H)


NKTR
0.048
4.902
3.00E-03
natural killer-tumor
202379_s_at






recognition sequence


BAZ2A
0.048
4.766
2.99E-03
bromodomain adjacent to
201353_s_at






zinc finger domain, 2A


DOCK2
0.048
0.100
2.96E-03
dedicator of cytokinesis
213160_at






2


FGR
0.048
0.088
3.02E-03
Gardner-Rasheed feline
208438_s_at






sarcoma viral (v-fgr)






oncogene homolog


ZCCHC2
0.048
0.080
2.98E-03
zinc finger, CCHC
219062_s_at






domain containing 2


QKI
0.049
29.983
3.12E-03
quaking homolog, KH
212263_at






domain RNA binding






(mouse)


SUCLA2
0.049
10.996
3.14E-03
succinate-CoA ligase,
202930_s_at






ADP-forming, beta






subunit


MATR3
0.049
0.124
3.11E-03
matrin3
200626_s_at


GABR1
0.049
0.117
3.09E-03
gamma-aminobutyric
203146_s_at






acid (GABA) B receptor,






1


SPN
0.049
0.102
3.14E-03
sialophorin (gpL115,
206057_x_at






leukosialin, CD43)


KIAA1536
0.049
0.073
3.10E-03
KIAA1536 protein
209002_s_at


PABPC3
0.049
0.038
3.06E-03
poly(A) binding protein,
215157_x_at






cytoplasmic 3


C3ORF4
0.049
5.543
3.17E-03
chromosome 3 open
208925_at






reading frame 4


CYLD
0.049
0.161
3.18E-03
cylindromatosis (turban
60084_at






tumor syndrome)


FLJ21347
0.049
0.098
3.18E-03
hypothetical protein
218164_at






FLJ21347


FBS1
0.049
0.031
3.17E-03
fibrosin 1
218255_s_at


AIM2
0.049
9.506
3.20E-03
absent in melanoma 2
206513_at


PTX1
0.049
9.051
3.20E-03
PTX1 protein
218135_at


CLN5
0.049
11.634
3.27E-03
ceroid-lipofuscinosis,
204084_s_at






neuronal 5


EPRS
0.049
9.17
3.28E-03
glutamyl-prolyl-tRNA
200842_s_at






synthetase


LRDD
0.049
0.219
3.27E-03
leucine-rich repeats and
219019_at






death domain containing


LOC283537
0.049
0.094
3.28E-03
hypothetical protein
214719_at






LOC283537


PEX3
0.050
9.809
3.31E-03
peroxisomal biogenesis
203972_s_at






factor 3


NCOA2
0.050
0.220
3.34E-03
nuclear receptor
212867_at






coactivator 2


ARHQ
0.050
41.236
3.38E-03
ras homolog gene family,
212119_at






member Q


PFKM
0.050
18.355
3.36E-03
phosphofructokinase,
210976_s_at






muscle


BHC80
0.050
0.124
3.37E-03
BRAF35/HDAC2
203278_s_at






complex (80 kDa)


CD2BP2
0.050
59.36
3.45E-03
CD2 antigen
202256_at






(cytoplasmic tail)






binding protein 2


WARS
0.050
23.882
3.48E-03
tryptophanyl-tRNA
200628_s_at






synthetase


FXC1
0.050
13.071
3.50E-03
fracture callus 1 homolog
217981_s_at






(rat)


TSTA3
0.050
6.918
3.49E-03
tissue specific
201644_at






transplantation antigen






P35B


ESPL1
0.050
6.537
3.54E-03
extra spindle poles like 1
204817_at






(S. cerevisiae)


PWP1
0.050
4.459
3.54E-03
nuclear phosphoprotein
201606_s_at






similar to S. cerevisiae






PWP1


KRAS2
0.050
3.71
3.54E-03
v-Ki-ras2 Kirsten rat
214352_s_at






sarcoma 2 viral oncogene






homolog


ZNF408
0.050
0.239
3.53E-03
zinc finger protein 408
219224_x_at


TCF7L2
0.050
0.223
3.51E-03
transcription factor 7-like
216035_x_at






2 (T-cell specific, HMG-






box)


RGS2
0.050
0.176
3.51E-03
regulator of G-protein
202388_at






signalling 2, 24kDa


PLEKHF2
0.050
0.154
3.54E-03
pleckstrin homology
218640_s_at






domain containing,






family F (with FYVE






domain) member 2


EDG6
0.050
0.144
3.51E-03
endothelial
206437_at






differentiation, G-






protein-coupled receptor 6


KIAA1076
0.050
0.117
3.55E-03
KIAA1076 protein
213153_at


DRE1
0.050
0.113
3.41E-03
DRE1 protein
221985_at


C14ORF32
0.050
0.097
3.52E-03
chromosome 14 open
212643_at






reading frame 32


MAP3K71P2
0.050
0.079
3.39E-03
mitogen-activated
212184_s_at






protein kinase kinase






kinase 7 interacting






protein 2


ARL4
0.050
0.063
3.44E-03
ADP-ribosylation factor-
205020_s_at






like 4A


RPA2
0.050
17.449
3.57E-03
replication protein A2,
201756_at






32kDa


NUP50
0.051
13.853
3.61E-03
nucleoporin 50kDa
218294_s_at


KIAA0555
0.051
7.853
3.61E-03
KIAA0555 gene product
205888_s_at


GAS7
0.051
0.091
3.60E-03
growth arrest-specific 7
202192_s_at


SSFA2
0.051
0.036
3.62E-03
sperm specific antigen 2
202506_at


GMEB2
0.051
0.097
3.68E-03
glucocorticoid
44146_at






modulatory element






binding protein 2


PIR51
0.051
9.238
3.70E-03
RAD51-interacting
204146_at






protein


C9ORF83
0.051
8.68
3.71E-03
chromosome 9 open
218085_at






reading frame 83


PRO1843
0.051
0.126
3.73E-03
hypothetical protein
219599_at






PRO1843


VEGF
0.052
0.124
3.78E-03
vascular endothelial
212171_x_at






growth factor


DNM1L
0.052
16.425
3.80E-03
dynamin 1-like
203105_s_at


RERE
0.052
0.093
3.82E-03
arginine-glutamic acid
200940_s_at






dipeptide (RE) repeats


ARID1A
0.052
11.487
3.83E-03
AT rich interactive
212152_x_at






domain 1A (SWI- like)


FLJ10815
0.052
9.617
3.83E-03
hypothetical protein
56821_at






FLJ10815


PSMA4
0.052
51.574
3.87E-03
proteasome (prosome,
203396_at






macropain) subunit,






alpha type, 4


GNL1
0.052
19.339
3.87E-03
guanine nucleotide
203307_at






binding protein-like 1


CIAO1
0.052
17.811
3.87E-03
WD40 protein Ciao1
203536_s_at


MNT
0.052
0.113
3.86E-03
MAX binding protein
204206_at


CXCL5
0.052
4.558
3.88E-03
chemokine (C-X-C
214974_x_at






motif) ligand 5


FLJ32731
0.052
0.180
3.90E-03
hypothetical protein
218017_s_at






FLJ32731


MYCBP
0.053
23.309
3.99E-03
c-myc binding protein
203359_s_at


KIAA0102
0.053
9.997
3.93E-03
KIAA0102 gene product
201240_s_at


PROSC
0.053
5.622
3.97E-03
proline synthetase co-
209385_s_at






transcribed homolog






(bacterial)


LYL1
0.053
0.235
3.97E-03
lymphoblastic leukemia
210044_s_at






derived sequence 1


DUSP10
0.053
0.099
3.97E-03
dual specificity
221563_at






phosphatase 10


MKRN1
0.053
0.095
3.98E-03
makorin, ring finger
209845_at






protein, 1


Unknown
0.053
0.081
3.99E-03
gene of unknown
65588_at






function


Unknown
0.053
0.271
4.01E-03
Unknown
211996_s_at


CKS2
0.053
5.629
4.02E-03
CDC28 protein kinase
204170_s_at






regulatory subunit 2


KMO
0.053
5.843
4.05E-03
kynurenine 3-
211138_s_at






monooxygenase






(kyrnurenine 3-






hydroxylase)


SGK
0.053
0.161
4.04E-03
serum/glucocorticoid
201739_at






regulated kinase


C20ORF104
0.053
0.091
4.05E-03
chromosome 20 open
209422_at






reading frame 104


ARS2
0.053
0.028
4.04E-03
arsenate resistance
201680_x_at






protein ARS2


ZNF259
0.053
26.34
4.12E-03
zinc finger protein 259
200054_at


SERP1
0.054
0.022
4.16E-03
stress-associated
200971_s_at






endoplasmic reticulum






protein 1


GC20
0.054
0.016
4.16E-03
translation factor sui1
201738_at






homolog


TRAPPC3
0.054
11.773
4.18E-03
trafficking protein
203511_s_at






particle complex 3


MSF
0.054
0.222
4.19E-03
MLL septin-like fusion
208657_s_at


CDC40
0.054
0.074
4.18E-03
cell division cycle 40
203377_s_at






homolog (yeast)


PPP3CA
0.054
5.417
4.21E-03
protein phosphatase 3
202425_x_at






(formerly 2B), catalytic






subunit, alpha isoform






(calcineurin A alpha)


FLJ14753
0.054
0.021
4.22E-03
hypothetical protein
211185_s_at






FLJ14753


PELI1
0.054
0.175
4.24E-03
pellino homolog 1
218319_at






(Drosophila)


PRKCSH
0.054
0.064
4.24E-03
protein kinase C
214080_x_at






substrate 80K-H


SPINT2
0.054
0.115
4.29E-03
serine protease inhibitor,
210715_s_at






Kunitz type, 2


PSARL
0.055
50.956
4.33E-03
presenilin associated,
218271_s_at






rhomboid-like


HT007
0.056
10.945
4.40E-03
uncharacterized
221622_s_at






hypothalamus protein






HT007


RAD51C
0.056
5.167
4.44E-03
RAD51 homolog C (S.
209849_s_at






cerevisiae)


TRIP-BR2
0.056
3.547
4.46E-03
SERTA domain
202656_s_at






containing 2


TRA1
0.056
12.843
4.49E-03
tumor rejection antigen
200598_s_at






(gp96) 1


DKFZP586D0919
0.056
25.287
4.52E-03
hepatocellularcarcinoma-
213861_s_at






associated antigen






HCA557a


CIC
0.056
0.300
4.52E-03
capicua homolog
212784_at






(Drosophila)


PIK3CA
0.056
0.075
4.51E-03
phosphoinositide-3-
204369_at






kinase, catalytic, alpha






polypeptide


HSPC051
0.057
12.156
4.53E-03
ubiquinol-cytochrome c
218190_s_at






reductase complex (7.2






kD)


ELAVL1
0.057
4.903
4.57E-03
ELAV (embryonic lethal,
201726_at






abnormal vision,






Drosophila)-like 1 (Hu






antigen R)


NADSYN1
0.057
0.182
4.56E-03
NAD synthetase 1
218840_s_at


CCL22
0.057
3.61
4.58E-03
chemokine (C-C motif)
207861_at






ligand 22


CCNB2
0.057
12.529
4.62E-03
cyclin B2
202705_at


C20ORF67
0.057
0.176
4.61E-03
chromosome 20 open
222044_at






reading frame 67


LOC51064
0.057
0.128
4.64E-03
glutathione S-transferase
217751_at






kappa 1


POLR2K
0.057
7.759
4.70E-03
polymerase (RNA) II
202635_s_at






(DNA directed)






polypeptide K, 7.0kDa


LRP8
0.057
7.185
4.71E-03
low density lipoprotein
205282_at






receptor-related protein






8, apolipoprotein e






receptor


FLJ20080
0.057
9.512
4.73E-03
aftiphilin protein
217939_s_at


ACADM
0.058
6.254
4.82E-03
acyl-Coenzyme A
202502_at






dehydrogenase, C-4 to C-






12 straight chain


JUND
0.058
0.059
4.82E-03
jun D proto-oncogene
203752_s_at


FLJ20534
0.058
12.83
4.86E-03
hypothetical protein
218646_at






FLJ20534


TOB1
0.058
0.172
4.87E-03
transducer of ERBB2, 1
202704_at


ACTG1
0.058
0.006
4.88E-03
actin, gamma 1
212988_x_at


FLJ10534
0.059
7.735
4.92E-03
hypothetical protein
221987_s_at






FLJ10534


CTPS
0.059
5.567
4.93E-03
CTP synthase
202613_at


TCP1
0.059
21.895
5.00E-03
t-complex 1
208778_s_at


D1S155E
0.059
0.073
5.00E-03
NRAS-related gene
219939_s_at


TIMELESS
0.059
6.145
5.02E-03
timeless homolog
203046_s_at






(Drosophila)


NCOR1
0.059
59.364
5.05E-03
nuclear receptor co-
200854_at






repressor 1


DDEF1
0.059
7.693
5.07E-03
development and
221039_s_at






differentiation enhancing






factor 1


UBE2L3
0.059
7.274
5.14E-03
ubiquitin-conjugating
200684_s_at






enzyme E2L 3


C9ORF40
0.059
7.261
5.11E-03
chromosome 9 open
218904_s_at






reading frame 40


PHF3
0.059
0.081
5.14E-03
PHD finger protein 3
217952_x_at


DKFZP564D0478
0.059
0.049
5.14E-03
hypothetical protein
52078_at






DKFZp564D0478


CSK
0.059
0.040
5.11E-03
c-src tyrosine kinase
202329_at


FBXL12
0.059
0.008
5.11E-03
F-box and leucine-rich
220127_s_at






repeat protein 12


CSNK2A1
0.059
9.961
5.17E-03
casein kinase 2, alpha 1
212075_s_at






polypeptide


KIAA0483
0.059
22.767
5.18E-03
F-box protein 28
202272_s_at


NEDD8
0.060
5.847
5.21E-03
neural precursor cell
201840_at






expressed,






developmentally down-






regulated 8


TNFRSF9
0.060
3.452
5.20E-03
tumor necrosis factor
207536_s_at






receptor superfamily,






member 9


KIAA0738
0.060
7.398
5.25E-03
KIAA0738 gene product
204403_x_at


ZNF161
0.060
0.086
5.24E-03
zinc finger protein 161
202171_at


SIAT9
0.060
0.018
5.29E-03
sialyltransferase 9 (CMP-
203217_s_at






NeuAc: lactosylceramide






alpha-2,3-






sialyltransferase; GM3






synthase)


MADH7
0.060
0.113
5.35E-03
SMAD, mothers against
204790_at






DPP homolog 7






(Drosophila)


USP3
0.060
0.051
5.36E-03
ubiquitin specific
221654_s_at






protease 3


KHDRBS1
0.060
7.898
5.39E-03
KH domain containing,
201488_x_at






RNA binding, signal






transduction associated 1


C5ORF6
0.061
0.047
5.42E-03
chromosome 5 open
218023_s_at






reading frame 6


GLG1
0.061
6.776
5.48E-03
golgi apparatus protein 1
207966_s_at


TCF8
0.061
0.300
5.47E-03
transcription factor 8
208078_s_at






(represses interleukin 2






expression)


RBAF600
0.061
0.011
5.50E-03
retinoblastoma-
211950_at






associated factor 600


SLC35D2
0.061
21.354
5.54E-03
solute carrier family 35,
213082_s_at






member D2


PIGA
0.061
0.156
5.55E-03
phosphatidylinositol
205281_s_at






glycan, class A






(paroxysmal nocturnal






hemoglobinuria)


DUSP3
0.061
9.995
5.57E-03
dual specificity
201536_at






phosphatase 3 (vaccinia






virus phosphatase VH1-






related)


DSCR1
0.061
26.47
5.59E-03
Down syndrome critical
208370_s_at






region gene 1


CGI-51
0.062
26.286
5.74E-03
CGI-51 protein
201570_at


TIP120A
0.062
14.465
5.68E-03
TBP-interacting protein
208839_s_at


MAC30
0.062
9.549
5.71E-03
hypothetical protein
212282_at






MAC30


PTMA
0.062
9.39
5.73E-03
prothymosin, alpha (gene
216384_x_at






sequence 28)


WDR12
0.062
6.497
5.69E-03
WD repeat domain 12
218512_at


POLE2
0.062
5.826
5.65E-03
polymerase (DNA
205909_at






directed), epsilon 2 (p59






subunit)


NRG1
0.062
0.308
5.70E-03
neuregulin 1
206343_s_at


SLC22A18
0.062
0.139
5.66E-03
solute carrier family 22
204981_at






(organic cation






transporter), member 18


VAMP2
0.062
0.092
5.71E-03
vesicle-associated
214792_x_at






membrane protein 2






(synaptobrevin 2)


Unknown
0.062
0.069
5.71E-03
no sequence similarity to
213215_at






any genes or proteins


TRAF6
0.062
0.049
5.76E-03
TNF receptor-associated
205558_at






factor 6


EV12B
0.062
0.109
5.81E-03
ecotropic viral
211742_s_at






integration site 2B


TIEG2
0.062
0.231
5.84E-03
TGFB inducible early
218486_at






growth response 2


COPS5
0.062
8.168
5.89E-03
COP9 constitutive
201652_at






photomorphogenic






homolog subunit 5






(Arabidopsis)


RNF139
0.062
0.143
5.92E-03
ring finger protein 139
209510_at


PCMT1
0.063
8.822
6.03E-03
protein-L-isoaspartate
205202_at






(D-aspartate) O-






methyltransferase


MPO
0.063
0.146
6.03E-03
myeloperoxidase
203949_at


KCNK4
0.063
6.788
6.07E-03
potassium channel,
219883_at






subfamily K, member 4


SSA2
0.063
5.063
6.09E-03
Sjogren syndrome
210438_x_at






antigen A2 (60kDa,






ribonucleoprotein






autoantigen SS-A/Ro)


Unknown
0.064
4.635
6.15E-03
no sequence similarity to
217586_x_at






any genes or proteins


DSIPI
0.064
0.237
6.17E-03
delta sleep inducing
208763_s_at






peptide, immunoreactor


KIAA0683
0.064
13.161
6.26E-03
KIAA0683 gene product
34260_at


POLD3
0.064
8.246
6.27E-03
polymerase (DNA-
212836_at






directed), delta 3,






accessory subunit


EEF2
0.064
0.180
6.27E-03
eukaryotic translation
204102_s_at






elongation factor 2


KIAA1002
0.064
0.133
6.25E-03
KIAA1002 protein
203831_at


NEIL1
0.064
0.090
6.26E-03
nei endonuclease VIII-
219396_s_at






like 1 (E. coli)


FLJ10099
0.065
25.454
6.34E-03
hypothetical protein
218008_at






FLJ10099


KIAA0582
0.065
6.047
6.34E-03
KIAA0582
212677_s_at


HADHSC
0.065
0.155
6.39E-03
L-3-hydroxyacyl-
201034_at






Coenzyme A






dehydrogenase, short






chain


C2ORF6
0.065
8.647
6.40E-03
MOB1, Mps One Binder
201298_s_at






kinase activator-like 1B






(yeast)


VIPR1
0.065
0.154
6.41E-03
vasoactive intestinal
205019_s_at






peptide receptor 1


SLC4A1AP
0.065
36.646
6.42E-03
solute carrier family 4
218682_s_at






(anion exchanger),






member 1, adaptor






protein


ARL7
0.065
0.187
6.44E-03
ADP-ribosylation factor-
202207_at






like 7


DXYS155E
0.065
0.032
6.44E-03
DNA segment on
203624_at






chromosome X and Y






(unique) 155 expressed






sequence


BIRC2
0.065
0.118
6.45E-03
baculoviral IAP repeat-
202076_at






containing 2


STAT5A
0.065
56.762
6.55E-03
signal transducer and
203010_at






activator of transcription






5A


PHB
0.065
8.89
6.56E-03
prohibitin
200659_s_at


MYLK
0.065
3.958
6.55E-03
myosin, light polypeptide
202555_s_at






kinase


ATP5L
0.065
0.096
6.57E-03
ATP synthase, H+
210453_x_at






transporting,






mitochondrial F0






complex, subunit g


KEAP1
0.066
7.403
6.58E-03
kelch-like ECH-
202417_at






associated protein 1


CAMKK2
0.066
3.454
6.63E-03
calcium/calmodulin-
213812_s_at






dependent protein kinase






kinase 2, beta


PRKCN
0.066
0.189
6.62E-03
protein kinase C, nu
218236_s_at


MARK4
0.066
0.196
6.65E-03
MAP/microtubule
55065_at






affinity-regulating kinase 4


CDK4
0.066
5.678
6.69E-03
cyclin-dependent kinase 4
202246_s_at


PAICS
0.066
5.533
6.71E-03
phosphoribosylaminoimi
201013_s_at






dazole carboxylase,






phosphoribosylaminoimi






dazole






succinocarboxamide






synthetase


CGI-90
0.066
9.14
6.77E-03
CGI-90 protein
218549_s_at


TNIP3
0.066
5.962
6.76E-03
TNFAIP3 interacting
220655_at






protein 3


NFKB1
0.066
5.354
6.75E-03
nuclear factor of kappa
209239_at






light polypeptide gene






enhancer in B-cells 1






(p105)


C1ORF9
0.066
0.058
6.77E-03
chromosome 1 open
203429_s_at






reading frame 9


POP5
0.067
8.456
6.83E-03
processing of precursor
204839_at






5, ribonuclease P/MRP






subunit (S. cerevisiae)


ILI2B
0.067
4.425
6.83E-03
interleukin 12B (natural
207901_at






killer cell stimulatory






factor 2, cytotoxic






lymphocyte maturation






factor 2, p40)


RUNX1
0.067
0.037
6.82E-03
runt-related transcription
208129_x_at






factor 1 (acute myeloid






leukemia 1; aml1






oncogene)


C20ORF121
0.067
0.125
6.84E-03
chromosome 20 open
221472_at






reading frame 121


EIF2B2
0.067
28.683
6.91E-03
eukaryotic translation
202461_at






initiation factor 2B,






subunit 2 beta, 39kDa


MGC4825
0.067
14.636
6.92E-03
hypothetical protein
221620_s_at






MGC4825


ILF3
0.067
11.625
6.86E-03
interleukin enhancer
217805_at






binding factor 3, 90kDa


MRPL19
0.067
7.652
6.91E-03
mitochondrial ribosomal
203465_at






protein L19


KIAA0982
0.067
0.170
6.86E-03
WD repeat domain 37
211383_s_at


CCR1
0.067
4.928
6.97E-03
chemokine (C-C motif)
205099_s_at






receptor 1


TNF
0.067
3.471
6.96E-03
tumor necrosis factor
207113_s_at






(TNF superfamily,






member 2)


LYRIC
0.067
0.074
6.95E-03
LYRIC/3D3
212248_at


FLJ21603
0.067
0.052
6.98E-03
zinc finger protein 552
219741_x_at


PRKAG1
0.067
8.572
7.02E-03
protein kinase, AMP-
201805_at






activated, gamma 1 non-






catalytic subunit


NISCH
0.067
0.054
7.00E-03
nischarin
201591_s_at


COX7C
0.067
0.116
7.13E-03
cytochrome c oxidase
201134_x_at






subunit VIIc


BRD3
0.067
0.076
7.13E-03
BRD3, bromodomain
212547_at






containing 3


SRF72
0.068
3.342
7.17E-03
signal recognition
208095_s_at






particle 72kDa


CA12
0.068
4.9
7.23E-03
carbonic anhydrase XII
203963_at


KLF3
0.068
0.147
7.26E-03
Kruppel-like factor 3
219657_s_at






(basic)


FLJ20546
0.068
9.408
7.35E-03
interphase cyctoplasmic
219122_s_at






foci protein 45


BLM
0.068
13.128
7.35E-03
Bloom syndrome
205733_at


PLAC8
0.069
0.161
7.39E-03
placenta-specific 8
219014_at


KIAA1012
0.069
32.462
7.42E-03
KIAA1012
207305_s_at


SRPK1
0.069
7.399
7.45E-03
SFRS protein kinase 1
202200_s_at


CLECSF12
0.069
0.260
7.47E-03
C-type (calcium
221698_s_at






dependent, carbohydrate-






recognition domain)






lectin, superfamily






member 12


COPS8
0.069
60.228
7.51E-03
COP9 constitutive
202142_at






photomorphogenic






homolog subunit 8






(Arabidopsis)


MGC11256
0.069
11.319
7.51E-03
hypothetical protein
218358_at






MGC11256


MRPS11
0.069
10.289
7.51E-03
mitochondrial ribosomal
211595_s_at






protein S11


SLC2A14
0.069
0.219
7.52E-03
solute carrier family 2
216236_s_at






(facilitated glucose






transporter), member 14


CUTL1
0.069
0.106
7.53E-03
cut-like 1, CCAAT
202367_at






displacement protein






(Drosophila)


PAFAH1B1
0.069
18.233
7.55E-03
platelet-activating factor
200816_s_at






acetylhydrolase, isoform






Ib, alpha subunit 45kDa


AKAP13
0.069
8.326
7.57E-03
A kinase (PRKA) anchor
209534_x_at






protein 13


HIST1H4C
0.071
4.545
7.77E-03
histone 1, H4c
205967_at


PSME4
0.071
0.102
7.90E-03
proteasome (prosome,
212219_at






macropain) activator






subunit 4


KIAA0152
0.072
23.99
7.98E-03
KIAA0152 gene product
200617_at


CINP
0.072
18.638
7.94E-03
cyclin-dependent kinase
218267_at






2-interacting protein


EIF5B
0.072
5.498
7.95E-03
eukaryotic translation
201027_s_at






initiation factor 5B


G0S2
0.072
0.266
7.97E-03
putative lymphocyte
213524_s_at






G0/G1 switch gene


SENP3
0.072
15.871
8.02E-03
SUMO1/sentrin/SMT3
203871_at






specific protease 3


HNRPR
0.072
10.584
8.12E-03
heterogeneous nuclear
208765_s_at






ribonucleoprotein R


FN5
0.072
10.552
8.11E-03
FN5 protein
219806_s_at


ATP6V1C1
0.072
6.714
8.11E-03
ATPase, H+ transporting,
202874_s_at






lysosomal 42kDa, V1






subunit C, isoform 1


COL18A1
0.072
0.057
8.11E-03
collagen, type XVIII,
209081_s_at






alpha 1


EEF1E1
0.072
9.813
8.14E-03
eukaryotic translation
204905_s_at






elongation factor 1






epsilon 1


TANK
0.072
6.588
8.15E-03
TRAF family member-
207616_s_at






associated NFKB






activator


IFIT5
0.073
0.128
8.23E-03
interferon-induced
203595_s_at






protein with






tetratricopeptide repeats 5


TUBA3
0.073
0.124
8.25E-03
tubulin, alpha 3
209118_s_at


UBE2J1
0.074
10.265
8.35E-03
ubiquitin-conjugating
217823_s_at






enzyme E2, J1 (UBC6






homolog, yeast)


PER1
0.074
0.118
8.35E-03
period homolog 1
36829_at






(Drosophila)


DGCR14
0.074
0.150
8.36E-03
DiGeorge syndrome
32032_at






critical region gene 14


CGI-49
0.074
15.54
8.39E-03
CGI-49 protein
201825_s_at


CEACAM8
0.074
0.244
8.43E-03
carcmoembryonic
206676_at






antigen-related cell






adhesion molecule 8


GNAI2
0.074
0.178
8.43E-03
guanine nucleotide
201040_at






binding protein (G






protein), alpha inhibiting






activity polypeptide 2


ACAT1
0.074
13.587
8.50E-03
acetyl-Coenzyme A
205412_at






acetyltransferase 1






(acetoacetyl Coenzyme






A thiolase)


GOT1
0.074
6.694
8.50E-03
glutamic-oxaloacetic
208813_at






transaminase 1, soluble






(aspartate






aminotransferase 1)


SMG1
0.074
0.096
8.48E-03
PI-3-kinase-related
208118_x_at






kinase SMG-1


13CDNA73
0.074
0.086
8.50E-03
hypothetical protein
204072_s_at






CG003


TUBB
0.074
0.250
8.55E-03
tubulin, beta polypeptide
204141_at


CHD4
0.075
12.6
8.60E-03
chromodomain helicase
201183_s_at






DNA binding protein 4


RGS10
0.075
4.866
8.70E-03
regulator of G-protein
214000_s_at






signalling 10


CAMP
0.075
0.253
8.74E-03
cathelicidin antimicrobial
210244_at






peptide


APOM
0.075
0.107
8.78E-03
apolipoprotein M
214910_s_at


FLJ21868
0.076
0.096
8.80E-03
transducer of regulated
218648_at






cAMP response element-






binding protein (CREB) 3


MCM10
0.076
4.325
8.83E-03
MCM10
220651_s_at






minichromosome






maintenance deficient 10






(S. cerevisiae)


C11ORF2
0.076
0.215
8.86E-03
chromosome 11 open
217969_at






reading frame2


Unknown
0.076
0.089
8.91E-03
gene of unknown
217625_x_at






function


MPZL1
0.076
26.093
8.96E-03
myelin protein zero-like 1
201874_at


MRPL48
0.077
15.923
9.04E-03
mitochondrial ribosomal
218281_at






protein L48


SET
0.077
10.103
9.11E-03
SET translocation
210231_x_at






(myeloid leukemia-






associated)


C16ORF35
0.077
0.221
9.11E-03
chromosome 16 open
214273_x_at






reading frame 35


RARA
0.077
0.123
9.07E-03
retinoic acid receptor,
203749_s_at






alpha


T1
0.077
0.084
9.11E-03
Tularik gene 1
56829_at


NCBP1
0.077
6.422
9.14E-03
nuclear cap binding
209520_s_at






protein subunit 1, 80kDa


INHBA
0.077
3.484
9.19E-03
inhibin, beta A (activin
210511_s_at






A, activin AB alpha






polypeptide)


NINJ2
0.077
0.217
9.19E-03
ninjurin 2
219594_at


NCOA1
0.077
0.124
9.15E-03
nuclear receptor
290105_at






coactivator 1


JMJD1
0.077
0.113
9.18E-03
jumonji domain
212689_s_at






containing 1A


UVRAG
0.077
0.043
9.17E-03
UV radiation resistance
203241_at






associated gene


CXCL13
0.077
4.537
9.22E-03
chemokine (C-X-C
205242_at






motif) ligand 13 (B-cell






chemoattractant)


CYB5
0.078
7.271
9.28E-03
cytochrome b-5
209366_x_at


KLRD1
0.079
13.813
9.47E-03
killer cell lectin-like
207796_x_at






receptor subfamily D,






member 1


APG12L
0.079
10.273
9.48E-03
APG12 autophagy 12-
213026_at






like (S. cerevisiae)


PLEKHB2
0.079
0.035
9.48E-03
pleckstrin homology
201410_at






domain containing,






family B (evectins)






member 2


TNPO1
0.079
16.302
9.50E-03
transportin 1
207657_x_at


PDPK1
0.079
6.544
9.55E-03
3-phosphoinositide
32029_at






dependent protein






kinase-1


SLCO3A1
0.079
0.139
9.54E-03
solute carrier organic
210542_s_at






anion transporter family,






member 3A1


YT521
0.079
0.097
9.52E-03
splicing factor YT521-B
212455_at


FOSL2
0.079
0.091
9.52E-03
FOS-like antigen 2
218881_s_at


NDUFB8
0.079
0.060
9.58E-03
NADH dehydrogenase
214241_at






(ubiquinone) 1 beta






subcomplex, 8, 19kDa


TRIM44
0.079
6.37
9.59E-03
tripartite motif-
217759_at






containing 44


UPS4
0.079
23.919
9.68E-03
ubiquitin specific
202682_s_at






protease 4 (proto-






oncogene)


SEC61A1
0.079
14.769
9.69E-03
Sec61 alpha 1 subunit (S.
217716_s_at






cerevisiae)


SF3B1
0.079
0.048
9.69E-03
splicing factor 3b,
201071_x_at






subunit 1, 155kDa


HA-1
0.080
0.193
9.73E-03
minor histocompatibility
212873_at






antigen HA-1


SMARCD3
0.080
0.202
9.78E-03
SWI/SNF related, matrix
204099_at






associated, actin






dependent regulator of






chromatin, subfamily d,






member 3


AP3D1
0.080
79.171
9.81E-03
adaptor-related protein
206592_s_at






complex 3, delta 1






subunit


EEG1
0.080
6.217
9.81E-03
solute carrier family 43,
213113_s_at






member 3


SIGIRR
0.080
0.267
9.80E-03
single Ig IL-1R-related
218921_at






molecule


Unknown
0.080
0.157
9.83E-03
gene of unknown
221988_at






function


S100A10
0.080
0.077
9.85E-03
S100 calcium binding
200872_at






protein A10 (annexin II






ligand, calpactin I, light






polypeptide (p11))


KIAA0553
0.080
4.734
9.87E-03
KIAA0553 protein
212487_at


PTMAP7
0.081
4.827
9.99E-03
prothymosin, alpha
208549_x_at






pseudogene 7


FLJ12671
0.081
11.68
1.00E-02
hypothetical protein
208114_s_at






FLJ12671


MELK
0.081
4.422
1.00E-02
maternal embryonic
204825_at






leucine zipper kinase


ELMO2
0.081
0.052
1.00E-02
engulfment and cell
55692_at






motility 2 (ced-12






homolog, C. elegans)


DDX41
0.081
10.888
1.01E-02
DEAD (Asp-Glu-Ala-
217840_at






Asp) box polypeptide 41


MGC39821
0.081
14.096
1.01E-02
hypothetical protein
216126_at






MGC39821


IMMT
0.081
24.233
1.02E-02
inner membrane protein,
200955_at






mitochondrial (mitofilin)


ASNA1
0.081
8.267
1.01E-02
arsA arsenite transporter,
202024_at






ATP-binding, homolog 1






(bacterial)


TM7SF1
0.081
7.167
1.01E-02
transmembrane 7
204137_at






superfamily member 1






(upregulated in kidney)


CDC2
0.081
5.354
1.02E-02
cell division cycle 2, G1
203213_at






to S and G2 to M


G3BP2
0.081
0.061
1.02E-02
Ras-GTPase activating
208840_s_at






protein SH3 domain-






binding protein 2


KIAA0143
0.081
24.981
1.02E-02
KIAA0143 protein
212150_at


CSNK1A1
0.081
33.586
1.02E-02
caseinkinase 1, alpha 1
213086_s_at


LOC203069
0.081
0.050
1.02E-02
hypothetical protein
35156_at






LOC203069


MRPL28
0.081
15.604
1.02E-02
mitochondrial ribosomal
204599_s_at






protein L28


FLJ20989
0.081
9.325
1.03E-02
hypothetical protein
218187_s_at






FLJ20989


SLC18A2
0.081
0.148
1.03E-02
solute carrier family 18
213549_at






(vesicular monoamine),






member 2


KIAA0399
0.081
0.142
1.03E-02
zinc finger, ZZ-type with
212601_at






EF hand domain 1


MTCP1
0.081
0.114
1.03E-02
mature T-cell
210212_x_at






proliferation 1


POT1
0.081
12.322
1.03E-02
protection of telomeres 1
204354_at


CUL4A
0.082
15.206
1.03E-02
cullin 4A
201423_s_at


LAPTM4B
0.082
4.808
1.04E-02
lysosomal associated
214039_s_at






protein transmembrane 4






beta


TYMS
0.082
3.794
1.04E-02
thymidylate synthetase
202589_at


ERCC1
0.082
0.089
1.04E-02
excision repair cross-
203719_at






complementing rodent






repair deficiency,






complementation group 1






(includes overlapping






antisense sequence)


PSMC3
0.082
3.801
1.04E-02
proteasome (prosome,
201267_s_at






macropain) 26S subunit,






ATPase, 3


PIAS1
0.082
5.994
1.05E-02
protein inhibitor of






activated STAT, 1


CDYL
0.083
14.787
1.07E-02
chromodomain protein,
203098_at






Y-like


ACAT2
0.083
6.322
1.07E-02
acetyl-Coenzyme A
209608_s_at






acetyltransferase 2






(acetoacetyl Coenzyme






A thiolase)


RBBP8
0.083
13.511
1.07E-02
retinoblastoma binding
203344_s_at






protein 8


ATF1
0.083
13.031
1.07E-02
activating transcription
222103_at






factor 1


CBX1
0.083
7.07
1.08E-02
chromobox homolog 1
201518_at






(HP1 beta homolog






Drosophila)


CLK1
0.084
0.090
1.08E-02
CDC-like kinase 1
210346_s_at


PBF
0.084
0.218
1.08E-02
zinc finger protein 395
218149_s_at


SLC2A4RG
0.084
0.080
1.09E-02
SLC2A4 regulator
218494_s_at


KAI1
0.084
6.32
1.09E-02
kangai 1 (suppression of
203904_x_at






tumorigenicity 6,






prostate; CD82 antigen






(R2 leukocyte antigen,






antigen detected by






monoclonal and antibody






IA4))


EP300
0.084
10.703
1.10E-02
E1A binding protein
213579_s_at






p300


DNAJB6
0.085
4.765
1.11E-02
DnaJ (Hsp40) homolog,
209015_s_at






subfamily B, member 6


UGDH
0.085
5.91
1.12E-02
UDP-glucose
203343_at






dehydrogenase


C20ORF9
0.085
56.405
1.12E-02
chromosome 20 open
218709_s_at






reading frame 9


NIT2
0.086
7.547
1.13E-02
Nit protein 2
218557_at


RBM5
0.086
13.578
1.13E-02
RNA binding motif
201394_s_at






protein 5


HNRPH3
0.086
0.088
1.14E-02
heterogeneous nuclear
210110_x_at






ribonucleoprotein H3






(2H9)


DHX40
0.087
0.073
1.15E-02
DEAH (Asp-Glu-Ala-
218277_s_at






His) box polypeptide 40


PSMB9
0.087
8.276
1.15E-02
proteasome (prosome,
204279_at






macropain) subunit, beta






type, 9 (large






multifunctional protease 2)


MSCP
0.087
7.256
1.15E-02
MSCP, mitochondrial
221155_x_at






solute carrier protein


C10ORF22
0.087
6.428
1.16E-02
chromosome 10 open
212500_at






reading frame 22


UROD
0.087
13.864
1.16E-02
uroporphyrinogen
208971_at






decarboxylase


MTSS1
0.087
16.897
1.16E-02
metastasis suppressor 1
203037_s_at


GAB2
0.088
0.115
1.17E-02
GRB2-associated
203853_s_at






binding protein 2


FLJ11856
0.089
15.457
1.19E-02
putative G-protein
218151_x_at






coupled receptor






GPCR41


SNX1
0.089
0.246
1.19E-02
sorting nexin 1
213364_s_at


CGI-94
0.089
12.952
1.19E-02
comparative gene
218235_s_at






identification transcript






94


ANAPC2
0.089
0.250
1.19E-02
anaphase promoting
218555_at






complex subunit 2


PPBP
0.090
7.479
1.21E-02
pro-platelet basic protein
214146_s_at






(chemokine (C-X-C






motif) ligand 7)


LOC51315
0.090
6.733
1.21E-02
hypothetical protein
218303_x_at






LOC51315


ARHGEF2
0.090
9.18
1.21E-02
rho/rac guanine
207629_s_at






nucleotide exchange






factor (GEF) 2


ANKRD10
0.090
0.121
1.22E-02
ankyrin repeat domain 10
218093_s_at


NUDC
0.091
10.798
1.23E-02
nuclear distribution gene
201173_x_at






C homolog (A. nidulans)


PFKP
0.091
6.294
1.24E-02
phosphofructokinase,
201037_at






platelet


NDUFAF1
0.091
5.219
1.24E-02
NADH dehydrogenase
204125_at






(ubiquinone) 1 alpha






subcomplex, assembly






factor 1


STX7
0.091
4.414
1.24E-02
STX7, syntaxin 7
212632_at


SLAMF8
0.091
3.465
1.24E-02
SLAM family member 8
219386_s_at


CPSF6
0.091
0.146
1.24E-02
cleavage and
202469_s_at






polyadenylation specific






factor 6, 68kDa


LSM2
0.091
8.984
1.25E-02
LSM2 homolog, U6
209449_at






small nuclear RNA






associated (S. cerevisiae)


LAP1B
0.091
0.054
1.25E-02
lamina-associated
212408_at






polypeptide 1B


RASA4
0.091
0.189
1.25E-02
RAS p21 protein
212707_s_at






activator 4


POGK
0.091
6.766
1.26E-02
pogo transposable
218229_s_at






element with KRAB






domain


SSR1
0.092
0.122
1.26E-02
signal sequence receptor,
200891_s_at






alpha (translocon-






associated protein alpha)


ATP1B1
0.092
5.102
1.27E-02
ATPase, Na+/K+
201242_s_at






transporting, beta 1






polypeptide


PA2G4
0.092
7.067
1.27E-02
proliferation-associated
208676_s_at






2G4, 38kDa


USF2
0.092
0.244
1.27E-02
upstream transcription
202152_x_at






factor 2, c-fos interacting


CASP3
0.092
14.85
1.27E-02
caspase 3, apoptosis-
202763_at






related cysteine protease


PPIB
0.092
5.353
1.28E-02
peptidylprolyl isomerase
200968_s_at






B (cyclophilin B)


SP2
0.093
0.135
1.29E-02
Sp2 transcription factor
204367_at


GRP58
0.093
15.372
1.29E-02
glucose regulated
208612_at






protein, 58kDa


KIAA0863
0.093
0.097
1.29E-02
KIAA0863 protein
203322_at


CD24
0.094
0.031
1.31E-02
CD24 antigen (small cell
266_s_at






lung carcinoma cluster 4






antigen)


TCFL1
0.094
19.522
1.31E-02
transcription factor-like 1
202261_at


MCM4
0.094
5.113
1.31E-02
MCM4 minichromosome
222037_at






maintenance deficient 4






(S. cerevisiae)


SULT1A1
0.094
0.047
1.31E-02
sulfotransferase family,
211385_x_at






cytosolic, 1A, phenol-






preferring, member 1


FARSLA
0.094
4.776
1.32E-02
phenylalanine-tRNA
216602_s_at






synthetase-like, alpha






subunit


PCF11
0.094
0.197
1.32E-02
pre-mRNA cleavage
203378_at






complex II protein Pcfl 1


TNS
0.094
0.064
1.32E-02
tensin
221747_at


HBP1
0.094
0.062
1.32E-02
HMG-box transcription
209102_s_at






factor 1


ILVBL
0.094
31.351
1.33E-02
ilvB (bacterial
210624_s_at






acetolactate synthase)-






like


ZNF24
0.094
24.761
1.33E-02
zinc finger protein 24
212534_at






(KOX 17)


DYRK1A
0.094
0.134
1.33E-02
dual-specificity tyrosine-
209033_s_at






(Y)-phosphorylation






regulated kinase 1A


GGA1
0.094
0.088
1.34E-02
golgi associated, gamma
45572_s_at






adaptin ear containing,






ARF binding protein 1


WDR26
0.094
0.037
1.33E-02
WD repeat domain 26
218107_at


HNRPAB
0.094
7.05
1.34E-02
heterogeneous nuclear
201277_s_at






ribonucleoprotein A/B


NOLC1
0.094
5.681
1.34E-02
nucleolar and coiled-
205895_s_at






body phosphoprotein 1


ASH2L
0.094
23.135
1.34E-02
ash2 (absent, small, or
209517_s_at






homeotic)-like






(Drosophila)


PPM1F
0.094
0.140
1.34E-02
protein phosphatase 1F
37384_at






(PP2C domain






containing)


SASH1
0.095
8.308
1.36E-02
SAM and SH3 domain
41644_at






containing 1


RPL13
0.095
0.167
1.36E-02
ribosomal protein L13
214351_x_at


RPS2
0.095
3.043
1.37E-02
DNA replication
221521_s_at






complex GINS protein






PSF2


TMEM14A
0.095
6.614
1.37E-02
transmembrane protein 14A
218477_at


FLJ35827
0.095
0.121
1.37E-02
hypothetical protein
212969_x_at






FLJ35827


REPIN1
0.096
0.114
1.38E-02
replication initiator 1
219041_s_at


EI24
0.096
14.543
1.39E-02
etoposide induced 2.4
208289_s_at






mRNA


MRPS7
0.096
9.557
1.39E-02
mitochondrial ribosomal
217932_at






protein S7


TRIM8
0.096
0.173
1.39E-02
tripartite motif-
221012_s_at






containing 8


ERP70
0.096
28.582
1.39E-02
protein disulfide
208658_at






isomerase related protein






(calcium-binding protein,






intestinal-related)


GABPB2
0.096
6.265
1.39E-02
GA binding protein
204618_s_at






transcription factor, beta






subunit 2, 47kDa


KIAA0763
0.096
0.150
1.40E-02
KIAA0763 gene product
203906_at


NGX6
0.096
0.125
1.40E-02
chromosome 9 open
207839_s_at






reading frame 127


CREBL2
0.097
15.292
1.41E-02
cAMP responsive
201989_s_at






element binding protein-






like 2


NEK7
0.097
9.293
1.41E-02
NIMA (never in mitosis
212530_at






gene a)-related kinase 7


CAMLG
0.097
0.165
1.41E-02
calcium modulating
203538_at






ligand


INSIG2
0.097
9.42
1.42E-02
insulin induced gene 2
209566_at


CCT7
0.097
4.279
1.42E-02
chaperonin containing
200812_at






TCP1, subunit 7 (eta)


CCNI
0.097
0.063
1.43E-02
cyclin 1


DAPP1
0.098
7.907
1.44E-02
dual adaptor of
219290_x_at






phosphotyrosine and 3-






phosphoinositides


RBM8A
0.098
4.954
1.45E-02
RNA binding motif
217857_s_at






protein 8A


USP21
0.098
0.207
1.45E-02
ubiquitin specific
218367_x_at






protease 21


PRKCI
0.098
6.662
1.45E-02
protein kinase C, iota
209678_s_at


WBP11
0.098
0.042
1.45E-02
WW domain binding
217822_at






protein 11


CCRL2
0.098
4.788
1.46E-02
chemokine (C-C motif)
211434_s_at






receptor-like 2


MRPL12
0.098
7.889
1.46E-02
mitochondrial ribosomal
203931_s_at






protein L12


PSMB5
0.099
5.428
1.47E-02
proteasome (prosome,
208799_at






macropain) subunit, beta






type, 5


EBI3
0.099
3.985
1.47E-02
Epstein-Barr virus
219424_at






induced gene 3


BCAP31
0.099
9.401
1.48E-02
B-cell receptor-
200837_at






associated protein 31


ZNF297B
0.099
6.866
1.48E-02
zinc finger protein 297B
204181_s_at


KNS2
0.099
5.27
1.49E-02
kinesin 2 60/70kDa
212878_s_at


SRD5A1
0.100
0.212
1.49E-02
steroid-5-alpha-
204675_at






reductase, alpha






polypeptide 1 (3-oxo-5






alpha-steroid delta 4-






dehydrogenase alpha 1)


MSH2
0.100
6.541
1.50E-02
mutS homolog 2, colon
209421_at






cancer, nonpolyposis






type 1 (E. coli)


EIF2B1
0.100
20.633
1.50E-02
eukaryotic translation
201632_at






initiation factor 2B,






subunit 1 alpha, 26kDa


ID3
0.100
0.052
1.50E-02
inhibitor of DNA binding
207826_s_at






3, dominant negative






helix-loop-helix protein


IRAK1BP1
0.100
0.191
1.51E-02
interleukin-1 receptor-
213074_at






associated kinase 1






binding protein 1
















TABLE 33










Annotation of Genes Associated with Meningoencephalitis














Ingenuity assignment to one of the






following functions: cell-cycle





(includes DNA synthesis, cell





growth and proliferation), cell





death, cell signaling and





interaction (includes cell signaling





and cell-to-cell signaling and





interaction), immune functions





(includes immune and lymphatic



Odds Ratio for

system development and function
Identified by



association with
FDR association
and immune response), protein
more than one


Gene
meningoencephalitis
meningoencephalitis
synthesis and trafficking
probeset














STAT1
230.416
0.004
Yes
Yes


NHP2L1
3136.203
0.010
Yes
No


C10ORF7
673.31
0.010
Yes
No


ZW10
470.958
0.010
Yes
No


ICMT
417.532
0.010
Yes
No


RABGAP1
303.809
0.010
Yes
No


BRD2
56.318
0.010
Yes
Yes


KPNB1
32.282
0.010
Yes
Yes


GZMB
31.809
0.010
Yes
No


KLF2
0.038
0.010
Yes
No


STK17B
0.025
0.010
Yes
No


FLJ11806
651.763
0.010
Not assigned to function by
Yes





Ingenuity


C12ORF22
459.155
0.010
Not assigned to function by
No





Ingenuity


SEC24C
66.791
0.010
Not assigned to function by
No





Ingenuity


FNBP3
13.972
0.010
Not assigned to function by
No





Ingenuity


JARID1B
0.006
0.010
Not assigned to function by
Yes





Ingenuity


TRAP240
68.675
0.010
No
No


STAT3
6.606
0.011
Yes
No


BTG2
0.033
0.011
Yes
No


MGC21416
8.373
0.011
Not assigned to function by
Yes





Ingenuity


OSBPL8
4.201
0.011
Not assigned to function by
No





Ingenuity


HEAB
0.001
0.011
Not assigned to function by
No





Ingenuity


UBE2D3
0.002
0.011
No
Yes


ATP6V1D
172.543
0.011
Not assigned to function by
Yes





Ingenuity


KIF5B
3.731
0.011
No
No


DC8
69.508
0.012
Not assigned to function by
No





Ingenuity


GLTSCR1
0.013
0.013
Not assigned to function by
No





Ingenuity


CD84
23.97
0.013
No
No


UGCG
14.445
0.013
Yes
No


SFRS2IP
57.281
0.014
Not assigned to function by
Yes





Ingenuity


MMP24
0.022
0.014
No
No


MBD4
8.79
0.014
Yes
No


TNPO3
21.713
0.014
Not assigned to function by
No





Ingenuity


GCDH
50.321
0.014
No
No


PABPC1
0.006
0.014
No
Yes


VDR
7.092
0.014
Yes
No


H2AFY
0.016
0.015
Not assigned to function by
No





Ingenuity


IL2RA
11.266
0.016
Yes
No


STAT5B
0.029
0.016
Yes
No


CBX6
34.482
0.016
Not assigned to function by
No





Ingenuity


TTC3
5.376
0.016
No
No


TRIP13
17.331
0.016
No
No


FLJ23441
17.419
0.016
No
No


STXBP2
0.095
0.016
No
No


LRRFIP1
18.564
0.016
No
Yes


PADI2
0.145
0.016
Not assigned to function by
No





Ingenuity


HNRPC
324.673
0.016
Yes
No


PTPRC
4.891
0.017
Yes
No


PTDSR
0.018
0.018
Yes
No


TPR
4.823
0.018
Yes
No


HUMGT198A
8.097
0.018
No
No


DUT
40.207
0.018
Yes
Yes


RAB1A
0.003
0.018
Yes
No


HMG2L1
5.679
0.019
Not assigned to function by
No





Ingenuity


RIN3
0.105
0.019
Not assigned to function by
No





Ingenuity


PDCD8
119.631
0.019
Yes
No


CSE1L
38.753
0.019
Yes
No


RNMT
0.050
0.019
Yes
No


TFE3
0.041
0.019
Yes
No


GLS
60.862
0.019
No
No


FLJ12788
167.936
0.020
Not assigned to function by
No





Ingenuity


MGAT2
20.774
0.020
No
No


CGI-37
10.964
0.021
Not assigned to function by
No





Ingenuity


C21ORF80
0.032
0.021
Not assigned to function by
No





Ingenuity


LUC7A
7.673
0.021
No
No


FBXW7
5.619
0.021
No
No


DICER1
0.073
0.021
No
No


UBCE71P5
0.036
0.021
No
No


TXNL2
152.265
0.021
Not assigned to function by
No





Ingenuity


PRKRA
0.027
0.022
Yes
No


BARD1
11.776
0.022
Yes
No


SH3BP5
11.205
0.022
Yes
No


OBRGRP
4.025
0.022
Not assigned to function by
No





Ingenuity


C1ORF33
12.564
0.023
Not assigned to function by
No





Ingenuity


M96
9.28
0.023
Not assigned to function by
Yes





Ingenuity


DNCL1
6.81
0.023
Yes
No


BAZ1A
6.808
0.023
Yes
No


NALP1
0.133
0.023
Yes
No


GNAS
0.071
0.023
Yes
Yes


IPO4
29.56
0.023
No
No


TH1L
13.185
0.024
Not assigned to function by
No





Ingenuity


IRS2
0.060
0.024
Yes
No


LTF
0.325
0.025
Yes
No


MIRAB13
0.109
0.026
Not assigned to function by
No





Ingenuity


BATF
9.718
0.026
Yes
No


FLN29
176.965
0.026
Not assigned to function by
No





Ingenuity


HAX1
34.12
0.026
No
No


MYO1B
18.41
0.026
No
No


SLC5A3
4.832
0.026
No
No


PADI4
0.108
0.026
No
No


STK10
0.052
0.026
Not assigned to function by
No





Ingenuity


RAB2
0.002
0.027
Yes
No


BPI
0.219
0.027
Yes
No


DEFA4
0.196
0.027
Not assigned to function by
No





Ingenuity


KPNA6
34.224
0.028
Yes
No


C19ORF10
45.058
0.028
Yes
No


DKFZP564G2022
11.966
0.028
Not assigned to function by
No





Ingenuity


SNRK
0.043
0.028
Not assigned to function by
No





Ingenuity


GBP1
5.53
0.028
Yes
Yes


ZFP36
0.108
0.029
Yes
No


SIPA1
0.053
0.029
Yes
No


ZNF238
0.120
0.029
No
No


CXCL10
7.825
0.029
Yes
No


RRM2
5.394
0.029
No
Yes


RAB31
3.04
0.029
Not assigned to function by
No





Ingenuity


USP36
0.071
0.029
Not assigned to function by
No





Ingenuity


PTP4A1
0.034
0.029
No
No


DPCK
156.071
0.029
No
No


ALDOC
11.591
0.029
No
No


ZFP36L1
0.036
0.030
Yes
No


PXMP3
39.115
0.030
No
No


CYLN2
0.060
0.030
Not assigned to function by
No





Ingenuity


STAU
0.078
0.031
No
Yes


PHF1
0.130
0.031
Not assigned to function by
No





Ingenuity


HN1
18.055
0.031
Not assigned to function by
No





Ingenuity


STOML2
6.512
0.031
Not assigned to function by
No





Ingenuity


ARID3B
0.149
0.031
Not assigned to function by
No





Ingenuity


IL19
8.869
0.031
Yes
No


WSX1
46.587
0.032
Yes
No


NFE2L1
33.502
0.032
Yes
No


TDE1
17.535
0.032
Yes
No


POLA
14.919
0.032
Yes
No


NALP2
16.21
0.032
Not assigned to function by
No





Ingenuity


CKLFSF6
13.746
0.032
Not assigned to function by
No





Ingenuity


SSH1
11.182
0.032
Not assigned to function by
No





Ingenuity


DKFZP434H132
0.143
0.032
Not assigned to function by
No





Ingenuity


JM5
0.114
0.032
Not assigned to function by
No





Ingenuity


FLJ13479
0.010
0.032
Not assigned to function by
No





Ingenuity


MINK
0.145
0.032
No
No


MK167
69.144
0.032
Yes
No


TIMM13
22.616
0.032
Yes
No


JUNB
0.108
0.032
Yes
No


RBX1
27.734
0.032
Not assigned to function by
No





Ingenuity


ECHDC1
16.161
0.032
Not assigned to function by
No





Ingenuity


KIAA0930
14.228
0.032
Not assigned to function by
No





Ingenuity


HEG
6.044
0.032
Not assigned to function by
No





Ingenuity


MASK
5.562
0.032
Not assigned to function by
No





Ingenuity


C9ORF28
0.037
0.032
Not assigned to function by
No





Ingenuity


RLF
0.028
0.032
Not assigned to function by
No





Ingenuity


AB026190
12.367
0.033
No
No


GTF2H5
8.729
0.033
Not assigned to function by
No





Ingenuity


RBMS1
5.153
0.033
Not assigned to function by
Yes





Ingenuity


ENIGMA
0.081
0.033
No
No


MIR
0.128
0.033
No
No


SRRM2
5.461
0.033
Not assigned to function by
No





Ingenuity


MCL1
0.058
0.033
Yes
Yes


SRR
15.068
0.033
No
No


FACL5
89.075
0.034
Yes
No


CPSF1
0.209
0.034
No
No


PTTG1IP
0.004
0.034
Yes
No


AK2
17.668
0.034
No
No


GTPBP1
0.032
0.034
Yes
No


UNG
10.732
0.035
Yes
No


RPS28
0.215
0.035
Yes
No


PAX5
8.402
0.035
Yes
No


PSMD8
11.013
0.035
Not assigned to function by
No





Ingenuity


NUDT1
10.67
0.035
No
No


SLC25A12
52.625
0.035
No
No


C1ORF24
12.539
0.036
Not assigned to function by
No





Ingenuity


HTATIP2
15.356
0.036
Yes
No


SRPK2
3.184
0.036
Not assigned to function by
No





Ingenuity


PRKAR1A
16.407
0.036
Yes
No


CD80
26.52
0.036
Yes
No


MGC3248
20.329
0.036
Not assigned to function by
No





Ingenuity


UBXD2
6.211
0.036
Not assigned to function by
No





Ingenuity


PDCD11
15.892
0.036
Yes
No


ISGF3G
7.836
0.036
Yes
No


RAB7
0.083
0.036
Yes
No


CDC42
0.051
0.036
Yes
Yes


GALNT1
36.544
0.036
No
No


STX18
23.897
0.036
No
No


NFATC1
80.225
0.036
Yes
No


NR3C1
11.05
0.036
Yes
Yes


CABIN1
0.162
0.036
Yes
No


NET1
0.146
0.036
Yes
No


NFIL3
0.116
0.036
Yes
No


MOAP1
0.115
0.036
Yes
No


SKP1A
0.113
0.036
Yes
No


G1P3
0.069
0.036
Yes
No


BNIP3L
0.044
0.036
Yes
No


PSMD1
19.918
0.036
Not assigned to function by
No





Ingenuity


PSMD11
5.523
0.036
Not assigned to function by
No





Ingenuity


H2AV
0.268
0.036
Not assigned to function by
No





Ingenuity


FLJ11127
0.069
0.036
Not assigned to function by
No





Ingenuity


C6ORF82
0.041
0.036
Not assigned to function by
No





Ingenuity


COL4A3BP
17.703
0.036
No
No


SEC63
7.604
0.036
No
No


XTP2
4.327
0.037
Not assigned to function by
Yes





Ingenuity


MBNL3
0.058
0.037
No
No


PDHB
33.983
0.037
No
No


CKS1B
16.085
0.038
Yes
No


GALNS
0.227
0.038
Not assigned to function by
No





Ingenuity


EIF5
8.566
0.038
Yes
Yes


USP12
48.047
0.038
Not assigned to function by
No





Ingenuity


KIAA0650
0.146
0.038
Not assigned to function by
Yes





Ingenuity


UQCRFS1
0.060
0.038
No
No


ACO1
49.485
0.038
Yes
No


MRPL13
9.48
0.038
Yes
No


SCGF
0.120
0.038
Yes
No


CHC1L
0.084
0.038
Not assigned to function by
No





Ingenuity


TRIAD3
29.384
0.039
Yes
No


RFP
35.742
0.039
Yes
Yes


ITGAV
12.837
0.039
Yes
No


RPA3
10.718
0.039
Yes
No


PSMD13
16.384
0.039
Not assigned to function by
Yes





Ingenuity


AGTPBP1
0.099
0.039
Not assigned to function by
No





Ingenuity


CGI-127
0.039
0.039
Not assigned to function by
No





Ingenuity


ACOX1
15.909
0.039
No
No


SEC23B
11.687
0.039
No
No


KIF7
7.918
0.039
No
No


KIAA0892
0.071
0.039
Not assigned to function by
No





Ingenuity


APLP2
0.155
0.039
No
Yes


IL7R
3.182
0.039
Yes
No


SR140
0.144
0.039
Not assigned to function by
No





Ingenuity


TDP1
10.611
0.040
Not assigned to function by
No





Ingenuity


HMGCL
11.109
0.040
No
No


VDAC3
7.789
0.040
No
No


HIPK1
0.025
0.040
Yes
No


CGI-01
9.62
0.040
Not assigned to function by
No





Ingenuity


FLJ11078
0.094
0.040
Not assigned to function by
No





Ingenuity


FLJ14639
38.716
0.040
No
No


CGI-128
54.238
0.041
Not assigned to function by
No





Ingenuity


IL9
9.187
0.041
Yes
No


CCNL1
0.153
0.041
Not assigned to function by
No





Ingenuity


GORASP2
0.100
0.041
Not assigned to function by
No





Ingenuity


NUP43
6.165
0.041
No
No


AP162
0.069
0.041
Not assigned to function by
No





Ingenuity


PLSCR3
0.029
0.041
Not assigned to function by
No





Ingenuity


NCOA3
13.528
0.042
Yes
No


TNFSF10
4.806
0.042
Yes
No


PPP6C
0.045
0.042
Yes
No


RNUT1
11.552
0.042
Not assigned to function by
No





Ingenuity


ALEX3
5.508
0.042
Not assigned to function by
No





Ingenuity


MGLL
14.896
0.042
No
No


CENPC1
0.106
0.042
Yes
No


NR1D1
0.197
0.042
Yes
No


FLJ12439
6.764
0.042
Not assigned to function by
No





Ingenuity


MTMR2
11.89
0.042
No
No


FDPS
11.053
0.042
No
No


TFEB
0.138
0.042
No
No


KIAA1332
0.061
0.042
Not assigned to function by
No





Ingenuity


C14ORF159
0.132
0.042
Not assigned to function by
No





Ingenuity


PSME2
10.064
0.042
Yes
No


MPHOSPH6
10.656
0.043
Yes
No


YWHAB
10.596
0.043
Yes
No


MCM7
7.75
0.043
Yes
No


PSMD2
334.893
0.043
Not assigned to function by
No





Ingenuity


AMPD2
0.122
0.043
No
No


CCNE1
6.88
0.044
Yes
No


MMP7
6.512
0.044
Yes
No


GTF2H1
12.954
0.044
Yes
No


FNBP1
5.151
0.044
No
No


UBD
7.847
0.044
Yes
No


FLJ38984
19.598
0.045
Not assigned to function by
No





Ingenuity


TLE4
0.108
0.045
No
No


ITM2B
0.032
0.045
Yes
No


HSD17B7
14.99
0.045
No
No


KIAA1115
33.455
0.047
Not assigned to function by
No





Ingenuity


COAS1
3.854
0.047
Not assigned to function by
No





Ingenuity


XRCC5
17.167
0.047
Yes
No


STMN1
11.125
0.047
Yes
No


CTLA4
8.016
0.047
Yes
No


STAG2
6.595
0.047
Not assigned to function by
No





Ingenuity


KIAA0404
0.144
0.047
Not assigned to function by
No





Ingenuity


SF3B4
0.180
0.047
No
No


CXCL9
6.108
0.047
Yes
No


ITGAX
0.032
0.047
No
No


FLJ14888
25.043
0.048
Not assigned to function by
No





Ingenuity


FLJ10803
31.56
0.048
Not assigned to function by
No





Ingenuity


PTEN
0.107
0.048
Yes
No


OSBPL9
288.036
0.048
Not assigned to function by
No





Ingenuity


EFHD2
0.128
0.048
Not assigned to function by
No





Ingenuity


PPIH
29.937
0.048
Yes
No


DOCK2
0.100
0.048
Yes
No


FGR
0.088
0.048
Yes
No


NKTR
4.902
0.048
Not assigned to function by
No





Ingenuity


ZCCHC2
0.080
0.048
Not assigned to function by
No





Ingenuity


BAZ2A
4.766
0.048
No
Yes


QKI
29.983
0.049
Yes
No


SPN
0.102
0.049
Yes
No


MATR3
0.124
0.049
Not assigned to function by
No





Ingenuity


KIAA1536
0.073
0.049
Not assigned to function by
No





Ingenuity


PABPC3
0.038
0.049
Not assigned to function by
No





Ingenuity


SUCLA2
10.996
0.049
No
No


GABBR1
0.117
0.049
No
No


FBS1
0.031
0.049
Yes
No


C3ORF4
5.543
0.049
Not assigned to function by
No





Ingenuity


CYLD
0.161
0.049
Not assigned to function by
Yes





Ingenuity


FLJ21347
0.098
0.049
Not assigned to function by
No





Ingenuity


AIM2
9.506
0.049
Yes
No


PTX1
9.051
0.049
Not assigned to function by
No





Ingenuity


LRDD
0.219
0.049
Yes
No


LOC283537
0.094
0.049
Not assigned to function by
No





Ingenuity


CLN5
11.634
0.049
No
No


EPRS
9.17
0.049
No
No


PEX3
9.809
0.050
No
No


NCOA2
0.220
0.050
No
No


BHC80
0.124
0.050
Not assigned to function by
No





Ingenuity


ARHQ
41.236
0.050
No
No


PFKM
18.355
0.050
No
No


WARS
23.882
0.050
Yes
Yes


ESPL1
6.537
0.050
Yes
No


KRAS2
3.71
0.050
Yes
No


RGS2
0.176
0.050
Yes
No


EDG6
0.144
0.050
Yes
No


MAP3K71P2
0.079
0.050
Yes
No


CD2BP2
59.36
0.050
Not assigned to function by
No





Ingenuity


ZNF408
0.239
0.050
Not assigned to function by
No





Ingenuity


PLEKHF2
0.154
0.050
Not assigned to function by
No





Ingenuity


KIAA1076
0.117
0.050
Not assigned to function by
No





Ingenuity


DRE1
0.113
0.050
Not assigned to function by
No





Ingenuity


C14ORF32
0.097
0.050
Not assigned to function by
No





Ingenuity


FXC1
13.071
0.050
No
No


TSTA3
6.918
0.050
No
No


PWP1
4.459
0.050
No
No


TCF7L2
0.223
0.050
No
No


ARL4
0.063
0.050
No
No


RPA2
17.449
0.050
Yes
No


GAS7
0.091
0.051
Yes
No


KIAA0555
7.853
0.051
Not assigned to function by
No





Ingenuity


SSFA2
0.036
0.051
Not assigned to function by
No





Ingenuity


NUP50
13.853
0.051
No
No


GMEB2
0.097
0.051
No
No


PIR51
9.238
0.051
Yes
No


C9ORF83
8.68
0.051
Not assigned to function by
No





Ingenuity


PRO1843
0.126
0.051
Not assigned to function by
No





Ingenuity


VEGF
0.124
0.052
Yes
Yes


RERE
0.093
0.052
Yes
No


DNM1L
16.425
0.052
No
No


ARID1A
11.487
0.052
Yes
No


FLJ10815
9.617
0.052
Not assigned to function by
No





Ingenuity


CIAO1
17.811
0.052
Yes
No


MNT
0.113
0.052
Yes
No


PSMA4
51.574
0.052
Not assigned to function by
No





Ingenuity


GNL1
19.339
0.052
No
No


CXCL5
4.558
0.052
Yes
No


FLJ32731
0.180
0.052
Not assigned to function by
No





Ingenuity


DUSP10
0.099
0.053
Yes
No


KIAA0102
9.997
0.053
Not assigned to function by
No





Ingenuity


PROSC
5.622
0.053
Not assigned to function by
No





Ingenuity


LYL1
0.235
0.053
Not assigned to function by
No





Ingenuity


MKRN1
0.095
0.053
Not assigned to function by
No





Ingenuity


MYCBP
23.309
0.053
No
No


CKS2
5.629
0.053
Yes
No


SGK
0.161
0.053
Yes
No


C20ORF104
0.091
0.053
Not assigned to function by
No





Ingenuity


KMO
5.843
0.053
No
No


ARS2
0.028
0.053
No
Yes


ZNF259
26.34
0.053
Yes
No


GC20
0.016
0.054
Yes
No


SERP1
0.022
0.054
No
No


MSF
0.222
0.054
Yes
No


TRAPPC3
11.773
0.054
Not assigned to function by
No





Ingenuity


CDC40
0.074
0.054
No
No


PPP3CA
5.417
0.054
Yes
No


FLJ14753
0.021
0.054
Not assigned to function by
No





Ingenuity


PELI1
0.175
0.054
Not assigned to function by
No





Ingenuity


PRKCSH
0.064
0.054
No
No


SPINT2
0.115
0.054
No
No


PSARL
50.956
0.055
Not assigned to function by
No





Ingenuity


HT007
10.945
0.056
Not assigned to function by
No





Ingenuity


RAD51C
5.167
0.056
Yes
No


TRIP-BR2
3.547
0.056
Not assigned to function by
No





Ingenuity


TRA1
12.843
0.056
Yes
No


PIK3CA
0.075
0.056
Yes
No


DKFZP586D0919
25.287
0.056
Not assigned to function by
No





Ingenuity


CIC
0.300
0.056
Not assigned to function by
No





Ingenuity


HSPC051
12.156
0.057
No
No


NADSYN1
0.182
0.057
Not assigned to function by
No





Ingenuity


ELAVL1
4.903
0.057
No
No


CCL22
3.61
0.057
Yes
No


C20ORF67
0.176
0.057
Not assigned to function by
No





Ingenuity


CCNB2
12.529
0.057
No
No


LOC51064
0.128
0.057
No
No


POLR2K
7.759
0.057
No
No


LRP8
7.185
0.057
No
No


FLJ20080
9.512
0.057
Not assigned to function by
No





Ingenuity


JUND
0.059
0.058
Yes
No


ACADM
6.254
0.058
No
No


FLJ20534
12.83
0.058
Not assigned to function by
No





Ingenuity


TOB1
0.172
0.058
Yes
No


ACTG1
0.006
0.058
No
Yes


FLJ10534
7.735
0.059
Not assigned to function by
Yes





Ingenuity


CTPS
5.567
0.059
No
No


TCP1
21.895
0.059
No
No


D1S155E
0.073
0.059
No
No


TIMELESS
6.145
0.059
Yes
No


NCOR1
59.364
0.059
No
No


CSK
0.040
0.059
Yes
No


C9ORF40
7.261
0.059
Not assigned to function by
No





Ingenuity


PHF3
0.081
0.059
Not assigned to function by
No





Ingenuity


DKFZP564D0478
0.049
0.059
Not assigned to function by
No





Ingenuity


DDEF1
7.693
0.059
No
No


UBE2L3
7.274
0.059
No
No


FBXL12
0.008
0.059
No
No


CSNK2A1
9.961
0.059
Yes
No


KIAA0483
22.767
0.059
Not assigned to function by
No





Ingenuity


TNFRSF9
3.452
0.060
Yes
No


NEDD8
5.847
0.060
No
No


ZNF161
0.086
0.060
Yes
No


KIAA0738
7.398
0.060
Not assigned to function by
No





Ingenuity


SIAT9
0.018
0.060
No
No


MADH7
0.113
0.060
Yes
No


USP3
0.051
0.060
No
No


KHDRBS1
7.898
0.060
Yes
No


C5ORF6
0.047
0.061
Not assigned to function by
No





Ingenuity


TCF8
0.300
0.061
Yes
No


GLG1
6.776
0.061
Not assigned to function by
No





Ingenuity


RBAF600
0.011
0.061
Not assigned to function by
No





Ingenuity


SLC35D2
21.354
0.061
Not assigned to function by
No





Ingenuity


PIGA
0.156
0.061
Yes
No


DUSP3
9.995
0.061
No
No


DSCR1
26.47
0.061
Yes
No


PTMA
9.39
0.062
Yes
Yes


POLE2
5.826
0.062
Yes
No


NRG1
0.308
0.062
Yes
No


TRAF6
0.049
0.062
Yes
No


CGI-51
26.286
0.062
Not assigned to function by
No





Ingenuity


TIP120A
14.465
0.062
No
No


MAC30
9.549
0.062
No
No


WDR12
6.497
0.062
No
No


SLC22A18
0.139
0.062
No
No


VAMP2
0.092
0.062
No
Yes


EVI2B
0.109
0.062
Not assigned to function by
No





Ingenuity


TIEG2
0.231
0.062
Yes
No


COPS5
8.168
0.062
Yes
No


RNF139
0.143
0.062
No
No


MPO
0.146
0.063
Yes
No


PCMT1
8.822
0.063
No
No


KCNK4
6.788
0.063
No
No


SSA2
5.063
0.063
No
No


Unknown
4.635
0.064
Not assigned to function by
No





Ingenuity


Unknown
0.315
0.064
Not assigned to function by
No





Ingenuity


Unknown
0.271
0.064
Not assigned to function by
No





Ingenuity


Unknown
0.157
0.064
Not assigned to function by
No





Ingenuity


Unknown
0.109
0.064
Not assigned to function by
No





Ingenuity


Unknown
0.089
0.064
Not assigned to function by
No





Ingenuity


Unknown
0.081
0.064
Not assigned to function by
No





Ingenuity


Unknown
0.069
0.064
Not assigned to function by
No





Ingenuity


Unknown
0.015
0.064
Not assigned to function by
No





Ingenuity


DSIPI
0.237
0.064
Yes
No


POLD3
8.246
0.064
Yes
No


EEF2
0.180
0.064
Yes
No


NEIL1
0.090
0.064
Yes
No


KIAA0683
13.161
0.064
Not assigned to function by
No





Ingenuity


KIAA1002
0.133
0.064
Not assigned to function by
No





Ingenuity


FLJ10099
25.454
0.064
Not assigned to function by
No





Ingenuity


KIAA0582
6.047
0.065
Not assigned to function by
No





Ingenuity


HADHSC
0.155
0.065
No
No


VIPR1
0.154
0.065
Yes
No


C2ORF6
8.647
0.065
Not assigned to function by
No





Ingenuity


SLC4A1AP
36.646
0.065
Not assigned to function by
No





Ingenuity


ARL7
0.187
0.065
Not assigned to function by
Yes





Ingenuity


DXYS155E
0.032
0.065
Not assigned to function by
No





Ingenuity


BIRC2
0.118
0.065
Yes
No


STAT5A
56.762
0.065
Yes
No


PHB
8.89
0.065
Yes
No


MYLK
3.958
0.065
Yes
No


ATP5L
0.096
0.065
Not assigned to function by
Yes





Ingenuity


KEAP1
7.403
0.066
Not assigned to function by
No





Ingenuity


CAMKK2
3.454
0.066
No
No


PRKCN
0.189
0.066
No
No


MARK4
0.196
0.066
Not assigned to function by
No





Ingenuity


CDK4
5.678
0.066
Yes
No


PAICS
5.533
0.066
No
Yes


NFKB1
5.354
0.066
Yes
No


CGI-90
9.14
0.066
Not assigned to function by
No





Ingenuity


TNIP3
5.962
0.066
Not assigned to function by
No





Ingenuity


C1ORF9
0.058
0.066
Not assigned to function by
No





Ingenuity


IL12B
4.425
0.067
Yes
No


RUNX1
0.037
0.067
Yes
No


POP5
8.456
0.067
No
No


C20ORF121
0.125
0.067
Not assigned to function by
No





Ingenuity


EIF2B2
28.683
0.067
Yes
No


MGC4825
14.636
0.067
Not assigned to function by
No





Ingenuity


MRPL19
7.652
0.067
Not assigned to function by
No





Ingenuity


KIAA0982
0.170
0.067
Not assigned to function by
No





Ingenuity


ILF3
11.625
0.067
No
No


CCR1
4.928
0.067
Yes
No


TNF
3.471
0.067
Yes
No


LYRIC
0.074
0.067
Not assigned to function by
Yes





Ingenuity


FLJ21603
0.052
0.067
Not assigned to function by
No





Ingenuity


PRKAG1
8.572
0.067
No
No


NISCH
0.054
0.067
No
No


BRD3
0.076
0.067
Not assigned to function by
No





Ingenuity


COX7C
0.116
0.067
No
Yes


SRP72
3.342
0.068
Not assigned to function by
No





Ingenuity


CA12
4.9
0.068
No
No


KLF3
0.147
0.068
No
No


FLJ20546
9.408
0.068
Not assigned to function by
No





Ingenuity


BLM
13.128
0.068
Yes
No


PLAC8
0.161
0.069
Not assigned to function by
No





Ingenuity


KIAA1012
32.462
0.069
Not assigned to function by
No





Ingenuity


SRPK1
7.399
0.069
No
No


CLECSF12
0.260
0.069
Yes
No


COPS8
60.228
0.069
Not assigned to function by
No





Ingenuity


MGC11256
11.319
0.069
Not assigned to function by
No





Ingenuity


MRPS11
10.289
0.069
Not assigned to function by
No





Ingenuity


SLC2A14
0.219
0.069
Not assigned to function by
No





Ingenuity


CUTL1
0.106
0.069
No
No


PAFAH1B1
18.233
0.069
Yes
No


AKAP13
8.326
0.069
Yes
Yes


HIST1H4C
4.545
0.071
Not assigned to function by
No





Ingenuity


PSME4
0.102
0.071
Not assigned to function by
No





Ingenuity


EIF5B
5.498
0.072
Yes
No


KIAA0152
23.99
0.072
Not assigned to function by
No





Ingenuity


CINP
18.638
0.072
Not assigned to function by
No





Ingenuity


G0S2
0.266
0.072
Not assigned to function by
No





Ingenuity


SENP3
15.871
0.072
No
No


COL18A1
0.057
0.072
Yes
No


FN5
10.552
0.072
Not assigned to function by
No





Ingenuity


HNRPR
10.584
0.072
No
No


ATP6V1C1
6.714
0.072
No
Yes


EEF1E1
9.813
0.072
Not assigned to function by
No





Ingenuity


TANK
6.588
0.072
No
No


IFIT5
0.128
0.073
Not assigned to function by
No





Ingenuity


TUBA3
0.124
0.073
Not assigned to function by
No





Ingenuity


UBE2J1
10.265
0.074
Not assigned to function by
No





Ingenuity


PER1
0.118
0.074
No
No


DGCR14
0.150
0.074
Not assigned to function by
No





Ingenuity


CGI-49
15.54
0.074
Not assigned to function by
No





Ingenuity


CEACAM8
0.244
0.074
Yes
No


GNAI2
0.178
0.074
Yes
No


13CDNA73
0.086
0.074
Not assigned to function by
No





Ingenuity


ACAT1
13.587
0.074
No
No


GOT1
6.694
0.074
No
No


SMG1
0.096
0.074
No
No


TUBB
0.250
0.074
Not assigned to function by
No





Ingenuity


CHD4
12.6
0.075
No
No


RGS10
4.866
0.075
No
No


CAMP
0.253
0.075
Yes
No


APOM
0.107
0.075
No
No


FLJ21868
0.096
0.076
Not assigned to function by
No





Ingenuity


MCM10
4.325
0.076
Not assigned to function by
No





Ingenuity


C11ORF2
0.215
0.076
Not assigned to function by
No





Ingenuity


MPZL1
26.093
0.076
No
No


MRPL48
15.923
0.077
Not assigned to function by
No





Ingenuity


SET
10.103
0.077
Yes
No


RARA
0.123
0.077
Yes
No


C16ORF35
0.221
0.077
Not assigned to function by
No





Ingenuity


T1
0.084
0.077
Not assigned to function by
No





Ingenuity


INHBA
3.484
0.077
Yes
No


NCOA1
0.124
0.077
Yes
No


JMJD1
0.113
0.077
Not assigned to function by
No





Ingenuity


UVRAG
0.043
0.077
Not assigned to function by
No





Ingenuity


NCBP1
6.422
0.077
No
No


NINJ2
0.217
0.077
No
No


CXCL13
4.537
0.077
Yes
No


CYB5
7.271
0.078
Yes
Yes


KLRD1
13.813
0.079
Yes
No


PLEKHB2
0.035
0.079
Not assigned to function by
No





Ingenuity


APG12L
10.273
0.079
No
No


TNPO1
16.302
0.079
Yes
No


PDPK1
6.544
0.079
Yes
No


FOSL2
0.091
0.079
Yes
No


SLCO3A1
0.139
0.079
Not assigned to function by
No





Ingenuity


YT521
0.097
0.079
No
No


NDUFB8
0.060
0.079
No
No


TRIM44
6.37
0.079
Not assigned to function by
No





Ingenuity


USP4
23.919
0.079
Not assigned to function by
No





Ingenuity


SEC61A1
14.769
0.079
Not assigned to function by
No





Ingenuity


SF3B1
0.048
0.079
Not assigned to function by
Yes





Ingenuity


HA-1
0.193
0.080
Not assigned to function by
No





Ingenuity


SMARCD3
0.202
0.080
No
No


AP3D1
79.171
0.080
Yes
No


EG1
6.217
0.080
Not assigned to function by
No





Ingenuity


SIGIRR
0.267
0.080
Not assigned to function by
No





Ingenuity


S100A10
0.077
0.080
Yes
No


KIAA0553
4.734
0.080
Not assigned to function by
No





Ingenuity


PTMAP7
4.827
0.081
Not assigned to function by
No





Ingenuity


FLJ12671
11.68
0.081
Not assigned to function by
No





Ingenuity


MELK
4.422
0.081
Not assigned to function by
No





Ingenuity


ELMO2
0.052
0.081
Not assigned to function by
No





Ingenuity


DDX41
10.888
0.081
Yes
No


MGC39821
14.096
0.081
No
No


CDC2
5.354
0.081
Yes
No


IMMT
24.233
0.081
Not assigned to function by
No





Ingenuity


TM7SF1
7.167
0.081
Not assigned to function by
No





Ingenuity


G3BP2
0.061
0.081
Not assigned to function by
No





Ingenuity


ASNA1
8.267
0.081
No
No


KIAA0143
24.981
0.081
Not assigned to function by
No





Ingenuity


CSNK1A1
33.586
0.081
No
Yes


LOC203069
0.050
0.081
Not assigned to function by
No





Ingenuity


MRPL28
15.604
0.081
Not assigned to function by
No





Ingenuity


SLC18A2
0.148
0.081
Yes
No


MTCP1
0.114
0.081
Yes
No


FLJ20989
9.325
0.081
Not assigned to function by
No





Ingenuity


KIAA0399
0.142
0.081
Not assigned to function by
Yes





Ingenuity


POT1
12.322
0.081
Not assigned to function by
No





Ingenuity


CUL4A
15.206
0.082
Not assigned to function by
No





Ingenuity


LAPTM4B
4.808
0.082
Not assigned to function by
No





Ingenuity


TYMS
3.794
0.082
Yes
No


ERCC1
0.089
0.082
Yes
No


PSMC3
3.801
0.082
Yes
No


PIAS1
5.994
0.082
Yes
No


CDYL
14.787
0.083
No
No


ACAT2
6.322
0.083
No
No


RBBP8
13.511
0.083
Not assigned to function by
No





Ingenuity


ATF1
13.031
0.083
Yes
No


CBX1
7.07
0.083
No
No


CLK1
0.090
0.084
Yes
Yes


PBF
0.218
0.084
Not assigned to function by
No





Ingenuity


SLC2A4RG
0.080
0.084
No
No


KAI1
6.32
0.084
Yes
No


EP300
10.703
0.084
Yes
No


DNAJB6
4.765
0.085
Yes
No


UGDH
5.91
0.085
No
No


C20ORF9
56.405
0.085
Not assigned to function by
No





Ingenuity


NIT2
7.547
0.086
Not assigned to function by
No





Ingenuity


RBM5
13.578
0.086
Yes
No


HNRPH3
0.088
0.086
No
No


DHX40
0.073
0.087
Not assigned to function by
No





Ingenuity


MSCP
7.256
0.087
Not assigned to function by
No





Ingenuity


PSMB9
8.276
0.087
No
No


C10ORF22
6.428
0.087
Not assigned to function by
No





Ingenuity


UROD
13.864
0.087
Not assigned to function by
No





Ingenuity


MTSS1
16.897
0.087
No
No


GAB2
0.115
0.088
Yes
No


FLJ11856
15.457
0.089
Not assigned to function by
No





Ingenuity


SNX1
0.246
0.089
No
No


CGI-94
12.952
0.089
Not assigned to function by
No





Ingenuity


ANAPC2
0.250
0.089
Yes
No


PPBP
7.479
0.090
Yes
No


LOC51315
6.733
0.090
Not assigned to function by
No





Ingenuity


ARHGEF2
9.18
0.090
Yes
No


ANKRD10
0.121
0.090
Not assigned to function by
No





Ingenuity


NUDC
10.798
0.091
Yes
No


STX7
4.414
0.091
Yes
No


NDUFAF1
5.219
0.091
Not assigned to function by
No





Ingenuity


SLAMF8
3.465
0.091
Not assigned to function by
No





Ingenuity


PFKP
6.294
0.091
No
No


CPSF6
0.146
0.091
No
No


LSM2
8.984
0.091
Not assigned to function by
No





Ingenuity


LAP1B
0.054
0.091
Not assigned to function by
No





Ingenuity


RASA4
0.189
0.091
Not assigned to function by
No





Ingenuity


POGK
6.766
0.091
Not assigned to function by
No





Ingenuity


SSR1
0.122
0.092
Yes
No


ATP1B1
5.102
0.092
No
No


PA2G4
7.067
0.092
Yes
No


USF2
0.244
0.092
Yes
No


CASP3
14.85
0.092
Yes
No


PPIB
5.353
0.092
Yes
No


SP2
0.135
0.093
Not assigned to function by
No





Ingenuity


GRP58
15.372
0.093
Yes
No


KIAA0863
0.097
0.093
Not assigned to function by
No





Ingenuity


CD24
0.031
0.094
Yes
No


TCFL1
19.522
0.094
No
No


MCM4
5.113
0.094
Yes
No


SULT1A1
0.047
0.094
No
No


FARSLA
4.776
0.094
Not assigned to function by
No





Ingenuity


PCF11
0.197
0.094
Not assigned to function by
No





Ingenuity


TNS
0.064
0.094
No
Yes


HBP1
0.062
0.094
No
No


GGA1
0.088
0.094
Yes
No


ILVBL
31.351
0.094
Not assigned to function by
No





Ingenuity


WDR26
0.037
0.094
Not assigned to function by
No





Ingenuity


ZNF24
24.761
0.094
No
No


DYRK1A
0.134
0.094
No
No


NOLC1
5.681
0.094
Yes
No


HNRPAB
7.05
0.094
No
No


PPM1F
0.140
0.094
Yes
No


ASH2L
23.135
0.094
Not assigned to function by
No





Ingenuity


SASH1
8.308
0.095
Not assigned to function by
No





Ingenuity


RPL13
0.167
0.095
Not assigned to function by
No





Ingenuity


PFS2
3.043
0.095
Not assigned to function by
No





Ingenuity


TMEM14A
6.614
0.095
Not assigned to function by
No





Ingenuity


FLJ35827
0.121
0.095
Not assigned to function by
No





Ingenuity


REPIN1
0.114
0.096
Yes
No


EI24
14.543
0.096
Yes
No


MRPS7
9.557
0.096
Not assigned to function by
No





Ingenuity


TRIM8
0.173
0.096
Not assigned to function by
No





Ingenuity


GABPB2
6.265
0.096
Yes
No


ERP70
28.582
0.096
No
No


KIAA0763
0.150
0.096
No
No


NGX6
0.125
0.096
Not assigned to function by
No





Ingenuity


CREBL2
15.292
0.097
No
No


NEK7
9.293
0.097
Not assigned to function by
No





Ingenuity


CAMLG
0.165
0.097
No
No


CCT7
4.279
0.097
Yes
No


INSIG2
9.42
0.097
No
No


CCNI
0.063
0.097
No
Yes


DAPP1
7.907
0.098
No
No


USP21
0.207
0.098
Yes
No


RBM8A
4.954
0.098
No
No


PRKCI
6.662
0.098
Yes
No


WBP11
0.042
0.098
Not assigned to function by
No





Ingenuity


CCRL2
4.788
0.098
Not assigned to function by
No





Ingenuity


MRPL12
7.889
0.098
Yes
No


PSMB5
5.428
0.099
Not assigned to function by
No





Ingenuity


EBI3
3.985
0.099
Not assigned to function by
No





Ingenuity


BCAP31
9.401
0.099
Yes
No


ZNF297B
6.866
0.099
Not assigned to function by
No





Ingenuity


KNS2
5.27
0.099
No
No


SRD5A1
0.212
0.100
No
No


MSH2
6.541
0.100
Yes
No


ID3
0.052
0.100
Yes
No


EIF2B1
20.633
0.100
No
No


IRAK1BP1
0.191
0.100
No
No
















TABLE 34










Meningoencephalitis-associated Genes Connected to Cell Death












Odds Ratio for






association with
Meningoencephalitis

Affymetrix


Gene name
meningoencephalitis
FDR
Pathway associations
identifier














HNRPC
324.673
0.016
Cell Death pathways:IgM
214737_x_at


STAT1
230.416
0.004
Cell Death pathways:TNF
209969_s_at





superfamily, TCR, p53


PDCD8
119.631
0.019
Cell Death pathways
205512_s_at


NFATC1
80.225
0.036
Cell Death pathways
210162_s_at


STAT5A
56.762
0.065
TNF superfamily
203010_at


BRD2
56.318
0.010
Cell Death pathways:cell
208686_s_at





cycle


IL27RA
46.587
0.032
Cell Death pathways
205926_at


DUT
40.207
0.018
Cell Death pathways:IGM
208955_at


CSE1L
38.753
0.019
Cell Death pathways:TNF
201112_s_at





superfamily


GZMB
31.809
0.010
Cell Death pathways:target
210164_at





cell killing


QKI
29.983
0.049
Cell Death pathways
212263_at


TRIAD3
29.384
0.039
Cell Death pathways:TNF
218426_s_at





superfamily


CD80
26.520
0.036
Cell Death pathways:TCR
207176_s_at





costimulation


DSCR1
26.470
0.061
Cell Death pathways
208370_s_at


PAFAH1B1
18.233
0.069
Cell Death pathways
200816_s_at


TDE1
17.535
0.032
Cell Death pathways
211769_x_at


XRCC5
17.167
0.047
Cell Death pathways
208643_s_at


PRKAR1A
16.407
0.036
Cell Death pathways
200604_s_at


PDCD11
15.892
0.036
Cell Death pathways:TNF
212424_at





superfamily


GRP58
15.372
0.093
Cell Death pathways
208612_at


HTATIP2
15.356
0.036
Cell Death pathways
207180_s_at


CASP3
14.850
0.092
Cell Death pathways:TNF
202763_at


EI24
14.543
0.096
Cell Death pathways
208289_s_at


UGCG
14.445
0.013
Cell Death pathways
204881_s_at


KLRD1
13.813
0.079
Cell Death pathways
207796_x_at


RBM5
13.578
0.086
Cell Death pathways:TNF
201394_s_at





superfamily


NCOA3
13.528
0.042
Cell Death pathways
207700_s_at


BLM
13.128
0.068
Cell Death pathways:p53
205733_at


ATF1
13.031
0.083
Cell Death
222103_at





pathways:apoptosis


TRA1
12.843
0.056
Cell Death pathways
200598_s_at


ITGAV
12.837
0.039
Cell Death pathways
202351_at


BARD1
11.776
0.022
Cell Death pathways:p53
205345_at


IL2RA
11.266
0.016
Cell Death pathways
211269_s_at


SH3BP5
11.205
0.022
Cell Death pathways
201810_s_at


STMN1
11.125
0.047
Cell Death pathways
200783_s_at


NR3C1
11.050
0.036
Cell Death pathways
201865_x_at


DDX41
10.888
0.081
Cell Death pathways
217840_at


UNG
10.732
0.035
Cell Death pathways
202330_s_at


EP300
10.703
0.084
Cell Death pathways
213579_s_at


CSNK2A1
9.961
0.059
Cell Death pathways
212075_s_at


AIM2
9.506
0.049
Cell Death pathways:Cell
206513_at





death, cell proliferation


BCAP31
9.401
0.099
Cell Death pathways
200837_at


PTMA
9.390
0.062
Cell Death pathways
216384_x_at


IL9
9.187
0.041
Cell Death pathways
208193_at


PHB
8.890
0.065
Cell Death pathways
200659_s_at


IL19
8.869
0.031
Cell Death pathways:TNF
220745_at





superfamily


MBD4
8.790
0.014
Cell Death pathways
209579_s_at


PAX5
8.402
0.035
Cell Death pathways
221969_at


CTLA4
8.016
0.047
Cell Death pathways:TCR
221331_x_at





costimulation


KHDRBS1
7.898
0.060
Cell Death pathways
201488_x_at


UBD
7.847
0.044
Cell Death pathways
205890_s_at


PPBP
7.479
0.090
Cell Death pathways
214146_s_at


CYB5
7.271
0.078
Cell Death pathways
209366_x_at


VDR
7.092
0.014
Cell Death pathways
204255_s_at


CCNE1
6.880
0.044
Cell Death pathways:p53
213523_at


DNCL1
6.810
0.023
Cell Death pathways
200703_at


PRKCI
6.662
0.098
Cell Death pathways:TNF
209678_s_at





superfamily, TGF





superfamily


STAT3
6.606
0.011
Cell Death pathways:TNF
208991_at





superfamily


PDPK1
6.544
0.079
Cell Death pathways
32029_at


MSH2
6.541
0.100
Cell Death pathways
209421_at


ESPL1
6.537
0.050
Cell Death pathways
204817_at


MMP7
6.512
0.044
Cell Death pathways:TNF
204259_at





superfamily


KAI1
6.320
0.084
Cell Death pathways
203904_x_at


GABPB2
6.265
0.096
Cell Death pathways
204618_s_at


PIAS1
5.994
0.082
Cell Death pathways:TNF
217864_s_at





superfamily, TCR, p53


CDK4
5.678
0.066
Cell Death pathways:p53
202246_s_at


RRM2
5.394
0.029
Cell Death pathways
209773_s_at


NFKB1
5.354
0.066
Cell Death pathways:TNF
209239_at





superfamily


CDC2
5.354
0.081
Cell Death pathways:p53
203213_at


RAD51C
5.167
0.056
Cell Death pathways
209849_s_at


CCR1
4.928
0.067
Cell Death pathways
205099_s_at


PTPRC
4.891
0.017
Cell Death pathways
212587_s_at


TNFSF10
4.806
0.042
Cell Death pathways:TNF
202688_at





superfamily


DNAJB6
4.765
0.085
Cell Death pathways
209015_s_at


IL12B
4.425
0.067
Cell Death pathways:TGFb,
207901_at





TNF


MYLK
3.958
0.065
Cell Death pathways
202555_s_at


TYMS
3.794
0.082
Cell Death pathways
202589_at


KRAS2
3.710
0.050
Cell Death pathways:TNF
214352_s_at





superfamily


INHBA
3.484
0.077
Cell Death
210511_s_at





pathways:INHBA:TGF





superfamily check


TNF
3.471
0.067
Cell Death pathways:TNF
207113_s_at





superfamily


TNFRSF9
3.452
0.060
Cell Death pathways:TNF
207536_s_at





superfamily


IL7R
3.182
0.039
Cell Death pathways
205798_at


NRG1
0.308
0.062
Cell Death pathways
206343_s_at


DSIPI
0.237
0.064
Cell Death pathways
208763_s_at


NCOA2
0.220
0.050
Cell Death pathways
212867_at


LRDD
0.219
0.049
Cell Death pathways:TNF
219019_at





superfamily, p53


BPI
0.219
0.027
Cell Death pathways
205557_at


NR1D1
0.197
0.042
Cell Death pathways
204760_s_at


CABIN1
0.162
0.036
Cell Death pathways:TCR
37652_at


SGK
0.161
0.053
Cell Death pathways
201739_at


VIPR1
0.154
0.065
Cell Death pathways
205019_s_at


SLC18A2
0.148
0.081
Cell Death pathways
213549_at


MPO
0.146
0.063
Cell Death pathways
203949_at


PPM1F
0.140
0.094
Cell Death pathways
37384_at


NALP1
0.133
0.023
Cell Death pathways
218380_at


VEGF
0.124
0.052
Cell Death pathways
212171_x_at


NCOA1
0.124
0.077
Cell Death pathways
209105_at


RARA
0.123
0.077
Cell Death pathways:TCR
203749_s_at


SCGF
0.120
0.038
Cell Death pathways
211709_s_at


BIRC2
0.118
0.065
Cell Death
202076_at





pathways:apoptosis


NFIL3
0.116
0.036
Cell Death pathways:p53
203574_at


MOAP1
0.115
0.036
Cell Death pathways
212508_at


SMAD7
0.113
0.060
Cell Death pathways:TGFb
204790_at


ZFP36
0.108
0.029
Cell Death pathways:TNF
201531_at





superfamily


JUNB
0.108
0.032
Cell Death pathways
201473_at


PTEN
0.107
0.048
Cell Death pathways:TNF
204054_at





superfamily, p53


SPN
0.102
0.049
Cell Death pathways
206057_x_at


GORASP2
0.100
0.041
Cell Death pathways
208843_s_at


DUSP10
0.099
0.053
Cell Death pathways:p53
221563_at


SMG1
0.096
0.074
Cell Death pathways:p53
208118_x_at


FOSL2
0.091
0.079
Cell Death pathways
218881_s_at


ERCC1
0.089
0.082
Cell Death pathways
203719_at


FGR
0.088
0.048
Cell Death pathways
208438_s_at


MAP3K7IP2
0.079
0.050
Cell Death pathways
212184_s_at


PIK3CA
0.075
0.056
Cell Death pathways:TNF
204369_at





superfamily, TGF





superfamily


GNAS
0.071
0.023
Cell Death pathways
200780_x_at


IRS2
0.060
0.024
Cell Death pathways
209185_s_at


JUND
0.059
0.058
Cell Death pathways:TGF
203752_s_at





superfamily


MCL1
0.058
0.033
Cell Death pathways
200797_s_at


COL18A1
0.057
0.072
Cell Death pathways
209081_s_at


ID3
0.052
0.100
Cell Death pathways:cell
207826_s_at





cycle, apoptosis


CDC42
0.051
0.036
Cell Death pathways:p53
210232_at


TRAF6
0.049
0.062
Cell Death pathways:TNF
205558_at





superfamily


BNIP3L
0.044
0.036
Cell Death
221478_at





pathways:apoptosis


KLF2
0.038
0.010
Cell Death pathways
219371_s_at


RUNX1
0.037
0.067
Cell Death pathways
208129_x_at


BTG2
0.033
0.011
Cell Death pathways
201236_s_at


ITM2B
0.032
0.045
Cell Death pathways
217732_s_at


CD24
0.031
0.094
Cell Death pathways
266_s_at


STAT5B
0.029
0.016
:TNF superfamily
212549_at


PRKRA
0.027
0.022
Cell Death pathways:TNF
209139_s_at





superfamily


STK17B
0.025
0.010
Cell Death
205214_at





pathways:apoptosis


HIPK1
0.025
0.040
Cell Death pathways
212291_at


PTDSR
0.018
0.018
Cell Death pathways
212723_at
















TABLE 35










Selection of Genes Associated with Risk of


Meningoencephalitis and Cell Death











Cell Death


Odds



pathways
Gene
FDR
Ratio
Description














IgM
HNRPC
0.016
324.673
heterogeneous nuclear ribonucleoprotein C






(C1/C2)


IgM
DUT
0.018
40.207
dUTP pyrophosphatase


P53
BARD1
0.022
11.776
BRCA1 associated RING domain 1


P53
CDC42
0.036
0.051
cell division cycle 42 (GTP binding protein,






25 kDa)


P53
NFIL3
0.036
0.116
nuclear factor, interleukin 3 regulated


P53
CCNE1
0.044
6.880
cyclin E1


P53
DUSP10
0.053
0.099
dual specificity phosphatase 10


P53
CDK4
0.066
5.678
cyclin-dependent kinase 4


P53
BLM
0.068
13.128
Bloom syndrome


P53
SMG1
0.074
0.096
PI-3-kinase-related kinase SMG-1


P53
CDC2
0.081
5.354
cell division cycle 2, G1 to S and G2 to M


target cell
GZMB
0.010
31.809
granzyme B (cytotoxic T-lymphocyte-


killing



associated serine esterase 1)


TCR
CABIN1
0.036
0.162
calcineurin binding protein 1


TCR
CD80
0.036
26.520
CD80 antigen (CD28 antigen ligand 1, B7-1


costimulation



antigen)


TCR
CTLA4
0.047
8.016
cytotoxic T-lymphocyte-associated protein 4


costimulation


TGF
SMAD7
0.060
0.113
SMAD7


TGF
INHBA
0.077
3.484
inhibin, beta A (activin A, activin AB alpha






polypeptide)


TGF
JUND
0.058
0.059
jun D proto-oncogene


TNF
CASP3
0.092
14.850
caspase 3, apoptosis-related cysteine






protease


TNF
STAT5B,
0.016
0.029
signal transducer and activator of



3′UTR


transcription 5,3″UTR


TNF
CSE1L
0.019
38.753
CSE1 chromosome segregation 1-like






(yeast)


TNF
PRKRA
0.022
0.027
protein kinase, interferon-inducible double






stranded RNA dependent


TNF
ZFP36
0.029
0.108
zinc finger protein 36, C3H type, homolog






(mouse)


TNF
IL19
0.031
8.869
interleukin 19


TNF
PDCD11
0.036
15.892
programmed cell death 11


TNF
TRIAD3
0.039
29.384
TRIAD3 protein


TNF
TNESF10
0.042
4.806
tumor necrosis factor (ligand) superfamily,






member 10


TNF
MMP7
0.044
6.512
matrix metalloproteinase 7 (matrilysin,






uterine)


TNF
KRAS2
0.050
3.710
v-Ki-ras2 Kirsten rat sarcoma 2 viral






oncogene homolog


TNF
TNFRSF9
0.060
3.452
tumor necrosis factor receptor superfamily,






member 9


TNF
TRAF6
0.062
0.049
TNF receptor-associated factor 6


TNF
STAT5A
0.065
56.762
signal transducer and activator of






transcription 5A


TNF
NFKB1
0.066
5.354
NFKB1 (p105)


TNF
TNF
0.067
3.471
tumor necrosis factor (TNF superfamily,






member 2)


TNF
RBM5
0.086
13.578
RNA binding motif protein 5


TNF, p53
PTEN
0.048
0.107
phosphatase and tensin homolog


TNF, p53
LRDD
0.049
0.219
leucine-rich repeats and death domain






containing


TNF, p53, TCR
STAT1
0.004
230.416
signal transducer and activator of






transcription 1, 91 kDa


TNF, p53, TCR
PIAS1
0.082
5.994
protein inhibitor of activated STAT, 1


TNF, p53, TGF
STAT3
0.011
6.606
signal transducer and activator of






transcription 3


TNF, TCR
RARA
0.077
0.123
retinoic acid receptor, alpha


TNF, TGF
PIK3CA
0.056
0.075
phosphoinositide-3-kinase, catalytic, alpha






polypeptide


TNF, TGF
IL12B
0.067
4.425
interleukin 12B


TNF, TGF
PRKCI
0.098
6.662
protein kinase C, iota
















TABLE 36










Optimal Classifier of Meningoencephalitis Patients Selected by GeneCluster











Permutation-based
Permutation-based
Odds Ratio



p value < 0.01 in
p value < 0.001 in
(logistic


Gene
GeneCluster
GeneCluster
regression)













STAT3
Yes
Yes
6.61


BRD2 - bromodomain
Yes
Yes
56.32


containing 2


KIF5B - kinesin family
Yes
Yes
3.73


member 5B


LRRFIP1 - leucine rich repeat
Yes
Yes
18.56


(in FLII) interacting


RAB2 - member RAS
Yes
No
0.002


oncogene family


ZNF408 - zinc finger protein
Yes
Yes
0.239


408


BTG2 - BTG family, member 2
Yes
Yes
0.033


Stat5B 3′UTR
Yes
Yes
0.029
























TABLE 37
















Resampling









Sum
FDR



Affymetrix
Gene

Affymetrix
Gene

(absolute
estimate


Pair
identifier
symbol
Description
identifier
symbol
Description
log-odds)
(q.value)























 1
213064_at
FLJ11806
nuclear protein UKp68
221718_s_at
AKAP13
A kinase (PRKA) anchor protein
16272922
<0.0007








13


 2
213064_at
FLJ11806
nuclear protein UKp68
211962_s_at
ZFP36L1
zinc finger protein 36, C3H type-
910473
<0.0007








like 1


 3
201730_s_at
TPR
translocated promoter
209969_s_at
STAT1
signal transducer and activator of
799523
<0.0007





region (to activated


transcription 1, 91 kDa





MET oncogene)


 4
212152_x_at
ARID1A
AT rich interactive
209969_s_at
STAT1
signal transducer and activator of
743094
<0.0007





domain 1A (SWI-like)


transcription 1, 91 kDa


 5
213064_at
FLJ11806
nuclear protein UKp68
221753_at
SSH1
slingshot homolog 1 (Drosophila)
615906
<0.0007


 6
211960_s_at
RAB7
RAB7, member RAS
209969_s_at
STAT1
signal transducer and activator of
595073
<0.0007





oncogene family


transcription 1, 91 kDa


 7
213064_at
FLJ11806
nuclear protein UKp68
202469_s_at
CPSF6
cleavage and polyadenylation
519469
<0.0007








specific factor 6, 68 kDa


 8
213064_at
FLJ11806
nuclear protein UKp68
210110_x_at
HNRPH3
heterogeneous nuclear
454540
<0.0007








ribonucleoprotein H3 (2H9)


 9
208657_s_at
MSF
MLL septin-like fusion
209969_s_at
STAT1
signal transducer and activator of
409646
<0.0007








transcription 1, 91 kDa


10
213064_at
FLJ11806
nuclear protein UKp68
205281_s_at
PIGA
phosphatidylinositol glycan, class
358825
<0.0007








A (paroxysmal nocturnal








hemoglobinuria)


11
221753_at
SSH1
slingshot homolog 1
209969_s_at
STAT1
signal transducer and activator of
325766
<0.0007





(Drosophila)


transcription 1, 91 kDa


12
211960_s_at
RAB7
RAB7, member RAS
213064_at
FLJ11806
nuclear protein UKp68
307504
<0.0007





oncogene family


13
202270_at
GBP1
guanylate binding
215823_x_at
PABPC1
poly(A) binding protein, cyto-
284704
<0.0007





protein 1, interferon-


plasmic 1





inducible, 67 kDa


14
209969_s_at
STAT1
signal transducer and
201394_s_at
RBM5
RNA binding motif protein 5
281277
<0.0007





activator of transcrip-





tion 1, 91 kDa


15
203159_at
GLS
glutaminase
209969_s_at
STAT1
signal transducer and activator of
270315
<0.0007








transcription 1, 91 kDa


16
202256_at
CD2BP2
CD2 antigen (cyto-
209969_s_at
STAT1
signal transducer and activator of
257425
<0.0007





plasmic tail) binding


transcription 1, 91 kDa





protein 2


17
209484_s_at
DC8
DKFZP566O1646
202256_at
CD2BP2
CD2 antigen (cytoplasmic tail)
240944
<0.0007





protein


binding protein 2


18
214911_s_at
BRD2
bromodomain contain-
209969_s_at
STAT1
signal transducer and activator of
239410
<0.0007





ing 2


transcription 1, 91 kDa


19
205988_at
CD84
CD84 antigen (leuko-
209969_s_at
STAT1
signal transducer and activator of
215312
<0.0007





cyte antigen)


transcription 1, 91 kDa


20
200626_s_at
MATR3
matrin 3
213064_at
FLJ11806
nuclear protein UKp68
197228
<0.0007








Claims
  • 1. A method for developing a genomically guided therapeutic product for treating Alzheimer's disease (AD), the method comprising the step of compiling pharmacogenomic information to associate a unique gene expression pattern of a patient sample with a particular clinical response to a treatment for AD.
  • 2. The method of claim 1, wherein the step of compiling comprises the following steps: (1) procuring at least one patient sample from a patient of a first population of patients and at least one patient sample from a patient of a second population of patients, wherein the first population consists of one or more patients who developed the particular clinical response to the treatment for AD and wherein the second population consists of one or more patients who did not develop the particular response to the treatment for AD; (2) acquiring a gene expression pattern from each procured patient sample; and (3) determining whether at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population, wherein a determination that at least most of the patient samples procured from the first population have a unique gene expression pattern not found in at least most of the patient samples procured from the second population results in associating the unique gene expression pattern with the particular clinical response to the treatment for AD.
  • 3. The method of claim 2, wherein the particular clinical response is an adverse clinical response.
  • 4. The method of claim 3, wherein the second population of one or more patients who did not develop the adverse clinical response to the treatment also developed a favorable clinical response.
  • 5. The method of claim 4, further comprising the step of excluding patients from the first population of patients who also developed a favorable clinical response to the treatment for AD.
  • 6. The method of claim 4, further comprising, after the step of procuring and before the step of acquiring, the step of culturing the procured patient samples.
  • 7. The method of claim 6, wherein the patient samples are peripheral blood mononuclear cells.
  • 8. The method of claim 7, wherein the gene expression pattern is selected from the group consisting of protein gene expression patterns and RNA gene expression patterns.
  • 9. The method of claim 1, wherein the treatment for AD comprises administering AN1792, and wherein the step of compiling comprises defining one or more gene expression patterns associated with the development of inflammation after administration of AN1792.
  • 10. The method of claim 3, wherein the treatment for AD comprises administering AN1792.
  • 11. The method of claim 10, wherein the adverse clinical response is inflammation.
  • 12. The method of claim 11, wherein inflammation is selected from the group consisting of encephalitis, meningoencephalitis, vasculitis, cellulitis, and nephritis.
  • 13. A gene expression pattern, wherein the gene expression pattern is associated with a particular clinical response to administration of AN1792.
  • 14. The gene expression pattern of claim 13, wherein the gene expression pattern comprises a panel of genes.
  • 15. The gene expression pattern of claim 14, wherein the panel of genes comprises one or more genes selected from the group consisting of the genes listed in Tables 10, the genes listed in Table 11, the genes listed in Table 12, the genes listed in Table 18, the genes listed in Table 24, the genes listed in Table 25, the genes listed in Table 26, the genes listed in Table 27, the genes listed in Table 28, the genes listed in Table 29, the genes listed in Table 30, the genes listed in Table 31, the genes listed in Table 32, the genes listed in Table 33, the genes listed in Table 34, the genes listed in Table 35, and the genes listed in Table 36.
  • 16. The gene expression pattern of claim 14, wherein the panel of genes comprises the genes listed in Table 36.
  • 17. The gene expression pattern of claim 14, wherein the panel of genes comprises a pair of genes.
  • 18. The gene expression pattern of claim 17, wherein the panel of genes comprises a pair of genes selected from the pairs of genes listed in Table 37.
  • 19. The gene expression pattern of claim 13, wherein the particular clinical response is an adverse clinical response.
  • 20. The gene expression pattern of claim 19, wherein the adverse clinical response is inflammation.
  • 21. The gene expression pattern of claim 20, wherein the gene expression pattern is selected from the group consisting of protein gene expression patterns and RNA gene expression patterns.
  • 22. A method for treating AD comprising: (1) predicting that a candidate patient will not have an adverse clinical response to a treatment for AD; and (2) administering the treatment for AD to the candidate patient.
  • 23. The method of claim 22, wherein the step of predicting comprises determining that the candidate patient does not have a gene expression pattern associated with an adverse clinical response to the treatment for AD.
  • 24. The method of claim 22, wherein the step of predicting comprises the following steps: (1) procuring a test sample from the candidate patient; and (2) determining whether the test sample from the candidate patient has a test gene expression pattern that is substantially similar to a reference gene expression pattern associated with an adverse clinical response, wherein if it is determined that the test sample does not have a test gene expression pattern that is substantially similar to the reference gene expression pattern, it may be predicted that the candidate patient will not develop the adverse clinical response.
  • 25. The method of claim 24, wherein the step of procuring a test sample from the candidate patient comprises the following steps: (1) collecting a blood sample from the patient; (2) isolating blood cells from the sample; (3) purifying total RNA from the cells, thereby producing an RNA sample; and (4) assaying RNA expression levels from the RNA sample to obtain a test gene expression pattern.
  • 26. The method of claim 24, wherein the treatment for AD comprises administering AN1792.
  • 27. The method of claim 26, wherein the adverse clinical response is inflammation.
  • 28. The method of claim 27, wherein inflammation is selected from the group consisting of encephalitis, meningoencephalitis, vasculitis, cellulitis, and nephritis.
  • 29. The method of claim 28, wherein the reference gene expression pattern associated with the adverse clinical response comprises an expression pattern of one or more genes selected from the group consisting of the genes listed in Table 32, the genes listed in Table 33, the genes listed in Table 34, the genes listed in Table 35, the genes listed in Table 36, and the genes listed in Table 37.
  • 30. The method of claim 28, further comprising after the step of isolating and before the step of purifying, the step of culturing the cells with AN1792.
  • 31. The method of claim 30, wherein the reference gene expression pattern associated with the adverse clinical response comprises an expression pattern of one or more genes selected from the group consisting of the genes listed in Table 10, the genes listed in Table 11, and the genes listed in Table 12.
Parent Case Info

This application claims the benefit of U.S. Provisional Application Ser. No. 60/589,877, filed Jul. 20, 2004, and U.S. Provisional Application Ser. No. 60/672,716, filed Apr. 18, 2005, both of which are incorporated herein by reference in their entireties.

Provisional Applications (2)
Number Date Country
60589877 Jul 2004 US
60672716 Apr 2005 US