ASSESSMENT OF THE P13K CELLULAR SIGNALING PATHWAY ACTIVITY USING MATHEMATICAL MODELLING OF TARGET GENE EXPRESSION

The present invention relates to a method comprising inferring activity of a PI3K cellular signaling pathway in a tissue and/or cells and/or a body fluid of a medical subject based at least on expression levels of one or more target gene(s) of the PI3K cellular signaling pathway measured in an extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject. The present invention further relates to an apparatus comprising a digital processor configured to perform such a method, a non-transitory storage medium storing instructions that are executable by a digital processing device to perform such a method, and a computer program comprising program code means for causing a digital processing device to perform such a method.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO PRIOR APPLICATIONS

This application is a Continuation of U.S. application Ser. No. 15/023,820, filed Mar. 22, 2016, which is the U.S. National Phase application under 35 U.S.C. § 371 of International Application No. PCT/EP2014/079468, filed on Dec. 30, 2014, which claims the benefit of European Patent Application No. 14150145.2, filed on Jan. 3, 2014. These applications are hereby incorporated by reference herein.

FIELD OF THE INVENTION

The present invention generally relates to the field of bioinformatics, genomic processing, proteomic processing, and related arts. More particularly, the present invention relates to a method comprising inferring activity of a PI3K cellular signaling pathway in a tissue and/or cells and/or a body fluid of a medical subject based at least on expression levels of one or more target gene(s) of the PI3K cellular signaling pathway measured in an extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject. The present invention further relates to an apparatus comprising a digital processor configured to perform such a method, a non-transitory storage medium storing instructions that are executable by a digital processing device to perform such a method, and a computer program comprising program code means for causing a digital processing device to perform such a method.

BACKGROUND OF THE INVENTION

Genomic and proteomic analyses have substantial realized and potential promise for clinical application in medical fields such as oncology, where various cancers are known to be associated with specific combinations of genomic mutations/variations and/or high or low expression levels for specific genes, which play a role in growth and evolution of cancer, e.g., cell proliferation and metastasis.

For example, screening for an over-expression of the HER2 receptor on the membrane of cells in breast cancer samples is currently the standard test performed for identifying patients that are eligible to HER2 inhibitors such as Trastuzumab. Over-expression of the ERBB2 gene, which results in an over-expression of the HER2 receptor on the cell membrane, occurs in approximately 25% to 30% of all breast cancers and is associated with an increased disease recurrence and a poor prognosis. However, the expression of the HER2 receptor is by no means a conclusive indictor for driving tumor growth as the signaling initiated by the HER2 receptor can for instance be dampened by the downstream cellular signaling pathway. This also seems to be reflected in the initial response rate of 26% in HER2-positive breast cancer patients treated with Trastuzumab (Charles L. Vogel, et al., “Efficacy and Safety of Trastuzumab as a Single Agent in First-Line Treatment of HER2-Overexpressing Metastatic Breast Cancer”, Journal of Clinical Oncology, Vol. 20, No. 3, February 2002, pages 719 to 726). Besides that, the cellular signaling pathway downstream of the HER2 receptor can also be activated by mutations/over-expression in proteins downstream of the HER2 receptor, resulting in (a) relatively aggressive tumor type(s) that will not be detected by measuring HER2 expression levels. It is therefore desirable to be able to improve the possibilities of characterizing patients that have a tumor, e.g., breast cancer, which is at least partially driven by effects occurring in the cellular signaling pathway downstream of the HER2 receptor.

SUMMARY OF THE INVENTION

The present invention provides new and improved methods and apparatuses as disclosed herein.

In accordance with a main aspect of the present invention, the above problem is solved by a method for inferring activity of a PI3K cellular signaling pathway using mathematical modelling of target gene expressions, namely a method comprising:

inferring activity of a PI3K cellular signaling pathway in a tissue and/or cells and/or a body fluid of a medical subject based at least on expression levels of one or more target gene(s) of the PI3K cellular signaling pathway measured in an extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject, wherein the inferring comprises:

determining a level of a FOXO transcription factor (TF) element in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject, the FOXO TF element controlling transcription of the one or more target gene(s) of the PI3K cellular signaling pathway, the determining being based at least in part on evaluating a mathematical model relating expression levels of the one or more target gene(s) of the PI3K cellular signaling pathway to the level of the FOXO TF element;

inferring the activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject based on the determined level of the FOXO TF element in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject, wherein the inferring is performed by a digital processing device using the mathematical model.

The present invention is based on the realization of the inventors that a suitable way of identifying effects occurring in the cellular signaling pathway downstream of the HER2 receptor, herein, the PI3K cellular signaling pathway, can be based on a measurement of the signaling output of the cellular signaling pathway, which is—amongst others—the transcription of the target genes by a transcription factor (TF), herein, the FOXO TF element, controlled by the cellular signaling pathway. The PI3K cellular signaling pathway targeted herein is not only linked to breast cancer, but is known to be inappropriately activated in many types of cancer (Jeffrey A. Engelman, “Targeting PI3K signalling in cancer: opportunities, challenges and limitations”, Nature Reviews Cancer, No. 9, August 2009, pages 550 to 562). It is thought to be regulated by the RTK receptor family, which also includes the HER-family. Subsequently, the PI3K cellular signaling pathway passes on its received signal(s) via a multitude of processes, of which the two main branches are the activation of the mTOR complexes and the inactivation of a family of transcription factors often referred to as FOXO (cf. the figure showing the PI3K cellular signaling pathway in the above article from Jeffrey A. Engelman). The present invention concentrates on the PI3K cellular signaling pathway and the FOXO TF family, the activity of which is substantially negatively correlated with the activity of the PI3K cellular signaling pathway, i.e., activity of FOXO is substantially correlated with inactivity of the PI3K cellular signaling pathway, whereas inactivity of FOXO is substantially correlated with activity of the PI3K cellular signaling pathway. The present invention makes it possible to determine the activity of the PI3K cellular signaling pathway in a tissue and/or cells and/or a body fluid of a medical subject by (i) determining a level of a FOXO TF element in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject, wherein the determining is based at least in part on evaluating a mathematical model relating expression levels of one or more target gene(s) of the PI3K cellular signaling pathway, the transcription of which is controlled by the FOXO TF element, to the level of the FOXO TF element, and by (ii) inferring the activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject based on the determined level of the FOXO TF element in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject. This preferably allows improving the possibilities of characterizing patients that have a tumor, e.g., breast cancer, which is at least partially driven by a deregulated PI3K cellular signaling pathway, and that are therefore likely to respond to inhibitors of the PI3K cellular signaling pathway.

Herein, a FOXO transcription factor (TF) element is defined to be a protein complex containing at least one of the FOXO TF family members, i.e., FOXO1, FOXO3A, FOXO4 and FOXO6, which is capable of binding to specific DNA sequences, thereby controlling transcription of target genes.

The mathematical model may be a probabilistic model, preferably a Bayesian network model, based at least in part on conditional probabilities relating the FOXO TF element and expression levels of the one or more target gene(s) of the PI3K cellular signaling pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject, or the mathematical model may be based at least in part on one or more linear combination(s) of expression levels of the one or more target gene(s) of the PI3K cellular signaling pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject. In particular, the inferring of the activity of the PI3K cellular signaling pathway may be performed as disclosed in the published international patent application WO 2013/011479 A2 (“Assessment of cellular signaling pathway activity using probabilistic modeling of target gene expression”) or as described in the published international patent application WO 2014/102668 A2 (“Assessment of cellular signaling pathway activity using linear combination(s) of target gene expressions”), the contents of which are herewith incorporated in their entirety.

The medical subject may be a human or an animal. Moreover, the tissue and/or the cells and/or the body fluid of the medical subject may be from a cell line and/or a tissue culture derived from a medical subject and, if applicable, cultivated in vitro in the lab (e.g., for regenerative purposes). Furthermore, the “target gene(s)” may be “direct target genes” and/or “indirect target genes” (as described herein).

Particularly suitable target genes are described in the following text passages as well as the examples below (see, e.g., Tables 1 to 3).

Thus, according to a preferred embodiment the target gene(s) is/are selected from the group consisting of the target genes listed in Table 3.

Particularly preferred is a method wherein the inferring comprises:

inferring the activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject based at least on expression levels of one or more, preferably at least three, target gene(s) of the PI3K cellular signaling pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject selected from the group consisting of: AGRP, BCL2L11, BCL6, BNIP3, BTG1, CAT, CAV1, CCND1, CCND2, CCNG2, CDKN1A, CDKN1B, ESR1, FASLG, FBXO32, GADD45A, INSR, MXI1, NOS3, PCK1, POMC, PPARGC1A, PRDX3, RBL2, SOD2 and TNFSF10.

Further preferred is a method, wherein the inferring is further based on expression levels of at least one target gene of the PI3K cellular signaling pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject selected from the group consisting of: ATP8A1, C10orf10, CBLB, DDB1, DYRK2, ERBB3, EREG, EXT1, FGFR2, IGF1R, IGFBP1, IGFBP3, LGMN, PPM1D, SEMA3C, SEPP1, SESN1, SLC5A3, SMAD4 and TLE4.

Further preferred is a method, wherein the inferring is further based on expression levels of at least one target gene of the PI3K cellular signaling pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject selected from the group consisting of: ATG14, BIRC5, IGFBP1, KLF2, KLF4, MYOD1, PDK4, RAG1, RAG2, SESN1, SIRT1, STK11 and TXNIP.

If the inferring is further based both on expression levels of at least one target gene selected from the group specified in the preceding paragraph and on expression levels of at least one target gene selected from the group specified in the paragraph preceding the preceding paragraph, the target genes IGFBP1 and SESN1, which are mentioned above with respect to both groups, may only be contained in one of the groups.

Another aspect of the present invention relates to a method (as described herein), further comprising:

determining whether the PI3K cellular signaling pathway is operating abnormally in the tissue and/or the cells and/or the body fluid of the medical subject based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject.

The present invention also relates to a method (as described herein) further comprising:

recommending prescribing a drug for the medical subject that corrects for abnormal operation of the PI3K cellular signaling pathway, wherein the recommending is performed only if the PI3K cellular signaling pathway is determined to be operating abnormally in the tissue and/or the cells and/or the body fluid of the medical subject based on the inferred activity of the PI3K cellular signaling pathway.

The present invention also relates to a method (as described herein), wherein the inferring comprises:

inferring the activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject based at least on expression levels of two, three or more target genes of a set of target genes of the PI3K cellular signaling pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject.

Preferably,

the set of target genes of the PI3K cellular signaling pathway includes at least nine, preferably all target genes selected from the group consisting of: AGRP, BCL2L11, BCL6, BNIP3, BTG1, CAT, CAV1, CCND1, CCND2, CCNG2, CDKN1A, CDKN1B, ESR1, FASLG, FBXO32, GADD45A, INSR, MXI1, NOS3, PCK1, POMC, PPARGC1A, PRDX3, RBL2, SOD2 and TNFSF10.

A method, wherein

the set of target genes of the PI3K cellular signaling pathway further includes at least one target gene selected from the group consisting of: ATP8A1, C10orf10, CBLB, DDB1, DYRK2, ERBB3, EREG, EXT1, FGFR2, IGF1R, IGFBP1, IGFBP3, LGMN, PPM1D, SEMA3C, SEPP1, SESN1, SLC5A3, SMAD4 and TLE4,

is particularly preferred.

A method, wherein

the set of target genes of the PI3K cellular signaling pathway further includes at least one target gene selected from the group consisting of: ATG14, BIRC5, IGFBP1, KLF2, KLF4, MYOD1, PDK4, RAG1, RAG2, SESN1, SIRT1, STK11 and TXNIP,

is also particularly preferred.

If the set of target genes further includes both at least one target gene selected from the group specified in the preceding paragraph and at least one target gene selected from the group specified in the paragraph preceding the preceding paragraph, the target genes IGFBP1 and SESN1, which are mentioned above with respect to both groups, may only be contained in one of the groups.

The sample(s) to be used in accordance with the present invention can be, e.g., a sample obtained from a cancer lesion, or from a lesion suspected for cancer, or from a metastatic tumor, or from a body cavity in which fluid is present which is contaminated with cancer cells (e.g., pleural or abdominal cavity or bladder cavity), or from other body fluids containing cancer cells, and so forth, preferably via a biopsy procedure or other sample extraction procedure. The cells of which a sample is extracted may also be tumorous cells from hematologic malignancies (such as leukemia or lymphoma). In some cases, the cell sample may also be circulating tumor cells, that is, tumor cells that have entered the bloodstream and may be extracted using suitable isolation techniques, e.g., apheresis or conventional venous blood withdrawal. Aside from blood, the body fluid of which a sample is extracted may be urine, gastrointestinal contents, or an extravasate. The term “extracted sample”, as used herein, also encompasses the case where tissue and/or cells and/or body fluid of the subject have been taken from the subject and, e.g., have been put on a microscope slide, and where for performing the claimed method a portion of this sample is extracted, e.g., by means of Laser Capture Microdissection (LCM), or by scraping off the cells of interest from the slide, or by fluorescence-activated cell sorting techniques.

In accordance with another disclosed aspect, an apparatus comprises a digital processor configured to perform a method according to the present invention as described herein.

In accordance with another disclosed aspect, a non-transitory storage medium stores instructions that are executable by a digital processing device to perform a method according to the present invention as described herein. The non-transitory storage medium may be a computer-readable storage medium, such as a hard drive or other magnetic storage medium, an optical disk or other optical storage medium, a random access memory (RAM), read only memory (ROM), flash memory, or other electronic storage medium, a network server, or so forth. The digital processing device may be a handheld device (e.g., a personal data assistant or smartphone), a notebook computer, a desktop computer, a tablet computer or device, a remote network server, or so forth.

In accordance with another disclosed aspect, a computer program comprises program code means for causing a digital processing device to perform a method according to the present invention as described herein. The digital processing device may be a handheld device (e.g., a personal data assistant or smartphone), a notebook computer, a desktop computer, a tablet computer or device, a remote network server, or so forth.

The present invention as described herein can, e.g., also advantageously be used in connection with:

diagnosis based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;

prognosis based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;

drug prescription based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;

prediction of drug efficacy based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;

prediction of adverse effects based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;

monitoring of drug efficacy;

drug development;

assay development;

pathway research;

cancer staging;

enrollment of the medical subject in a clinical trial based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;

selection of subsequent test to be performed; and

selection of companion diagnostics tests.

Further advantages will be apparent to those of ordinary skill in the art upon reading and understanding the attached figures, the following description and, in particular, upon reading the detailed examples provided herein below.

It shall be understood that the method of claim 1, the apparatus of claim 13, the non-transitory storage medium of claim 15, and the computer program of claim 15 have similar and/or identical preferred embodiments, in particular, as defined in the dependent claims.

It shall be understood that a preferred embodiment of the present invention can also be any combination of the dependent claims or above embodiments with the respective independent claim.

These and other aspects of the invention will be apparent from and elucidated with reference to the embodiments described hereinafter.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 shows schematically and exemplarily a mathematical model, herein, a Bayesian network model, used to model the transcriptional program of the PI3K cellular signaling pathway.

FIG. 2 shows training results of the exemplary Bayesian network model based on (A.) the evidence curated list of target genes of the PI3K cellular signaling pathway (cf. Table 1), (B.) the database-based list of target genes of the PI3K cellular signaling pathway (cf. Table 2), and (C.) the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3).

FIG. 3 shows test results of the exemplary Bayesian network model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for breast (cancer) samples of GSE17907.

FIG. 4 shows test results of the exemplary Bayesian network model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for a number of healthy colon samples (group 1) and adenomatous polyps (group 2) published as the GSE8671 dataset.

FIG. 5 shows test results of the exemplary Bayesian network model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for colon (cancer) samples of GSE20916.

FIG. 6 shows test results of the exemplary Bayesian network model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for prostate (cancer) cells published in the GSE17951 dataset.

FIG. 7 illustrates a prognosis of ER+ breast cancer patients (GSE6532 & GSE9195) depicted in a Kaplan-Meier plot.

FIG. 8 shows training results of the exemplary linear model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3).

FIG. 9 shows test results of the exemplary linear model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for breast (cancer) samples of GSE17907.

FIG. 10 shows test results of the exemplary linear model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for prostate (cancer) samples of GSE17951.

DETAILED DESCRIPTION OF EMBODIMENTS

The following examples merely illustrate particularly preferred methods and selected aspects in connection therewith. The teaching provided therein may be used for constructing several tests and/or kits, e.g., to detect, predict and/or diagnose the abnormal activity of one or more cellular signaling pathways. Furthermore, upon using methods as described herein drug prescription can advantageously be guided, drug prediction and monitoring of drug efficacy (and/or adverse effects) can be made, drug resistance can be predicted and monitored, e.g., to select subsequent test(s) to be performed (like a companion diagnostic test). The following examples are not to be construed as limiting the scope of the present invention.

Example 1: Mathematical Model Construction

As described in detail in the published international patent application WO 2013/011479 A2 (“Assessment of cellular signaling pathway activity using probabilistic modeling of target gene expression”), by constructing a probabilistic model, e.g., a Bayesian network model, and incorporating conditional probabilistic relationships between expression levels of one or more target gene(s) of a cellular signaling pathway, herein, the PI3K cellular signaling pathway, and the level of a transcription factor (TF) element, herein, the FOXO TF element, the TF element controlling transcription of the one or more target gene(s) of the cellular signaling pathway, such a model may be used to determine the activity of the cellular signaling pathway with a high degree of accuracy. Moreover, the probabilistic model can be readily updated to incorporate additional knowledge obtained by later clinical studies, by adjusting the conditional probabilities and/or adding new nodes to the model to represent additional information sources. In this way, the probabilistic model can be updated as appropriate to embody the most recent medical knowledge.

In another easy to comprehend and interpret approach described in detail in the published international patent application WO 2014/102668 A2 (“Assessment of cellular signaling pathway activity using linear combination(s) of target gene expressions”), the activity of a cellular signaling pathway, herein, the PI3K cellular signaling pathway, may be determined by constructing and evaluating a linear or (pseudo-)linear model incorporating relationships between expression levels of one or more target gene(s) of the cellular signaling pathway and the level of a transcription factor (TF) element, herein, the FOXO TF element, the TF element controlling transcription of the one or more target gene(s) of the cellular signaling pathway, the model being based at least in part on one or more linear combination(s) of expression levels of the one or more target gene(s).

In both approaches, the expression levels of the one or more target gene(s) may preferably be measurements of the level of mRNA, which can be the result of, e.g., (RT)-PCR and microarray techniques using probes associated with the target gene(s) mRNA sequences, and of RNA-sequencing. In another embodiment the expression levels of the one or more target gene(s) can be measured by protein levels, e.g., the concentrations of the proteins encoded by the target genes.

The aforementioned expression levels may optionally be converted in many ways that might or might not suit the application better. For example, four different transformations of the expression levels, e.g., microarray-based mRNA levels, may be:

“continuous data”, i.e., expression levels as obtained after preprocessing of microarrays using well known algorithms such as MAS5.0 and fRMA,

“z-score”, i.e., continuous expression levels scaled such that the average across all samples is 0 and the standard deviation is 1,

“discrete”, i.e., every expression above a certain threshold is set to 1 and below it to 0 (e.g., the threshold for a probeset may be chosen as the median of its value in a set of a number of positive and the same number of negative clinical samples),

“fuzzy”, i.e., the continuous expression levels are converted to values between 0 and 1 using a sigmoid function of the following format: 1/(1+exp((thr−expr)/se)), with expr being the continuous expression levels, thr being the threshold as mentioned before and se being a softening parameter influencing the difference between 0 and 1.

One of the simplest linear models that can be constructed is a model having a node representing the transcription factor (TF) element, herein, the FOXO TF element, in a first layer and weighted nodes representing direct measurements of the target gene(s) expression intensity levels, e.g., by one probeset that is particularly highly correlated with the particular target gene, e.g., in microarray or (q)PCR experiments, in a second layer. The weights can be based either on calculations from a training data set or based on expert knowledge. This approach of using, in the case where possibly multiple expression levels are measured per target gene (e.g., in the case of microarray experiments, where one target gene can be measured with multiple probesets), only one expression level per target gene is particularly simple. A specific way of selecting the one expression level that is used for a particular target gene is to use the expression level from the probeset that is able to separate active and passive samples of a training data set the best. One method to determine this probeset is to perform a statistical test, e.g., the t-test, and select the probeset with the lowest p-value. The training data set's expression levels of the probe with the lowest p-value is by definition the probe with the least likely probability that the expression levels of the (known) active and passive samples overlap. Another selection method is based on odds-ratios. In such a model, one or more expression level(s) are provided for each of the one or more target gene(s) and the one or more linear combination(s) comprise a linear combination including for each of the one or more target gene(s) a weighted term, each weighted term being based on only one expression level of the one or more expression level(s) provided for the respective target gene. If the only one expression level is chosen per target gene as described above, the model may be called a “most discriminant probesets” model.

In an alternative to the “most discriminant probesets” model, it is possible, in the case where possibly multiple expression levels are measured per target gene, to make use of all the expression levels that are provided per target gene. In such a model, one or more expression level(s) are provided for each of the one or more target gene(s) and the one or more linear combination(s) comprise a linear combination of all expression levels of the one or more expression level(s) provided for the one or more target gene(s). In other words, for each of the one or more target gene(s), each of the one or more expression level(s) provided for the respective target gene may be weighted in the linear combination by its own (individual) weight. This variant may be called an “all probesets” model. It has an advantage of being relatively simple while making use of all the provided expression levels.

Both models as described above have in common that they are what may be regarded as “single-layer” models, in which the level of the TF element is calculated based on a linear combination of expression levels.

After the level of the TF element, herein, the FOXO TF element, has been determined by evaluating the respective model, the determined TF element level can be thresholded in order to infer the activity of the cellular signaling pathway, herein, the PI3K cellular signaling pathway. A method to calculate such an appropriate threshold is by comparing the determined TF element level wlc of training samples known to have a passive pathway and training samples with an active pathway. A method that does so and also takes into account the variance in these groups is given by using a threshold

thr = σ w l c pas μ w l c a c t + σ w l c a c t μ w l c pas σ w l c pas + σ w l c a c t ( 1 )

where σ and μ are the standard deviation and the mean of the training samples. In case only a small number of samples are available in the active and/or passive training samples, a pseudocount may be added to the calculated variances based on the average of the variances of the two groups:

v ~ = v w l c act + v w l c pas 2 v ~ w l c αct = x v ~ + ( n act - 1 ) v w l c act x + n act - 1 v ~ w l c pas = xv + ( n pas - 1 ) v w l c pas x + n pas - 1 ( 2 )

where ν is the variance of the groups and x a positive pseudocount. The standard deviation σ can next be obtained by taking the square root of the variance ν.

The threshold can be subtracted from the determined level of the TF element wlc for ease of interpretation, resulting in the cellular signaling pathway's activity score, such that negative values corresponds to a passive cellular signaling pathway and positive values to an active cellular signaling pathway.

As an alternative to the above-described “single-layer” models, a “two-layer” model may also be used in an example. In such a model, a summary value is calculated for every target gene using a linear combination based on the measured intensities of its associated probesets (“first (bottom) layer”). The calculated summary value is subsequently combined with the summary values of the other target genes of the cellular signaling pathway using a further linear combination (“second (upper) layer”). Again, the weights can be either learned from a training data set or based on expert knowledge or a combination thereof. Phrased differently, in the “two-layer” model, one or more expression level(s) are provided for each of the one or more target gene(s) and the one or more linear combination(s) comprise for each of the one or more target gene(s) a first linear combination of all expression levels of the one or more expression level(s) provided for the respective target gene (“first (bottom) layer”). The model is further based at least in part on a further linear combination including for each of the one or more target gene(s) a weighted term, each weighted term being based on the first linear combination for the respective target gene (“second (upper) layer”

The calculation of the summary values can, in a preferred version of the “two-layer” model, include defining a threshold for each target gene using the training data and subtracting the threshold from the calculated linear combination, yielding the target gene summary. Here the threshold may be chosen such that a negative target gene summary value corresponds to a down-regulated target gene and that a positive target gene summary value corresponds to an up-regulated target gene. Also, it is possible that the target gene summary values are transformed using, e.g., one of the above-described transformations (fuzzy, discrete, etc.), before they are combined in the “second (upper) layer”. Next the determined target genes summary values are summed to get the TF summary level.

After the level of the TF element has been determined by evaluating the “two-layer” model, the determined TF element level can be thresholded in order to infer the activity of the cellular signaling pathway, as described above.

In the following, the models described above are collectively denoted as “(pseudo-) linear” models. A more detailed description of the training and use of probabilistic models, e.g., a Bayesian network model, and of (pseudo-)linear models is provided in Example 3 below.

Example 2: Selection of Target Genes

A transcription factor (TF) is a protein complex (i.e., a combination of proteins bound together in a specific structure) or a protein that is able to regulate transcription from target genes by binding to specific DNA sequences, thereby controlling the transcription of genetic information from DNA to mRNA. The mRNA directly produced due to this action of the transcription complex is herein referred to as a “direct target gene” (of the transcription factor). Cellular signaling pathway activation may also result in more secondary gene transcription, referred to as “indirect target genes”. In the following, Bayesian network models (as exemplary mathematical models) comprising or consisting of direct target genes as direct links between cellular signaling pathway activity and mRNA level, are preferred, however the distinction between direct and indirect target genes is not always evident. Herein, a method to select direct target genes using a scoring function based on available scientific literature data is presented. Nonetheless, an accidental selection of indirect target genes cannot be ruled out due to limited information as well as biological variations and uncertainties. In order to select the target genes, two repositories of currently available scientific literature were employed to generate two lists of target genes.

The first list of target genes was generated based on scientific literature retrieved from the MEDLINE database of the National Institute of Health accessible at “www.ncbi.nlm.nih.gov/pubmed” and herein further referred to as “Pubmed”. Publications containing putative FOXO target genes were searched for by using queries such as (FOXO AND “target gene”) in the period of the first quarter of 2013. The resulting publications were further analyzed manually following the methodology described in more detail below. Specific cellular signaling pathway mRNA target genes were selected from the scientific literature, by using a ranking system in which scientific evidence for a specific target gene was given a rating, depending on the type of scientific experiments in which the evidence was accumulated. While some experimental evidence is merely suggestive of a gene being a target gene, like for example an mRNA increasing on an microarray of an cell line in which it is known that the PI3K cellular signaling axis is active, other evidence can be very strong, like the combination of an identified cellular signaling pathway TF binding site and retrieval of this site in a chromatin immunoprecipitation (ChIP) assay after stimulation of the specific cellular signaling pathway in the cell and increase in mRNA after specific stimulation of the cellular signaling pathway in a cell line.

Several types of experiments to find specific cellular signaling pathway target genes can be identified in the scientific literature:

1. ChIP experiments in which direct binding of a cellular signaling pathway-TF to its binding site on the genome is shown. Example: By using chromatin immunoprecipitation (ChIP) technology subsequently putative functional FOXO TF binding sites in the DNA of cell lines with and without active induction of the PI3K cellular signaling pathway were identified, as a subset of the binding sites recognized purely based on nucleotide sequence. Putative functionality was identified as ChIP-derived evidence that the TF was found to bind to the DNA binding site.
2. Electrophoretic Mobility Shift (EMSA) assays which show in vitro binding of a TF to a fragment of DNA containing the binding sequence. Compared to ChIP-based evidence EMSA-based evidence is less strong, since it cannot be translated to the in vivo situation.
3. Stimulation of the cellular signaling pathway and measuring mRNA profiles on a microarray or using RNA sequencing, using cellular signaling pathway-inducible cell lines and measuring mRNA profiles measured several time points after induction—in the presence of cycloheximide, which inhibits translation to protein, thus the induced mRNAs are assumed to be direct target genes.
4. Similar to 3, but using quantitative PCR to measure the amounts of mRNAs.
5. Identification of TF binding sites in the genome using a bioinformatics approach. Example for the FOXO TF element: Using the conserved FOXO binding motif 5′-TTGTTTAC-3′, a software program was run on the human genome sequence, and potential binding sites were identified, both in gene promoter regions and in other genomic regions.
6. Similar as 3, only in the absence of cycloheximide.
7. Similar to 4, only in the absence of cycloheximide.
8. mRNA expression profiling of specific tissue or cell samples of which it is known that the cellular signaling pathway is active, however in absence of the proper negative control condition.

In the simplest form one can give every potential target mRNA 1 point for each of these experimental approaches in which the target mRNA was identified.

Alternatively, points can be given incrementally, meaning one technology 1 point, a second technology adds a second point, and so on. Using this relatively ranking strategy, one can make a list of most reliable target genes.

Alternatively, ranking in another way can be used to identify the target genes that are most likely to be direct target genes, by giving a higher number of points to the technology that provides most evidence for an in vivo direct target gene, in the list above this would mean 8 points for experimental approach 1), 7 for 2), and going down to 1 point for experimental approach 8). Such a list may be called a “general target gene list”.

Despite the biological variations and uncertainties, the inventors assumed that the direct target genes are the most likely to be induced in a tissue-independent manner. A list of these target genes may be called an “evidence curated list of target genes”. Such an evidence curated list of target genes has been used to construct computational models of the PI3K cellular signaling pathway that can be applied to samples coming from different tissue sources.

The following will illustrate exemplary how the selection of an evidence curated target gene list specifically was constructed for the PI3K cellular signaling pathway.

For the purpose of selecting PI3K target genes used as input for the “model”, the following three criteria were used:

1. Gene promoter/enhancer region contains a FOXO binding motif:
a. The FOXO binding motif should be proven to respond to an activity of the PI3K cellular signaling pathway, e.g., by means of a transient transfection assay in which the specific FOXO motif is linked to a reporter gene, and
b. The presence of the FOXO motif should be confirmed by, e.g., an enriched motif analysis of the gene promoter/enhancer region.
2. FOXO (differentially) binds in vivo to the promoter/enhancer region of the gene in question, demonstrated by, e.g., a ChIP/CHIP experiment or another chromatin immunoprecipitation technique:
a. FOXO is proven to bind to the promoter/enhancer region of the gene when the PI3K cellular signaling pathway is not active, and
b. (preferably) does not bind (or weakly binds) to the gene promoter/enhancer region of the gene when the PI3K cellular signaling pathway is active.
3. The gene is differentially transcribed when the activity of the PI3K cellular signaling pathway is changed, demonstrated by, e.g.,
a. fold enrichment of the mRNA of the gene in question through real time PCR, or microarray experiment, or
b. the demonstration that RNA Pol II binds to the promoter region of the gene through an immunoprecipitation assay.

The selection was performed by defining as target genes of the PI3K cellular signaling pathway the genes for which enough and well documented experimental evidence was gathered proving that all three criteria mentioned above were met. A suitable experiment for collecting evidence of PI3K differential binding is to compare the results of, e.g., a ChIP/CHIP experiment in a cancer cell line that expresses activity of the PI3K cellular signaling pathway in response to tamoxifen (e.g., a cell line transfected with a tamoxifen-inducible FOXO construct, such as FOXO.A3.ER), when exposed or not exposed to tamoxifen. The same holds for collecting evidence of mRNA transcription.

The foregoing discusses the generic approach and a more specific example of the target gene selection procedure that has been employed to select a number of target genes based upon the evidence found using the above mentioned approach. The lists of target genes used in the Bayesian network models for the PI3K cellular signaling pathway is shown in Table 1.

TABLE 1 Evidence curated list of target genes of the PI3K cellular signaling pathway used in the Bayesian network models and associated probesets used to measure the mRNA expression level of the target genes. Target gene Probeset ATP8A1 1569773_at 210192_at 213106_at BCL2L11 1553088_a_at 1553096_s_at 1555372_at 1558143_a_at 208536_s_at 222343_at 225606_at BNIP3 201848_s_at 201849_at BTG1 1559975_at 200920_s_at 20092_s_at C10orf10 209182_s_at 209183_s_at CAT 201432_at 211922_s_at 215573_at CBLB 208348_s_at 209682_at CCND1 208711_s_at 208712_at 214019_at CCND2 200951_s_at 200952_s_at 200953_s_at 231259_s_at 1555056_at 202769_at 202770_s_at 211559_s_at CDKN1B 209112_at DDB1 208619_at DYRK2 202968_s_at 202969_at 202970_at 202971_s_at ERBB3 1563252_at 1563253_s_at 202454_s_at 215638_at 226213_at EREG 1569583_at 205767_at ESR1 205225_at 211233_x_at 211234_x_at 211235_s_at 211627_x_at 215551_at 215552_s_at 217190_x_at 207672_at EXT1 201995_at FASLG 210865_at 211333_s_at FGFR2 203638_s_at 203639_s_at 208225_at 208228_s_at 208229_at 208234_x_at 211398_at 211399_at 211400_at 211401_s_at 240913_at GADD45A 203725_at IGF1R 203627_at 203628_at 208441_at 225330_at 243358_at IGFBP1 205302_at IGFBP3 210095_s_at 212143_s_at INSR 207851_s_at 213792_s_at 226212_s_at 226216_at 226450_at LGMN 201212_at MXI1 202364_at PPM1D 204566_at 230330_at SEMA3C 203788_s_at 203789_s_at SEPP1 201427_s_at 231669_at SESN1 218346_s_at SLC5A3 1553313_s_at 212944_at 213167_s_at 213164_at SMAD4 1565702_at 1565703_at 202526_at 202527_s_at 235725_at SOD2 215078_at 215223_s_at 216841_s_at 221477_s_at TLE4 204872_at 214688_at 216997_x_at 233575_s_at 235765_at TNFSF10 202687_s_at 202688_at 214329_x_at

The second list of target genes was generated using the manually-curated database of scientific publications provided within Thomson-Reuters' Metacore (last accessed: 14th May, 2013). The database was queried for genes that are transcriptionally regulated directly downstream of the family of human FOXO transcription factors (i.e., FOXO1, FOXO3A, FOXO4 and FOXO6). This query resulted in 336 putative FOXO target genes that were further analyzed as follows. First all putative FOXO target genes that only had one supporting publication were pruned. Next a scoring function was introduced that gave a point for each type of experimental evidence, such as ChIP, EMSA, differential expression, knock down/out, luciferase gene reporter assay, sequence analysis, that was reported in a publication. The same experimental evidence is sometimes mentioned in multiple publications resulting in a corresponding number of points, e.g., two publications mentioning a ChIP finding results in twice the score that is given for a single ChIP finding. Further analysis was performed to allow only for genes that had diverse types of experimental evidence and not only one type of experimental evidence, e.g., differential expression. Finally, an evidence score was calculated for all putative FOXO target genes and all putative FOXO target genes with an evidence score of 6 or more were selected (shown in Table 2). The cut-off level of 6 was chosen heuristically as it was previously shown that approximately 30 target genes suffice largely to determine pathway activity.

A list of these target genes may be called a “database-based list of target genes”. Such a curated target gene list has been used to construct computational models that can be applied to samples coming from different tissue sources.

TABLE 2 Database-based list of target genes of the PI3K cellular signaling pathway used in the Bayesian network models and associated probesets used to measure the mRNA expression level of the target genes. Target gene Probeset AGRP 207193_at ATG14 204568_at BCL2L11 1553088_a_at 1553096_s_at 1555372_at 1558143_a_at 208536_s_at 222343_at 225606_at BCL6 203140_at 215990_s_at BIRC5 202094_at 202095_s_at 210334_x_at BNIP3 201848_s_at 201849_at CAT 201432_at 211922_s_at 215573_at CAV1 203065_s_at 212097_at CCNG2 1555056_at 202769_at 202770_s_at 211559_s_at 228081_at CDKN1A 1555186_at 202284_s_at CDKN1B 209112_at FASLG 210865_at 211333_s_at FBXO32 225801_at 225803_at 225345_s_at 225328_at GADD45A 203725_at IGFBP1 205302_at KLF2 219371_s_at 226646_at KLF4 220266_s_at 221841_s_at MYOD1 206656_s_at 206657_s_at NOS3 205581_s_at PCK1 208383_s_at PDK4 1562321_at 205960_at 225207_at POMC 205720_at PPARGC1A 1569141_a_at 219195_at PRDX3 201619_at 209766_at RAG1 1554994_at 206591_at RAG2 215117_at RBL2 212331_at 212332_at SESN1 218346_s_at SIRT1 218878_s_at SOD2 215078_at 215223_s_at 216841_s_at 221477_s_at STK11 204292_x_at 231017_at 41657_at TNFSF10 202687_s_at 202688_at 214329_x_at TXNIP 201008_s_at 201009_s_at 201010_s_at

The third list of target genes was generated on the basis of the two aforementioned lists, i.e., the evidence curated list (cf. Table 1) and the database-based list (cf. Table 2). Three criteria have been used to further select genes from these two lists. The first criterion is related to the function attributed to the target genes. Functions attributed to genes can be found in scientific literature, but are often available in public databases such as the OMIM database of the NIH (accessible via “http://www.ncbi.nlm.nih.gov/omim”). Target genes from the evidence curated list in Table 1 and the database-based list in Table 2 that were found to be attributed to be involved in processes essential to cancer, such as apoptosis, cell cycle, tumor suppression/progression, DNA repair, differentiation, were selected in the third list. Lastly, target genes that were found to have a high differential expression in cell line experiments with known high PI3K/low FOXO activity versus known low PI3K/high FOXO activity were selected. Herein, target genes that had a minimum expression difference of 20.5 (herein: on a probeset level) between the “on” and “off” state of FOXO transcription averaged over multiple samples were included in the third list. The third criterion was especially aimed at selecting the most discriminative target genes. Based on the expression levels in cell line experiments with multiple samples with known high PI3K/low FOXO activity and multiple samples with known low PI3K/high FOXO activity, an odds ratio (OR) was calculated. Herein, the odds ratio was calculated per probeset using the median value as a cut-off and a soft boundary representing uncertainty in the measurement. Target genes from the evidence curated list and the database-based list were ranked according to the “soft” odds ratio and the highest ranked (OR>2) and lowest ranked (OR<½, i.e., negatively regulated target genes) target genes were selected for the third list of target genes.

Taking into account the function of the gene, the differential expression in “on” versus “off” signaling and a higher odds ratio, a set of target genes was found (shown in Table 3) that was considered to be more probative in determining the activity of the PI3K signaling pathway. Such a list of target genes may be called a “shortlist of target genes”. Hence, the target genes reported in Table 3 are particularly preferred according to the present invention. Nonetheless, given the relative ease with which acquisition technology such as microarrays can acquire expression levels for large sets of genes, it is contemplated to utilize some or all of the target genes of Table 3, and optionally additionally use on, two, some, or all of the remaining target genes of Table 1 and Table 2.

TABLE 3 Shortlist of target genes of the PI3K cellular signaling pathway based on the evidence curated list of target genes and the database-based list of target genes. Target gene AGRP BCL2L11 BCL6 BNIP3 BTG1 CAT CAV1 CCND1 CCND2 CCNG2 CDKN1A CDKN1B ESR1 FASLG FBXO32 GADD45A INSR MXI1 NOS3 PCK1 POMC PPARGC1A PRDX3 RBL2 SOD2 TNFSF10

Example 3: Training and Using the Mathematical Model

Before the mathematical model can be used to infer the activity of the cellular signaling pathway, herein, the PI3K cellular signaling pathway, in a tissue and/or cells and/or a body fluid of a medical subject, the model must be appropriately trained.

If the mathematical model is a probabilistic model, e.g., a Bayesian network model, based at least in part on conditional probabilities relating the FOXO TF element and expression levels of the one or more target gene(s) of the PI3K cellular signaling pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject, the training may preferably be performed as described in detail in the published international patent application WO 2013/011479 A2 (“Assessment of cellular signaling pathway activity using probabilistic modeling of target gene expression”).

If the mathematical model is based at least in part on one or more linear combination(s) of expression levels of the one or more target gene(s) of the PI3K cellular signaling pathway measured in the extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject, the training may preferably be performed as described in detail in the published international patent application WO 2014/102668 A2 (“Assessment of cellular signaling pathway activity using linear combination(s) of target gene expressions”).

a) Exemplary Bayesian Network Model

Herein, an exemplary Bayesian network model as shown in FIG. 1 was first used to model the transcriptional program of the PI3K cellular signaling pathway in a simple manner. The model consists of three types of nodes: (a) a transcription factor (TF) element in a first layer 1; (b) target gene(s) TG1, TG2, TGn in a second layer 2, and, in a third layer 3; (c) measurement nodes linked to the expression levels of the target gene(s). These can be microarray probesets PS1a, PS1b, PS1c, PS2a, PSna, PSnb, as preferably used herein, but could also be other gene expression measurements such as RNAseq or RT-qPCR.

A suitable implementation of the mathematical model, herein, the exemplary Bayesian network model, is based on microarray data. The model describes (i) how the expression levels of the target gene(s) depend on the activation of the TF element, and (ii) how probeset intensities, in turn, depend on the expression levels of the respective target gene(s). For the latter, probeset intensities may be taken from fRMA pre-processed Affymetrix HG-U133Plus2.0 microarrays, which are widely available from the Gene Expression Omnibus (GEO, www.ncbi.nlm.nih.gov/geo) and ArrayExpress (www. ebi.ac.uk/arrayexpress).

As the exemplary Bayesian network model is a simplification of the biology of a cellular signaling pathway, herein, the PI3K cellular signaling pathway, and as biological measurements are typically noisy, a probabilistic approach was opted for, i.e., the relationships between (i) the TF element and the target gene(s), and (ii) the target gene(s) and their respective probesets, are described in probabilistic terms. Furthermore, it was assumed that the activity of the oncogenic cellular signaling pathway which drives tumor growth is not transiently and dynamically altered, but long term or even irreversibly altered. Therefore the exemplary Bayesian network model was developed for interpretation of a static cellular condition. For this reason complex dynamic cellular signaling pathway features were not incorporated into the model.

Once the exemplary Bayesian network model is built and calibrated (see below), the model can be used on microarray data of a new sample by entering the probeset measurements as observations in the third layer 3, and inferring backwards in the model what the probability must have been for the TF element to be “present”. Here, “present” is considered to be the phenomenon that the TF element is bound to the DNA and is controlling transcription of the cellular signaling pathway's target genes, and “absent” the case that the TF element is not controlling transcription. This latter probability is hence the primary read-out that may be used to indicate activity of the cellular signaling pathway, herein, the PI3K cellular signaling pathway, which can next be translated into the odds of the cellular signaling pathway being active by taking the ratio of the probability of being active vs. being inactive (i.e., the odds are given by p/(1−p) if p is the predicted probability of the cellular signaling pathway being active).

In the exemplary Bayesian network model, the probabilistic relations have been made quantitative to allow for a quantitative probabilistic reasoning. In order to improve the generalization behavior across tissue types, the parameters describing the probabilistic relationships between (i) the TF element and the target gene(s) have been carefully hand-picked. If the TF element is “absent”, it is most likely that the target gene is “down”, hence a probability of 0.95 is chosen for this, and a probability of 0.05 for the target gene being “up”. The latter (non-zero) probability is to account for the (rare) possibility that the target gene is regulated by other factors or accidentally observed “up” (e.g. because of measurement noise). If the TF element is “present”, then with a probability of 0.70 the target gene is considered “up”, and with a probability of 0.30 the target gene is considered “down”. The latter values are chosen this way, because there can be several reasons why a target gene is not highly expressed even though the TF element is present, for instance, because the gene's promoter region is methylated. In the case that a target gene is not up-regulated by the TF element, but down-regulated, the probabilities are chosen in a similar way, but reflecting the down-regulation upon presence of the TF element. The parameters describing the relationships between (ii) the target gene(s) and their respective probesets have been calibrated on experimental data. For the latter, in this example, microarray data was used from cell line experiments with defined active and inactive pathway settings, but this could also be performed using patient samples with known cellular signaling pathway activity status.

Herein, publically available data on the expression of a HUVEC cell line with a stable transfection of a FOXO construct that is inducible upon stimulation with 4OHT (GSE16573 available from the Gene Expression Omnibus) was used as an example. The cell lines with the inducible FOXO construct that were stimulated for 12 hours with 4OHT were considered as the FOXO active samples (n=3), whereas the passive FOXO samples were the cell lines with the construct without 4OHT stimulation (n=3).

FIG. 2 shows training results of the exemplary Bayesian network model based on (A.) the evidence curated list of target genes of the PI3K cellular signaling pathway (cf. Table 1), (B.) the database-based list of target genes of the PI3K cellular signaling pathway (cf. Table 2), and (C.) the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3). In the diagram, the vertical axis indicates the odds that the FOXO TF element is “present” resp. “absent”, which corresponds to the PI3K cellular signaling pathway being inactive resp. active, wherein values above the horizontal axis correspond to the FOXO TF element being more likely “present”/active and values below the horizontal axis indicate that the odds that the FOXO TF element is “absent”/inactive are larger than the odds that it is “present”/active.

The third group 3 of three samples encompassing the cell lines that were not stimulated with tamoxifen and that are thus FOXO inactive was assigned a passive FOXO label, whereas the fourth group 4 encompassing the samples stimulated with 4OHT, which are thus FOXO active, was assigned an active label. In the same dataset, the first, second and fifth group 1, 2, 5 were correctly predicted to have a passive PI3K cellular signaling pathway. The last group 6 consists of cell lines transfected with a mutation variant of the FOXO that is expected to be insensitive towards 4OHT stimulation. Nevertheless, some activity was found in the second model (B.) and in the third model (C.). The model based on the evidence curated list of target genes of the PI3K cellular signaling pathway correctly predicts the PI3K cellular signaling pathway to be passive in the last group 6, whereas the other two lists predicted it to be active with a relative low probability. (Legend: 1—Primary HUVECs infected with empty vector; 2—Primary HUVECs with empty vector+12 h stimulation with OHT; 3—Primary HUVECs infected with FOXO.A3.ER vector; 4—Primary HUVECs with FOXO.A3.ER vector+12 h stimulation with OHT; 5—Primary HUVECs infected with FOXO.A3.ER. H212R vector, 6—Primary HUVECs with FOXO.A3.ER.H212R vector+12 h stimulation with OHT)

In the following, test results of the exemplary Bayesian network model are shown in FIGS. 3 to 6.

FIG. 3 show test results of the exemplary Bayesian network model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for breast (cancer) samples of GSE17907. In the diagram, the vertical axis indicates the odds that the FOXO TF element is “present” resp. “absent”, which corresponds to the PI3K cellular signaling pathway being inactive resp. active, wherein values above the horizontal axis correspond to the FOXO TF element being more likely “present”/active and values below the horizontal axis indicate that the odds that the FOXO TF element is “absent”/inactive are larger than the odds that it is “present”/active. The model correctly predicts an active FOXO TF element in the normal breast samples (group 5) as it is known from the literature. The majority of the samples predicted to have a passive FOXO TF element are found in the ERBB2/HER2 subgroup (group 3), which is not unexpectedly as an over-amplification of the ERBB2 gene, which encodes for HER2, is scientifically linked to an activity of the PI3K cellular signaling pathway and, consequently, in the translocation of FOXO out of the nucleus resulting in inhibition of FOXO-regulated transcription. The breast cancer sample with the molecular subtype basal (group 2) is, as expected, predicted to have an inactive FOXO TF element, since it is known that basal breast cancers typically lack HER2 expression and are therefore not likely to have an active PI3K cellular signaling pathway. (Legend: 1—Unknown, 2—Basal, 3—ERBB2/HER2, 4—Luminal A, 5—Normal breast, 6—Normal like).

FIG. 4 shows test results of the exemplary Bayesian network model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for a number of healthy colon samples (group 1) and adenomatous polyps (group 2) published as the GSE8671 dataset. In the diagram, the vertical axis indicates the odds that the FOXO TF element is “present” resp. “absent”, which corresponds to the PI3K cellular signaling pathway being inactive resp. active, wherein values above the horizontal axis correspond to the FOXO TF element being more likely “present”/active and values below the horizontal axis indicate that the odds that the FOXO TF element is “absent”/inactive are larger than the odds that it is “present”/active. The model correctly predicts an active PI3K cellular signaling pathway in the normal samples (group 1), where the PI3K cellular signaling pathway is expected to be working normally. With respect to the adenomatous polyps (group 2), it is known from the literature that they express an increased activity of the PI3K cellular signaling pathway as a result of mutation therein. Philips and colleagues have shown that up to 86% of the colorectal tumors in their study had an increased activity of the PI3K cellular signaling pathway (Wayne A. Philips, et al., “Increased levels of phosphatidylinositol 3-kinase activity in colorectal tumors”, Cancer, Vol. 83, No. 1, July 1998, pages 41 to 47). All but three of the adenoma samples were predicted by the model as being FOXO passive, and, hence, PI3K active, which nicely correlates with the number found in the literature. (Legend: 1—Normal, 2—Adenoma).

FIG. 5 shows test results of the exemplary Bayesian network model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for colon (cancer) samples of GSE20916. In the diagram, the vertical axis indicates the odds that the FOXO TF element is “present” resp. “absent”, which corresponds to the PI3K cellular signaling pathway being inactive resp. active, wherein values above the horizontal axis correspond to the FOXO TF element being more likely “present”/active and values below the horizontal axis indicate that the odds that the FOXO TF element is “absent”/inactive are larger than the odds that it is “present”/active. The model, again, correctly predicts the normal samples to have an active FOXO TF element (groups 1 and 3), with the exception of the micro-dissected samples of the crypt epithelial cells (group 2), which likely have an active PI3K cellular signaling pathway and a passive FOXO TF element as a result of their continuous proliferation and more stem cell-like behaviour (Patrick Laprise, et al., “Phosphatidylinositol 3-kinase controls human intestinal epithelial cell differentiation by promoting adherens junction assembly and p38 MAPK activation”, Journal of Biological Chemistry, Vol. 277, No. 10, March 2002, pages 8226 to 8234). Unsurprisingly other FOXO passive samples are found in cancerous tissue (adenomas and carcinomas; groups 8 to 11). (Legend: 1—Normal colon (mucosa), 2—Normal colon (crypt), 3—Normal colon (surgery), 4—Distant normal colon (mucosa), 5—Distant normal colon (crypt), 6—Adenoma (mucosa), 7—Adenoma (crypt), 8—Adenocarcinoma (surgery), 9—Carcinoma (mucosa), 10—Carcinoma (crypt), 11—Carcinoma (surgery))

FIG. 6 shows test results of the exemplary Bayesian network model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for prostate (cancer) cells published in the GSE17951 dataset. In the diagram, the vertical axis indicates the odds that the FOXO TF element is “present” resp. “absent”, which corresponds to the PI3K cellular signaling pathway being inactive resp. active, wherein values above the horizontal axis correspond to the FOXO TF element being more likely “present”/active and values below the horizontal axis indicate that the odds that the FOXO TF element is “absent”/inactive are larger than the odds that it is “present”/active. All normal cells of the control group (group 2) are predicted to have an active FOXO TF element, whereas a small fraction of the samples in the tumour group (group 3) and the biopsy group (group 1) are predicted to have FOXO transcription silenced. In the literature, activity of the PI3K cellular signaling pathway in prostate cancer is reported (e.g., Mari Kaarbø, et al., “PI3K-AKT-mTOR pathway is dominant over androgen receptor signaling in prostate cancer cells”, Cellular Oncology, Vol. 32, No. 1-2, 2010, pages 11 to 27). (Legend: 1—Biopsy, 2—Control, 3—Tumor)

FIG. 7 illustrates a prognosis of ER+ breast cancer patients (GSE6532 & GSE9195) depicted in a Kaplan-Meier plot. In the diagram, the vertical axis indicates the recurrence free survival as a fraction of the patient group and the horizontal axis indicates a time in years. The plot indicates that an active FOXO TF element (indicated by the less steep slope of the curve that the curve ending above the other curve on the right side of the plot), which correlates with a passive PI3K cellular signaling pathway, is protective for recurrence, whereas having a passive FOXO TF element and, thus, an abnormally active PI3K cellular signaling pathway, is associated with a high risk of recurrence. (The patient group with a predicted active FOXO TF element consisted of 114 patients, whereas the patient group with a predicted passive FOXO TF element consisted of 50 patients). This result is also demonstrated in the hazard ratio of the predicted probability of FOXO transcription activity (using the probability of FOXO activity based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) as predictor): 0.45 (95% CI: 0.20-1.0, p<0.03).

b) Exemplary (Pseudo-)Linear Model

Before the (pseudo-)linear models as exemplary described herein can be used to infer pathway activity in a test sample the weights indicating the sign and magnitude of the association between the nodes and a threshold to call whether a node is either “absent” or present” need to be determined. One can use expert knowledge to fill in the weights and threshold a priori, but typically models are trained using a representative set of training samples, of which preferably the ground truth is known. E.g. expression data of probesets in samples with a known present transcription factor complex (=active pathway) or absent transcription factor complex (=passive pathway). However, it is impractical to obtain training samples from many different kinds of cancers, of which it is known what the activation status of the pathway to be modeled is. As a result, available training sets consist of a limited number of samples, typically from one type of cancer only. Herein a method is described to determine the parameters necessary to classify test samples as having an active or passive pathway.

Known in the field are a multitude of training algorithms (e.g. regression) that take into account the model topology and changes the model parameters, here weight and threshold, such that the model output, here weighted linear score, is optimized. Herein we demonstrate two exemplary methods that can be used to calculate the weights directly from the expression levels without the need of an optimization algorithm.

The first method, defined here as “black and white”-method boils down to a ternary system with the weighting factors being an element of {−1, 0, 1}. If we would put this in the biological context, the −1 and 1 corresponds to genes or probes that are down- and upregulated in case of PI3K cellular signaling pathway activity, respectively. In case a probe or gene cannot be statistically proven to be either up- or downregulated, it receives a weight of 0. Here one can use a left-sided and right-sided, two sample t-test of the expression levels of the active PI3K cellular signaling pathway samples versus the expression levels of the samples with a passive PI3K cellular signaling pathway to determine whether a probe or gene is up- or downregulated given the used training data. In cases where the average of the active samples is statistically larger than the passive samples, i.e. the p-value is below a certain threshold, e.g. 0.3, the probeset or target gene is determined to be upregulated. Conversely, in cases where the average of the active samples is statistically lower than the passive samples this probeset or target gene is determined to be downregulated upon activation of the PI3K cellular signaling pathway. In case the lowest p-value (left- or right-sided) exceeds the aforementioned threshold, the weight of this probe or gene can be defined to be 0.

An alternative method to come to weights and threshold(s) is based on the logarithm (e.g. base e) of the odds ratio, and therefore called “log odds”-weights. The odds ratio for each probe or gene is calculated based on the number of positive and negative training samples for which the probe/gene level is above and below a corresponding threshold, e.g. the median of all training samples (equation 3 in WO 2014/102668 A2). A pseudo-count can be added to circumvent divisions by zero (equation 4 in WO 2014/102668 A2). A further refinement is to count the samples above/below the threshold in a somewhat more probabilistic manner, by assuming that the probe/gene levels are e.g. normally distributed around its observed value with a certain specified standard deviation (e.g. 0.25 on a 2-log scale), and counting the probability mass above and below the threshold (equation 5 in WO 2014/102668 A2).

Alternatively, one can employ optimization algorithms known in the field such as regression to determine the weights and the threshold(s) of the (pseudo-) linear models described herein.

One has to take special attention to the way the parameters are determined for the (pseudo-)linear models to generalize well. Alternatively, one can use other machine learning methods such as Bayesian networks that are known in the field to be able to generalize quite well by taking special measures during training procedures.

With reference to FIG. 8, an exemplary “two-layer” (pseudo-)linear model of the PI3K cellular signaling pathway using all target genes from the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) and all probesets of these target genes on the first and second layer, respectively, was trained using continuous data on the expression of a HUVEC cell line with a stable transfection of a FOXO construct that is inducible upon stimulation with 4OHT (GSE16573 available from the Gene Expression Omnibus) (cf. also the above description for the exemplary Bayesian network model). The cell lines with the inducible FOXO construct that were stimulated for 12 hours with 4OHT were considered as the FOXO active samples (n=3), whereas the passive FOXO samples were the cell lines with the construct without 4OHT stimulation (n=3). The training encompassed calculating the weights of the connections between the target genes expression levels, here represented by means of probeset intensities, and the target genes nodes using the “log odds”-method with a pseudocount of 10, as described herein. Subsequently, the activity score of the FOXO TF element was calculated by summation of the calculated target genes expression scores multiplied by either 1 or −1 for upregulated or downregulated target genes, respectively.

In the diagram shown in FIG. 8, the vertical axis shows the weighted linear score, wherein a positive resp. negative score indicates that the FOXO TF element is “present” resp. “absent”, which corresponds to the PI3K cellular signaling pathway being inactive resp. active. The third group 3 of three samples encompassing the cell lines that were not stimulated with tamoxifen and that are thus FOXO inactive was assigned a passive FOXO label, whereas the fourth group 4 encompassing the samples stimulated with 4OHT, which are thus FOXO active, was assigned an active label. In the same dataset, the first, second and fifth group 1, 2, 5 were correctly predicted to have a passive PI3K cellular signaling pathway. The last group 6 consists of cell lines transfected with a mutation variant of the FOXO that is expected to be insensitive towards 4OHT stimulation. Nevertheless, some activity was also found in the sixth group using the trained (pseudo-)linear model. (Legend: 1—Primary HUVECs infected with empty vector, 2—Primary HUVECs with empty vector+12 h stimulation with OHT, 3—Primary HUVECs infected with FOXO.A3.ER vector, 4—Primary HUVECs with FOXO.A3.ER vector+12 h stimulation with OHT, 5—Primary HUVECs infected with FOXO.A3.ER. H212R vector, 6—Primary HUVECs with FOXO.A3.ER.H212R vector+12 h stimulation with OHT)

In the following, test results of the exemplary (pseudo-)linear model are shown in FIGS. 9 and 10.

FIG. 9 shows test results of the exemplary (pseudo-)linear model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for breast (cancer) samples of GSE17907. In the diagram, the vertical axis indicates the score that the FOXO TF element is “present” resp. “absent”, which corresponds to the PI3K cellular signaling pathway being inactive resp. active, wherein values above the horizontal axis correspond to the FOXO TF element being more likely “present”/active and values below the horizontal axis indicate that the odds that the FOXO TF element is “absent”/inactive are larger than the odds that it is “present”/active. The model correctly predicts an active FOXO TF element in the normal breast samples (group 5), as it is known from the literature. The majority of the samples predicted to have a passive FOXO TF element are found in the ERBB2/HER2 group (group 3), which is not unexpectedly, as an over-amplification of the ERBB2 gene, which encodes for HER2, is scientifically linked to an activity of the PI3K cellular signaling pathway and, consequently, in the translocation of FOXO out of the nucleus resulting in inhibition of FOXO-regulated transcription. (Legend: 1—Unknown, 2—Basal, 3—ERBB2/HER2, 4—Luminal A, 5—Normal breast, 6—Normal like)

FIG. 10 shows test results of the exemplary (pseudo-)linear model based on the shortlist of target genes of the PI3K cellular signaling pathway (cf. Table 3) for prostate (cancer) samples of GSE17951. In the diagram, the vertical axis indicates the score that the FOXO TF element is “present” resp. “absent”, which corresponds to the PI3K cellular signaling pathway being inactive resp. active, wherein values above the horizontal axis correspond to the FOXO TF element being more likely “present”/active and values below the horizontal axis indicate that the odds that the FOXO TF element is “absent”/inactive are larger than the odds that it is “present”/active. All normal cells of the control group (group 2) are predicted to have an active FOXO TF element, whereas a small fraction of the samples in the tumor group (group 3) and a larger fraction in the biopsy group (group 1) are predicted to have FOXO transcription silenced, corresponding to an increased activity of the PI3K cellular signaling pathway. In the literature, activity of the PI3K cellular signaling pathway in prostate cancer is reported (e.g., Mari Kaarbø, et al., “PI3K-AKT-mTOR pathway is dominant over androgen receptor signaling in prostate cancer cells”, Cellular Oncology, Vol. 32, No. 1-2, 2010, pages 11 to 27) which is confirmed in these results. (Legend: 1—Biopsy, 2—Control, 3—Tumor)

Instead of applying the mathematical model, e.g., the exemplary Bayesian network model or the (pseudo-)linear model, on mRNA input data coming from microarrays or RNA sequencing, it may be beneficial in clinical applications to develop dedicated assays to perform the sample measurements, for instance on an integrated platform using qPCR to determine mRNA levels of target genes. The RNA/DNA sequences of the disclosed target genes can then be used to determine which primers and probes to select on such a platform.

Validation of such a dedicated assay can be done by using the microarray-based mathematical model as a reference model, and verifying whether the developed assay gives similar results on a set of validation samples. Next to a dedicated assay, this can also be done to build and calibrate similar mathematical models using mRNA-sequencing data as input measurements.

The set of target genes which are found to best indicate specific pathway activity, based on microarray/RNA sequencing based investigation using the mathematical model, e.g., the exemplary Bayesian network model or the (pseudo-)linear model, can be translated into a multiplex quantitative PCR assay to be performed on an extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject and/or a computer to interpret the expression measurements and/or to infer the activity of the PI3K cellular signaling pathway. To develop such a test (e.g., FDA-approved or a CLIA waived test in a central service lab) for cellular signaling pathway activity, development of a standardized test kit is required, which needs to be clinically validated in clinical trials to obtain regulatory approval.

The present invention relates to a method comprising inferring activity of a PI3K cellular signaling pathway in a tissue and/or cells and/or a body fluid of a medical subject based at least on expression levels of one or more target gene(s) of the PI3K cellular signaling pathway measured in an extracted sample of the tissue and/or the cells and/or the body fluid of the medical subject. The present invention further relates to an apparatus comprising a digital processor configured to perform such a method, a non-transitory storage medium storing instructions that are executable by a digital processing device to perform such a method, and a computer program comprising program code means for causing a digital processing device to perform such a method.

The method may be used, for instance, in diagnosing an (abnormal) activity of the PI3K cellular signaling pathway, in prognosis based on the inferred activity of the PI3K cellular signaling pathway, in the enrollment of a medical subject in a clinical trial based on the inferred activity of the PI3K cellular signaling pathway, in the selection of subsequent test(s) to be performed, in the selection of companion diagnostics tests, in clinical decision support systems, or the like. In this regard, reference is made to the published international patent application WO 2013/011479 A2 (“Assessment of cellular signaling pathway activity using probabilistic modeling of target gene expression”) and to the published international patent application WO 2014/102668 A2 (“Assessment of cellular signaling pathway activity using linear combination(s) of target gene expressions”), which describe these applications in more detail.

SEQUENCE LISTING: Seq. No.: Gene: Seq. 1 AGRP Seq. 2 ATG14 Seq. 3 ATP8A1 Seq. 4 BCL2L11 Seq. 5 BCL6 Seq. 6 BIRC5 Seq. 7 BNIP3 Seq. 8 BTG1 Seq. 9 C10orf10 Seq. 10 CAT Seq. 11 CAV1 Seq. 12 CBLB Seq. 13 CCND1 Seq. 14 CCND2 Seq. 15 CCNG2 Seq. 16 CDKN1A Seq. 17 CDKN1B Seq. 18 DDB1 Seq. 19 DYRK2 Seq. 20 ERBB3 Seq. 21 EREG Seq. 22 ESR1 Seq. 23 EXT1 Seq. 24 FASLG Seq. 25 FBXO32 Seq. 26 FGFR2 Seq. 27 GADD45A Seq. 28 IGF1R Seq. 29 IGFBP1 Seq. 30 IGFBP3 Seq. 31 INSR Seq. 32 KLF2 Seq. 33 KLF4 Seq. 34 LGMN Seq. 35 MXI1 Seq. 36 MYOD1 Seq. 37 NOS3 Seq. 38 PCK1 Seq. 39 PDK4 Seq. 40 POMC Seq. 41 PPARGC1A Seq. 42 PPM1D Seq. 43 PRDX3 Seq. 44 RAG1 Seq. 45 RAG2 Seq. 46 RBL2 Seq. 47 SEMA3C Seq. 48 SEPP1 Seq. 49 SESN1 Seq. 50 SIRT1 Seq. 51 SLC5A3 Seq. 52 SMAD4 Seq. 53 SOD2 Seq. 54 STK11 Seq. 55 TLE4 Seq. 56 TNFSF10 Seq. 57 TXNIP

Claims

1. A kit for determining abnormal activity of a PI3K cellular signaling pathway in a subject, comprising:

one or more components for determining expression levels of three or more target genes of the PI3K cellular signaling pathway in a sample of the subject, wherein the one or more components are primers and probes for detecting the expression levels of the three or more target genes, and wherein the three or more target genes are selected from the group consisting of: AGRP, BCL2L11, BCL6, BNIP3, BTG1, CAT, CAV1, CCND1, CCND2, CCNG2, CDKN1A, CDKN1B, ESR1, FASLG, FBXO32, GADD45A, INSR, MXI1, NOS3, PCK1, POMC, PPARGC1A, PRDX3, RBL2, SOD2 and TNFSF10; and
a digital processing device configured to determine the abnormal activity of the PI3K cellular signaling pathway in the subject, wherein the inferring comprises: determining a level of a FOXO transcription factor (TF) element in the sample, the FOXO TF element controlling transcription of the three or more target genes of the PI3K cellular signaling pathway, the determining being based at least in part on evaluating a mathematical model relating expression levels of the three or more target genes of the PI3K cellular signaling pathway to the level of the FOXO TF element; inferring the activity of the PI3K cellular signaling pathway subject based on the determined level of the FOXO TF element in the sample; and determining that the PI3K cellular signaling pathway is operating abnormally in the subject based on the inferred activity of the PI3K cellular signaling pathway in the sample.

2. The kit according to claim 1, further comprising primers and probes for detecting the expression levels of an additional target gene selected from the group consisting of: ATP8A1, C10orf10, CBLB, DDB1, DYRK2, ERBB3, EREG, EXT1, FGFR2, IGF1R, IGFBP1, IGFBP3, LGMN, PPM1D, SEMA3C, SEPP1, SESN1, SLC5A3, SMAD4 and TLE4.

3. The kit according to claim 1, further comprising primers and probes for detecting the expression levels of an additional target gene selected from the group consisting of: ATG14, BIRC5, IGFBP1, KLF2, KLF4, MYOD1, PDK4, RAG1, RAG2, SESN1, SIRT1, STK11 and TXNIP.

4. The kit according to claim 1, wherein the kit comprises primers and probes for determining the expression levels of at least nine target genes selected from the group consisting of: AGRP, BCL2L11, BCL6, BNIP3, BTG1, CAT, CAV1, CCND1, CCND2, CCNG2, CDKN1A, CDKN1B, ESR1, FASLG, FBXO32, GADD45A, INSR, MXI1, NOS3, PCK1, POMC, PPARGC1A, PRDX3, RBL2, SOD2 and TNFSF10.

5. The kit according to claim 4, further comprising primers and probes for detecting the expression levels of an additional target gene selected from the group consisting of: ATP8A1, C10orf10, CBLB, DDB1, DYRK2, ERBB3, EREG, EXT1, FGFR2, IGF1R, IGFBP1, IGFBP3, LGMN, PPM1D, SEMA3C, SEPP1, SESN1, SLC5A3, SMAD4 and TLE4.

6. The kit according to claim 4, further comprising primers and probes for detecting the expression levels of an additional target gene selected from the group consisting of: ATG14, BIRC5, IGFBP1, KLF2, KLF4, MYOD1, PDK4, RAG1, RAG2, SESN1, SIRT1, STK11 and TXNIP.

7. The kit according to claim 1, wherein the kit comprises primers and probes for determining the expression levels of all target genes selected from the group consisting of: AGRP, BCL2L11, BCL6, BNIP3, BTG1, CAT, CAV1, CCND1, CCND2, CCNG2, CDKN1A, CDKN1B, ESR1, FASLG, FBXO32, GADD45A, INSR, MXI1, NOS3, PCK1, POMC, PPARGC1A, PRDX3, RBL2, SOD2 and TNFSF10.

8. The kit according to claim 1, wherein the kit is for determining the abnormal activity of the PI3K cellular signaling pathway in an extracted sample of a tissue and/or cells and/or body fluid of the subject.

9. The kit according to claim 1, wherein determining further comprises:

recommending prescribing a drug for the subject that corrects for the determined abnormal operation of the PI3K cellular signaling pathway.

10. The kit according to claim 1, wherein determining further comprises one or more of:

diagnosis based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;
prognosis based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;
drug prescription based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;
prediction of drug efficacy based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;
prediction of adverse effects based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;
monitoring of drug efficacy;
drug development;
assay development;
pathway research;
cancer staging;
enrollment of the medical subject in a clinical trial based on the inferred activity of the PI3K cellular signaling pathway in the tissue and/or the cells and/or the body fluid of the medical subject;
selection of subsequent test to be performed; and
selection of companion diagnostics tests.

11. The kit according to claim 1, wherein the determined abnormal operation of the PI3K cellular signaling pathway is overactive

12. The kit according to claim 11, wherein determining further comprises recommending prescribing a specific treatment for the subject that inhibits or deregulates the activity of the PI3K cellular signaling pathway.

13. The kit according to claim 11, wherein determining further comprises providing, based on the determined abnormal operation of the PI3K cellular signaling pathway, a recommendation for medical intervention configured to inhibit or deregulate the activity of the PI3K cellular signaling pathway.

14. The kit according to claim 13, wherein the recommendation for medical intervention configured to inhibit or deregulate the activity of the PI3K cellular signaling pathway comprises recommending or selecting a drug that inhibits or downregulates the activity of the PI3K cellular signaling pathway.

15. The kit according to claim 1, further comprising a non-transitory storage medium storing instructions that are executable by the digital processing device to determine the abnormal activity of the PI3K cellular signaling pathway in the subject.

16. The kit according to claim 1, further comprising a computer program comprising program code means for causing the digital processing device to determine the abnormal activity of the PI3K cellular signaling pathway in the subject.

Patent History
Publication number: 20220259666
Type: Application
Filed: Jan 17, 2022
Publication Date: Aug 18, 2022
Inventors: Hendrik Jan VAN OOIJEN (EINDHOVEN), Wilhelmus Franciscus Johannes VERHAEGH (EINDHOVEN), Anja VAN DE STOLPE (EINDHOVEN)
Application Number: 17/577,078
Classifications
International Classification: C12Q 1/6886 (20060101); G16B 25/00 (20060101); G16C 99/00 (20060101); G16B 25/10 (20060101);