IMMUNOGLOBULIN EXPRESSION LEVELS AS BIOMARKER FOR PROTEASOME INHIBITOR RESPONSE

Information

  • Patent Application
  • 20230235406
  • Publication Number
    20230235406
  • Date Filed
    September 06, 2022
    2 years ago
  • Date Published
    July 27, 2023
    a year ago
Abstract
Methods of treating a tumor in a subject and methods of determining a treatment regimen for a subject with a tumor are provided herein. In exemplary aspects, the methods comprise measuring the level of expression of immunoglobulin, FCGR2B, a gene listed in Table 4, or a combination thereof. In exemplary aspects, the subject is a subject from which a sample was obtained, wherein the level of immunoglobulin, FCGR2B, a gene listed in Table 4, or a combination thereof, has been measured from the sample. Related kits, computer readable-storage media, systems, and methods implemented by a processor in a computer are further provided.
Description
INCORPORATION BY REFERENCE OF MATERIAL SUBMITTED ELECTRONICALLY

Incorporated by reference in its entirety is a computer-readable nucleotide/amino acid sequence listing submitted concurrently herewith and identified as follows: 46,919,680 bytes ASCII (Text) file named “40058A_SeqListing.txt,” created on Aug. 8, 2014.


TECHNICAL FIELD

This invention relates to the fields of molecular biology and cancer treatment. In some aspects, the invention relates to determining (e.g. predicting) a tumor's (e.g., a hematological tumor) sensitivity to treatment with a proteasome inhibitor. In some other aspects, the invention relates to methods useful for diagnosing, classifying, profiling, and treating cancer.


BACKGROUND

Multiple myeloma (MM) is an incurable malignancy that originates in the antibody-secreting bone marrow plasma cells. MM comprises approximately 10% of all hematologic malignancies. The progression of the tumor is well understood, and it can be diagnosed by the presence of multiple myeloma cells in the bone marrow and monitored by the amount of antibody secretion from the clonal population of plasma cells. With conventional treatment, median survival is approximately 3 to 4 years, but the clinical course is highly variable and difficult to predict. Several therapies for MM are now approved and many more are in development, promising improved outcomes for patients with this incurable cancer. With expanding treatment options, however, comes a pressing need to pair each patient with the most efficacious and safe treatment. With the narrow therapeutic index and the toxic potential of many available cancer therapies, such differential responses potentially contribute to patients undergoing unnecessary ineffective and even potentially harmful therapy regimens. If a designed therapy could be optimized to treat individual patients, such situations could be reduced or even eliminated. Furthermore, targeted designed therapy may provide more focused, successful patient therapy overall. Therefore, there is a need to better define patient-specific treatment strategies for the use of both standard and novel therapies.


Proteasome inhibition has emerged as an important strategy in cancer treatment, including in the treatment of multiple myeloma. By way of background, proteasomes are large, multienzyme complexes that play a key role in protein breakdown. The average human cell contains about 30,000 proteasomes, each of which contains several protein-digesting proteases. The proteasome mediates the proteasomal degradation pathway which is necessary to rid cells of excess and misfolded proteins. Proteasomal complexes help regulate a whole host of functions including transcription, viral infection, oncogenesis, cell cycle, stress response, ribosome biogenesis, abnormal protein catabolism, neural and muscular degeneration, antigen processing, DNA repair, and cellular differentiation. Proteasome activity is exquisitely controlled; when it becomes either overzealous (degrading more proteins than it should) or underachieving (neglecting to degrade certain proteins) disease can develop. Proteasome inhibitors (PIs), such as carfilzomib (marketed as Kyprolis® by Onyx Pharmaceuticals) and bortezomib (marketed as Velcade® by Millennium Pharmaceuticals), have become a standard therapy across all lines of MM therapy. Carfilzomib is a tetrapeptide epoxyketone, a selective proteasome inhibitor, and is approved for the treatment of patients with multiple myeloma who have already received at least two other treatments including bortezomib and an immunomodulatory agent (e.g., lenalidomide and/or thalidomide), and whose disease has progressed on their last therapy or within 60 days of their last therapy. Despite extensive study, the mechanism of selective tumor cell death following proteasome inhibition is poorly understood. Many patients have disease that does not respond to PIs, whereas others develop resistance, suggesting the need to better define patient-specific treatment strategies for the use of PI therapies.


SUMMARY

Provided herein are methods of treating a tumor in a subject. In exemplary embodiments, the method comprises (a) measuring the level of expression of (i) immunoglobulin (Ig), including gene or gene product, (ii) Fc gamma receptor 2B (FCGR2B), including gene or gene product, or (iii) both Ig and FCGR2B, in a sample obtained from the subject, wherein the sample comprises a cell from the tumor; and (b) administering to the subject an effective amount of a proteasome inhibitor, when the level of Ig expression and/or FCGR2B expression in the sample is greater than a reference level. When the level of Ig expression and/or FCGR2B expression in the sample is less than this reference level, then the patient is administered an alternative anti-tumor therapy that is not a proteasome inhibitor. In alternative or additional embodiments, the method comprises (a) measuring the level of expression of one or more genes or gene products listed in Table 4, in a sample obtained from the subject, wherein the sample comprises a cell from the tumor; and (b) administering to the subject an effective amount of a proteasome inhibitor, when (i) the level of expression of the one or more genes listed in Table 4 in the sample is greater than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “up”, or (ii) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is less than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “down”, or (iii) both (i) and (ii).


In exemplary embodiments, the method comprises the step of administering to the subject an effective amount of a proteasome inhibitor. In exemplary aspects, the subject is a subject from which a sample was obtained and the level of expression of (i) immunoglobulin (Ig), (ii) Fc gamma receptor 2B (FCGR2B), or (iii) both Ig and FCGR2B, has been measured from the sample. In exemplary aspects, the proteasome inhibitor is administered, when the level of expression is greater than a reference level.


In alternative or additional aspects, the subject is a subject from which a sample was obtained and the level of expression of one or more genes or gene products listed in Table 4 has been measured from the sample. In exemplary aspects, the proteasome inhibitor is administered, when (i) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is greater than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “up”, or (ii) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is less than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “down”, or (iii) both (i) and (ii).


Also provided herein are methods of determining a treatment regimen for a subject with a tumor. In exemplary embodiments, the method comprises (a) measuring the level of expression of (i) immunoglobulin (Ig), (ii) Fc gamma receptor 2B (FCGR2B), or (iii) both Ig and FCGR2B, in a sample obtained from the subject, wherein the sample comprises a cell from the tumor, and (b) selecting a treatment regimen comprising administration of a proteasome inhibitor, when the level of Ig expression and/or FCGR2B expression in the sample is greater than a reference level. When the level of Ig expression and/or FCGR2B expression in the sample is less than this reference level, then the treatment regimen preferably includes administration of an alternative anti-tumor therapy that is not a proteasome inhibitor. In exemplary embodiments, the method comprises (a) measuring the level of expression of one or more genes or gene products listed in Table 4 in a sample obtained from the subject, wherein the sample comprises a cell from the tumor, and (b) selecting a treatment regimen comprising administration of a proteasome inhibitor, when (i) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is greater than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “up”, or (ii) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is less than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “down”, or (iii) both (i) and (ii). When (i) the level of expression of the one or more genes or gene products denoted in Table 4 as“up” is less than a reference level, or (ii) the level of expression of the one or more genes or gene products denoted in Table 4 as “down” is greater than a reference level, or both, then the treatment regimen preferably includes administration of an alternative anti-tumor therapy that is not a proteasome inhibitor.


Further provided herein are kits. In exemplary embodiments, the kit comprises one or more binding agents to an Ig gene or gene product, optionally an IgH, IgK or IgL gene segment or gene segment product, and a binding agent to FCGR2B gene or gene product. In exemplary embodiments, the kits comprises (i) one or more binding agents to an Ig gene or gene product, optionally an IgH, IgK or IgL gene segment or gene segment product, or a binding agent to FCGR2B gene or gene product and (ii) at least one binding agent to a gene or gene product listed in Table 4. In exemplary embodiments, the kit comprises at least a first binding agent and a second binding agent, wherein the first binding agent binds to a first gene or gene product encoded by a first gene listed in Table 4, wherein the second binding agent binds to a second gene or gene product encoded by a second gene listed in Table 4, wherein the first gene is different from the second gene.


Computer readable-storage media are furthermore provided herein. In exemplary embodiments, the computer readable storage medium is one having stored thereon a plurality of reference levels or ranges of reference levels, each reference level or range of reference levels corresponding to (i) an expression level of Ig or (ii) an expression level of FCGR2B, or (iii) an expression level of a gene listed in Table 4, or (iv) a combination thereof; and a data value that is an expression level of Ig and/or an expression level of FCGR2B and/or an expression level of a gene listed in Table 4, measured from a cell from a sample from a patient. In exemplary aspects, the the computer readable storage medium is one having stored thereon (I) a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of Ig determined from a sample obtained from a responder and each data value of the second set is an expression level of Ig determined from a sample obtained from a non-responder; (II) a receiver operating characteristic (ROC) curve based on a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of Ig determined from a sample obtained from a responder and each data value of the second set is an expression level of Ig determined from a sample obtained from a non-responder; or (III) a table listing (a) a plurality of cut-off points on the ROC curve of (II), (b) the % sensitivity associated with each cut-off point of (a), and (c) the % specificity associated with each cut-off point of (a).


In exemplary embodiments, the computer readable storage medium is one having stored thereon (I) a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of FCGR2B determined from a sample obtained from a responder and each data value of the second set is an expression level of FCGR2B determined from a sample obtained from a non-responder; (II) a receiver operating characteristic (ROC) curve based on a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of FCGR2B determined from a sample obtained from a responder and each data value of the second set is an expression level of FCGR2B determined from a sample obtained from a non-responder; or (III) a table listing (a) a plurality of cut-off points on the ROC curve of (II), (b) the % sensitivity associated with each cut-off point of (a), and (c) the % specificity associated with each cut-off point of (a).


In exemplary embodiments, the computer readable storage medium is one having stored thereon (I) a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of a gene listed in Table 4 determined from a sample obtained from a responder and each data value of the second set is an expression level of a gene listed in Table 4 determined from a sample obtained from a non-responder; (II) a receiver operating characteristic (ROC) curve based on a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of a gene listed in Table 4 determined from a sample obtained from a responder and each data value of the second set is an expression level of a gene listed in Table 4 determined from a sample obtained from a non-responder; or (III) a table listing (a) a plurality of cut-off points on the ROC curve of (II), (b) the % sensitivity associated with each cut-off point of (a), and (c) the % specificity associated with each cut-off point of (a). In exemplary aspects, the computer readable storage medium comprises two or more of the foregoing media.


In exemplary embodiments, the computer readable storage medium is one having stored thereon machine-readable instructions executable by a processor, comprising: (a) instructions for receiving a data value, α, relating to a test level of Ig expression from a sample obtained from a test subject; and (b) instructions for displaying an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the computer readable storage medium is one having stored thereon machine-readable instructions executable by a processor, comprising: (a) instructions for receiving a data value, a, relating to a test level of FCGR2B expression from a sample obtained from a test subject; and (b) instructions for displaying an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the computer readable storage medium is one having stored thereon machine-readable instructions executable by a processor, comprising: (a) instructions for receiving a data value, α, relating to a test level of expression of a gene or gene product listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “up”; and (b) instructions for displaying an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the computer readable storage medium is one having stored thereon machine-readable instructions executable by a processor, comprising: (a) instructions for receiving a data value, α, relating to a test level of expression of a gene or gene product listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “down”; and (b) instructions for displaying an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when α is less than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


The invention additionally provides systems comprising: a processor; a memory device coupled to the processor, and machine readable instructions stored on the memory device. In exemplary embodiments, the machine readable instructions, when executed by the processor, cause the processor to: (i.) receive a data value, α, relating to a test level of Ig expression from a sample obtained from a test subject; and (ii) display an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the machine readable instructions, when executed by the processor, cause the processor to: (i) receive a data value, α, relating to a test level of FCGR2B expression from a sample obtained from a test subject; and (ii) display an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the machine readable instructions, when executed by the processor, cause the processor to: (i) receive a data value, α, relating to a test level of expression of a gene or gene product listed in Table 4, wherein the change in gene expression level for the gene is denoted in Table 4 as “up”, from a sample obtained from a test subject; and (ii) display an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when a is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the machine readable instructions, when executed by the processor, cause the processor to: (i) receive a data value, α, relating to a test level of expression of a gene or gene product listed in Table 4, wherein the change in gene expression level for the gene is denoted in Table 4 as “down”, from a sample obtained from a test subject; and (ii) display an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when α is less than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


The invention further provides methods implemented by a processor in a computer. In exemplary embodiments, the method comprises the steps of: (a) receiving a data value, α, relating to a test level of Ig expression from a sample obtained from a test subject; and (b) displaying an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve. In exemplary embodiments, the method comprises the steps of: (a) receiving a data value, α, relating to a test level of FCGR2B expression from a sample obtained from a test subject; and (b) displaying an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve. In exemplary embodiments, the method comprises the steps of: (a) receiving a data value, α, relating to a test level of expression of a gene or gene product listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “up”; and (b) displaying an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve. In exemplary embodiments, the method comprises the steps of: (a) receiving a data value, α, relating to a test level of expression of a gene or gene product listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “down”; and (b) displaying an output relating to treating a patient for multiple myeloma with a proteasome inhibitor, when α is less than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.





BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A and 1B are graphs demonstrating the association between IG expression and bortezomib response. IE, response not evaluable; PD, progressive disease; no change, NC; minimal response, MR; partial response, PR; complete response, CR.



FIGS. 2A and 2B are graphs demonstrating the lack of association between IG expression and dexamethasone response. IE, response not evaluable; PD, progressive disease; no change, NC; minimal response, MR; partial response, PR; complete response, CR.



FIGS. 3A and 3B are graphs demonstrating the association between IGH expression and carfilzomib response. PD, progressive disease; stable disease, SD; minimal response, MR; partial response, PR; very good partial response, VGPR.



FIG. 4 is a graph demonstrating the time-to-progression (TTP) for IGH-High and IGH-Low patients treated with carfilzomib.



FIG. 5 is graph demonstrating the expression level of IG genes from multiple myeloma cell line (U266) continuously exposed to either bortezomib (BTZ) or carfilzomib (CFZ) for 24 hours.



FIG. 6 is graphical representation of the transcriptional profiling data of tumors collected from patients subsequently treated with carfilzomib. Highlighted is a cluster of genes encoding proteins of the Ig structural superfamily, which includes FCGR2B



FIGS. 7A and 7B are graphs demonstrating the association between FCGR2b expression and carfilzomib response.



FIG. 8 is a is a graph demonstrating the time-to-progression (TTP) for IGH+FCGR2B positive an IGH+FCGR2G negative patients treated with carfilzomib.



FIG. 9 is a graph containing three receiver operating characteristic (ROC) curves, each curve plotting cutoff values corresponding to a % specificity and % sensitivity: IGH, FCGR2B, and the combination of IGH and FCGR2B.



FIG. 10 is a schematic of an exemplary embodiment 101 of a system 100 for determining a therapeutic regimen for a subject with a tumor.



FIG. 11A is a graph of the immunoglobulin expression (ng IgG protein per μg total cellular protein) by cells of Line A or Line B, as measured by ELISA. SN, supernatant of cell culture; L, cell lysate.



FIG. 11B is a graph of the viability of cells of Line A and Line B upon treatment with carfilzomib (CFZ). Viability is expressed as % viable cells relative to untreated control.



FIG. 12 is a graph of the amount of FCGR2B RNA as measured by qPCR. The FCGR2B RNA amount is expressed as normalized threshold cycle (Ct).





DETAILED DESCRIPTION

Methods of Treating a Tumor


The invention provides methods of treating a tumor in a subject. In exemplary embodiments, the method comprises (a) measuring the level of expression of (i) immunoglobulin (Ig), including gene or gene product, (ii) Fc gamma receptor 2B (FCGR2B), including gene or gene product or (iii) both Ig and FCGR2B, in a sample obtained from the subject, wherein the sample comprises a cell from the tumor; and (b) administering to the subject an effective amount of a proteasome inhibitor when the level of Ig and/or FCGR2B expression in the sample is greater than a reference level.


In alternative or additional embodiments, the method comprises (a) measuring the level of expression of one or more genes or gene products listed in Table 4, in a sample obtained from the subject, wherein the sample comprises a cell from the tumor; and (b) administering to the subject an effective amount of a proteasome inhibitor, when (i) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is greater than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “up”, or (ii) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is less than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “down”, or (iii) both (i) and (ii).


The invention additional provides methods of treating a tumor in a subject, wherein the method comprises the step of administering to the subject an effective amount of a proteasome inhibitor and the subject is a subject from which a sample was obtained and the level of expression of (i) immunoglobulin (Ig), (ii) Fc gamma receptor 2B (FCGR2B), or (iii) both Ig and FCGR2B, has been measured from the sample. In exemplary aspects, the proteasome inhibitor is administered, when the level of expression is greater than a reference level.


In alternative or additional embodiments, the method of treating a tumor in a subject comprises the step of administering to the subject an effective amount of a proteasome inhibitor and the subject is a subject from which a sample was obtained and the level of expression of one or more genes or gene products listed in Table 4 has been measured from the sample. In exemplary aspects, the proteasome inhibitor is administered, when (i) the level of expression of the one or more genes listed in Table 4 in the sample is greater than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “up”, or (ii) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is less than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “down”, or (iii) both (i) and (ii).


Methods of Determining a Treatment Regimen for a Subject with a Tumor


Also provided herein are methods of determining a treatment regimen for a subject with a tumor. In exemplary embodiments, the method comprises (a) measuring the level of expression of (i) immunoglobulin (Ig), (ii) Fc gamma receptor 2B (FCGR2B), or (iii) both Ig and FCGR2B, in a sample obtained from the subject, wherein the sample comprises a cell from the tumor, and (b) selecting a treatment regimen comprising administration of a proteasome inhibitor, when the level of Ig expression and/or FCGR2B expression in the sample is greater than a reference level. In additional or alternative embodiments, the method of determining a treatment regimen for a subject with a tumor comprises (a) measuring the level of expression of one or more genes or gene products listed in Table 4 in a sample obtained from the subject, wherein the sample comprises a cell from the tumor, and (b) selecting a treatment regimen comprising administration of a proteasome inhibitor, when (i) the level of expression of the one or more genes listed in Table 4 in the sample is greater than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “up”, or (ii) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is less than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “down”, or (iii) both (i) and (ii).


Measurement of Expression Levels


In the methods of the invention, gene expression level(s) or gene segment expression level(s) is/are measured in a sample obtained from the subject. In exemplary aspects, the method comprises measuring the level, concentration, or amount of RNA, e.g., mRNA, encoded by the gene or gene segments in the sample. Levels of RNA, e.g., mRNA, may be measured by any technique known in the art, including but not limited to northern blotting or quantitative PCR (qPCR), including methods such as reverse transcription qPCR, real time qPCR, and end-point qPCR. Alternatively, “tag based” technologies, such as Serial analysis of gene expression (SAGE) and RNA-Seq, may be carried out to provide a relative measure of the cellular concentration of different mRNAs. Exemplary methods of measuring levels of RNA (e.g., mRNA) are also described herein at Example 5.


In alternative or additional aspects, the method comprises measuring the level, concentration, or amount of the protein product encoded by the gene or gene segments in the sample. Suitable methods of determining expression levels of protein products are known in the art and include immunoassays (e.g., Western blotting, an enzyme-linked immunosorbent assay (ELISA), a radioimmunoassay (MA), and immunohistochemical assay. See, e.g., U.S. Patent Application Publication No. 2007/0212723 A1, Shang et al., Circulation Research 101: 1146-1154 (2007); and International Patent Application Publication Nos. WO/2012/094651 and WO/2010/129964. Exemplary methods of determining expression levels of protein products are also described herein at Example 6.


In alternative or additional aspects, the level of protein product is represented by a level of the protein product's biological activity, e.g., enzymatic activity. In exemplary aspects, the protein level is reflected by the levels of the substrate or product of the enzymatic reaction catalyzed by the protein product. Methods of assaying for the level of biological activity, e.g., enzymatic activity, are known in the art.


In some aspects, the level of the protein product is represented by the level of biological activity of a related protein, e.g., a protein which acts upstream or downstream of the protein product. For example, if the protein product is a phosphorylated protein in the active state, then, in some embodiments, the level of the protein product may be represented by the activity level of the kinase which phosphorylates the protein product. In other aspects, if the protein product is a transcription factor which activates expression of a gene, then, in some embodiments, the level of the protein product may be represented by the expression levels of the gene activated by the protein product.


In exemplary aspects, the expression level that is measured, may be normalized or calibrated to a level of a housekeeping gene. The housekeeping gene in some aspects is β-actin or GAPDH. In exemplary aspects, the housekeeping gene is any one of those set forth in the table below.

















HGNC Gene



Ensemble Gene ID
ID









ENSG00000097007
ABL1



ENSG00000218739
AC007390.5



ENSG00000132842
AP3B1



ENSG00000065802
ASBI



ENSG00000108591
DRG2



ENSG00000181090
EHMT1



ENSG00000108262
GIT1



ENSG00000089022
MAPKAPK5



ENSG00000007047
MARK4



ENSG00000075975
MKRN2



ENSG00000198646
NCOA6



ENSG00000126653
NSRP1



ENSG00000107960
OBFC1



ENSG00000175470
PPP2R2D



ENSG00000269277
PPP2R2D



ENSG00000113456
RAD1



ENSG00000107185
RGP1



ENSG00000173456
RNF26



ENSG00000147164
SNX12



ENSG00000167182
SP2



ENSG00000110066
SUV420H1



ENSG00000149930
TAOK2



ENSG00000107021
TBC1D13



ENSG00000068354
TBC1D25



ENSG00000269499
TBC1D25



ENSG00000103671
TRIP4



ENSG00000130939
UBE4B



ENSG00000170473
WIBG



ENSG00000073050
XRCC1



ENSG00000121766
ZCCHC17



ENSG00000023041
ZDHHC6










In exemplary aspects, the expression level that is measured, is not normalized or calibrated to a level of a housekeeping gene. In exemplary aspects, the expression level that is measured may represent an average expression level or a mean expression level based on more than one measurement of the expression level. In exemplary aspects, the measured expression level is an average or mean of several measurements of expression levels of the same sample. In exemplary aspects, the measured expression level is an average or mean of several measurements of expression levels of different samples containing the same components obtained from the same subject. In exemplary aspects, the measured expression level is quantile normalized, as is done in RNA Seq techniques.


Immunoglobulin (Ig)


In exemplary aspects, the sample obtained from the subject is measured for the expression level of immunoglobulins. Ig molecules comprise heavy chains and light chains, each of which have a constant region and a variable region. The genes encoding the segments of an Ig molecule are located at three loci within the human genome: the Ig heavy (IGH) locus, the Ig kappa (IGK) locus, and the Ig lambda (IGL) locus. The IGH locus is located on chromosome 14 (at ch. 14q32.33) and contains gene segments encoding Ig heavy chains. The IGK locus is located on chromosome 2 (at ch. 2p11.2) and contains gene segments encoding Ig light chains. The IGL locus is located on chromosome 22 (at ch. 22q11.2) and contains gene segments for Ig light chains. Each heavy chain and light chain gene contains multiple copies of different types of gene segments for the variable regions of the Ig molecule. For example, the immunoglobulin heavy chain region contains 44 Variable (V) gene segments[Matsuda et al., J Expmtal Med 188: 2151-2162 (1998)] plus 27 Diversity (D) gene segments and 6 Joining (J) gene segments.[Li et al., Blood 103: 4602-4609 (2004)]. Likewise, the light chain region possesses numerous V and J gene segments. However, light chain genes do not have D gene segments. DNA rearrangement causes one copy of each type of gene segment to be selected in any given lymphocyte, generating an enormous antibody repertoire; roughly 3×1011 combinations are possible.


With regard to the inventive methods, when the sample obtained from the subject is measured for the expression level of immunoglobulins, the method may comprise measuring the expression level of any gene segment of the IGH locus, the IGK locus, or the IGL locus. In alternative or additional aspects, the method may comprise measuring the expression level of any IGH orphon gene segment which is not located at the IGH locus, any IGK orphon gene segment which is not located at the IGK locus, or any IGL orphon gene segment which is not located at the IGL locus. In exemplary aspects, the method comprises measuring the level of expression of one or more gene segments of the IGH locus, IGK locus, or IGL locus, or any IGH orphon gene segment, IGK orphon gene segment, or IGL orhon gene segment. In exemplary aspects, the method comprises measuring the level of expression of a combination of gene segments of at least two of the IGH locus, IGK locus, and IGL locus, or an orphon gene segment thereof. In exemplary aspects, the method comprises measuring the level of expression of a combination gene segments at each of the IGH locus, IGK locus, and IGL locus or at each of the IGH and IGK loci or at each of the IGH and IGL loci, or at each of the IGK and IGL loci. In alternative or additional aspects, the method comprises measuring the level of expression of a combination of IGH orphon gene segment(s), IGK orphon gene segment(s), and IGL orphon gene segment(s) or a combination of IGH orphon gene segments and IGK orphon gene segments or a combination of IGH orphon gene segments and IGL orphon gene segments or a combination of IGK orphon gene segments and IGL orphon gene segments.


In exemplary aspects, the method comprises measuring the level of expression of one or more gene segments at the IGH locus. In exemplary aspects, the one or more gene segments is selected from the group consisting of: IGHA1, IGHA2, IGHD, IGHD1-1, IGHD1-14, IGHD1-20, IGHD1-26, IGHD1-7, IGHD2-15, IGHD2-2, IGHD2-21, IGHD2-8, IGHD3-10, IGHD3-16, IGHD3-22, IGHD3-3, IGHD3-9, IGHD4-11, IGHD4-17, IGHD4-23, IGHD4-4, IGHD5-12, IGHD5-18, IGHD5-24, IGHD5-5, IGHD6-13, IGHD6-19, IGHD6-25, IGHD6-6, IGHD7-27, IGHE, IGHEP1, IGHEP2, IGHG1, IGHG2, IGHG3, IGHG4, IGHGP, IGHJ1, IGHJ1P, IGHJ2, IGHJ2P, IGHJ3, IGHJ3P, IGHJ4, IGHJ5, IGHJ6, IGHM, IGHMBP2, IGHV1-12, IGHV1-14, IGHV1-17, IGHV1-18, IGHV1-2, IGHV1-24, IGHV1-3, IGHV1-45, IGHV1-46, IGHV1-58, IGHV1-67, IGHV1-68, IGHV1-69, IGHV1-8, IGHV1OR21-1, IGHV2-10, IGHV2-26, IGHV2-5, IGHV2-70, IGHV2OR16-5, IGHV3-11, IGHV3-13, IGHV3-15, IGHV3-16, IGHV3-19, IGHV3-20, IGHV3-21, IGHV3-22, IGHV3-23, IGHV3-25, IGHV3-29, IGHV3-30, IGHV3-30-2, IGHV3-32, IGHV3-33, IGHV3-33-2, IGHV3-35, IGHV3-36, IGHV3-37, IGHV3-38, IGHV3-41, IGHV3-42, IGHV3-43, IGHV3-47, IGHV3-48, IGHV3-49, IGHV3-50, IGHV3-52, IGHV3-53, IGHV3-54, IGHV3-57, IGHV3-6, IGHV3-60, IGHV3-62, IGHV3-63, IGHV3-64, IGHV3-65, IGHV3-66, IGHV3-7, IGHV3-71, IGHV3-72, IGHV3-73, IGHV3-74, IGHV3-75, IGHV3-76, IGHV3-79, IGHV3-9, IGHV3OR16-8, IGHV4-28, IGHV4-31, IGHV4-34, IGHV4-39, IGHV4-4, IGHV4-55, IGHV4-59, IGHV4-61, IGHV4-80, IGHV5-51, IGHV5-78, IGHV6-1, IGHV7-27, IGHV7-34-1, IGHV7-40, IGHV7-56, IGHV7-81, IGHVII-1-1, IGHVII-15-1, IGHVII-20-1, IGHVII-22-1, IGHVII-26-2, IGHVII-28-1, IGHVII-30-1, IGHVII-31-1, IGHVII-33-1, IGHVII-40-1, IGHVII-43-1, IGHVII-44-2, IGHVII-46-1, IGHVII-49-1, IGHVII-51-2, IGHVII-53-1, IGHVII-60-1, IGHVII-62-1, IGHVII-65-1, IGHVII-67-1, IGHVII-74-1, IGHVII-78-1, IGHVIII-11-1, IGHVIII-13-1, IGHVIII-16-1, IGHVIII-2-1, IGHVIII-22-2, IGHVIII-25-1, IGHVIII-26-1, IGHVIII-38-1, IGHVIII-44, IGHVIII-47-1, IGHVIII-5-1, IGHVIII-51-1, IGHVIII-5-2, IGHVIII-67-2, IGHVIII-67-3, IGHVIII-67-4, IGHVIII-76-1, IGHVIII-82, and IGHVIV-44-1. In exemplary aspects, the one or more gene segments at the IgH locus comprises a sequence selected from the group consisting of SEQ ID NOs: 1-174. In exemplary aspects, the one or more gene segments at the IGH locus is one listed in the following table.





















Approved
Approved
Previous


Accession


HGNC ID
Symbol
Name
Symbols
Synonyms
Chromosome
Numbers





HGNC: 5487
IGHD1OR15-1A
immunoglobulin

IGHD1/OR15-1A,
15q11.2
X55575




heavy diversity

IGHD1OR151A




1/OR15-1A




(non-functional)


HGNC: 5488
IGHD1OR15-1B
immunoglobulin

IGHD1/OR15-1B,
15q11.2
X55576




heavy diversity

IGHD1OR151B




1/OR15-1B




(non-functional)


HGNC: 5493
IGHD2OR15-2A
immunoglobulin

IGHD2/OR15-2A,
15q11.2
X55577




heavy diversity

IGHD2OR152A




2/OR15-2A




(non-functional)


HGNC: 5494
IGHD2OR15-2B
immunoglobulin

IGHD2/OR15-2B,
15q11.2
X55578




heavy diversity

IGHD2OR152B




2/OR15-2B




(non-functional)


HGNC: 5500
IGHD3OR15-3A
immunoglobulin

IGHD3/OR15-3A,
15q11.2
X55579




heavy diversity

IGHD3OR153A




3/OR15-3A




(non-functional)


HGNC: 5501
IGHD3OR15-3B
immunoglobulin

IGHD3/OR15-3B,
15q11.2
X55580




heavy diversity

IGHD3OR153B




3/OR15-3B




(non-functional)


HGNC: 5506
IGHD4OR15-4A
immunoglobulin

IGHD4/OR15-4A,
15q11.2
X55581




heavy diversity

IGHD4OR154A




4/OR15-4A




(non-functional)


HGNC: 5507
IGHD4OR15-4B
immunoglobulin

IGHD4/OR15-4B,
15q11.2
X55582




heavy diversity

IGHD4OR154B




4/OR15-4B




(non-functional)


HGNC: 5512
IGHD5OR15-5A
immunoglobulin

IGHD5/OR15-5A,
15q11.2
X55583




heavy diversity

IGHD5OR155A




5/OR15-5A




(non-functional)


HGNC: 5513
IGHD5OR15-5B
immunoglobulin

IGHD5/OR15-5B,
15q11.2
X55584




heavy diversity

IGHD5OR155B




5/OR15-5B




(non-functional)


HGNC: 5524
IGHEP2
immunoglobulin


9p24.1
K01241




heavy constant




epsilon P2




(pseudogene)


HGNC: 5563
IGHV1OR15-1
immunoglobulin

IGHV1/OR15-1
15q11.2
Z29631




heavy variable




1/OR15-1




(non-functional)


HGNC: 5564
IGHV1OR15-2
immunoglobulin

IGHV1/OR15-2
15q11.1
L25543




heavy variable




1/OR15-2




(pseudogene)


HGNC: 5565
IGHV1OR15-3
immunoglobulin

IGHV1/OR15-3
15q11.2
Z29595




heavy variable




1/OR15-3




(pseudogene)


HGNC: 5566
IGHV1OR15-4
immunoglobulin

IGHV1/OR15-4
15q11.2
Z29596




heavy variable




1/OR15-4




(pseudogene)


HGNC: 5567
IGHV1OR15-5
immunoglobulin

IGHV1/OR15-5
15q11.2
Z29633




heavy variable




1/OR15-5




(non-functional)


HGNC: 5568
IGHV1OR15-6
immunoglobulin

IGHV1/OR15-6
15q11.2
Z29634




heavy variable




1/OR15-6




(pseudogene)


HGNC: 5569
IGHV1OR15-9
immunoglobulin
VSIG7
IGHV1/OR15-9,
15q11.1
L25542




heavy variable

IGHV1OR159




1/OR15-9




(non-functional)


HGNC: 5570
IGHV1OR16-1
immunoglobulin

IGHV1/OR16-1
16p11.2
Z29599




heavy variable




1/OR16-1




(pseudogene)


HGNC: 5571
IGHV1OR16-2
immunoglobulin

IGHV1/OR16-2
16p11.2
Z29600




heavy variable




1/OR16-2




(pseudogene)


HGNC: 5572
IGHV1OR16-3
immunoglobulin

IGHV1/OR16-3
16p11.2
Z29639




heavy variable




1/OR16-3




(pseudogene)


HGNC: 5573
IGHV1OR16-4
immunoglobulin

IGHV1/OR16-4
16p11.2
Z17397




heavy variable




1/OR16-4




(pseudogene)


HGNC: 38040
IGHV1OR21-1
immunoglobulin

IGHV1/OR21-1
21p11.2




heavy variable




1/OR21-1




(non-functional)


HGNC: 5579
IGHV2OR16-5
immunoglobulin

IGHV2/OR16-5
16p11.2
L25544




heavy variable




2/OR16-5




(non-functional)


HGNC: 5633
IGHV3OR15-7
immunoglobulin

IGHV3/OR15-7
15q11.2
Z29597




heavy variable




3/OR15-7




(pseudogene)


HGNC: 5641
IGHV3OR16-6
immunoglobulin

IGHV3/OR16-6
16p11.2
L25545




heavy variable




3/OR16-6




(pseudogene)


HGNC: 5642
IGHV3OR16-7
immunoglobulin

IGHV3/OR16-7
16p11.2
Z29604




heavy variable




3/OR16-7




(pseudogene)


HGNC: 5643
IGHV3OR16-8
immunoglobulin

IGHV3/OR16-8
16p11.2
Z29605




heavy variable




3/OR16-8




(non-functional)


HGNC: 5644
IGHV3OR16-9
immunoglobulin

IGHV3/OR16-9
16p11.2
Z29606




heavy variable




3/OR16-9




(non-functional)


HGNC: 5634
IGHV3OR16-10
immunoglobulin

IGHV3/OR16-10
16p11.2
Z29607




heavy variable




3/OR16-10




(non-functional)


HGNC: 5635
IGHV3OR16-11
immunoglobulin

IGHV3/OR16-11
16p11.2
Z29608




heavy variable




3/OR16-11




(pseudogene)


HGNC: 5636
IGHV3OR16-12
immunoglobulin

IGHV3/OR16-12
16p11.2
Z29609




heavy variable




3/OR16-12




(non-functional)


HGNC: 5637
IGHV3OR16-13
immunoglobulin

IGHV3/OR16-13
16p11.2
Z29610




heavy variable




3/OR16-13




(non-functional)


HGNC: 5638
IGHV3OR16-14
immunoglobulin

IGHV3/OR16-14
16p11.2
Z29611




heavy variable




3/OR16-14




(pseudogene)


HGNC: 5639
IGHV3OR16-15
immunoglobulin

IGHV3/OR16-15
16p11.2
L25546




heavy variable




3/OR16-15




(pseudogene)


HGNC: 5640
IGHV3OR16-16
immunoglobulin

IGHV3/OR16-16
16p11.2
Z29613




heavy variable




3/OR16-16




(pseudogene)


HGNC: 5658
IGHV4OR15-8
immunoglobulin
VSIG6
IGHV4/OR15-8,
15q11.2
Z29598




heavy variable

IGHV4OR158




4/OR15-8




(non-functional)

















Gene Family
Gene family



HGNC ID
RefSeq IDs
Tag
description







HGNC: 5487

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5488

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5493

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5494

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5500

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5501

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5506

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5507

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5512

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5513

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5524
NG_003254
IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5563

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5564

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5565

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5566

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5567
NG_016978
IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5568

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5569

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5570

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5571

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5572

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5573

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 38040
NG_011680
IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5579

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5633

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5641

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5642

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5643

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5644

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5634

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5635

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5636

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5637
NG_011771
IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5638

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5639

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5640

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5658

IGHO
“Immunoglobulins/IGH orphons”










In exemplary aspects, the method comprises measuring the level of expression of one or more gene segments at the IGK locus. In exemplary aspects, the one or more gene segments is selected from the group consisting of: IGKC, IGKJ1, IGKJ2, IGKJ3, IGKJ4, IGKJ5, IGKV1-12, IGKV1-13, IGKV1-16, IGKV1-17, IGKV1-22, IGKV1-27, IGKV1-32, IGKV1-33, IGKV1-35, IGKV1-37, IGKV1-39, IGKV1-5, IGKV1-6, IGKV1-8, IGKV1-9, IGKV1D-12, IGKV1D-13, IGKV1D-16, IGKV1D-17, IGKV1D-22, IGKV1D-27, IGKV1D-32, IGKV1D-33, IGKV1D-35, IGKV1D-37, IGKV1D-39, IGKV1D-42, IGKV1D-43, IGKV1D-8, IGKV1OR22-1, IGKV2-10, IGKV2-14, IGKV2-18, IGKV2-19, IGKV2-23, IGKV2-24, IGKV2-26, IGKV2-28, IGKV2-29, IGKV2-30, IGKV2-36, IGKV2-38, IGKV2-4, IGKV2-40, IGKV2D-10, IGKV2D-14, IGKV2D-18, IGKV2D-19, IGKV2D-23, IGKV2D-24, IGKV2D-26, IGKV2D-28, IGKV2D-29, IGKV2D-30, IGKV2D-36, IGKV2D-38, IGKV2D-40, IGKV2OR22-3, IGKV2OR22-4, IGKV3-11, IGKV3-15, IGKV3-20, IGKV3-25, IGKV3-31, IGKV3-34, IGKV3-7, IGKV3D-11, IGKV3D-15, IGKV3D-20, IGKV3D-25, IGKV3D-31, IGKV3D-34, IGKV3D-7, IGKV3OR22-2, IGKV4-1, IGKV5-2, IGKV6-21, IGKV6D-21, IGKV6D-41, IGKV7-3. In exemplary aspects, the one or more gene segments at the IgK locus comprises a sequence selected from the group consisting of SEQ ID NOs: 175-260. In exemplary aspects, the one or more gene segments at the IGK locus is one listed in the following table.





















Approved
Approved
Previous


Accession


HGNC ID
Symbol
Name
Symbols
Synonyms
Chromosome
Numbers





HGNC: 5715
IGK
immunoglobulin
IGK@

2p11.2




kappa locus


HGNC: 5716
IGKC
immunoglobulin

HCAK1
2p11.2
J00241




kappa constant


HGNC: 5719
IGKJ1
immunoglobulin

J1
2p11.2
J00242




kappa joining 1


HGNC: 5720
IGKJ2
immunoglobulin

J2
2p11.2
J00242




kappa joining 2


HGNC: 5721
IGKJ3
immunoglobulin


2p11.2
J00242




kappa joining 3


HGNC: 5722
IGKJ4
immunoglobulin


2p11.2
J00242




kappa joining 4


HGNC: 5723
IGKJ5
immunoglobulin


2p11.2
J00242




kappa joining 5


HGNC: 5741
IGKV1-5
immunoglobulin


2p11.2
Z00001




kappa variable 1-5


HGNC: 5742
IGKV1-6
immunoglobulin


2p11.2
M64858




kappa variable 1-6


HGNC: 5743
IGKV1-8
immunoglobulin

IGKV18, L9
2p11.2
Z00014




kappa variable 1-8


HGNC: 5744
IGKV1-9
immunoglobulin

IGKV19, L8
2p11.2
Z00013




kappa variable 1-9


HGNC: 5730
IGKV1-12
immunoglobulin

IGKV112, L19
2p11.2
V01577




kappa variable 1-12


HGNC: 5731
IGKV1-13
immunoglobulin


2p11.2
Z00010




kappa variable 1-13




(gene/pseudogene)


HGNC: 5732
IGKV1-16
immunoglobulin

IGKV116, L1
2p11.2
J00248




kappa variable 1-16


HGNC: 5733
IGKV1-17
immunoglobulin

IGKV117, A30
2p11.2
X72808




kappa variable 1-17


HGNC: 5734
IGKV1-22
immunoglobulin


2p11.2
X71885




kappa variable 1-22




(pseudogene)


HGNC: 5735
IGKV1-27
immunoglobulin

IGKV127, A20
2p11.2
X63398




kappa variable 1-27


HGNC: 5736
IGKV1-32
immunoglobulin


2p11.2
X71883




kappa variable 1-32




(pseudogene)


HGNC: 5737
IGKV1-33
immunoglobulin

IGKV133, O18
2p11.2
M64856




kappa variable 1-33


HGNC: 5738
IGKV1-35
immunoglobulin


2p11.2
X71890




kappa variable 1-35




(pseudogene)


HGNC: 5739
IGKV1-37
immunoglobulin

IGKV137, O14
2p11.2
X59316




kappa variable 1-37




(non-functional)


HGNC: 5740
IGKV1-39
immunoglobulin


2p11.2
X59315




kappa variable 1-39




(gene/pseudogene)


HGNC: 5759
IGKV1D-8
immunoglobulin


2p11.2
Z00008




kappa variable 1D-8


HGNC: 5746
IGKV1D-12
immunoglobulin


2p11.2
X17263




kappa variable 1D-12


HGNC: 5747
IGKV1D-13
immunoglobulin


2p11.2
X17262




kappa variable 1D-13


HGNC: 5748
IGKV1D-16
immunoglobulin


2p11.2
K01323




kappa variable 1D-16


HGNC: 5749
IGKV1D-17
immunoglobulin


2p11.2
X63392




kappa variable 1D-17


HGNC: 5750
IGKV1D-22
immunoglobulin


2p11.2
X71887




kappa variable 1D-22




(pseudogene)


HGNC: 5751
IGKV1D-27
immunoglobulin


2p11.2
Z00004




kappa variable 1D-27




(pseudogene)


HGNC: 5752
IGKV1D-32
immunoglobulin


2p11.2
X71896




kappa variable 1D-32




(pseudogene)


HGNC: 5753
IGKV1D-33
immunoglobulin


2p11.2
M64855




kappa variable 1D-33


HGNC: 5754
IGKV1D-35
immunoglobulin


2p11.2
X71894




kappa variable 1D-35




(pseudogene)


HGNC: 5755
IGKV1D-37
immunoglobulin

IGKV1D37, O4
2p11.2
X71893




kappa variable 1D-37




(non-functional)


HGNC: 5756
IGKV1D-39
immunoglobulin


2p11.2
X59312




kappa variable 1D-39


HGNC: 5757
IGKV1D-42
immunoglobulin


2p11.2
X72816




kappa variable 1D-42




(non-functional)


HGNC: 5758
IGKV1D-43
immunoglobulin


2p11.2
X72817




kappa variable 1D-43


HGNC: 5788
IGKV2-4
immunoglobulin


2p11.2
X72814




kappa variable 2-4




(pseudogene)


HGNC: 5776
IGKV2-10
immunoglobulin


2p11.2
Z00012




kappa variable 2-10




(pseudogene)


HGNC: 5777
IGKV2-14
immunoglobulin


2p11.2
X72810




kappa variable 2-14




(pseudogene)


HGNC: 5778
IGKV2-18
immunoglobulin


2p11.2
X63400




kappa variable 2-18




(pseudogene)


HGNC: 5779
IGKV2-19
immunoglobulin


2p11.2
X12692




kappa variable 2-19




(pseudogene)


HGNC: 5780
IGKV2-23
immunoglobulin


2p11.2
X71885




kappa variable 2-23




(pseudogene)


HGNC: 5781
IGKV2-24
immunoglobulin


2p11.2
X12684




kappa variable 2-24


HGNC: 5782
IGKV2-26
immunoglobulin


2p11.2
X71884




kappa variable 2-26




(pseudogene)


HGNC: 5783
IGKV2-28
immunoglobulin


2p11.2
X63397




kappa variable 2-28


HGNC: 5784
IGKV2-29
immunoglobulin


2p11.2
X63396




kappa variable 2-29




(gene/pseudogene)


HGNC: 5785
IGKV2-30
immunoglobulin


2p11.2
X63403




kappa variable 2-30


HGNC: 5786
IGKV2-36
immunoglobulin


2p11.2
X71889




kappa variable 2-36




(pseudogene)


HGNC: 5787
IGKV2-38
immunoglobulin


2p11.2
X71888




kappa variable 2-38




(pseudogene)


HGNC: 5789
IGKV2-40
immunoglobulin


2p11.2
X59314




kappa variable 2-40


HGNC: 5792
IGKV2D-10
immunoglobulin


2p11.2
X17265




kappa variable 2D-10




(pseudogene)


HGNC: 5793
IGKV2D-14
immunoglobulin


2p11.2
X72811




kappa variable 2D-14




(pseudogene)


HGNC: 5794
IGKV2D-18
immunoglobulin


2p11.2
X63395




kappa variable 2D-18




(pseudogene)


HGNC: 5795
IGKV2D-19
immunoglobulin


2p11.2
X71882




kappa variable 2D-19




(pseudogene)


HGNC: 5796
IGKV2D-23
immunoglobulin


2p11.2
X71887




kappa variable 2D-23




(pseudogene)


HGNC: 5797
IGKV2D-24
immunoglobulin


2p11.2
X63401




kappa variable 2D-24




(non-functional)


HGNC: 5798
IGKV2D-26
immunoglobulin


2p11.2
X12689




kappa variable 2D-26


HGNC: 5799
IGKV2D-28
immunoglobulin


2p11.2
X12691




kappa variable 2D-28


HGNC: 5800
IGKV2D-29
immunoglobulin


2p11.2
M31952




kappa variable 2D-29


HGNC: 5801
IGKV2D-30
immunoglobulin


2p11.2
X63402




kappa variable 2D-30


HGNC: 5802
IGKV2D-36
immunoglobulin


2p11.2
X71893




kappa variable 2D-36




(pseudogene)


HGNC: 5803
IGKV2D-38
immunoglobulin


2p11.2
X71892




kappa variable 2D-38




(pseudogene)


HGNC: 5804
IGKV2D-40
immunoglobulin


2p11.2
X59311




kappa variable 2D-40


HGNC: 5821
IGKV3-7
immunoglobulin


2p11.2
X02725




kappa variable 3-7




(non-functional)


HGNC: 5815
IGKV3-11
immunoglobulin


2p11.2
X01668




kappa variable 3-11


HGNC: 5816
IGKV3-15
immunoglobulin


2p11.2
M23090




kappa variable 3-15


HGNC: 5817
IGKV3-20
immunoglobulin


2p11.2
X12686




kappa variable 3-20


HGNC: 5818
IGKV3-25
immunoglobulin


2p11.2
X06583




kappa variable 3-25




(pseudogene)


HGNC: 5819
IGKV3-31
immunoglobulin


2p11.2
X71883




kappa variable 3-31




(pseudogene)


HGNC: 5820
IGKV3-34
immunoglobulin


2p11.2
X71891




kappa variable 3-34




(pseudogene)


HGNC: 5829
IGKV3D-7
immunoglobulin


2p11.2
X72820




kappa variable 3D-7


HGNC: 5823
IGKV3D-11
immunoglobulin


2p11.2
X17264




kappa variable 3D-11


HGNC: 5824
IGKV3D-15
immunoglobulin


2p11.2
X72815




kappa variable 3D-15




(gene/pseudogene)


HGNC: 5825
IGKV3D-20
immunoglobulin


2p11.2
X12687




kappa variable 3D-20


HGNC: 5826
IGKV3D-25
immunoglobulin


2p11.2
X71886




kappa variable 3D-25




(pseudogene)


HGNC: 5827
IGKV3D-31
immunoglobulin


2p11.2
X71896




kappa variable 3D-31




(pseudogene)


HGNC: 5828
IGKV3D-34
immunoglobulin


2p11.2
X71895




kappa variable 3D-34




(pseudogene)


HGNC: 5834
IGKV4-1
immunoglobulin

IGKV41, B3
2p11.2
Z00023




kappa variable 4-1


HGNC: 5835
IGKV5-2
immunoglobulin

IGKV52, B2
2p11.2
X02485




kappa variable 5-2


HGNC: 5836
IGKV6-21
immunoglobulin

IGKV621, A26
2p11.2
X63399




kappa variable 6-21




(non-functional)


HGNC: 5837
IGKV6D-21
immunoglobulin

IGKV6D21, A10
2p11.2
X12683




kappa variable 6D-21




(non-functional)


HGNC: 5838
IGKV6D-41
immunoglobulin


2p11.2
X12688




kappa variable 6D-41




(non-functional)


HGNC: 5839
IGKV7-3
immunoglobulin


2p11.2
X12682




kappa variable 7-3




(pseudogene)

















Gene Family
Gene family



HGNC ID
RefSeq IDs
Tag
description







HGNC: 5715
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5716
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5719
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5720
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5721
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5722
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5723
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5741
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5742
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5743
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5744
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5730
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5731
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5732
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5733
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5734
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5735
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5736
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5737
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5738
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5739
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5740
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5759
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5746
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5747
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5748
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5749
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5750
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5751
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5752
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5753
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5754
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5755
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5756
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5757
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5758
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5788
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5776
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5777
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5778
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5779
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5780
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5781
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5782
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5783
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5784
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5785
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5786
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5787
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5789
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5792
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5793
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5794
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5795
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5796
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5797
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5798
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5799
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5800
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5801
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5802
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5803
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5804
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5821
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5815
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5816
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5817
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5818
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5819
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5820
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5829
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5823
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5824
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5825
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5826
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5827
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5828
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5834
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5835
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5836
NG_000834
IGK
“Immunoglobulins/IGK locus”



HGNC: 5837
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5838
NG_000833
IGK
“Immunoglobulins/IGK locus”



HGNC: 5839
NG_000834
IGK
“Immunoglobulins/IGK locus”










In exemplary aspects, the method comprises measuring the level of expression of one or more gene segments at the IGL locus. In exemplary aspects, the one or more gene segments is selected from the group consisting of: IGLC1, IGLC2, IGLC3, IGLC4, IGLC5, IGLC6, IGLC7, IGLCOR22-1, IGLJ1, IGLJ2, IGLJ3, IGLJ4, IGLJ5, IGLJ6, IGLJ7, IGLL1, IGLL3, IGLON5, IGLV10-54, IGLV10-67, IGLV11-55, IGLV1-36, IGLV1-40, IGLV1-41, IGLV1-44, IGLV1-47, IGLV1-50, IGLV1-51, IGLV1-62, IGLV2-11, IGLV2-14, IGLV2-18, IGLV2-23, IGLV2-28, IGLV2-33, IGLV2-34, IGLV2-5, IGLV2-8, IGLV3-1, IGLV3-10, IGLV3-12, IGLV3-13, IGLV3-15, IGLV3-16, IGLV3-17, IGLV3-19, IGLV3-2, IGLV3-21, IGLV3-22, IGLV3-24, IGLV3-25, IGLV3-26, IGLV3-27, IGLV3-29, IGLV3-30, IGLV3-31, IGLV3-32, IGLV3-4, IGLV3-6, IGLV3-7, IGLV3-9, IGLV4-3, IGLV4-60, IGLV4-69, IGLV5-37, IGLV5-45, IGLV5-48, IGLV5-52, IGLV6-57, IGLV7-35, IGLV7-43, IGLV7-46, IGLV8-61, IGLV9-49, IGLVI-20, IGLVI-38, IGLVI-42, IGLVI-56, IGLVI-63, IGLVI-68, IGLVI-70, IGLVIV-53, IGLVIV-59, IGLVIV-64, IGLVIV-65, IGLVIV-66-1, IGLVV-58, IGLVV-66, IGLVVI-22-1, IGLVVI-25-1, and IGLVVII-41-1. In exemplary aspects, the one or more gene segments at the IgL locus comprises a sequence selected from the group consisting of SEQ ID NOs: 261-350. In exemplary aspects, the one or more gene segments at the IGL locus is one listed in the following table.





















Approved
Approved
Previous


Accession


HGNC ID
Symbol
Name
Symbols
Synonyms
Chromosome
Numbers





HGNC: 5853
IGL
immunoglobulin
IGL@

22q11.2




lambda locus


HGNC: 5855
IGLC1
immunoglobulin
IGLC

22q11.2
J00252




lambda constant 1




(Mcg marker)


HGNC: 5856
IGLC2
immunoglobulin
IGLC

22q11.2
J00253




lambda constant 2




(Kern − Oz − marker)


HGNC: 5857
IGLC3
immunoglobulin
IGLC

22q11.2
J00254




lambda constant 3




(Kern − Oz + marker)


HGNC: 5858
IGLC4
immunoglobulin
IGLC

22q11.2
J03009




lambda constant 4




(pseudogene)


HGNC: 5859
IGLC5
immunoglobulin
IGLC

22q11.2
J03010




lambda constant 5




(pseudogene)


HGNC: 5860
IGLC6
immunoglobulin
IGLC

22q11.2
J03011




lambda constant 6




(Kern + Oz − marker,




gene/pseudogene)


HGNC: 5861
IGLC7
immunoglobulin


22q11.2
X51755




lambda constant 7


HGNC: 5863
IGLJ1
immunoglobulin


22q11.2
X04457




lambda joining 1


HGNC: 5864
IGLJ2
immunoglobulin


22q11.2
M15641




lambda joining 2


HGNC: 5865
IGLJ3
immunoglobulin


22q11.2
M15642




lambda joining 3


HGNC: 5866
IGLJ4
immunoglobulin


22q11.2
X51755




lambda joining 4




(non-functional)


HGNC: 5867
IGLJ5
immunoglobulin


22q11.2
X51755




lambda joining 5




(non-functional)


HGNC: 5868
IGLJ6
immunoglobulin


22q11.2
M18338




lambda joining 6


HGNC: 5869
IGLJ7
immunoglobulin


22q11.2
X51755




lambda joining 7


HGNC: 5876
IGLV1-36
immunoglobulin


22q11.2
Z73653




lambda variable 1-36


HGNC: 5877
IGLV1-40
immunoglobulin


22q11.2
M94116




lambda variable 1-40


HGNC: 5878
IGLV1-41
immunoglobulin


22q11.2
M94118




lambda variable 1-41




(pseudogene)


HGNC: 5879
IGLV1-44
immunoglobulin


22q11.2
Z73654




lambda variable 1-44


HGNC: 5880
IGLV1-47
immunoglobulin


22q11.2
Z73663




lambda variable 1-47


HGNC: 5881
IGLV1-50
immunoglobulin


22q11.2
M94112




lambda variable 1-50




(non-functional)


HGNC: 5882
IGLV1-51
immunoglobulin


22q11.2
Z73661




lambda variable 1-51


HGNC: 5883
IGLV1-62
immunoglobulin


22q11.2
D87022




lambda variable 1-62




(pseudogene)


HGNC: 5894
IGLV2-5
immunoglobulin


22q11.2
Z73641




lambda variable 2-5




(pseudogene)


HGNC: 5895
IGLV2-8
immunoglobulin


22q11.2
X97462




lambda variable 2-8


HGNC: 5887
IGLV2-11
immunoglobulin


22q11.2
Z73657




lambda variable 2-11


HGNC: 5888
IGLV2-14
immunoglobulin


22q11.2
Z73664




lambda variable 2-14


HGNC: 5889
IGLV2-18
immunoglobulin


22q11.2
Z73642




lambda variable 2-18


HGNC: 5890
IGLV2-23
immunoglobulin


22q11.2
X14616




lambda variable 2-23


HGNC: 5891
IGLV2-28
immunoglobulin


22q11.2
X97466




lambda variable 2-28




(pseudogene)


HGNC: 5892
IGLV2-33
immunoglobulin


22q11.2
Z73643




lambda variable 2-33




(non-functional)


HGNC: 5893
IGLV2-34
immunoglobulin


22q11.2
D87013




lambda variable 2-34




(pseudogene)


HGNC: 5896
IGLV3-1
immunoglobulin


22q11.2
X57826




lambda variable 3-1


HGNC: 5904
IGLV3-2
immunoglobulin


22q11.2
X97468




lambda variable 3-2




(pseudogene)


HGNC: 5915
IGLV3-4
immunoglobulin


22q11.2
D87024




lambda variable 3-4




(pseudogene)


HGNC: 5916
IGLV3-6
immunoglobulin


22q11.2
X97465




lambda variable 3-6




(pseudogene)


HGNC: 5917
IGLV3-7
immunoglobulin


22q11.2
X97470




lambda variable 3-7




(pseudogene)


HGNC: 5918
IGLV3-9
immunoglobulin


22q11.2
X97473




lambda variable 3-9




(gene/pseudogene)


HGNC: 5897
IGLV3-10
immunoglobulin


22q11.2
X97464




lambda variable 3-10


HGNC: 5898
IGLV3-12
immunoglobulin


22q11.2
Z73658




lambda variable 3-12


HGNC: 5899
IGLV3-13
immunoglobulin


22q11.2
X97463




lambda variable 3-13




(pseudogene)


HGNC: 5900
IGLV3-15
immunoglobulin


22q11.2
D87015




lambda variable 3-15




(pseudogene)


HGNC: 5901
IGLV3-16
immunoglobulin


22q11.2
X97471




lambda variable 3-16


HGNC: 5902
IGLV3-17
immunoglobulin


22q11.2
X97472




lambda variable 3-17




(pseudogene)


HGNC: 5903
IGLV3-19
immunoglobulin


22q11.2
X56178




lambda variable 3-19


HGNC: 5905
IGLV3-21
immunoglobulin


22q11.2
X71966




lambda variable 3-21


HGNC: 5906
IGLV3-22
immunoglobulin


22q11.2
Z73666




lambda variable 3-22




(gene/pseudogene)


HGNC: 5907
IGLV3-24
immunoglobulin


22q11.2
X71968




lambda variable 3-24




(pseudogene)


HGNC: 5908
IGLV3-25
immunoglobulin


22q11.2
X97474




lambda variable 3-25


HGNC: 5909
IGLV3-26
immunoglobulin


22q11.2
X97467




lambda variable 3-26




(pseudogene)


HGNC: 5910
IGLV3-27
immunoglobulin


22q11.2
D86994




lambda variable 3-27


HGNC: 5911
IGLV3-29
immunoglobulin


22q11.2
Z73644




lambda variable 3-29




(pseudogene)


HGNC: 5912
IGLV3-30
immunoglobulin


22q11.2
Z73646




lambda variable 3-30




(pseudogene)


HGNC: 5913
IGLV3-31
immunoglobulin


22q11.2
X97469




lambda variable 3-31




(pseudogene)


HGNC: 5914
IGLV3-32
immunoglobulin


22q11.2
Z73645




lambda variable 3-32




(non-functional)


HGNC: 5919
IGLV4-3
immunoglobulin


22q11.2
X57828




lambda variable 4-3


HGNC: 5920
IGLV4-60
immunoglobulin


22q11.2
Z73667




lambda variable 4-60


HGNC: 5921
IGLV4-69
immunoglobulin


22q11.2
Z73648




lambda variable 4-69


HGNC: 5922
IGLV5-37
immunoglobulin


22q11.2
Z73672




lambda variable 5-37


HGNC: 5923
IGLV5-39
immunoglobulin


22q11.2
Z73668




lambda variable 5-39


HGNC: 5924
IGLV5-45
immunoglobulin


22q11.2
Z73670




lambda variable 5-45


HGNC: 5925
IGLV5-48
immunoglobulin


22q11.2
Z73649




lambda variable 5-48




(non-functional)


HGNC: 5926
IGLV5-52
immunoglobulin


22q11.2
Z73669




lambda variable 5-52


HGNC: 5927
IGLV6-57
immunoglobulin


22q11.2
Z73673




lambda variable 6-57


HGNC: 5928
IGLV7-35
immunoglobulin


22q11.2
Z73660




lambda variable 7-35




(pseudogene)


HGNC: 5929
IGLV7-43
immunoglobulin


22q11.2
X14614




lambda variable 7-43


HGNC: 5930
IGLV7-46
immunoglobulin


22q11.2
Z73674




lambda variable 7-46




(gene/pseudogene)


HGNC: 5931
IGLV8-61
immunoglobulin


22q11.2
Z73650




lambda variable 8-61


HGNC: 5933
IGLV9-49
immunoglobulin


22q11.2
Z73675




lambda variable 9-49


HGNC: 5884
IGLV10-54
immunoglobulin


22q11.2
Z73676




lambda variable 10-54


HGNC: 5885
IGLV10-67
immunoglobulin


22q11.2
Z73651




lambda variable 10-67




(pseudogene)


HGNC: 5886
IGLV11-55
immunoglobulin


22q11.2
D86996




lambda variable 11-55




(non-functional)


HGNC: 5934
IGLVI-20
immunoglobulin

IGLV(I)-20
22q11.2
D87007




lambda variable (I)-20




(pseudogene)


HGNC: 5935
IGLVI-38
immunoglobulin

IGLV(I)-38
22q11.2
D87009




lambda variable (I)-38




(pseudogene)


HGNC: 5936
IGLVI-42
immunoglobulin

IGLV(I)-42
22q11.2
X14613




lambda variable (I)-42




(pseudogene)


HGNC: 5937
IGLVI-56
immunoglobulin

IGLV(I)-56
22q11.2
D86996




lambda variable (I)-56




(pseudogene)


HGNC: 5938
IGLVI-63
immunoglobulin

IGLV(I)-63
22q11.2
D87022




lambda variable (I)-63




(pseudogene)


HGNC: 5939
IGLVI-68
immunoglobulin

IGLV(I)-68
22q11.2
D86993




lambda variable (I)-68




(pseudogene)


HGNC: 5940
IGLVI-70
immunoglobulin

IGLV(I)-70
22q11.2
D86993




lambda variable




(I)-70




(pseudogene)


HGNC: 5941
IGLVIV-53
immunoglobulin

IGLV(IV)-53
22q11.2
D86996




lambda variable (IV)-53




(pseudogene)


HGNC: 5942
IGLVIV-59
immunoglobulin

IGLV(IV)-59
22q11.2
D87000




lambda variable (IV)-59




(pseudogene)


HGNC: 5943
IGLVIV-64
immunoglobulin

IGLV(IV)-64
22q11.2
D87022




lambda variable (IV)-64




(pseudogene)


HGNC: 5944
IGLVIV-65
immunoglobulin

IGLV(IV)-65
22q11.2
D87022




lambda variable (IV)-65




(pseudogene)


HGNC: 15692
IGLVIV-66-1
immunoglobulin

IGLV(IV)-66-1
22q11.2
D87004




lambda variable (IV)-66-1




(pseudogene)


HGNC: 5945
IGLVV-58
immunoglobulin

IGLV(V)-58
22q11.2
D87000




lambda variable (V)-58




(pseudogene)


HGNC: 5946
IGLVV-66
immunoglobulin

IGLV(V)-66
22q11.2
D87004




lambda variable (V)-66




(pseudogene)


HGNC: 15689
IGLVVI-22-1
immunoglobulin

IGLV(VI)-22-1
22q11.2
X71351




lambda variable (VI)-22-1




(pseudogene)


HGNC: 15690
IGLVVI-25-1
immunoglobulin

IGLV(VI)-25-1
22q11.2
D86994




lambda variable (VI)-25-1




(pseudogene)


HGNC: 15691
IGLVVII-41-1
immunoglobulin

IGLV(VII)-41-1
22q11.2
X99568




lambda variable (VII)-41-1




(pseudogene)

















Gene Family
Gene family



HGNC ID
RefSeq IDs
Tag
description







HGNC: 5853
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5855
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5856
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5857
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5858
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5859
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5860
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5861
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5863
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5864
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5865
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5866
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5867
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5868
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5869
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5876
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5877
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5878
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5879
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5880
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5881
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5882
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5883
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5894
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5895
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5887
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5888
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5889
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5890
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5891
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5892
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5893
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5896
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5904
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5915
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5916
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5917
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5918
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5897
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5898
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5899
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5900
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5901
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5902
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5903
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5905
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5906
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5907
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5908
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5909
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5910
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5911
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5912
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5913
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5914
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5919
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5920
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5921
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5922
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5923

IGL
“Immunoglobulins/IGL locus”



HGNC: 5924
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5925
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5926
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5927
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5928
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5929
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5930
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5931
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5933
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5884
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5885
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5886
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5934
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5935
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5936
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5937
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5938
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5939
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5940
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5941
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5942
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5943
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5944
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 15692
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5945
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 5946
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 15689
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 15690
NG_000002
IGL
“Immunoglobulins/IGL locus”



HGNC: 15691
NG_000002
IGL
“Immunoglobulins/IGL locus”










In exemplary aspects, the method comprises measuring the expression of one or more IGH, IGK, or IGL orphon gene segments, and optionally, the one or more gene segments is one listed in the following table.





















Approved
Approved
Previous


Accession


HGNC ID
Symbol
Name
Symbols
Synonyms
Chromosome
Numbers





HGNC: 5487
IGHD1OR15-1A
immunoglobulin

IGHD1/OR15-1A,
15q11.2
X55575




heavy diversity

IGHD1OR151A




1/OR15-1A




(non-functional)


HGNC: 5488
IGHD1OR15-1B
immunoglobulin

IGHD1/OR15-1B,
15q11.2
X55576




heavy diversity

IGHD1OR151B




1/OR15-1B




(non-functional)


HGNC: 5493
IGHD2OR15-2A
immunoglobulin

IGHD2/OR15-2A,
15q11.2
X55577




heavy diversity

IGHD2OR152A




2/OR15-2A




(non-functional)


HGNC: 5494
IGHD2OR15-2B
immunoglobulin

IGHD2/OR15-2B,
15q11.2
X55578




heavy diversity

IGHD2OR152B




2/OR15-2B




(non-functional)


HGNC: 5500
IGHD3OR15-3A
immunoglobulin

IGHD3/OR15-3A,
15q11.2
X55579




heavy diversity

IGHD3OR153A




3/OR15-3A




(non-functional)


HGNC: 5501
IGHD3OR15-3B
immunoglobulin

IGHD3/OR15-3B,
15q11.2
X55580




heavy diversity

IGHD3OR153B




3/OR15-3B




(non-functional)


HGNC: 5506
IGHD4OR15-4A
immunoglobulin

IGHD4/OR15-4A,
15q11.2
X55581




heavy diversity

IGHD4OR154A




4/OR15-4A




(non-functional)


HGNC: 5507
IGHD4OR15-4B
immunoglobulin

IGHD4/OR15-4B,
15q11.2
X55582




heavy diversity

IGHD4OR154B




4/OR15-4B




(non-functional)


HGNC: 5512
IGHD5OR15-5A
immunoglobulin

IGHD5/OR15-5A,
15q11.2
X55583




heavy diversity

IGHD5OR155A




5/OR15-5A




(non-functional)


HGNC: 5513
IGHD5OR15-5B
immunoglobulin

IGHD5/OR15-5B,
15q11.2
X55584




heavy diversity

IGHD5OR155B




5/OR15-5B




(non-functional)


HGNC: 5524
IGHEP2
immunoglobulin


9p24.1
K01241




heavy constant




epsilon P2




(pseudogene)


HGNC: 5563
IGHV1OR15-1
immunoglobulin

IGHV1/OR15-1
15q11.2
Z29631




heavy variable




1/OR15-1




(non-functional)


HGNC: 5564
IGHV1OR15-2
immunoglobulin

IGHV1/OR15-2
15q11.1
L25543




heavy variable




1/OR15-2




(pseudogene)


HGNC: 5565
IGHV1OR15-3
immunoglobulin

IGHV1/OR15-3
15q11.2
Z29595




heavy variable




1/OR15-3




(pseudogene)


HGNC: 5566
IGHV1OR15-4
immunoglobulin

IGHV1/OR15-4
15q11.2
Z29596




heavy variable




1/OR15-4




(pseudogene)


HGNC: 5567
IGHV1OR15-5
immunoglobulin

IGHV1/OR15-5
15q11.2
Z29633




heavy variable




1/OR15-5




(non-functional)


HGNC: 5568
IGHV1OR15-6
immunoglobulin

IGHV1/OR15-6
15q11.2
Z29634




heavy variable




1/OR15-6




(pseudogene)


HGNC: 5569
IGHV1OR15-9
immunoglobulin
VSIG7
IGHV1/OR15-9,
15q11.1
L25542




heavy variable

IGHV1OR159




1/OR15-9




(non-functional)


HGNC: 5570
IGHV1OR16-1
immunoglobulin

IGHV1/OR16-1
16p11.2
Z29599




heavy variable




1/OR16-1




(pseudogene)


HGNC: 5571
IGHV1OR16-2
immunoglobulin

IGHV1/OR16-2
16p11.2
Z29600




heavy variable




1/OR16-2




(pseudogene)


HGNC: 5572
IGHV1OR16-3
immunoglobulin

IGHV1/OR16-3
16p11.2
Z29639




heavy variable




1/OR16-3




(pseudogene)


HGNC: 5573
IGHV1OR16-4
immunoglobulin

IGHV1/OR16-4
16p11.2
Z17397




heavy variable




1/OR16-4




(pseudogene)


HGNC: 38040
IGHV1OR21-1
immunoglobulin

IGHV1/OR21-1
21p11.2




heavy variable




1/OR21-1




(non-functional)


HGNC: 5579
IGHV2OR16-5
immunoglobulin

IGHV2/OR16-5
16p11.2
L25544




heavy variable




2/OR16-5




(non-functional)


HGNC: 5633
IGHV3OR15-7
immunoglobulin

IGHV3/OR15-7
15q11.2
Z29597




heavy variable




3/OR15-7




(pseudogene)


HGNC: 5641
IGHV3OR16-6
immunoglobulin

IGHV3/OR16-6
16p11.2
L25545




heavy variable




3/OR16-6




(pseudogene)


HGNC: 5642
IGHV3OR16-7
immunoglobulin

IGHV3/OR16-7
16p11.2
Z29604




heavy variable




3/OR16-7




(pseudogene)


HGNC: 5643
IGHV3OR16-8
immunoglobulin

IGHV3/OR16-8
16p11.2
Z29605




heavy variable




3/OR16-8




(non-functional)


HGNC: 5644
IGHV3OR16-9
immunoglobulin

IGHV3/OR16-9
16p11.2
Z29606




heavy variable




3/OR16-9




(non-functional)


HGNC: 5634
IGHV3OR16-10
immunoglobulin

IGHV3/OR16-10
16p11.2
Z29607




heavy variable




3/OR16-10




(non-functional)


HGNC: 5635
IGHV3OR16-11
immunoglobulin

IGHV3/OR16-11
16p11.2
Z29608




heavy variable




3/OR16-11




(pseudogene)


HGNC: 5636
IGHV3OR16-12
immunoglobulin

IGHV3/OR16-12
16p11.2
Z29609




heavy variable




3/OR16-12




(non-functional)


HGNC: 5637
IGHV3OR16-13
immunoglobulin

IGHV3/OR16-13
16p11.2
Z29610




heavy variable




3/OR16-13




(non-functional)


HGNC: 5638
IGHV3OR16-14
immunoglobulin

IGHV3/OR16-14
16p11.2
Z29611




heavy variable




3/OR16-14




(pseudogene)


HGNC: 5639
IGHV3OR16-15
immunoglobulin

IGHV3/OR16-15
16p11.2
L25546




heavy variable




3/OR16-15




(pseudogene)


HGNC: 5640
IGHV3OR16-16
immunoglobulin

IGHV3/OR16-16
16p11.2
Z29613




heavy variable




3/OR16-16




(pseudogene)


HGNC: 5658
IGHV4OR15-8
immunoglobulin
VSIG6
IGHV4/OR15-8,
15q11.2
Z29598




heavy variable

IGHV4OR158




4/OR15-8




(non-functional)


HGNC: 5761
IGKV1OR-2
immunoglobulin
IGKVPZ2
IGKV1/OR-2
9q21.11
X64640




kappa variable




1/OR-2




(pseudogene)


HGNC: 5762
IGKV1OR-3
immunoglobulin
IGKVPZ3
IGKV1/OR-3
9q12
X64641




kappa variable




1/OR-3




(pseudogene)


HGNC: 5763
IGKV1OR-4
immunoglobulin
IGKVPZ4
IGKV1/OR-4
reserved
X64642




kappa variable




1/OR-4




(pseudogene)


HGNC: 5764
IGKV1OR1-1
immunoglobulin
IGKVP1
IGKV1/OR1-1
1
M20809




kappa variable




1/OR1-1




(pseudogene)


HGNC: 5766
IGKV1OR2-0
immunoglobulin

IGKV1/OR2-0
2p11.2
Y08392




kappa variable




1/OR2-0




(non-functional)


HGNC: 5760
IGKV1OR2-1
immunoglobulin
IGKVPZ1,
IGKV1/OR-1,
2p11.1
Z12367




kappa variable
IGKV1OR-1
IGKV1/OR2-1




1/OR2-1




(pseudogene)


HGNC: 5769
IGKV1OR2-3
immunoglobulin

IGKV1/OR2-3
2q11.2
X05102




kappa variable




1/OR2-3




(pseudogene)


HGNC: 5770
IGKV1OR2-6
immunoglobulin

IGKV1/OR2-6
2q11.2
X05103




kappa variable




1/OR2-6




(pseudogene)


HGNC: 5771
IGKV1OR2-9
immunoglobulin

IGKV1/OR2-9
2q11.2
X51879




kappa variable




1/OR2-9




(pseudogene)


HGNC: 5768
IGKV1OR2-11
immunoglobulin

IGKV1/OR2-11
2q11.2
X51885




kappa variable




1/OR2-11




(pseudogene)


HGNC: 5767
IGKV1OR2-108
immunoglobulin

IGKV1/OR2-108,
2q12-q14
X51887




kappa variable

IGKV1OR2108, IGO1




1/OR2-108




(non-functional)


HGNC: 37488
IGKV1OR2-118
immunoglobulin

IGKV1/OR2-118
2p11.1




kappa variable




1/OR2-118




(pseudogene)


HGNC: 44978
IGKV1OR10-1
immunoglobulin

IGKV1/OR10-1
10q11.21




kappa variable




1/OR10-1




(pseudogene)


HGNC: 5765
IGKV1OR15-118
immunoglobulin
IGKVP2
IGKV1/OR-118,
15




kappa variable

IGKV1/OR15-118




1/OR15-118




(pseudogene)


HGNC: 5772
IGKV1OR22-1
immunoglobulin
IGKVP5
IGKV1/OR22-1
22q11
Z00040




kappa variable




1/OR22-1




(pseudogene)


HGNC: 5773
IGKV1OR22-5
immunoglobulin
IGKVP7,
IGKV1/OR22-5,
22q11
Z00003




kappa variable
IGKV1OR22-5A
IGKV1/OR22-5A




1/OR22-5




(pseudogene)


HGNC: 37729
IGKV1ORY-1
immunoglobulin

IGKV1/ORY-1
Yq11.21




kappa variable




1/ORY-1




(pseudogene)


HGNC: 5805
IGKV2OR2-1
immunoglobulin
IGKV2OR2-1A
IGKV2/OR2-1,
2q11.2
X05101




kappa variable

IGKV2/OR2-1A




2/OR2-1




(pseudogene)


HGNC: 5808
IGKV2OR2-2
immunoglobulin

IGKV2/OR2-2
2q11.2
X51884




kappa variable




2/OR2-2




(pseudogene)


HGNC: 5809
IGKV2OR2-4
immunoglobulin

IGKV2/OR2-4
2q11.2
X51883




kappa variable




2/OR2-4




(pseudogene)


HGNC: 5810
IGKV2OR2-7
immunoglobulin

IGKV2/OR2-7
2q11.2
X51881




kappa variable




2/OR2-7




(pseudogene)


HGNC: 37489
IGKV2OR2-7D
immunoglobulin

IGKV2/OR2-7D
2q11.2
X51881




kappa variable




2/OR2-7D




(pseudogene)


HGNC: 5811
IGKV2OR2-8
immunoglobulin

IGKV2/OR2-8
2q11.2
X51880




kappa variable




2/OR2-8




(pseudogene)


HGNC: 5806
IGKV2OR2-10
immunoglobulin

IGKV2/OR2-10
2q11.2
X51886




kappa variable




2/OR2-10




(pseudogene)


HGNC: 5812
IGKV2OR22-3
immunoglobulin
IGKVP4
IGKV2/OR22-3
22q11
Z00041




kappa variable




2/OR22-3




(pseudogene)


HGNC: 5813
IGKV2OR22-4
immunoglobulin
IGKVP6
IGKV2/OR22-4
22q11
M20707




kappa variable




2/OR22-4




(pseudogene)


HGNC: 5832
IGKV3OR2-5
immunoglobulin

IGKV3/OR2-5
2q11.2
X51882




kappa variable




3/OR2-5




(pseudogene)


HGNC: 5830
IGKV3OR2-268
immunoglobulin
IGKV268,
IGKV3/OR2-268,
2p12
X74459




kappa variable
IGKV3OR2-268A
IGKV3/OR2-268A




3/OR2-268




(non-functional)


HGNC: 5833
IGKV3OR22-2
immunoglobulin
IGKVP3
IGKV3/OR22-2
22q11
Z00042




kappa variable




3/OR22-2




(pseudogene)


HGNC: 15696
IGLCOR22-1
immunoglobulin

IGLC/OR22-1
22q12.2-q12.3
AL008723




lambda




constant/OR22-1




(pseudogene)


HGNC: 15697
IGLCOR22-2
immunoglobulin

IGLC/OR22-2
22q12.2-q12.3
AL021937




lambda




constant/OR22-2




(pseudogene)


HGNC: 28614
IGLJCOR18
immunoglobulin

IGLJ-COR18,
18p11.31
J00255




lambda joining-

IGLJ-C/OR18




constant/OR18




(pseudogene)


HGNC: 5932
IGLV8OR8-1
immunoglobulin

IGLV8/OR8-1
8q11.2
Y08831




lambda variable




8/OR8-1




(pseudogene)


HGNC: 15694
IGLVIVOR22-1
immunoglobulin

IGLV(IV)/OR22-1
22q11.2-q12.1
AL008721




lambda variable




(IV)/OR22-1




(pseudogene)


HGNC: 15695
IGLVIVOR22-2
immunoglobulin

IGLV(IV)/OR22-2
22q12.2-q12.3
AL021937




lambda variable




(IV)/OR22-2




(pseudogene)

















Gene Family
Gene family



HGNC ID
RefSeq IDs
Tag
description







HGNC: 5487

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5488

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5493

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5494

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5500

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5501

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5506

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5507

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5512

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5513

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5524
NG_003254
IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5563

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5564

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5565

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5566

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5567
NG_016978
IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5568

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5569

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5570

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5571

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5572

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5573

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 38040
NG_011680
IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5579

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5633

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5641

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5642

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5643

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5644

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5634

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5635

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5636

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5637
NG_011771
IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5638

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5639

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5640

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5658

IGHO
“Immunoglobulins/IGH orphons”



HGNC: 5761
NG_011657
IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5762

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5763

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5764
NG_011766
IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5766

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5760

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5769

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5770

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5771

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5768

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5767

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 37488
NG_011659
IGKO
“Immunoglobulins/IGK orphons”



HGNC: 44978

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5765

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5772
NG_011658
IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5773

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 37729

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5805

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5808

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5809

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5810
NG_011671
IGKO
“Immunoglobulins/IGK orphons”



HGNC: 37489

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5811
NG_011662
IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5806
NG_011661
IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5812

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5813

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5832

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5830

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 5833

IGKO
“Immunoglobulins/IGK orphons”



HGNC: 15696

IGLO
“Immunoglobulins/IGL orphons”



HGNC: 15697

IGLO
“Immunoglobulins/IGL orphons”



HGNC: 28614
XM_497569
IGLO
“Immunoglobulins/IGL orphons”



HGNC: 5932

IGLO
“Immunoglobulins/IGL orphons”



HGNC: 15694

IGLO
“Immunoglobulins/IGL orphons”



HGNC: 15695

IGLO
“Immunoglobulins/IGL orphons”










In exemplary aspects of the inventive methods provided herein, the method comprises measuring the level of expression of all the gene segments at the IGH, IGK, and IGL loci and all the IGH orhpon gene segments, all the IGK orphon gene segments, and all the IGL orphon gene segments. In exemplary aspects, the level of expression is the sum of the expression levels of more than one gene segment of the IgH locus, IgK locus, and/or IgL locus. In exemplary aspects, the level of Ig expression is the sum of the expression levels of all the gene segments of the IgH locus and optionally all the IGH orphon gene segments. In exemplary aspects, the level of Ig expression is the sum of the expression levels of all the gene segments of the IgK locus and optionally all the IGK orphon gene segments. In exemplary aspects, the level of Ig expression is the sum of the expression levels of all the gene segments of the IgL locus and optionally all the IGL orphon gene segments. In exemplary aspects, the level of expression of Ig is the sum of (i) the levels of expression of all the gene segments of the IgH locus, (ii) the levels of expression of all the gene segments of the IgK locus, and (iii) the levels of expression of all the gene segments of the IgL locus, and optionally, all the IgH orphon gene segments, all the IgK orphon gene segments, and all the IgL orphon gene segments.


FCGR2B


In exemplary aspects, the sample obtained from the subject is measured for the expression level of FCGR2B. FCGR2B is also known as CD32 of the Fc fragmen of IgG, low affinity IIb, receptor. The gene encoding FCGR2B is located at ch. 1q23. Exemplary sequences encoding FCGR2B are provided herein as SEQ ID NO: 351 and 352, but are also known in the art. The FCGR2B gene is Entrez Gene No. 2213. The nucleotide sequence and amino acid sequence are available in the NCBI's nucleotide database as Accession No. NM_004001 and NP_003992.3 (SEQ ID NOs: 352-353, respectively).


Genes and Gene Segments of Table 4


In exemplary aspects, the sample obtained from the subject is measured for the expression level of one or more genes (or gene segments) listed in Table 4 set forth below. As used herein, the term “gene” refers to both a gene and gene segments. Table 4 includes, for each gene (or gene segment): (i) the HUGO gene symbol, if available, (ii) the Ensembl Gene Name, (iii) gene expression level data, and (iv) statistical data: the P-value and Q-value. The HUGO Gene Symbol is a name determined and approved by the HUGO Gene Nomenclature Committee (HGNC). The HGNC approves both a short-form abbreviation known as a gene symbol, and also a longer and more descriptive name. Each gene symbol is unique and the HGNC ensures that each gene is only given one approved gene symbol. This unique gene symbol allows for clear and unambiguous reference to genes in scientific communications, and facilitates electronic data retrieval from databases and publications. Gene symbols also maintain parallel construction for different members of a gene family and can also be used for orthologous genes in other vertebrate species. A record for each gene symbol listed in Table 4 is accessible by the public via the HGNC database. The HGNC database is a curated online repository of HGNC-approved gene nomenclature, gene families, and associated resources including links to genomic, proteomic and phenotypic information. The HGNC database contains records for over 38,000 gene symbols is accessible to the public on the internet at http://www.genenames.org. The Ensemble Gene Name provided in Table 4 is the one listed in the HGNC database record for the indicated gene or gene segment. The Ensembl project produces genome databases for vertebrates and other eukaryotic species, and makes this information freely available on the internet at http://uswest.ensembl.org/index.html. When this Ensembl gene name or accession number is entered in the search window at the above web address, the sequence of the gene, as well as other structural information of the gene, may be accessed.


Reference Levels and Reference Values


In exemplary embodiments of the inventive methods, the expression level of the indicated gene(s) or gene segment(s) is/are compared to a reference level or reference value. As used herein, the term “reference level” is a cutoff or threshold against which the measured expression level is compared, which correlates with a pre-determined % specificity and/or pre-determined % sensitivity, as determined by a receiver operative characteristics (ROC) curve. In exemplary aspects, the ROC curve is based on the distribution of biomarker expression levels of a population of responders and the distribution of biomarker expression levels of a population of non-responders. In exemplary aspects, the reference level is a cutoff which correlates with X % specificity and Y % sensitivity, as determined by an ROC curve, wherein each of X and Y is selected from 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, and 99.


In exemplary aspects, when the level of expression of an Ig is measured or has been measured, the reference level is a cutoff correlative with a pre-determined % specificity and/or pre-determined % sensitivity, as determined by a receiver operative characteristics (ROC) curve. In exemplary aspects, the ROC curve is based on the distribution of Ig expression levels of a population of responders and the distribution of Ig expression levels of a population of non-responders. Exemplary definitions of responders and non-responders are found herein at Example 2. In exemplary aspects, the reference level is the point on the ROC curve at which the % specificity is X %, wherein X is selected from 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, and 99. In exemplary aspects, the reference level is the point on the ROC curve at which the % sensitivity is Y %, wherein Y is selected from 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, and 99. In exemplary aspects, the reference level is a cut off correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by the ROC curve. In exemplary aspects, the reference level is a cut off correlative with a % specificity of at least 75% and a % sensitivity of at least 75%, as determined by the ROC curve. In exemplary aspects, the reference level is a cut off correlative with a % specificity of at least 85% and a % sensitivity of at least 85%, as determined by the ROC curve. In exemplary aspects, the reference level is a cut off correlative with a % specificity of at least 90% and a % sensitivity of at least 90%, as determined by the ROC curve.


In exemplary aspects, when the level of expression of FCGR2B is measured or has been measured, the reference level is a cutoff correlative with a pre-determined % specificity and/or pre-determined % sensitivity, as determined by an ROC curve. In exemplary aspects, the ROC curve is based on the distribution of FCGR2B expression levels of a population of responders and the distribution of FCGR2B expression levels of a population of non-responders. Exemplary definitions of responders and non-responders are found herein at Example 2. In exemplary aspects, the reference level is the point on the ROC curve at which the % specificity is X %, wherein X is selected from 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, and 99. In exemplary aspects, the reference level is the point on the ROC curve at which the % sensitivity is Y %, wherein Y is selected from 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, and 99. In exemplary aspects, the reference level is a cut off correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by the ROC curve. In exemplary aspects, the reference level is a cut off correlative with a % specificity of at least 75% and a % sensitivity of at least 75%, as determined by the ROC curve. In exemplary aspects, the reference level is a cut off correlative with a % specificity of at least 85% and a % sensitivity of at least 85%, as determined by the ROC curve. In exemplary aspects, the reference level is a cut off correlative with a % specificity of at least 90% and a % sensitivity of at least 90%, as determined by the ROC curve.


In exemplary aspects, when the level of expression of a gene or gene segment listed in Table 4 is measured or has been measured, the reference level is a cutoff correlative with a pre-determined % specificity and/or pre-determined % sensitivity, as determined by an ROC curve. In exemplary aspects, the ROC curve is based on the distribution of expression levels of the gene or gene segment listed in Table 4 of a population of responders and the distribution of expression levels of the a gene or gene segment listed in Table 4 of a population of non-responders. Exemplary definitions of responders and non-responders are found herein at Example 2. In exemplary aspects, the reference level is the point on the ROC curve at which the % specificity is X %, wherein X is selected from 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, and 99. In exemplary aspects, the reference level is the point on the ROC curve at which the % sensitivity is Y %, wherein Y is selected from 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, and 99. In exemplary aspects, the reference level is a cut off correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by the ROC curve. In exemplary aspects, the reference level is a cut off correlative with a % specificity of at least 75% and a % sensitivity of at least 75%, as determined by the ROC curve. In exemplary aspects, the reference level is a cut off correlative with a specificity of at least 85% and a % sensitivity of at least 85%, as determined by the ROC curve. In exemplary aspects, the reference level is a cut off correlative with a % specificity of at least 90% and a % sensitivity of at least 90%, as determined by the ROC curve.


In exemplary aspects, the expression level measured in the sample is above the reference level or reference value. In exemplary aspects, when the expression level measured in the sample is above the reference level or reference value and the expression level measured is that of an Ig, FCGR2B, or a gene or gene segment listed in Table 4 and labeled as having a change in gene expression level of “up”, the method comprises administering to the subject an effective amount of a proteasome inhibitor or selecting a treatment regimen comprising administration of a proteasome inhibitor. In exemplary aspects, when the expression level measured in the sample is below the reference level or reference value and the expression level measured is that of an Ig, FCGR2B, or a gene or gene segment listed in Table 4 and labeled as having a change in gene expression level of “up”, an effective amount of a proteasome inhibitor is not administered to the subject or a treatment regimen comprising administration of a proteasome inhibitor is not selected or a treatment regimen lacking administration of a proteasome inhibitor is selected.


In exemplary aspects, the expression level measured in the sample is below the reference level or reference value. In exemplary aspects, when the expression level measured in the sample is below the reference level or reference value and the expression level measured is that of a gene or gene segment listed in Table 4 and labeled as having a change in gene expression level of “down”, the method comprises administering to the subject an effective amount of a proteasome inhibitor or selecting a treatment regimen comprising administration of a proteasome inhibitor. In exemplary aspects, when the expression level measured in the sample is above the reference level or reference value and the expression level measured is that of a gene or gene segment listed in Table 4 and labeled as having a change in gene expression level of “down”, an effective amount of a proteasome inhibitor is not administered to the subject or a treatment regimen comprising administration of a proteasome inhibitor is not selected or a treatment regimen lacking administration of a proteasome inhibitor is selected.


In exemplary aspects, the expression level measured in the sample is greater than or above the reference level or reference value. The extent to which the measured expression level is above the reference level or reference value may be to any extent. In exemplary aspects, the measured expression level is at least or about 10% greater than the reference level (e.g., at least or about 15% greater than the reference level, at least or about 20% greater than the reference level, at least or about 25% greater than the reference level, at least or about 30% greater than the reference level, at least or about 35% greater than the reference level, at least or about 40% greater than the reference level, at least or about 45% greater than the reference level, at least or about 50% greater than the reference level, at least or about 55% greater than the reference level, at least or about 60% greater than the reference level, at least or about 65% greater than the reference level, at least or about 70% greater than the reference level, at least or about 75% greater than the reference level, at least or about 80% greater than the reference level, at least or about 85% greater than the reference level, at least or about 90% greater than the reference level, at least or about 95% greater than the reference level). In exemplary aspects, the measured expression level is at least 2-fold greater than the reference level, at least 3-fold greater than the reference level, at least 4-fold greater than the reference level, at least 5-fold greater than the reference level, at least 6-fold greater than the reference level, at least 7-fold greater than the reference level, at least 8-fold greater than the reference level, at least 9-fold greater than the reference level, or at least 10-fold greater than the reference level.


In exemplary aspects, the expression level measured in the sample is below or less than the reference level or reference value. The extent to which the measured expression level is below the reference level or reference value may be to any extent. In exemplary aspects, the measured expression level is at least or about 10% less than the reference level, at least or about 15% less than the reference level, at least or about 20% less than the reference level, at least or about 25% less than the reference level, at least or about 30% less than the reference level, at least or about 35% less than the reference level, at least or about 40% less than the reference level, at least or about 45% less than the reference level, at least or about 50% less than the reference level, at least or about 55% less than the reference level, at least or about 60% less than the reference level, at least or about 65% less than the reference level, at least or about 70% less than the reference level, at least or about 75% less than the reference level, at least or about 80% less than the reference level, at least or about 85% less than the reference level, at least or about 90% less than the reference level, at least or about 95% less than the reference level. In exemplary aspects, the measured expression level is at least 2-fold less than the reference level, at least 3-fold less than the reference level, at least 4-fold less than the reference level, at least 5-fold less than the reference level, at least 6-fold less than the reference level, at least 7-fold less than the reference level, at least 8-fold less than the reference level, at least 9-fold less than the reference level, or at least 10-fold less than the reference level.


In exemplary aspects, the reference level is normalized to a housekeeping gene, such as, the β-actin gene or GADPH gene. The levels may be normalized to another housekeeping gene, such as any of those described herein. In exemplary aspects, the reference level is not normalized to a housekeeping gene. In exemplary aspects, the reference level is normalized wherein the measured expression level is normalized or not normalized when the measured expression level is not normalized.


Responders and Non-Responders


As used herein, the term “responder” refers to one who has multiple myeloma, has been treated with the referenced drug, e.g., proteasome inhibitor, and has responded to treatment with the referenced drug, wherein response to treatment is as defined by the International Myeloma Working Group in Durie et al., “International uniform response criteria for multiple myeloma” Leukemia, Volume 20, No. 10, (2006). In exemplary aspects, a responder is one who has demonstrated a complete response (CR), a stringent complete response (sCR), a very good partial response (VGPR), or a partial response (PR). The definitions for a CR, sCR, VGPR, and PR are known in the art. See, Durie et al., 2006, supra. In exemplary aspects, a responder is one who has demonstrated a CR, sCR, VGPR, PR or a minimal response (MR). A response who has demonstrated an MR is one who has demonstrated at least a 25% decrease in paraprotein levels upon treatment.


As used herein, the term “non-responder” refers to one who has multiple myeloma, has been treated with the referenced drug, e.g., proteasome inhibitor, and has not responded to treatment with the reference drug, wherein response treatment is defined by the International Myeloma Working Group in Durie et al., 2006, supra. In exemplary aspects, a non-responder is one does not meet the criteria for a responder. In exemplary aspects, a non-responder is one who has demonstrated progressive disease (PD). In exemplary aspects, a non-responder is one who has demonstrated PD or stable disease (SD).


Additional Steps


In exemplary aspects, the method may include additional steps. For example, the method may include repeating one or more of the recited step(s) of the method. Accordingly, in exemplary aspects, the method comprises measuring the level of expression of Ig, FCGR2B, and/or one or more genes listed in Table 4 more than one time. In exemplary aspects, the method comprises measuring samples obtained from the subject every 6 to 12 months, wherein the measurement is based on a different biological sample obtained from the same subject.


In exemplary aspects, the method comprises measuring the sample for more than one expression level. For example, the method may comprise measuring the sample for Ig expression level and FCGR2B. In additional or alternative aspects, the method may comprise measuring the sample for at least one gene listed in Table 4. In exemplary aspects, the method may comprise measuring the sample for Ig expression and at least one gene listed in Table 4 or may comprise measuring the sample for FCGR2B expression and at least one gene listed in Table 4. In exemplary aspects, the method comprises measuring the expression level of more than one, more than two, more than three, more than four, more than five, more than six, more than seven, more than eight, more than nine, more than 10, more than 11, more than 12, more than 13, more than 14, more than 15, more than 16, more than 17, more than 18, more than 19, more than 20, more than 21, more than 22, more than 23, more than 24, more than 25, more than 26, more than 27, more than 28, more than 29, or more than 30 genes listed in Table 4. In exemplary aspects, the method comprises measuring the expression level of more than 100, more than 200, more than 300, more than 400 genes listed in Table 4. In exemplary aspects, the method comprises measuring the expression levels of all of the genes listed in Table 4.


In exemplary aspects, the method comprises measuring the RNA expression level of Ig, FCGR2B, or the one or more genes listed in Table 4, and comprises measuring the protein expression level of Ig, FCGR2B, or the one or more genes listed in Table 4.


In exemplary aspects, the subject's medical history is analyzed for expression levels of Ig, FCGR2B, and/or a gene or gene segment listed in Table 4.


In exemplary aspects, the method comprises sample preparation steps. For example, in some aspects, the method comprises selecting a specific cell population from the sample obtained from the subject. In exemplary aspects, the method comprises selecting for CD138-positive cells from the sample. The selection step may be carried out by any means known in the art, including, but not limited to FACS or chromatography. In exemplary aspects, wherein RNA expression levels are measured, the method may further comprise a step to extract or isolate the RNA from the cells of the sample. In exemplary aspects, the method comprises extracting RNA from CD138-positive tumor cells.


In exemplary aspects, wherein the method comprises measuring expression levels by measuring nucleic acids, e.g., RNA, mRNA, encoded by the Ig gene segment, the FCGR2B gene, and/or the gene listed in Table 4, the method further comprises amplifying at least a fragment of the nucleic acids to be measured. In exemplary aspects, the amplification is carried out via PCR or RT-PCR.


In exemplary aspects, the method comprises measuring the Ig expression level in the cell using a microarray platform that map to genes encoding Ig-related proteins. In exemplary aspects, the method comprises measuring the Ig protein load in the cells with an anti-human Ig antibody. In exemplary aspects, the measuring comprises measuring the presence, absence, or amount of a human Ig protein in the sample


In exemplary aspects of the inventive methods of determining a treatment regimen for a subject with a tumor, the method may optionally include an administering step, wherein a therapeutic agent or device is administered to the subject, when the expression level of Ig, FCGR2B, and/or a gene or gene segment listed in Table 4 having a change in gene expression level denoted in Table 4 as “up” is increased. For example, the methods described herein may optionally comprise a step of providing an appropriate therapy (administering a pharmaceutical agent or implementing a standard of care) to the subject determined to have a need therefor. In exemplary aspects, the therapeutic agent is a proteasome inhibitor, including those discussed herein. In exemplary aspects, the therapeutic agent is carfilzomib, bortezomib, disulfiram, or oprozomib. The therapeutic agent may be administered to the subject by any suitable route of administration known in the art, some routes of which are described herein below.


Any and all possible combinations of the steps described herein are contemplated for purposes of the inventive methods.


Tumors and Cancer


As used herein, the term “tumor” refers to an abnormal mass of tissue that results when cells divide at a higher rate than a healthy cell and/or when the cells do not die. In exemplary aspects, the tumor is a malignant tumor. In exemplary aspects, the tumor is a carcinoma, sarcoma, lymphoma, leukemia, germ cell tumor, or a blastoma. In exemplary aspects, the tumor is a hematological tumor, and in further exemplary aspects, the hematological tumor is derived from lymphoid cells. In alternative aspects, the hematological tumor is derived from myeloid cells. In exemplary aspects, the hematological tumor is a lymphoma, e.g., a Hodgkin's lymphoma or a non-Hodgkin's lymphoma. In exemplary aspects, the non-Hodgkin's lymphoma is mantle cell lymphoma. In exemplary aspects, the hematological tumor is a multiple myeloma, including, but not limited to, smouldering myeloma, relapsed multiple myeloma, or refractory myeloma. The multiple myeloma may be of any stage of the International Staging System, including Stage I, Stage II, and Stage III (Greipp et al., J Clin Oncol 23: 3412-3420 (2005). In exemplary aspects, the multiple myeloma is a Stage I, Stage II, or Stage III multiple myeloma according to the Durie-Salmon staging system (Durie et al., Cancer 36:842-854 (1975).


Samples


With regard to the methods disclosed herein, in exemplary embodiments, the sample obtained from the subject comprises a bodily fluid, including, but not limited to, blood, plasma, serum, lymph, breast milk, saliva, mucous, semen, vaginal secretions, cellular extracts, inflammatory fluids, cerebrospinal fluid, feces, vitreous humor, or urine obtained from the subject. In some aspects, the sample is a composite panel of at least two of the foregoing samples. In exemplary aspects, the sample comprises blood or a fraction thereof (e.g., plasma, serum, fraction obtained via leukopheresis). In exemplary aspects, the sample comprises white blood cells obtained from the subject. In exemplary aspects, the sample comprises a cell or cells from the tumor being treated. The tumor may be any of those described herein, including but not limited to, a hematological tumor, e.g., multiple myeloma tumor, mantle cell lymphoma. In exemplary aspects, the sample comprises bone marrow cells, e.g., intact bone marrow cells. In exemplary aspects, the sample comprises intact bone marrow cells and the method comprises contacting antibodies specific for FCGR2B or for Ig with the sample. In exemplary aspects, the sample comprises blood, serum, a biopsy sample, or bone marrow cells. In exemplary aspects, the sample comprises CD138-positive tumor cells. In exemplary aspects, the sample is a sample obtained from any of the subjects described herein. In exemplary aspects, the sample is a bone marrow aspirate.


Subjects


With regard to the methods disclosed herein, the subject in exemplary aspects is a mammal, preferably a human.


In exemplary aspects, the subject is a subject with a tumor. In exemplary aspects, the tumor is any of those mentioned herein. In exemplary aspects, the subject has cancer. In exemplary aspects, the cancer is any of those mentioned herein. In exemplary aspects, the subject has previously been treated for multiple myeloma. In exemplary aspects, the subject has previously been diagnosed with multiple myeloma. In exemplary aspects, the subject is a human patient having or suspected of having multiple myeloma, refractory multiple myeloma, or relapsed multiple myeloma. In alternative aspects, the subject has never been treated for multiple myeloma. In exemplary aspects, the subject has been newly diagnosed for multiple myeloma.


Proteasome Inhibitors


As used herein, the term “proteasome inhibitor” refers to any drug that blocks the action of proteasomes. In exemplary aspects, the proteasome inhibitor is lactacystin, bortezomib, disulfiram, epigallocatechin-3-gallate, salinosporamide A, carfilzomib, ONX0912, CEP-18770, MLN9708, epoxomicin, MG132, and the like. In exemplary aspects, the proteasome inhibitor is carfilzomib, bortezomib, disulfiram, and oprozomib. In exemplary aspects, the proteasome inhibitor is carfilzomib or bortezomic or a structural analog thereof. In exemplary aspects, the proteasome inhibitor is carfilzomib.


Formulations and Routes of Administration


With regard to the administration of a therapeutic agent, e.g., proteasome inhibitor, the agent may be administered through any suitable means, compositions and routes known in the art.


Kits


The invention further provides kits. In exemplary embodiments, the kit comprises one or more binding agents to an Ig gene or gene segment, or a gene product thereof. In exemplary aspects, the kit comprises a binding agent which specifically binds to an IgH, IgK or IgL gene segment (including orphon gene segments) or a product encoded thereby, and a binding agent to an FCGR2B gene or gene product. In exemplary embodiments, the kits comprises (i) one or more binding agents to an Ig gene segment or a product encoded thereby, optionally an IgH, IgK or IgL gene segment or product encoded thereby, or a binding agent to FCGR2B gene or gene product and (ii) at least one binding agent to a gene listed in Table 4 or a gene product encoded thereby. In exemplary embodiments, the kit comprises at least a first binding agent and a second binding agent, wherein the first binding agent binds to a first gene or gene product encoded by a first gene listed in Table 4, wherein the second binding agent binds to a second gene or gene product encoded by a second gene listed in Table 4, wherein the first gene is different from the second gene.


In exemplary aspects, the kit comprises a proteasome inhibitor, e.g., any of those described herein. In exemplary aspects, the kit comprises a container suitable for holding a blood sample. In exemplary aspects, the kit comprises a vial, a tube, a microtiter plate, a dish, a flask, or the like. In exemplary aspects, the container holds about 5 mL of fluid, or less. In exemplary aspects, the kit comprises heparin to prevent the blood from clotting. In exemplary aspects, the kit comprises reagents suitable for isolating RNA or proteins from tumor cells. In exemplary aspects, the kit comprises reagents suitable for reverse transcribing the RNA into complimentary DNA (cDNA) and for amplification of the cDNA. In exemplary aspects, the kit comprises a reagent that produces a signal indicative of a reference level


In exemplary aspects, the product encoded by said gene or gene segment is a nucleic acid molecule, e.g., an mRNA. In exemplary aspects, the binding agent is a nucleic acid probe. In exemplary aspects, the product encoded by said gene or gene segment is a protein, polypeptide, or peptide. In exemplary aspects, the binding agent is an antibody or an antigen-binding fragment thereof or a derivative thereof. In exemplary aspects, the kit comprises both nucleic acid probes and antibodies, or antigen binding fragments or derivatives thereof.


Binding Agents: Nucleic Acid Molecules


In exemplary embodiments, the binding agent is a nucleic acid molecule, e.g., a nucleic acid probe which specifically binds to (i) at least a portion of an Ig gene segment, a FCGR2B gene, or a gene or gene segment listed in Table 4, or (ii) at least a portion of a product encoded by the Ig gene segment, FCGR2B, or the gene or gene segment listed in Table 4, which product comprises nucleic acids. In exemplary aspects, the binding agent is a nucleic acid molecule which is about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45 or about 50 nucleotides in length. In exemplary aspects, the nucleic acid molecule is about 15 to about 30 nucleotides in length or about 20 to 30 nucleotides in length or about 25 to 30 nucleotides in length. In exemplary aspects, the nucleic acid molecule is about 25 nucleotides in length.


In exemplary aspects, the nucleic acid molecule comprises DNA or RNA. In exemplary aspects, the nucleic acid molecule comprises at least one non-naturally-occurring nucleotide and/or at least one non-naturally-occurring internucleotide linkage and/or one or more modified nucleotides, all of which are well known in the art. Binding Agents: Antibodies and derivatives


Any polynucleotide or polypeptide that binds the gene product may be used to detect its expression levels. In some embodiments, the polypeptide is a fragment of a receptor or ligand of the gene product. In some embodiments of the invention, the binding agent is an antibody that binds to a protein product encoded by an Ig gene segment, an FCGR2B gene, or a gene or gene segment listed in Table 4. The antibody may be any type of immunoglobulin known in the art. In exemplary embodiments, the antibody is an antibody of isotype IgA, IgD, IgE, IgG, or IgM. Also, the antibody in some embodiments is a monoclonal antibody. In other embodiments, the antibody is a polyclonal antibody.


In some embodiments, the antibody is a naturally-occurring antibody, e.g., an antibody isolated and/or purified from a mammal, or produced by a hybridoma generated from a mammalian cell. Methods of producing antibodies are well known in the art.


In some embodiments, the antibody is a genetically-engineered antibody, e.g., a single chain antibody, a humanized antibody, a chimeric antibody, a CDR-grafted antibody, a humaneered antibody, a bispecific antibody, a trispecific antibody, and the like. Genetic engineering techniques also provide the ability to make fully human antibodies in a non-human source.


In some aspects, the antibody is in polymeric, oligomeric, or multimeric form. In certain embodiments in which the antibody comprises two or more distinct antigen binding regions fragments, the antibody is considered bispecific, trispecific, or multi-specific, or bivalent, trivalent, or multivalent, depending on the number of distinct epitopes that are recognized and bound by the antibody.


Antigen Binding Fragments


In some aspects of the invention, the binding agent is an antigen binding fragment of an antibody. The antigen binding fragment (also referred to herein as “antigen binding portion”) may be an antigen binding fragment of any of the antibodies described herein. The antigen binding fragment can be any part of an antibody that has at least one antigen binding site, including, but not limited to, Fab, F(ab′)2, dsFv, sFv, diabodies, triabodies, bis-scFvs, fragments expressed by a Fab expression library, domain antibodies, VhH domains, V-NAR domains, VH domains, VL domains, and the like.


Computer Related Inventions


Computer readable-storage media are furthermore provided herein. In exemplary embodiments, the computer readable storage medium is one having stored thereon a plurality of reference levels or ranges of reference levels, each reference level or range of reference levels corresponding to (i) an expression level of Ig or (ii) an expression level of FCGR2B, or (iii) an expression level of a gene listed in Table 4, or (iv) a combination thereof; and a data value that is an expression level of Ig and/or an expression level of FCGR2B and/or an expression level of a gene listed in Table 4, measured from a cell from a sample from a patient. In exemplary aspects, the data value that is the expression level of Ig is the sum of the expression levels of more than one gene segment of the IgH, IgK, and/or IgL locus, optionally, wherein the expression level of Ig is indicative of a responder or non-responder In exemplary aspects, the computer readable storage medium is one having stored thereon (I) a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of Ig determined from a sample obtained from a responder and each data value of the second set is an expression level of Ig determined from a sample obtained from a non-responder; (II) a receiver operating characteristic (ROC) curve based on a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of Ig determined from a sample obtained from a responder and each data value of the second set is an expression level of Ig determined from a sample obtained from a non-responder; or (III) a table listing (a) a plurality of cut-off points on the ROC curve of (II), (b) the % sensitivity associated with each cut-off point of (a), and (c) the % specificity associated with each cut-off point of (a).


In exemplary embodiments, the computer readable storage medium is one having stored thereon (I) a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of FCGR2B determined from a sample obtained from a responder and each data value of the second set is an expression level of FCGR2B deteremined from a sample obtained from a non-responder; (II) a receiver operating characteristic (ROC) curve based on a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of FCGR2B determined from a sample obtained from a responder and each data value of the second set is an expression level of FCGR2B deteremined from a sample obtained from a non-responder; or (III) a table listing (a) a plurality of cut-off points on the ROC curve of (II), (b) the % sensitivity associated with each cut-off point of (a), and (c) the % specificity associated with each cut-off point of (a).


In exemplary embodiments, the computer readable storage medium is one having stored thereon (I) a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of a gene listed in Table 4 determined from a sample obtained from a responder and each data value of the second set is an expression level of a gene listed in Table 4 determined from a sample obtained from a non-responder; (II) a receiver operating characteristic (ROC) curve based on a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of a gene listed in Table 4 determined from a sample obtained from a responder and each data value of the second set is an expression level of a gene listed in Table 4 determined from a sample obtained from a non-responder; or (III) a table listing (a) a plurality of cut-off points on the ROC curve of (II), (b) the % sensitivity associated with each cut-off point of (a), and (c) the % specificity associated with each cut-off point of (a). In exemplary aspects, the computer readable storage medium comprises two or more of the foregoing media.


In exemplary embodiments, the computer readable storage medium is one having stored thereon machine-readable instructions executable by a processor, comprising: (a) instructions for receiving a data value, α, relating to a test level of Ig expression from a sample obtained from a test subject, e.g., a level of Ig expression measured from a sample obtained from a test subject; and (b) instructions for displaying an output relating to treating the test subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the computer readable storage medium is one having stored thereon machine-readable instructions executable by a processor, comprising: (a) instructions for receiving a data value, α, relating to a test level of FCGR2B expression from a sample obtained from a test subject, e.g., a level of FCGR2B expression measured from a sample obtained from a test subject; and (b) instructions for displaying an output relating to treating the test subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the computer readable storage medium is one having stored thereon machine-readable instructions executable by a processor, comprising: (a) instructions for receiving a data value, α, relating to a test level of expression of a gene listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “up”, e.g., a level of expression of the gene listed in Table 4 measured from a sample obtained from a test subject; and (b) instructions for displaying an output relating to treating the test subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the computer readable storage medium is one having stored thereon machine-readable instructions executable by a processor, comprising: (a) instructions for receiving a data value, α, relating to a test level of expression of a gene listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “down”, e.g., a level of expression of the gene listed in Table 4 measured from a sample obtained from a test subject; and (b) instructions for displaying an output relating to treating the test subjectfor multiple myeloma with a proteasome inhibitor, when α is less than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


The invention additionally provides systems comprising: a processor; a memory device coupled to the processor, and machine readable instructions stored on the memory device. In exemplary embodiments, the machine readable instructions, when executed by the processor, cause the processor to: (i.) receive a data value, α, relating to a test level of Ig expression from a sample obtained from a test subject, e.g., a level of expression of Ig measured from a sample obtained from a test subject; and (ii) display an output relating to treating the test subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the machine readable instructions, when executed by the processor, cause the processor to: (i) receive a data value, α, relating to a test level of FCGR2B expression from a sample obtained from a test subject, e.g., a level of expression of FCGR2B measured from a sample obtained from a test subject; and (ii) display an output relating to treating the test subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the machine readable instructions, when executed by the processor, cause the processor to: (i) receive a data value, α, relating to a test level of expression of a gene listed in Table 4, wherein the change in gene expression level for the gene is denoted in Table 4 as “up”, from a sample obtained from a test subject, e.g., a level of expression of the gene listed in Table 4 measured from a sample obtained from a test subject; and (ii) display an output relating to treating the test subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


In exemplary embodiments, the machine readable instructions, when executed by the processor, cause the processor to: (i) receive a data value, α, relating to a test level of expression of a gene listed in Table 4, wherein the change in gene expression level for the gene is denoted in Table 4 as “down”, from a sample obtained from a test subject e.g., a level of expression of the gene listed in Table 4 measured from a sample obtained from a test subject; and (ii) display an output relating to treating the test subject for multiple myeloma with a proteasome inhibitor, when α is less than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


The invention further provides methods implemented by a processor in a computer. In exemplary embodiments, the method comprises the steps of: (a) receiving a data value, α, relating to a test level of Ig expression from a sample obtained from a test subject e.g., a level of expression of Ig measured from a sample obtained from a test subject; and (b) displaying an output relating to treating the test subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve. In exemplary embodiments, the method comprises the steps of: (a) receiving a data value, α, relating to a test level of FCGR2B expression from a sample obtained from a test subject e.g., a level of expression of FCGR2B measured from a sample obtained from a test subject; and (b) displaying an output relating to treating the test subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve. In exemplary embodiments, the method comprises the steps of: (a) receiving a data value, α, relating to a test level of expression of a gene listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “up” e.g., a level of expression of the gene listed in Table 4 measured from a sample obtained from a test subject; and (b) displaying an output relating to treating the test subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve. In exemplary embodiments, the method comprises the steps of: (a) receiving a data value, α, relating to a test level of expression of a gene listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “down” e.g., a level of expression of the gene listed in Table 4 measured from a sample obtained from a test subject; and (b) displaying an output relating to treating the test subject for multiple myeloma with a proteasome inhibitor, when α is less than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.



FIG. 10 illustrates an exemplary embodiment 101 of a system 100 for determining a therapeutic regimen for a subject with a tumor. Generally, the system 100 may include one or more client devices 102, a network 104, and a database 108. Each client device 102 may be communicatively coupled to the network 104 by one or more wired or wireless network connections 112, which may be, for example, a connection complying with a standard such as one of the IEEE 802.11 standards (“Wi-Fi”), the Ethernet standard, or any other appropriate network connection. Similarly, the database 108 may be communicatively coupled to the network 104 via one or more connections 114. (Of course, the database could alternatively be internal to one or more of the client devices 102.) The database 108 may store data related to the determination of the therapeutic regimen for a subject with a tumor including, but not limited to, data of a sample obtained from the subject, data of a sample obtained from a subject from a non-responder category or from a responder category, etc. The data of the samples may be, for example, related to one or more of a level of expression of an Ig gene segment, FCGR2B, or a gene or gene segment listed in Table 4.


As will be understood, the network 104 may be a local area network (LAN) or a wide-area network (WAN). That is, network 104 may include only local (e.g., intra-organization) connections or, alternatively, the network 104 may include connections extending beyond the organization and onto one or more public networks (e.g., the Internet). In some embodiments, for example, the client device 102 and the database 108 may be within the network operated by a single company (Company A). In other embodiments, for example, the client device(s) 102 may be on a network operated by Company A, while the database 108 may be on a network operated by a second company (Company B), and the networks of Company A and Company B may be coupled by a third network such as, for example, the Internet.


Referring still to FIG. 10, the client device 102 includes a processor 128 (CPU), a RAM 130, and a non-volatile memory 132. The non-volatile memory 132 may be any appropriate memory device including, by way of example and not limitation, a magnetic disk (e.g., a hard disk drive), a solid state drive (e.g., a flash memory), etc. Additionally, it will be understood that, at least with regard to FIG. 10, the database 108 need not be separate from the client device 102. Instead, in some embodiments, the database 108 is part of the non-volatile memory 132 and the data 122, 124, 126 may be stored as data within the memory 132. For example, the data 122 may be included as data in a spreadsheet file stored in the memory 132, instead of as data in the database 108. In addition to storing the records of the database 108 (in some embodiments), the memory 132 stores program data and other data necessary to analyze data of one or more sample and/or control populations, determine a mean of the data, determine a threshold against which data of the subject may be compared, and/or determine the therapeutic regimen for a subject with a tumor. For example, in an embodiment, the memory 132 stores a first routine 134, a second routine 136, and a third routine 138. The first routine 134 may receive data values related to one or more sample and/or control populations, and determine a mean of the data values received by the routine 134. The second routine 136 may compute one or more statistical parameters of the data collected by the first routine 134, such as determining a mean value, a standard deviation value, etc. Additionally and/or alternatively, the second routine 136 may set a first cutoff against which data from one or more subjects may be compared in order to determine the therapeutic regiment for a subject with a tumor. The third routine 138 may, for example, receive data for one or more subjects, compare the data of the one or more subjects to the cutoff value(s) determined by the second routine 136, and/or determine the therapeutic regimen for a subject with a tumor according to the comparison of the subject's data to the cutoff value. Regardless, each of the routines is executable by the processor 128 and comprises a series of compiled or compilable machine-readable instructions stored in the memory 132. Additionally, the memory 132 may store generated reports or records of data output by one of the routines 134 or 136. Alternatively, the reports or records may be output to the database 108. One or more display/output devices 140 (e.g., printer, display, etc.) and one or more input devices 142 (e.g., mouse, keyboard, tablet, touch-sensitive interface, etc.) may also be coupled to the client device 102, as is generally known.


As will be understood, although individual operations of one or more methods are illustrated and described as separate operations, one or more of the individual operations may be performed concurrently, and nothing requires that the operations be performed in the order illustrated. Structures and functionality presented as separate components in example configurations may be implemented as a combined structure or component. Similarly, structures and functionality presented as a single component may be implemented as separate components. These and other variations, modifications, additions, and improvements fall within the scope of the subject matter herein.


For example, the network 104 may include but is not limited to any combination of a LAN, a MAN, a WAN, a mobile, a wired or wireless network, a private network, or a virtual private network. Moreover, while only two clients 102 are illustrated in FIG. 10 to simplify and clarify the description, it is understood that any number of client computers are supported and can be in communication with one or more servers (not shown).


Additionally, certain embodiments are described herein as including logic or a number of routines. Routines may constitute either software routines (e.g., code embodied on a machine-readable medium or in a transmission signal) or hardware routines. A hardware routine is tangible unit capable of performing certain operations and may be configured or arranged in a certain manner. In example embodiments, one or more computer systems (e.g., a standalone, client or server computer system) or one or more hardware routines of a computer system (e.g., a processor or a group of processors) may be configured by software (e.g., an application or application portion) as a hardware routine that operates to perform certain operations as described herein.


Similarly, the methods or routines described herein may be at least partially processor-implemented. For example, at least some of the operations of a method may be performed by one or processors or processor-implemented hardware modules. The performance of certain of the operations may be distributed among the one or more processors, not only residing within a single machine, but deployed across a number of machines. In some example embodiments, the processor or processors may be located in a single location (e.g., within a home environment, an office environment or as a server farm), while in other embodiments the processors may be distributed across a number of locations.


The performance of certain of the operations may be distributed among the one or more processors, not only residing within a single machine, but deployed across a number of machines. In some example embodiments, the one or more processors or processor-implemented modules may be located in a single geographic location (e.g., within a home environment, an office environment, or a server farm). In other example embodiments, the one or more processors or processor-implemented modules may be distributed across a number of geographic locations.


Some embodiments may be described using the expression “coupled” and “connected” along with their derivatives. For example, some embodiments may be described using the term “coupled” to indicate that two or more elements are in direct physical or electrical contact. The term “coupled,” however, may also mean that two or more elements are not in direct contact with each other, but yet still co-operate or interact with each other. The embodiments are not limited in this context.


As used herein, the terms “comprises,” “comprising,” “includes,” “including,” “has,” “having” or any other variation thereof, are intended to cover a non-exclusive inclusion. For example, a process, method, article, or apparatus that comprises a list of elements is not necessarily limited to only those elements but may include other elements not expressly listed or inherent to such process, method, article, or apparatus. Further, unless expressly stated to the contrary, “or” refers to an inclusive or and not to an exclusive or. For example, a condition A or B is satisfied by any one of the following: A is true (or present) and B is false (or not present), A is false (or not present) and B is true (or present), and both A and B are true (or present).


In addition, use of the “a” or “an” are employed to describe elements and components of the embodiments herein. This is done merely for convenience and to give a general sense of the description. This description should be read to include one or at least one and the singular also includes the plural unless it is obvious that it is meant otherwise.


ADDITIONAL EMBODIMENTS

The inventions described herein are based, in part, on the discovery that (i) high levels of immunoglobulin expression in cancer cells (e.g., hematological tumor cells) correlate with response to one or more proteasome inhibitors (e.g., carfilzomib, bortezomib, oprozomib); (ii) increased levels of FCGR2B expression in cancer cells (e.g., hematological tumor cells) correlate with response to one or more proteasome inhibitors (e.g., carfilzomib, bortezomib, oprozomib); and/or (iii) decreased levels of FCGR2B expression in cancer cells (e.g., hematological tumor cells) correlate with non-response to proteasome inhibitors (e.g., carfilzomib, bortezomib, oprozomib).


Provided herein are methods for treatment of tumors and/or determining efficacy of a treatment of a tumor with a proteasome inhibitor (e.g., carfilzomib, bortezomib, oprozomib) in a subject by determining the level of Ig expression or FCGR2B expression in a sample obtained from the tumor. These methods require detecting the level of Ig expression or overall Ig protein load in a sample or FCGR2B expression or overall FCGR2B protein load in a sample.


The disclosed methods can be employed to determine the efficacy of treatments for multiple myeloma in subjects who are undergoing carfilzomib therapy and/or in subjects who are undergoing therapy with other chemotherapeutic agents including, but not limited to, other proteasome inhibitors (e.g., oprozomib, bortezomib). Also provided are methods of selecting a subject for participation in a clinical study that include determining the level of Ig expression or FCGR2B expression in a sample (e.g., a biological sample) obtained form a subject having a tumor, or at risk for having a tumor.


The invention also provides additional methods of treating a tumor. In exemplary aspects, the method includes detecting the level of Ig expression or FCGR2B expression in a sample (e.g., a biological sample) obtained from a subject identified as having, or at risk for having a tumor, wherein a difference in the Ig expression level or FCGR2B expression level in the tumor cell compared to a reference level is an indication of the subject's responsiveness to treatment with a proteasome inhibitor. In one embodiment, the Ig expression level or FCGR2B expression level in the tumor cell is elevated as compared to the reference level, and the elevated levels indicate that the tumor is sensitive (e.g., susceptible) to the therapy with one or more proteasome inhibitors. According to one aspect, the methods disclosed herein comprise administering to subjects having elevated levels of Ig expression or FCGR2B expression in the tumor cell as compared to the reference level an effective amount of one or more proteasome inhibitors (e.g. carfilzomib, bortezomib or oprozomib). In one embodiment, the subject is administered an effective amount of carfilzomib.


In another embodiment, the Ig expression level or FCGR2B expression level in the tumor cell is reduced as compared to the reference level, and the reduced levels indicate that the tumor is not sensitive to the therapy with one or more proteasome inhibitors. According to one aspect, the methods disclosed herein comprise administering to subjects having reduced levels of Ig expression or FCGR2B expression in the tumor cell as compared to the reference level an effective amount of a chemotherapeutic agent other than a proteasome inhibitor. In one embodiment, the subject is administered an effective amount of a chemotherapeutic agent other than a carfilzomib. In some embodiments, the tumor is a hematologic tumor (e.g., a myeloma).


In exemplay aspects, the method of treating a tumor in a subject comprises identifying a subject having a tumor, or at risk for having a tumor; providing a sample comprising a cell from a tumor; detecting the level of immunoglobulin (Ig) expression in the sample; identifying a subject with elevated levels of Ig expression in a cell of the tumor as compared to a reference level; and administering to the identified subject an effective amount of a proteasome inhibitor. In exemplary aspects, the method comprises identifying a subject having a tumor, or at risk for having a tumor; providing a sample comprising a cell from a tumor; detecting the level of immunoglobulin (Ig) in the sample; identifying a subject with reduced levels of Ig in a cell of the tumor as compared to a reference level; and administering to the subject a chemotherapeutic agent other than a proteasome inhibitor.


In exemplary aspects, the method of treating a tumor in a subject comprises identifying a subject having a tumor, or at risk for having a tumor; providing a sample comprising a cell from a tumor; detecting the level of Fc gamma receptor 2B (FCGR2B) expression in the sample; identifying a subject with elevated levels of FCGR2B expression in a cell of the tumor as compared to a reference level; and administering to the identified subject an effective amount of a proteasome inhibitor. In exemplary aspects, the method comprises identifying a subject having a tumor, or at risk for having a tumor; providing a sample comprising a cell from a tumor; detecting the level of FCGR2B in the sample; identifying a subject with reduced levels of FCGR2B in a cell of the tumor as compared to a reference level; and administering to the subject a chemotherapeutic agent other than a proteasome inhibitor.


The invention also provides a method for determining whether to treat a subject having a tumor, e.g., a hematological tumor, with a proteasome inhibitor, e.g., carfilzomib. In exemplary aspects, the method of determining whether to treat a subject comprises identifying a subject having a hematological tumor, or at risk for having a hematological tumor. In one aspect, the hematological tumor is multiple myeloma. The level of Ig expression or FCGR2B expression in a sample (e.g., a biological sample) obtained from a subject is detected, and compared to a reference level. According to one aspect, the methods include determining to treat the subject with carfilzomib if the sample has elevated levels of Ig or FCGR2B, as compared to a reference level. In one embodiment, the subject is administered an effective amount of carfilzomib. Conversely, the methods include determining to treat the subject with a chemotherapeutic agent other than carfilzomib if the sample has reduced levels of Ig or FCGR2B as compared to a reference level. In exemplary aspects, the method comprises identifying a subject having a tumor, or at risk for having a tumor; providing a sample comprising a cell from the tumor; detecting the level of immunoglobulin (Ig) expression in the sample; and determining to treat a subject with a proteasome inhibitor, e.g., carfilzomib, if the subject has elevated levels of Ig in the sample as compared to a reference level. In exemplary aspects, the method further includes the step of administering to subject an effective amount of a proteasome inhibitor, e.g., carfilzomib.


In alternative or additional aspects, the method comprises identifying a subject having, at risk for, having a tumor, e.g., a hematological tumor; providing a sample comprising a cell from the tumor; detecting the level of FCGR2B expression in the sample; and determining to treat a subject with a proteasome inhibitor, e.g, carfilzomib, if the subject has elevated levels of FCGR2B in the sample as compared to a reference level. In exemplary aspects, the method further includes the step of administering to subject an effective amount of a proteasome inhibitor, e.g., carfilzomib.


The invention further provides a method of predicting the sensitivity of a tumor (e.g., a hematological tumor) to treatment with a proteasome inhibitor. In exemplary embodiments, the methods comprise identifying a subject having a tumor, or at risk for having a tumor. In one aspect, the tumor is multiple myeloma. These methods include detecting the level of Ig expression or FCGR2B expression in a sample (e.g., a biological sample) obtained from a subject identified as having, or at risk for having a tumor, and predicting that the tumor will be sensitive to (e.g., susceptible to) treatment with proteasome inhibitor if the sample has elevated levels of Ig or FCGR2B in the sample relative to a reference level. In one embodiment, the Ig expression level or the FCGR2B expression level in the sample is elevated as compared to the reference level, and the elevated levels indicate that the tumor is sensitive (e.g., susceptible) to the therapy with a proteasome inhibitor. According to one aspect, the methods disclosed herein comprise administering to subjects having elevated levels of Ig expression or FCGR2B expression in the sample as compared to the reference level an effective amount of proteasome inhibitor, including carfilzomib. In exemplary aspects, the method comprises identifying a subject having a tumor, or at risk for having a tumor; providing a sample comprising a cell from the tumor; detecting the level of Ig expression in the sample; comparing the level of Ig expressiong in the sample with a reference level; predicting that the tumor will be sensitive to treatment with a proteasome inhibitor if the sample has elevated levels of Ig in the sample relative to a reference level. In exemplary aspects, the method further includes the step of administering to the subject having a tumor predicted to be sensitive to treatment with a proteasome inhibitor, e.g., carfilzomib, an effective amount of a proteasome inhibitor, e.g., carfilzomib. In alternative or additional aspects, the method comprises identifying a subject having a tumor, or at risk for having a tumor; providing a sample comprising a cell from the tumor; detecting the level of FCGR2B expression in the sample; comparing the level of FCGR2B expression in the sample with a reference level; predicting that the tumor will be sensitive to treatment with a proteasome inhibitor if the sample has elevated levels of FCGR2B in the sample relative to a reference level. In exemplary aspects, the method further includes the step of administering to the subject having a tumor predicted to be sensitive to treatment with proteasome inhibitor, e.g., carfilzomib, an effective amount of a proteasome inhibitor, e.g., carfilzomib.


In another embodiment, the methods include detecting the level of Ig expression or FCGR2B expression in a sample (e.g., a biological sample) obtained from a subject identified as having, or at risk for having a tumor, and predicting that the tumor will not be sensitive (e.g., susceptible to) to treatment with proteasome inhibitor if the sample has reduced levels of Ig or FCGR2B in the sample relative to a reference level. According to one aspect, the methods disclosed herein comprise administering to subjects having reduced levels of Ig expression or FCGR2B expression in the tumor cell as compared to the reference level an effective amount of a chemotherapeutic agent other than a proteasome inhibitor. In one embodiment, the methods further comprise administering to the subject an effective amount of a chemotherapeutic agent other than a proteasome inhibitor (e.g., a chemotherapeutic agent other than carfilzomib).


Some embodiments, where the treatment has been indicated to be ineffective in the subject, further include administering, recommending, or prescribing an alternate treatment to the subject. In some embodiments, the alternate treatment can be a different therapeutic agent or a different combination of one or more therapeutic agents. In some embodiments, the alternate treatment can be an increased dosage of one or more therapeutic agents currently being taken by the subject, an increase in the frequency of administration of one or more therapeutic agents currently being taken by the subject, or an alteration in the route of delivery of one or more therapeutic agents being currently taken by the subject.


In exemplary aspects of the foregoing methods, the proteasome inhibitor is selected from the group consisting of carfilzomib, bortezomib and oprozomib. In exemplary aspects, the tumor is a hematological tumor, including, but not limited to multiple myeloma. In exemplary aspecst, the the cell is a CD138+ tumor cell.


In exemplary aspects of the foregoing methods comprising detecting the level of immunoglobulin (Ig) in the sample, the detecting comprises amplifying a fragment of a human Ig mRNA. In one embodiment, the detecting comprises measuring the Ig expression level in the cell using a microarray platform that map to genes encoding Ig-related proteins. In yet another embodiment, the detecting comprises measuring the Ig expression level in the cells using an anti-Ig antibody (e.g., an anti-human Ig antibody). In exemplary aspects of the foregoing methods comprising detecting the level of immunoglobulin (Ig) in the sample, the detecting comprises (i) amplifying a fragment of a human Ig mRNA; (ii) measuring the Ig expression level in the cell using a microarray platform that map to genes encoding Ig-related proteins; or (iii) measuring the Ig protein load in the cells with an anti-human Ig antibody.


In exemplary aspects of the foregoing methods comprising detecting the level of FCGR2B in the sample, the detecting comprises amplifying a fragment of a human FCGR2B mRNA. In one embodiment, the detecting comprises measuring the FCGR2B expression level in the cell using a microarray platform that map to genes encoding FCGR2B. In yet another embodiment, the detecting comprises measuring the Ig expression level in the cells using an anti-FCGR2B antibody (e.g., an anti-human FCGR2B antibody). In exemplary aspects of the foregoing methods comprising detecting the level of FCGR2B in the sample, the detecting comprises (i) amplifying a fragment of a human FCGR2B mRNA; (ii) measuring the FCGR2B expression level in the cell using a microarray platform that map to genes encoding FCGR2B; or (iii) measuring the FCGR2B protein load in the cells with an anti-human FCGR2B antibody.


The invention moreover provides a method of inhibiting proliferation of multiple myeloma cells in a subject. In exemplary aspects, the methods include detecting the level of Ig expression or FCGR2B expression level in multiple myeloma cells obtained from a subject, wherein a difference in the Ig expression level or FCGR2B expression level in the multiple myeloma cells compared to a reference level is an indication that treating the subject with a proteasome inhibitor will inhibit proliferation of multiple myeloma cells in the subject. In an exemplary embodiment, the Ig expression level or FCGR2B expression level in the multiple myeloma cells is elevated as compared to the reference level, and the elevated levels indicate that treating the subject with a proteasome inhibitor will inhibit proliferation of multiple myeloma cells in the subject. According to one aspect, the methods disclosed herein comprise administering to the subject an effective amount of a proteasome inhibitor. In one embodiment, the subject is administered an effective amount of carfilzomib. In another embodiment, the methods disclosed herein include designing a chemotherapeutic regimen comprising carfilzomib effective to inhibit proliferation of the tumor cells; and administering carfilzomib to the subject thereby treating the tumor.


In another embodiment, the Ig expression level or FCGR2B expression level in the multiple myeloma cells is reduced as compared to the reference level, and the reduced levels indicate that treating the subject with a proteasome inhibitor may not inhibit proliferation of multiple myeloma cells in the subject. According to one aspect, the methods disclosed herein comprise administering to the subjects an effective amount of a chemotherapeutic agent other than a proteasome inhibitor. In one embodiment, the subject is administered an effective amount of a chemotherapeutic agent other than a carfilzomib.


In exemplary aspects, the method of inhibiting proliferation of multiple myeloma cells in a subject comprises identifying a subject having, at risk for, or suspected of having multiple myeloma; detecting the level of Ig expression in multiple myeloma cells obtained from the subject; comparing the level of Ig expression in the multiple myeloma cells with a control sample; and a) administering to the subject an effective amount of a proteasome inhibitor to the subject if it has been determined that the multiple myeloma cells have elevated Ig expression relative to a reference level; orb) administering to the subject an effective amount of a chemotherapeutic agent other than a proteasome inhibitor to the subject if it has been determined that the multiple myeloma cells have reduced Ig expression relative to a reference level. In alternative or additional aspects, the method comprises identifying a subject having, at risk for, or suspected of having multiple myeloma; detecting the level of FCGR2B expression in multiple myeloma cells obtained from the subject; comparing the level of FCGR2B expression in the multiple myeloma cells with a control sample; and a) administering to the subject an effective amount of a proteasome inhibitor to the subject if it has been determined that the multiple myeloma cells have elevated FCGR2B expression relative to a reference level; or b) administering to the subject an effective amount of a chemotherapeutic agent other than a proteasome inhibitor to the subject if it has been determined that the multiple myeloma cells have reduced or unchanged FCGR2B expression relative to a reference level.


In exemplary aspects, the methods provided herein, (e.g., the method of determining whether to treat a subject), further comprises designing a chemotherapeutic regimen comprising carfilzomib effective to inhibit proliferation of the tumor cells (e.g., multiple myeloma cells); and administering carfilzomib to the subject thereby treating the tumor.


In exemplary aspects of any of the foregoing methods, the reference level of Ig is based on the Ig expression level determined from a plasma cell obtained from a healthy individual or the reference level of FCGR2B is based on the FCGR2B expression level determined from a plasma cell obtained from a healthy individual. In exemplary aspects, the reference level of Ig expression is based on the Ig expression level determined from a plasma cell obtained from an individual classified as non-responsive to therapy with a proteasome inhibitor. In exemplary embodiments, the reference level of FCGR2B expression is based on the FCGR2B expression level determined from a plasma cell obtained from a healthy individual. In another embodiment, the reference level of FCGR2B expression is based on the FCGR2B expression level determined from a plasma cell obtained from an individual classified as non-responsive to therapy with a proteasome inhibitor.


In exemplary aspects of any of the foregoing methods, the subject has previously been treated for multiple myeloma or has previously been diagnosed with multiple myeloma. In exemplary aspects of any of the foregoing methods, the sample is a blood, serum, or biopsy sample.


Also, in exemplary aspects of any of the foregoing methods, the detecting comprises amplifying a fragment of a human Ig mRNA or amplifying a fragment of a human FCGR2B mRNA. Optionally, the amplifying is by polymerase chain reaction (PCR) or RT-PCR. In exemplary aspects, the amplifying employs a detectably-labeled primer or probe.


With regard to the foregoing methods, the Ig expression levels or FCGR2B expression levels in a biological sample can be determined, for example, by using one or more oligonucleotides that are specific for genes encoding Ig-related proteins or for FCGR2B. For example, the levels of mRNA corresponding to a human Ig or human FCGR2B can be detected using oligonucleotides in Southern hybridizations, in situ hybridizations, and quantitative real-time PCR amplification (qRT-PCR). A plurality of oligonucleotides specific for a plurality genes encoding Ig-related proteins can be employed in an array format wherein each oligonucleotide is immobilized at a pre-determined location on a substrate, such as nitrocellulose membrane. Methods for performing such assays are well known to those of skill in the art.


The oligonucleotides employed in such methods are generally single-stranded molecules, such as synthetic antisense molecules or cDNA fragments, and are, for example, 6-60 nt, 15-30 or 20-25 nt in length.


Oligonucleotides specific for a polynucleotide encoding Ig-related proteins are prepared using techniques well known to those of skill in the art. For example, oligonucleotides can be designed using known computer algorithms to identify oligonucleotides of a defined length that are unique to the polynucleotide, have a GC content within a range suitable for hybridization, and lack predicted secondary structure that may interfere with hybridization. Oligonucleotides can be synthesized using methods well known to those in the art. For use in array formats, the oligonucleotides may be synthesized directly on the surface of a substrate. Oligonucleotides specific for the prostate cancer biomarkers disclosed herein are known in the art and are commercially available.


In certain embodiments, the oligonucleotides are labeled using one or more detectable moieties. DNA or mRNA isolated from a biological sample is contacted with the labeled oligonucleotides under conditions that allow for formation of hybridization complexes, and the amount of label associated with the hybridization complexes is measured and compared to a standard value.


In alternative or additional aspects, the detecting comprises measuring the presence, absence, or amount of a human Ig protein or FCGR2B protein in the test sample. In exemplary aspects, the measuring uses an antibody that specifically binds to a human Ig protein or a human FCGR2B protein. Antibodies that bind to human Ig employed in the present methods, together with ELISA kits that employ such antibodies for the detection of human Ig employed herein, are well known to those of skill in the art and are available commercially. Optionally, the measuring is by an ELISA assay, a western blot assay, or an immunohistochemical assay.


In certain embodiments, the Ig or FCGR2B, expression level is determined using a binding agent, such as a protein, antibody or antibody fragment, that specifically binds to the human Ig or FCGR2B, for example in an enzyme-linked immunosorbent assay (ELISA), radioimmunoassay, antibody array, Western blot, immunohistochemical, immunoprecipitation or immunofluoresence assay. Methods for performing such assays are well known to those of skill in the art.


Solid phase substrates, or carriers, that can be effectively employed in such assays are well known to those of skill in the art and include, for example, 96 well microtiter plates, glass, paper, and microporous membranes constructed, for example, of nitrocellulose, nylon, polyvinylidene difluoride, polyester, cellulose acetate, mixed cellulose esters and polycarbonate.


With regard to the foregoing methods, in exemplary aspects, the subject is a human patient having or suspected of having multiple myeloma, refractory multiple myeloma, or relapsed multiple myeloma.


In exemplary embodiments, the method further comprises administering to the subject one or more chemotherapeutic agents other than a proteasome inhibitor (e.g., carfilzomib, bortezomib or oprozomib). In some embodiments, the subject is a participant in a clinical trial. In alternative embodiments, the method further comprises administering to the subject one or more chemotherapeutic agents other than a proteasome inhibitor.


In exemplary embodiments, the difference is an increase in the level of Ig expression or FCGR2B expression in the tumor cell compared to the reference level and the increase is prognostic for an improved overall survival of the subject undergoing the therapy, compared to individuals afflicted with multiple myeloma that do not have the increase in the expression level of Ig or FCGR2B in the tumor cell compared to a reference level.


In one embodiment, the difference is a decrease in the level of Ig expression in the tumor cell in the subject compared to the reference level and the decrease indicates that the multiple myeloma is resistant to therapy with one or more proteasome inhibitors. In one embodiment, the difference is a decrease in the level of Ig expression in the tumor cell compared to the reference level and the decrease is prognostic for a diminished overall survival of the subject compared to individuals that do not have the decrease in the level of Ig expression in the tumor cell. In exemplary aspects, the difference is an decrease in the level of FCGR2B expression in the tumor cell compared to the reference level and the decrease is prognostic for an diminished overall survival of the subject undergoing the therapy, compared to individuals afflicted with multiple myeloma that do not have an decrease in the expression level of FCGR2B in the tumor cell compared to a reference level.


In exemplary embodiments, the method further includes determining whether the patient will be a candidate for therapy with one or more proteasome inhibitors, prior to the administering, wherein an increase in a level of Ig expression or FCGR2B expression in a tumor cell from the patient compared to a reference level indicates that the patient is a candidate for the foregoing therapies.


Provided herein are methods for determining whether to treat a patient having a tumor (e.g., a hematological tumor) with a proteasome inhibitor (e.g., carfilzomib) that include identifying a subject having, at risk for, having a hematological tumor, providing a sample comprising a cell from the tumor, detecting the level of Ig expression in the sample, determining to treat the patient with a proteasome inhibitor (e.g., carfilzomib) if the level of Ig expression in a cell of the tumor is greater than a predetermined reference level. In some embodiments, the hematological tumor is a myeloma. In some embodiments, the myeloma is multiple myeloma.


Provided herein are methods for determining whether to treat a patient having a tumor (e.g., a hematological tumor, such as multiple myeloma) with a proteasome inhibitor (e.g., carfilzomib, oprozomib, or bortezomib) that include identifying a subject having a tumor, or at risk for having a tumor, providing a sample comprising a cell from the tumor, detecting the level of FCGR2B expression in the sample, determining to treat the patient with a chemotherapeutic agent other than a proteasome inhibitor when if the level of FCGR2B expression in a cell of the tumor is less than a predetermined reference level. In some embodiments, the hematological tumor is a myeloma. In some embodiments, the myeloma is multiple myeloma.


Some embodiments further include assessing, or alternatively obtaining, providing, or using previously determined information regarding, the level of Ig expression or FCGR2B expression from samples taken from a control population of normal, or healthy (disease-free) subjects.


Some embodiments further include recording the results of these methods in the subject's medical records (e.g., recording the results in a computer readable medium) or performing a diagnostic test. In some embodiments, these methods can be performed by a medical professional (e.g., a physician, a physician's assistant, a nurse, a nurse's assistant, or a laboratory technician).


In one aspect, the disclosure generally provides compositions, which include therapeutic agents and pharmaceutically acceptable carriers therefor.


In this regard, the disclosure provides for one or more therapeutic agents, e.g., proteasome inhibitors, which are administered to a MM patient. The therapeutic agents can be administered to a patient prior to, during, or after other conventional chemotherapeutic treatments. In one embodiment, the therapeutic agents are administered to a patient subsequent to determining that the patient is a candidate for such treatment. In this respect, the therapeutic agents are administered to a subject, prior to, or in combination with, conventional chemotherapeutic treatments.


In one aspect, the therapeutic agents, alone or in combination, are administered to a patient in an effective amount, e.g., a therapeutically effective dose of proteasome inhibitor or other chemotherapeutic agent. A therapeutic dose may vary depending upon the type of therapeutic agent, route of administration, and dosage form. Dosage unit forms generally contain between from about 1 mg to about 500 mg of an active ingredient. The preferred composition or compositions is a formulation that exhibits a high therapeutic index. The therapeutic index is the dose ratio between toxic and therapeutic effects which can be expressed as the ratio between LD50 and ED50. The LD50 is the dose lethal to 50% of the population and the ED50 is the dose therapeutically effective in 50% of the population. The LD50 and ED50 are determined by standard pharmaceutical procedures in animal cell cultures or experimental animals.


Specific dosages may be adjusted depending on conditions of disease, the age, body weight, general health conditions, sex, and diet of the subject, dose intervals, administration routes, excretion rate, and combinations of drugs. Any of the above dosage forms containing effective amounts are well within the bounds of routine experimentation.


In the compositions for treating multiple myeloma described herein, the therapeutically effective amount of the proteasome inhibitor can range from about 0.001 mg/kg to about 30 mg/kg body weight of the subject. In some embodiments, the therapeutically effective amount of the agent can range from about 0.05 mg/kg to about 30 mg/kg, from about 0.1 mg/kg to about 30 mg/kg, from about 1 mg/kg to about 25 mg/kg, from about 1 mg/kg to about 20 mg/kg, or from about 1 or 2 mg/kg to about 15 mg/kg.


The methods described herein include the manufacture and use of pharmaceutical compositions, which include compounds (e.g., proteasome inhibitors and/or other chemotherapeutic agents) identified by a method described herein as active ingredients. Also included are the pharmaceutical compositions themselves.


Pharmaceutical compositions typically include a pharmaceutically acceptable carrier. As used herein the language “pharmaceutically acceptable carrier” includes saline, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Supplementary active compounds can also be incorporated into the compositions, e.g., chemotherapeutic agents.


Pharmaceutical compositions are typically formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.


Methods of formulating suitable pharmaceutical compositions are known in the art, see, e.g., Remington: The Science and Practice of Pharmacy, 21st ed., 2005; and the books in the series Drugs and the Pharmaceutical Sciences: a Series of Textbooks and Monographs (Dekker, N.Y.). For example, solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfate; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.


Pharmaceutical compositions suitable for injectable use can include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, aluminum monostearate and gelatin.


Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle, which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying, which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.


Oral compositions generally include an inert diluent or an edible carrier. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules, e.g., gelatin capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.


In one embodiment, the therapeutic compounds are prepared with carriers that will protect the therapeutic compounds against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Such formulations can be prepared using standard techniques, or obtained commercially, e.g., from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to selected cells with monoclonal antibodies to cellular antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.


The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.


With regard to the inventions described in the section titled “Additional Embodiments” the following applies:


As disclosed herein, the term “immunoglobulin expression” or “Ig expression” refers to the expression level of one or more of the known immunoglobulin classes including IgA, IgG, IGM, IgE, and IgD.


As used herein, the term “immunoglobulin” refers to a protein consisting of one or more polypeptide(s) substantially encoded by immunoglobulin genes. The recognized immunoglobulin genes include the different constant region genes as well as the myriad immunoglobulin variable region genes. Immunoglobulins may exist in a variety of formats, including, for example, Fv, Fab, and F(ab)2 as well as single chains (scFv) or diabodies. Immunoglobulins can come in different varieties known as isotypes or classes. In placental mammals there are five antibody isotypes known as IgA, IgD, IgE, IgG and IgM. They are each named with an “Ig” prefix that stands for immunoglobulin, another name for antibody, and differ in their biological properties, functional locations and ability to deal with different antigens.


As used herein, the term “antibody” refers to an immunoglobulin and any antigen-binding portion of an immunoglobulin, e.g., IgG, IgD, IgA, IgM and IgE, or a polypeptide that contains an antigen binding site, which specifically binds or “immunoreacts with” an antigen. Antibodies can comprise at least one heavy (H) chain and at least one light (L) chain inter-connected by at least one disulfide bond. The term “VH” refers to a heavy chain variable region of an antibody. The term “VL” refers to a light chain variable region of an antibody. In exemplary embodiments, the term “antibody” specifically covers monoclonal and polyclonal antibodies. A “polyclonal antibody” refers to an antibody which has been derived from the sera of animals immunized with an antigen or antigens. A “monoclonal antibody” refers to an antibody produced by a single clone of hybridoma cells.


As used herein, the term “biomarker” refers to a molecule that is associated either quantitatively or qualitatively with a biological change. Examples of biomarkers include: polypeptides, proteins or fragments of a polypeptide or protein; polynucleotides, such as a gene product, RNA or RNA fragment; and other body metabolites.


As disclosed herein, the term “hematological malignancy” or “hematological tumor” refers to cancers that affect blood and bone marrow.


The term “myeloma” as used herein means any tumor or cancer composed of cells derived from the hemopoietic tissues of the bone marrow. For example, myeloma includes multiple myeloma.


The “proteasome” as used herein refers to a multimeric enzymatic complex involved in the degradation of protein.


As disclosed herein, the term “proteasome inhibitor” is intended to include compounds which target, decrease or inhibit the activity of the proteasome. Compounds which target, decrease or inhibit the activity of the proteasome include, but are not limited to, carfilzomib (Kyprolis), oprozomib and bortezomib (Velcade).


As used herein, to “inhibit” or “suppress” or “reduce” a function or activity, such as proteasomal activity, is to reduce the function or activity when compared to otherwise same conditions except for a condition or parameter of interest, or alternatively, as compared to another condition.


As used herein, the term “reference level” refers to a level of a substance which may be of interest for comparative purposes. In one embodiment, a reference level may be the expression level of a protein or nucleic acid expressed as an average of the level of the expression level of a protein or nucleic acid from samples taken from a control population of normal, or healthy (disease-free) subjects. In another embodiment, the reference level may be the level in the same subject at a different time, e.g., before the present assay, such as the level determined prior to the subject developing the disease or prior to initiating therapy. In general, samples are normalized by a common factor. For example, body fluid samples are normalized by volume body fluid and cell-containing samples are normalized by protein content or cell count. In another embodiment, the reference level may also refer to the level of expression of the same biomarker in a corresponding control sample or control group of subjects which do not respond to PI treatment (e.g., treatment with carfilzomib, oprozomib or bortezomib).


As used herein, the term “subject” refers to a mammal, preferably a human, who may or may not have cancer. Typically, the terms “subject” and “patient” are used interchangeably herein in reference to a human subject. The subject may be a patient undergoing proteasome inhibition s (e.g., carfilzomib, oprozomib, bortezomib or other related agent) therapy using a sole therapeutic agent. The subject may be a patient undergoing proteasome inhibition s (e.g., carfilzomib, oprozomib, bortezomib or other related agent) therapy using a therapeutic agent in conjunction with another agent (e.g., a chemotherapeutic agent).


As used herein, the term “sample” or “test sample” refers to any liquid or solid material, specimen or culture obtained from any source containing nucleic acids or proteins. In suitable embodiments, a test sample is obtained from a biological source, e.g., a “biological sample”. Biological samples include blood products (such as plasma, serum, whole blood and peripheral blood mononuclear cells (PBMCs)), urine, saliva, blood, serum, or biopsy sample and the like. Biological samples also include tissue samples, such as biopsy tissues or pathological tissues that have previously been fixed (e.g., formalin, snap frozen, cytological processing, etc.). In an exemplary embodiment, the sample is a tumor sample.


The terms “detecting”, “determining,” “measuring,” “assessing,” and “assaying” are used interchangeably and include both quantitative and qualitative determinations. These terms refer to any form of measurement, and include determining if a characteristic, trait, or feature is present or not.


As used herein, the phrase “difference of the level” refers to differences in the quantity of a particular marker, such as a biomarker protein or nucleic acid, in a sample as compared to a control or reference level. For example, the quantity of particular protein or nucleic acid may be present at an elevated amount or at a decreased amount in samples of patients with a disease compared to a reference level. In one embodiment, a “difference of a level” may be a difference between the level of biomarker present in a sample as compared to a control of at least about 1%, at least about 2%, at least about 3%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 75%, at least about 80% or more. In one embodiment, a “difference of a level” may be a statistically significant difference between the level of the biomarker present in a sample as compared to a control. For example, a difference may be statistically significant if the measured level of the biomarker falls outside of about 1.0 standard deviations, about 1.5 standard deviations, about 2.0 standard deviations, or about 2.5 standard deviations of the mean of any control or reference group.


The term “elevated levels”, “increased levels” or “higher levels” as used herein refers to levels of a biomarker protein or nucleic acid that are higher than what would normally be observed in a comparable sample from control or normal subjects, e.g., a reference value. In some embodiments, “control levels”, e.g., normal levels, refer to a range of biomarker protein or nucleic acid levels that would normally be expected to be observed in a sample from a mammal that does not have a disease. A control level may be used as a reference level for comparative purposes. “Elevated levels” refer to biomarker protein or nucleic acid levels that are above the range of reference levels (e.g, control levels). The ranges accepted as “elevated levels” or “reference levels” are dependent on a number of factors. For example, one laboratory may routinely determine the level of biomarker protein or nucleic acid in a sample that are different than the level obtained for the same sample by another laboratory. Also, different assay methods may achieve different value ranges. Value ranges may also differ in various sample types, for example, different body fluids or by different treatments of the sample. One of ordinary skill in the art is capable of considering the relevant factors and establishing appropriate reference ranges for “control values” and “elevated values” of the present disclosure. For example, a series of samples from control subjects and subjects diagnosed with cancer can be used to establish ranges that are “normal” or “control” levels and ranges that are “elevated” or “higher” than the control range. In one embodiment, expression/amount of a gene or biomarker (e.g., Ig expression levels) in a sample is at an “elevated level” compared with a reference value if the expression level/amount of the gene or biomarker in the sample is at least about 1.5×, 1.75×, 2×, 3×, 4×, 5×, 6×, 7×, 8×, 9× or 10× the expression level/amount of the gene or biomarker in reference value.


Similarly, “reduced levels” or “lower levels” as used herein refer to levels of a biomarker protein or nucleic acid that are lower than what would normally be observed in a comparable sample from control or normal subjects, e.g., a reference value. In some embodiments, “control levels”, e.g., normal levels, refer to a range of biomarker protein or nucleic acid levels that would be normally be expected to be observed in a mammal that does not have a disease and “reduced levels” refer to biomarker protein or nucleic acid levels that are below the range of control levels.


As used herein, the terms “gene expression” or “expression” refer to the process of converting genetic information encoded in a gene into RNA, e.g., mRNA, rRNA, tRNA, or snRNA, through transcription of the gene, e.g., via the enzymatic action of an RNA polymerase, and for protein encoding genes, into protein through translation of mRNA. Gene expression can be regulated at many stages in the process.


As used herein, the term “diagnosis” means detecting a disease or disorder or determining the stage or degree of a disease or disorder. Usually, a diagnosis of a disease or disorder is based on the evaluation of one or more factors and/or symptoms that are indicative of the disease. That is, a diagnosis can be made based on the presence, absence or amount of a factor which is indicative of presence or absence of the disease or condition. Each factor or symptom that is considered to be indicative for the diagnosis of a particular disease does not need be exclusively related to the particular disease, e.g. there may be differential diagnoses that can be inferred from a diagnostic factor or symptom. Likewise, there may be instances where a factor or symptom that is indicative of a particular disease is present in an individual that does not have the particular disease. The term “diagnosis” also encompasses determining the therapeutic effect of a drug therapy, or predicting the pattern of response to a drug therapy. The diagnostic methods may be used independently, or in combination with other diagnosing and/or staging methods known in the medical arts for a particular disease or disorder, e.g., MM.


The term “prognosis” as used herein refers to a prediction of the probable course and outcome of a clinical condition or disease. A prognosis is usually made by evaluating factors or symptoms of a disease that are indicative of a favorable or unfavorable course or outcome of the disease. The phrase “determining the prognosis” as used herein refers to the process by which the skilled artisan can predict the course or outcome of a condition in a patient. The term “prognosis” does not refer to the ability to predict the course or outcome of a condition with 100% accuracy. Instead, the skilled artisan will understand that the term “prognosis” refers to an increased probability that a certain course or outcome will occur; that is, that a course or outcome is more likely to occur in a patient exhibiting a given condition, when compared to those individuals not exhibiting the condition. The terms “favorable prognosis” and “positive prognosis,” or “unfavorable prognosis” and “negative prognosis” as used herein are relative terms for the prediction of the probable course and/or likely outcome of a condition or a disease. A favorable or positive prognosis predicts a better outcome for a condition than an unfavorable or negative prognosis. In a general sense, a “favorable prognosis” is an outcome that is relatively better than many other possible prognoses that could be associated with a particular condition, whereas an unfavorable prognosis predicts an outcome that is relatively worse than many other possible prognoses that could be associated with a particular condition. Typical examples of a favorable or positive prognosis include a better than average remission rate, a lower propensity for metastasis, a longer than expected life expectancy, differentiation of a benign process from a cancerous process, and the like.


As used herein, the term “effective amount” or “pharmaceutically effective amount” or “therapeutically effective amount” refers to is a quantity of the compound(s) in a preparation which, when administered as part of a dosage regimen (to a mammal, e.g., a human) sufficient to achieve a desired therapeutic and/or prophylactic effect, e.g., an amount which alleviates a symptom, ameliorates a condition, or slows the onset of disease conditions according to clinically acceptable standards for the disorder or condition to be treated. The amount of a composition administered to the subject will depend on the type and severity of the disease and on the characteristics of the individual, such as general health, age, sex, body weight and tolerance to drugs. It will also depend on the degree, severity and type of disease. The skilled artisan will be able to determine appropriate dosages depending on these and other factors. The compositions can also be administered in combination with one or more additional therapeutic compounds and/or treatments.


As used herein, the term “treating” or “treatment” includes reversing, reducing, or arresting the symptoms, clinical signs, and underlying pathology of a condition in manner to improve or stabilize a subject's condition.


As used herein, “microarray” or “gene expression array” or “array” or “tissue microarray” refers to an arrangement of a collection of nucleic acids, e.g., nucleotide sequences in a centralized location. Arrays can be on a solid substrate, such as a glass slide, or on a semi-solid substrate, such as nitrocellulose membrane. The nucleotide sequences can be DNA, RNA, or any combination or permutations thereof. The nucleotide sequences can also be partial sequences or fragments from a gene, primers, whole gene sequences, non-coding sequences, coding sequences, published sequences, known sequences, or novel sequences.


Exemplary embodiments of the invention include:

    • 1. A method of treating a tumor in a subject, comprising:
      • a) measuring the level of expression of (i) immunoglobulin (Ig), (ii) Fc gamma receptor 2B (FCGR2B), or (iii) both Ig and FCGR2B, in a sample obtained from the subject, wherein the sample comprises a cell from the tumor; and
      • b) administering to the subject an effective amount of a proteasome inhibitor when the level of Ig expression and/or FCGR2B expression in the sample is greater than a reference level.
    • 2. A method of treating a tumor in a subject from which a sample was obtained, wherein the level of expression of (i) immunoglobulin (Ig), (ii) Fc gamma receptor 2B (FCGR2B), or (iii) both Ig and FCGR2B, has been measured from the sample, the method comprising the step of administering to the subject an effective amount of a proteasome inhibitor when the level of expression is greater than a reference level.
    • 3. A method of determining a treatment regimen for a subject with a tumor, comprising:
      • a) measuring the level of expression of (i) immunoglobulin (Ig), (ii) Fc gamma receptor 2B (FCGR2B), or (iii) both Ig and FCGR2B, in a sample obtained from the subject, wherein the sample comprises a cell from the tumor, and
      • b) selecting a treatment regimen comprising administration of a proteasome inhibitor, when the level of Ig expression and/or FCGR2B expression in the sample is greater than a reference level.
    • 4. The method of any one of claims 1-3, wherein the proteasome inhibitor is selected from the group consisting of carfilzomib, bortezomib, disulfiram, and oprozomib.
    • 5. The method of any one of claims 1-4, wherein the tumor is a hematological tumor, optionally, a hematological tumor derived from lymphoid cells.
    • 6. The method of claim 5, wherein the hematological tumor is a lymphoma, optionally a non-Hodgkin's lymphoma.
    • 7. The method of claim 6, wherein the non-Hodgkin's lymphoma is mantle cell lymphoma.
    • 8. The method of claim 5, wherein the hematological tumor is a multiple myeloma.
    • 9. The method of claim 8, wherein the multiple myeloma is smouldering myeloma, relapsed multiple myeloma, or refractory myeloma.
    • 10. The method of any one of claims 1-9, wherein the level of expression of (i) Ig, (ii) FCGR2B, or (iii) both Ig and FCGR2B, is measured in CD138-positive tumor cells obtained from the subject.
    • 11. The method of claim 10, comprising extracting RNA from the CD138-positive tumor cells.
    • 12. The method of any one of claims 1-11, wherein the sample comprises bone marrow cells, blood, serum, or a biopsy sample.
    • 13. The method of claim 12, comprising contacting antibodies specific for FCGR2B or for Ig with the sample comprising intact bone marrow cells.
    • 14. The method of any one of claims 1-13, comprising measuring the level of expression of one or more gene segments of the IgH locus, IgK locus, or IgL locus, or an IgH orphon gene segment, an IgK orphon gene segment, or an IgL orphon gene segment, or a combination thereof
    • 15. The method of claim 14, wherein the one or more gene segments at the IgH locus or the IgH orphon gene segment is selected from the group consisting of: IGHA1, IGHA2, IGHD, IGHD1-1, IGHD1-14, IGHD1-20, IGHD1-26, IGHD1-7, IGHD2-15, IGHD2-2, IGHD2-21, IGHD2-8, IGHD3-10, IGHD3-16, IGHD3-22, IGHD3-3, IGHD3-9, IGHD4-11, IGHD4-17, IGHD4-23, IGHD4-4, IGHD5-12, IGHD5-18, IGHD5-24, IGHD5-5, IGHD6-13, IGHD6-19, IGHD6-25, IGHD6-6, IGHD?-27, IGHE, IGHEP1, IGHEP2, IGHG1, IGHG2, IGHG3, IGHG4, IGHGP, IGHJ1, IGHJ1P, IGHJ2, IGHJ2P, IGHJ3, IGHJ3P, IGHJ4, IGHJ5, IGHJ6, IGHM, IGHMBP2, IGHV1-12, IGHV1-14, IGHV1-17, IGHV1-18, IGHV1-2, IGHV1-24, IGHV1-3, IGHV1-45, IGHV1-46, IGHV1-58, IGHV1-67, IGHV1-68, IGHV1-69, IGHV1-8, IGHV1OR21-1, IGHV2-10, IGHV2-26, IGHV2-5, IGHV2-70, IGHV2OR16-5, IGHV3-11, IGHV3-13, IGHV3-15, IGHV3-16, IGHV3-19, IGHV3-20, IGHV3-21, IGHV3-22, IGHV3-23, IGHV3-25, IGHV3-29, IGHV3-30, IGHV3-30-2, IGHV3-32, IGHV3-33, IGHV3-33-2, IGHV3-35, IGHV3-36, IGHV3-37, IGHV3-38, IGHV3-41, IGHV3-42, IGHV3-43, IGHV3-47, IGHV3-48, IGHV3-49, IGHV3-50, IGHV3-52, IGHV3-53, IGHV3-54, IGHV3-57, IGHV3-6, IGHV3-60, IGHV3-62, IGHV3-63, IGHV3-64, IGHV3-65, IGHV3-66, IGHV3-7, IGHV3-71, IGHV3-72, IGHV3-73, IGHV3-74, IGHV3-75, IGHV3-76, IGHV3-79, IGHV3-9, IGHV3OR16-8, IGHV4-28, IGHV4-31, IGHV4-34, IGHV4-39, IGHV4-4, IGHV4-55, IGHV4-59, IGHV4-61, IGHV4-80, IGHV5-51, IGHV5-78, IGHV6-1, IGHV7-27, IGHV7-34-1, IGHV7-40, IGHV7-56, IGHV7-81, IGHVII-1-1, IGHVII-15-1, IGHVII-20-1, IGHVII-22-1, IGHVII-26-2, IGHVII-28-1, IGHVII-30-1, IGHVII-31-1, IGHVII-33-1, IGHVII-40-1, IGHVII-43-1, IGHVII-44-2, IGHVII-46-1, IGHVII-49-1, IGHVII-51-2, IGHVII-53-1, IGHVII-60-1, IGHVII-62-1, IGHVII-65-1, IGHVII-67-1, IGHVII-74-1, IGHVII-78-1, IGHVIII-11-1, IGHVIII-13-1, IGHVIII-16-1, IGHVIII-2-1, IGHVIII-22-2, IGHVIII-25-1, IGHVIII-26-1, IGHVIII-38-1, IGHVIII-44, IGHVIII-47-1, IGHVIII-5-1, IGHVIII-51-1, IGHVIII-5-2, IGHVIII-67-2, IGHVIII-67-3, IGHVIII-67-4, IGHVIII-76-1, IGHVIII-82, and IGHVIV-44-1,
    • wherein the one or more gene segments at the IgK locus or the IgK orphon gene segment is selected from the group consisting of: IGKC, IGKJ1, IGKJ2, IGKJ3, IGKJ4, IGKJ5, IGKV1-12, IGKV1-13, IGKV1-16, IGKV1-17, IGKV1-22, IGKV1-27, IGKV1-32, IGKV1-33, IGKV1-35, IGKV1-37, IGKV1-39, IGKV1-5, IGKV1-6, IGKV1-8, IGKV1-9, IGKV1D-12, IGKV1D-13, IGKV1D-16, IGKV1D-17, IGKV1D-22, IGKV1D-27, IGKV1D-32, IGKV1D-33, IGKV1D-35, IGKV1D-37, IGKV1D-39, IGKV1D-42, IGKV1D-43, IGKV1D-8, IGKV1OR22-1, IGKV2-10, IGKV2-14, IGKV2-18, IGKV2-19, IGKV2-23, IGKV2-24, IGKV2-26, IGKV2-28, IGKV2-29, IGKV2-30, IGKV2-36, IGKV2-38, IGKV2-4, IGKV2-40, IGKV2D-10, IGKV2D-14, IGKV2D-18, IGKV2D-19, IGKV2D-23, IGKV2D-24, IGKV2D-26, IGKV2D-28, IGKV2D-29, IGKV2D-30, IGKV2D-36, IGKV2D-38, IGKV2D-40, IGKV2OR22-3, IGKV2OR22-4, IGKV3-11, IGKV3-15, IGKV3-20, IGKV3-25, IGKV3-31, IGKV3-34, IGKV3-7, IGKV3D-11, IGKV3D-15, IGKV3D-20, IGKV3D-25, IGKV3D-31, IGKV3D-34, IGKV3D-7, IGKV3OR22-2, IGKV4-1, IGKV5-2, IGKV6-21, IGKV6D-21, IGKV6D-41, and IGKV7-3,
    • wherein the one or more gene segments at the IgT locus or the IgL orphon gene segment is selected from the group consisting of: IGLC1, IGLC2, IGLC3, IGLC4, IGLC5, IGLC6, IGLC7, IGLCOR22-1, IGLJ1, IGLJ2, IGLJ3, IGLJ4, IGLJ5, IGLJ6, IGLJ7, IGLL1, IGLL3, IGLON5, IGLV10-54, IGLV10-67, IGLV11-55, IGLV1-36, IGLV1-40, IGLV1-41, IGLV1-44, IGLV1-47, IGLV1-50, IGLV1-51, IGLV1-62, IGLV2-11, IGLV2-14, IGLV2-18, IGLV2-23, IGLV2-28, IGLV2-33, IGLV2-34, IGLV2-5, IGLV2-8, IGLV3-1, IGLV3-10, IGLV3-12, IGLV3-13, IGLV3-15, IGLV3-16, IGLV3-17, IGLV3-19, IGLV3-2, IGLV3-21, IGLV3-22, IGLV3-24, IGLV3-25, IGLV3-26, IGLV3-27, IGLV3-29, IGLV3-30, IGLV3-31, IGLV3-32, IGLV3-4, IGLV3-6, IGLV3-7, IGLV3-9, IGLV4-3, IGLV4-60, IGLV4-69, IGLV5-37, IGLV5-45, IGLV5-48, IGLV5-52, IGLV6-57, IGLV7-35, IGLV7-43, IGLV7-46, IGLV8-61, IGLV9-49, IGLVI-20, IGLVI-38, IGLVI-42, IGLVI-56, IGLVI-63, IGLVI-68, IGLVI-70, IGLVIV-53, IGLVIV-59, IGLVIV-64, IGLVIV-65, IGLVIV-66-1, IGLVV-58, IGLVV-66, IGLVVI-22-1, IGLVVI-25-1, and IGLVVII-41-1.
    • 16. The method of claim 14, wherein the one or more gene segments at the IgH locus or the IgH orphon gene segment comprises a sequence selected from the group consisting of SEQ ID NOs: 1-174, wherein the one or more gene segments at the IgK locus or the IgK orphon gene segment comprises a sequence selected from the group consisting of SEQ ID NOs: 175-260, or wherein the one or more gene segments at the IgL locus or the IgL orphon gene segment comprises a sequence selected from the group consisting of SEQ ID NOs: 261-350.
    • 17. The method of any one of claims 1-16, wherein the level of expression of Ig is the sum of the expression levels of more than one gene segment of the IgH locus, IgK locus, and/or IgL locus and/or more than one IgH orphon gene segment, IgK orphon gene segment, and/or IgK orphon gene segment.
    • 18. The method of claim 17, wherein the level of expression of Ig is the sum of the expression levels of all the gene segments of the IgH locus and all the IgH orphon gene segments.
    • 19. The method of claim 17, wherein the level of expression of Ig is the sum of (i) the levels of expression of all the gene segments of the IgH locus and all the IgH orphon gene segments, (ii) the levels of expression of all the gene segments of the IgK locus and all the IgK orphon gene segments, and (iii) the levels of expression of all the gene segments of the IgL locus and all the IgL orphon gene segments.
    • 20. The method of any one of claims 1-19, comprising measuring the level of expression of Ig and the level of expression of FCGR2B in the sample.
    • 21. The method of any one of claims 1-20, further comprising measuring the level of expression of one of more genes listed in Table 4, optionally, wherein the level of expression of two, three, four, five, six, seven, eight, nine, ten, or more genes listed in Table 4 are measured.
    • 22. The method of any one of claims 1-21, wherein the reference level is a reference level of Ig expression.
    • 23. The method of any one of claims 1-22, wherein the measured level of Ig expression and/or the measured level of FCGR2B expression is at least 2-fold greater than the reference level.
    • 24. The method of any one of claims 1-23, wherein the measured level of Ig expression and/or the measured level of FCGR2B expression is at least 3-fold greater than the reference level.
    • 25. The method of any one of claims 1-24, wherein the measured level of Ig expression and/or the measured level of FCGR2B expression is at least 4-fold greater than the reference level.
    • 26. The method of any one of claims 1-22, wherein the reference level is a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve, optionally, wherein the ROC curve is based on (i) the distribution of Ig expression levels and/or FCGR2B expression levels of responders and (ii) the distribution of Ig expression levels and/or FCGR2B expression levels of non-responders.
    • 27. The method of any one of claims 1-22, wherein the reference level is a cutoff correlative with a % specificity of at least 75% and a % sensitivity of at least 75%, as determined by a receiver operating characteristic (ROC) curve, optionally, wherein the ROC curve is based on (i) the distribution of Ig expression levels and/or FCGR2B expression levels of responders and (ii) the distribution of Ig expression levels and/or FCGR2B expression levels of non-responders.
    • 28. The method of any one of claims 1-27, wherein the subject (i) has previously been treated for multiple myeloma or (ii) has previously been diagnosed with multiple myeloma or (iii) is a human patient having or suspected of having multiple myeloma, refractory multiple myeloma, or relapsed multiple myeloma.
    • 29. The method of any one of claims 1-28, wherein measuring the level of expression of Ig and/or FCGR2B in a sample comprises (i) amplifying a fragment of a human Ig mRNA or human FCGR2B mRNA; (ii) measuring the Ig expression level in the cell using a microarray platform that map to genes encoding Ig-related proteins; or (iii) measuring the Ig protein load in the cells with an anti-human Ig antibody.
    • 30. The method of any one of claims 1-29, wherein the measuring comprises amplifying a fragment of a human Ig mRNA and/or human FCGR2B mRNA.
    • 31. The method of claim 30, wherein the amplifying is by polymerase chain reaction (PCR) or RT-PCR.
    • 32. The method of any one of claims 1-31, wherein the measuring comprises measuring the presence, absence, or amount of a human Ig protein in the sample.
    • 33. The method of claim 32, wherein the measuring uses an antibody that specifically binds to a human Ig protein, a human FCGR2B, or a human protein encoded by a gene listed in Table 4.
    • 34. The method of claim 33, wherein the measuring is by an ELISA assay, a western blot assay, or an immunohistochemical assay.
    • 35. A kit comprising one or more binding agents to an Ig gene or gene product, optionally an IgH, IgK or IgL gene segment or gene segment product, or an IgH, IgK, or IgL orphon gene segment or gene segment product, and a binding agent to FCGR2B gene or gene product.
    • 36. A kit comprising (i) one or more binding agents to an Ig gene or gene product, optionally an IgH, IgK or IgL gene segment or gene segment product, or an IgH, IgK, or IgL orphon gene segment or gene segment product, or a binding agent to FCGR2B gene or gene product and (ii) at least one binding agent to a gene or gene product listed in Table 4.
    • 37. The kit of claim 35, further comprising at least one binding agent to a gene or gene product listed in Table 4.
    • 38. The kit of any one of claims 35 to 37, further comprising a proteasome inhibitor.
    • 39. The kit of any one of claims 35-38, further comprising a reagent that produces a signal indicative of a reference level.
    • 40. The kit of any one of claims 35-39, wherein the binding agent is a compound that binds to a nucleic acid molecule, optionally, wherein the binding agent is a nucleic acid molecule.
    • 41. The kit of any one of claims 35 to 40, wherein the binding agent is a compound that binds to a protein, optionally, wherein the binding agent is an antibody, an antigen binding fragment thereof, or an antibody derivative.
    • 42. A method of treating a tumor in a subject, comprising:
      • a) measuring the level of expression of one or more genes listed in Table 4, in a sample obtained from the subject, wherein the sample comprises a cell from the tumor; and
      • b) administering to the subject an effective amount of a proteasome inhibitor, when (i) the level of expression of the one or more genes listed in Table 4 in the sample greater than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “up”, or (ii) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is less than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “down”, or (iii) both (i) and (ii).
    • 43. A method of treating a tumor in a subject from which a sample was obtained, wherein the level of expression of one or more genes or gene products listed in Table 4 has been measured from the sample, the method comprising the step of administering to the subject an effective amount of a proteasome inhibitor, when (i) the level of expression of the one or more genes listed in Table 4 in the sample is greater than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “up”, or (ii) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is less than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “down”, or (iii) both (i) and (ii).
    • 44. A method of determining a treatment regimen for a subject with a tumor, comprising:
      • a) measuring the level of expression of one or more genes or gene products listed in Table 4 in a sample obtained from the subject, wherein the sample comprises a cell from the tumor, and
      • b) selecting a treatment regimen comprising administration of a proteasome inhibitor, when (i) the level of expression of the one or more genes listed in Table 4 in the sample is greater than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “up”, or (ii) the level of expression of the one or more genes or gene products listed in Table 4 in the sample is less than a reference level, and the change in gene expression level for the one or more genes or gene products is denoted in Table 4 as “down”, or (iii) both (i) and (ii).
    • 45. The method of any one of claims 42-44, wherein the proteasome inhibitor is selected from the group consisting of carfilzomib, bortezomib, disulfiram, and oprozomib.
    • 46. The method of any one of claims 42-45, wherein the tumor is a hematological tumor, optionally, a hematological tumor derived from lymphoid cells.
    • 47. The method of claim 46, wherein the hematological tumor is a lymphoma, optionally, a non-Hodgkin's lymphoma.
    • 48. The method of claim 47, wherein the a non-Hodgkin's lymphoma is mantel cell lymphoma.
    • 49. The method of claim 46, wherein the hematological tumor is a multiple myeloma.
    • 50. The method of claim 49, wherein the multiple myeloma is smouldering myeloma, relapsed multiple myeloma, or refractory myeloma.
    • 51. The method of any one of claims 42-50, wherein the level of expression of the one or more genes or gene products in Table 4 is measured in CD138-positive tumor cells obtained from the subject.
    • 52. The method of claim 51, comprising extracting RNA from the CD138-positive tumor cells.
    • 53. The method of any one of claims 42-52, wherein the sample comprises bone marrow cells, blood, serum, or a biopsy sample.
    • 54. The method of claim 53, comprising contacting antibodies specific for a gene product encoded by the one or more genes in Table 4 with the sample comprising bone marrow cells.
    • 55. The method of any one of claims 42-54, further comprising measuring the level of expression of Ig and/or FCGF2B in the sample.
    • 56. The method of claim 55, further comprising measuring the level of expression of one or more gene segments of the IgH locus, IgK locus, or IgL locus, or an IgH orphon gene segment, an IgK orphon gene segment, or an IgL orphon gene segment, or a combination thereof
    • 57. The method of claim 56, wherein the one or more genes of the IgH locus or the IgH orphon gene segment is/are selected from the group consisting of the group of gene segments of the IgH locus in claim 15, wherein the one or more gene segments of the IgK locus or the IgK orphon gene segment is/are selected from the group consisting of the group of gene segments of the IgK locus in claim 15, or wherein the one or more gene segments at the IgL locus or the IgL orphon gene segment is/are selected from the group consisting of the group of gene segments of the IgL locus in claim 15.
    • 58. The method of claim 56, wherein the one or more gene segments at the IgH locus or the IgH orphon gene segment comprises a sequence selected from the group consisting of SEQ ID NOs: 1-174, wherein the one or more gene segments at the IgK locus or the IgK orphon gene segment comprises a sequence selected from the group consisting of SEQ ID NOs: 175-260, or wherein the one or more gene segments at the IgL locus or the IgL orphon gene segment comprises a sequence selected from the group consisting of SEQ ID NOs: 261-350.
    • 59. The method of any one of claims 42 to 58, wherein the level of expression of Ig is the sum of the expression levels of more than one gene segment of the IgH locus, IgK locus, and/or IgL locus and/or more than one IgH orphon gene segment, IgK orphon gene segment, and/or IgK orphon gene segment.
    • 60. The method of claim 59, wherein the level of expression of Ig is the sum of the expression levels of all the gene segments of the IgH locus and all the IgH orphon gene segments.
    • 61. The method of any one of claims 42-60, wherein the level of expression of Ig is the sum of (i) the levels of expression of all the gene segments of the IgH locus and all the IgH orphon gene segments, (ii) the levels of expression of all the gene segments of the IgK locus and all the IgK orphon gene segments, and (iii) the levels of expression of all the gene segments of the IgL locus and all the IgL orphon gene segments.
    • 62. The method of any one of claims 42 to 61, comprising measuring the level of expression of two of more genes listed in Table 4, optionally, wherein the level of expression of three, four, five, six, seven, eight, nine, ten, or more genes listed in Table 4 are measured.
    • 63. The method of any one of claims 42-62, wherein the reference level is a reference value of expression level of the one or more genes.
    • 64. The method of any one of claims 42-63, wherein the measured level of expression of the one or more genes is at least 2-foldless than or greater than the reference level.
    • 65. The method of any one of claims 42-64, wherein the measured level of expression of the one or more genes is at least 3-fold less than or greater than, the reference level.
    • 66. The method of any one of claims 42-65, wherein the measured level of expression of the one or more genes is at least 4-fold less than or greater than the reference level.
    • 67. The method of any one of claims 42-66, wherein the reference level is a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve, optionally, wherein the ROC curve is based on (i) the distribution of expression levels of responders, wherein the expression levels are the expression levels of the one or more genes listed in Table 4 and (ii) the distribution of expression levels of non-responders, wherein the expression levels are the expression levels of the one or more genes listed in Table 4.
    • 68. The method of any one of claims 42-67, wherein the reference level is a cutoff correlative with a % specificity of at least 75% and a % sensitivity of at least 75%, as determined by a receiver operating characteristic (ROC) curve, optionally, wherein the ROC curve is based on (i) the distribution of expression levels of responders, wherein the expression levels are the expression levels of the one or more genes listed in Table 4 and (ii) the distribution of expression levels of non-responders, wherein the expression levels are the expression levels of the one or more genes listed in Table 4.
    • 69. The method of any one of claims 42-67, wherein the subject (i) has previously been treated for multiple myeloma or (ii) has previously been diagnosed with multiple myeloma or (iii) is a human patient having or suspected of having multiple myeloma, refractory multiple myeloma, or relapsed multiple myeloma.
    • 70. The method any one of claims 42-69, comprising (i) amplifying a fragment of a human mRNA encoded by the one or more genes; (ii) measuring the expression level in the cell using a microarray platform that map to the one or more genes; or (iii) measuring the protein load in the cells with an antibody specific for the protein product encoded by the one or more genes.
    • 71. The method of any one of claims 42-70, comprising amplifying a fragment of a human mRNA encoded by the one or more genes.
    • 72. The method of claim 71, wherein the amplifying is by polymerase chain reaction (PCR) or RT-PCR.
    • 73. The method of any one of claims 42-72, comprising measuring the presence, absence, or amount of a human protein encoded by the one or more genes in the test sample.
    • 74. The method of claim 73, comprising the use of an antibody that specifically binds to a gene produce encoded by a gene listed in Table 4.
    • 75. The method of claim 74, comprising measuring by an ELISA assay, a western blot assay, or an immunohistochemical assay.
    • 76. A kit comprising at least a first binding agent and a second binding agent, wherein the first binding agent binds to a first gene or gene product encoded by a first gene listed in Table 4, wherein the second binding agent binds to a second gene or gene product encoded by a second gene listed in Table 4, wherein the first gene is different from the second gene.
    • 77. The kit of claim 76 further comprising a proteasome inhibitor.
    • 78. The kit of claim 76 or 77, further comprising a binding agent to an Ig or FCGR2B.
    • 79. The kit of any one of claims 76 to 78 further comprising a reagent that produces a signal indicative of a reference level.
    • 80. The kit of any one of claims 76 to 79, wherein the binding agent is a compound that binds to a nucleic acid molecule, optionally, wherein the binding agent is a nucleic acid molecule.
    • 81. The kit of any one of claims 76 to 80, wherein the binding agent is a compound that binds to a protein, optionally, wherein the binding agent is an antibody, an antigen binding fragment thereof, or an antibody derivative.
    • 82. A computer readable-storage medium having stored thereon a plurality of reference levels or ranges of reference levels, each reference level or range of reference levels corresponding to (i) an expression level of Ig or (ii) an expression level of FCGR2B, or (iii) an expression level of a gene listed in Table 4, or (iv) a combination thereof; and a data value that is an expression level of Ig and/or an expression level of FCGR2B and/or an expression level of a gene listed in Table 4, measured from a cell from a sample from a patient.
    • 83. The computer readable-storage medium of claim 82, wherein the data value that is the expression level of Ig is the sum of the expression levels of more than one gene segment of the IgH, IgK, and/or IgL locus, optionally, wherein the expression level of Ig is indicative of a responder or non-responder.
    • 84. A computer readable-storage medium having stored thereon (I) a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of Ig determined from a sample obtained from a responder and each data value of the second set is an expression level of Ig determined from a sample obtained from a non-responder; (II) a receiver operating characteristic (ROC) curve based on a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of Ig determined from a sample obtained from a responder and each data value of the second set is an expression level of Ig determined from a sample obtained from a non-responder; or (III) a table listing (a) a plurality of cut-off points on the ROC curve of (II), (b) the % sensitivity associated with each cut-off point of (a), and (c) the % specificity associated with each cut-off point of (a).
    • 85. A computer readable-storage medium having stored thereon (I) a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of FCGR2B determined from a sample obtained from a responder and each data value of the second set is an expression level of FCGR2B deteremined from a sample obtained from a non-responder; (II) a receiver operating characteristic (ROC) curve based on a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of FCGR2B determined from a sample obtained from a responder and each data value of the second set is an expression level of FCGR2B deteremined from a sample obtained from a non-responder; or (III) a table listing (a) a plurality of cut-off points on the ROC curve of (II), (b) the % sensitivity associated with each cut-off point of (a), and (c) the % specificity associated with each cut-off point of (a).
    • 86. A computer readable-storage medium having stored thereon (I) a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of a gene listed in Table 4 determined from a sample obtained from a responder and each data value of the second set is an expression level of a gene listed in Table 4 determined from a sample obtained from a non-responder; (II) a receiver operating characteristic (ROC) curve based on a first set of data values and a second set of data values, wherein each data value of the first set is an expression level of a gene listed in Table 4 determined from a sample obtained from a responder and each data value of the second set is an expression level of a gene listed in Table 4 determined from a sample obtained from a non-responder; or (III) a table listing (a) a plurality of cut-off points on the ROC curve of (II), (b) the % sensitivity associated with each cut-off point of (a), and (c) the % specificity associated with each cut-off point of (a).
    • 87. A computer readable storage medium comprising two or more of the computer storage media of claims 84-86.
    • 88. A system comprising: a processor; a memory device coupled to the processor, and machine readable instructions stored on the memory device, wherein the machine readable instructions, when executed by the processor, cause the processor to:
      • i. receive a data value, α, relating to a test level of Ig expression from a sample obtained from a test subject; and
      • ii. display an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
    • 89. A system comprising: a processor; a memory device coupled to the processor, and machine readable instructions stored on the memory device, wherein the machine readable instructions, when executed by the processor, cause the processor to:
      • i. receive a data value, α, relating to a test level of FCGR2B expression from a sample obtained from a test subject; and
      • ii. display an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
    • 90. A system comprising: a processor; a memory device coupled to the processor, and machine readable instructions stored on the memory device, wherein the machine readable instructions, when executed by the processor, cause the processor to:
      • i. receive a data value, α, relating to a test level of expression of a gene listed in Table 4, wherein the change in gene expression level for the gene is denoted in Table 4 as “up”, from a sample obtained from a test subject; and
      • ii. display an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
    • 91. A system comprising: a processor; a memory device coupled to the processor, and machine readable instructions stored on the memory device, wherein the machine readable instructions, when executed by the processor, cause the processor to:
      • i. receive a data value, α, relating to a test level of expression of a gene listed in Table 4, wherein the change in gene expression level for the gene is denoted in Table 4 as “down”, from a sample obtained from a test subject; and
      • ii. display an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when α is less than β, a cutoff correlative with a % specificity of at least 50% and a sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
    • 92. A computer-readable storage medium having stored thereon machine-readable instructions executable by a processor, comprising:
      • i. instructions for receiving a data value, α, relating to a test level of Ig expression from a sample obtained from a test subject; and
      • ii. instructions for displaying an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when a is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
    • 93. A computer-readable storage medium having stored thereon machine-readable instructions executable by a processor, comprising:
      • i. instructions for receiving a data value, α, relating to a test level of FCGR2B expression from a sample obtained from a test subject; and
      • ii. instructions for displaying an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
    • 94. A computer-readable storage medium having stored thereon machine-readable instructions executable by a processor, comprising:
      • i. instructions for receiving a data value, α, relating to a test level of expression of a gene listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “up”; and
      • ii. instructions for displaying an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when a is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
    • 95. A computer-readable storage medium having stored thereon machine-readable instructions executable by a processor, comprising:
      • (i) instructions for receiving a data value, α, relating to a test level of expression of a gene listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “down”; and
      • (ii) instructions for displaying an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when α is less than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
    • 96. A method implemented by a processor in a computer, the method comprising the steps of:
      • i. receiving a data value, α, relating to a test level of Ig expression from a sample obtained from a test subject; and
      • ii. displaying an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
    • 97. A method implemented by a processor in a computer, the method comprising the steps of:
      • i. receiving a data value, α, relating to a test level of FCGR2B expression from a sample obtained from a test subject; and
      • ii. displaying an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
    • 98. A method implemented by a processor in a computer, the method comprising the steps of:
      • i. receiving a data value, α, relating to a test level of expression of a gene listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “up”; and
      • ii. displaying an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when α is greater than β, a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
    • 99. A method implemented by a processor in a computer, the method comprising the steps of:
      • (i) receiving a data value, α, relating to a test level of expression of a gene listed in Table 4 from a sample obtained from a test subject, wherein the change in gene expression level for the gene is denoted in Table 4 as “down”; and
      • (ii) displaying an output relating to treating the subject for multiple myeloma with a proteasome inhibitor, when α is less than β, a cutoff correlative with a % specificity of at least 50% and a sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.


The following examples serve only to illustrate the invention or provide background information relating to the invention. The following examples are not intended to limit the scope of the invention in any way.


EXAMPLES

Several therapies for multiple myeloma (MM) are now approved and many more are in development, promising improved outcomes for patients with this incurable cancer. With expanding treatment options, however, comes a pressing need to pair each patient with the most efficacious and safe treatment. Proteasome inhibitors (PIs), such as carfilzomib and bortezomib, have become a standard therapy across all lines of MM therapy. Despite extensive study, the mechanism of selective tumor cell death following proteasome inhibition is poorly understood. However, the uniquely high sensitivity of myeloma cells to PI, the uniquely high burden of protein (immunoglobulin) secretion these cells experience, and the key role of the proteasome in maintaining protein homeostasis, together point toward a unifying model in which protein load drives PI sensitivity. This simple model is supported by published studies of murine and human myeloma cell lines (Meister et al. & Bianchi et al.). As part of company-sponsored Phase II & III clinical trials of PIs, CD138+ tumor cells collected during patient screening were banked for comprehensive genomic analyses. Patient samples banked on bortezomib trials were utilized in now-published microarray-based RNA studies (Mulligan et al.), while samples from carfilzomib trials are currently being used for NGS-based DNA and RNA studies. Here, examining the early carfilzomib data along with publically-available bortezomib data, the inventors demonstrate a strong association between higher immunoglobulin expression and sensitivity to each compound (Wilcoxon P-value=3×10−3 and P-value=2×10−4, respectively). In fact, using IGH expression alone, the inventors were able to classify response with 55% sensitivity and 91% specificity for the carfilzomib training data set. As expected for a bona fide predictive biomarker of PI, an association between IG expression and response was not found in patients treated with single agent dexamethasone (Wilcoxon P-value=0.82). Median time to progression for IGH-high carfilzomib patients was 6-fold longer than for IGH-low carfilzomib patients (7.6 months vs. 1.4 months; log-rank P-value=0.003). This is the first report that high levels of IG expression correlate with response to PIs and therefore IG expression represents, to our knowledge, the first validated biomarker for this important class of anti-tumor agents.


Example 1: Bortezomib Data Analysis

Publicly available RNA microarray data in tab-delimited text format were downloaded from the Gene Expression Omnibus (GEO ID: GSE9782). These data, originally reported in a publication by Mulligan et al., represent normalized, probe-level measurements of mRNA abundance in multiple myeloma tumor cells (CD138+ selected) collected from patients prior to treatment with either single-agent bortezomib or single-agent dexamethasone on Millennium's Phase 2 and Phase 3 clinical trials.


A list of 55 probes from this Affymetrix microarray platform that map to genes encoding immunoglobulin (Ig)-related proteins was obtained from Rody et al. Addition File 5. This example tested for an association between normalized intensity of each of these 55 probes and bortezomib response, which was labeled as “PGx_Responder=R” in the dataset. Of these, 13 showed a significant correlation (Wilcoxon P-value<=0.05) between higher expression values and response on the bortezomib arms of the studies: 211798_x_at, 211881_x_at, 216365_x_at, 216560_x_at, 217148_x_at, 217179_x_at, 217227_x_at, 217258_x_at, 211639_x_at, 216491_x_at, 211649_x_at, 216510_x_at, and 211637_x_at. The finding of 13 significant associations is far more than expected by chance (Binomial P-value=2×10−6). None of the 55 probes showed the opposite effect. It is not surprising that many probes are not associated, given that some map to rarer, patient-specific portions of the Ig loci and that others map to regions that are unusually highly-expressed, and therefore not well-measured with microarray technology (i.e., their fluorescence is likely to be saturated).


The 13 significantly-associated probes were each normalized to a [0, 1] scale and the mean was computed, yielding our combined Ig expression score. The combined score is very strongly associated with bortezomib response (Wilcoxon P-value=2×10−4; FIGS. 1A and 1B), but exhibits no association with response to dexamethasone (Wilcoxon P-value=0.82; FIGS. 2A and 2B). Patients were classified as achieving complete response (CR), partial response (PR), minimal response (MR), no change (NC), or progressive disease (PD), using European Group for Bone Marrow Transplantation criteria. In brief, PD requires 25% increase in paraprotein, whereas MR, PR, and CR require at least 25%, 50%, and 100% decreases, respectively. Further illustrating the specificity of this marker for predicting PI response, of the 55 Ig probes selected, only one showed an association with dexamethasone response (which is roughly what is expected by chance; Binomial P-value=0.23) and the P-value itself was only marginally significant (Wilcoxon P-value=0.0495).


Various mean IG expression cutoffs were considered to maximize the clinical utility of a potential diagnostic test. Ultimately, a cutoff of 0.29 (defining “IG-High”>=0.29 and “IG-Low”<0.29) was chosen, predicting response in the training data with 71% sensitivity and 62% specificity. This same cutoff was then applied to time-to-progression (TTP) data for these patients, finding that IG-High patients in this cohort have a 1.4-fold longer median TTP than IG-Low patients (8.4 months vs. 6.0 months; log-rank P-value=0.025).


Example 2: Carfilzomib Data Analysis

Bone marrow samples were aspirated from patients enrolled on Onyx-sponsored Phase 2 trials (NCT00511238, NCT00530816, and NCT00721734) prior to treatment with single-agent carfilzomib. Myeloma tumor cells were isolated by EasySep® immunomagnetic bead-based CD138+ selection (StemCell Technologies), re-suspended in TRIzol (Life Technologies) and frozen at −80° F. RNA was extracted from these samples with the PureLink RNA kit from Life Technologies (Cat #12183018A), following the recommended protocol for cells in TRIzol suspension, including the optional on-column DNase treatment. Elution volumes ranged from 75 μl-30 μl depending on the total number of cells going into isolation. ERCC control sequences from Life Technologies (ERCC ExFold RNA Spike-In Mix; Cat #4456739) were spiked in to all total RNA samples of sufficient quantity (Nanodrop yield >=150 ng) and quality (Bioanalyzer RIN>=7.0). For 500 ng input libraries, 1 μl of a 1:100 dilution of Mix 1 was added to each total RNA sample and for 150 ng input libraries, 3 μl of a 1:1000 dilution of Mix 1 was added to each total RNA sample. The resulting material was used to construct RNA-Seq libraries with Illumina's TruSeq RNA sample prep kit v2 (Cat #RS-122-2001), with PolyA selection included as the first step. Libraries were sequenced on Illumina's HiSeq 2000 with a target of 70 million fragments using 100×100 bp paired-end sequencing to generate 140 million reads per sample and the resulting raw data was QC-ed with metrics implemented in OmicSoft Array Studio v6.1.


Raw sequence reads were aligned and expression of genes & isoforms were quantified with a customized pipeline also built in Array Studio v6.1 [Jun]. This pipeline accepts Illumina adapter-stripped, paired-end reads that are trimmed at the 5′ end if a base reaches PHRED quality score Q2 or lower. All reads are aligned to the transcriptome, which consists of the RefGene annotation of human hg19 supplemented with the more rich Ig loci annotations available from ENSEMBL. Reads aligned with mismatches and unaligned reads are subsequently aligned to the entire human genome, searching for novel exon junctions. Reads that remain unaligned are then aligned to the newly identified exon junctions. Alignments to the different references are compared and the highest scoring alignment is retained, or in the event of a tie, the transcriptome alignment is preferentially kept. Finally, all transcriptome alignment locations are translated to genomic coordinates to estimate the expected number of mappings per gene/isoform using the EM algorithm [Dempster]. The EM algorithm assigns reads with multiple alignment locations to an isoform by calculating the conditional probability of a read aligning to a specific isoform, given all other alignments. Weighting the total number of aligned reads with this probability yields posterior expected read counts for each transcript. The EM counts are normalized by the length of the genes/isoforms and number of reads in a library to yield FPKM values (Fragments Per Kilobase per Million reads). Between-sample normalization is achieved by 75th quantile normalization, where each sample is scaled by the median of 75th quantiles from all samples to yield quantile-normalized FPKM or “FPKQ” values.


The FPKQ values corresponding to each “gene” of the three Ig loci, IGH (immunoglobulin heavy locus), IGK (immunoglobulin kappa locus) and IGL (immunoglobulin lambda locus), were summed to produce locus-level expression estimates.


For IGH, this included the following genes: IGHA1, IGHA2, IGHD, IGHD1-1, IGHD1-14, IGHD1-20, IGHD1-26, IGHD1-7, IGHD2-15, IGHD2-2, IGHD2-21, IGHD2-8, IGHD3-10, IGHD3-16, IGHD3-22, IGHD3-3, IGHD3-9, IGHD4-11, IGHD4-17, IGHD4-23, IGHD4-4, IGHD5-12, IGHD5-18, IGHD5-24, IGHD5-5, IGHD6-13, IGHD6-19, IGHD6-25, IGHD6-6, IGHD7-27, IGHE, IGHEP1, IGHEP2, IGHG1, IGHG2, IGHG3, IGHG4, IGHGP, IGHJ1, IGHJ1P, IGHJ2, IGHJ2P, IGHJ3, IGHJ3P, IGHJ4, IGHJ5, IGHJ6, IGHM, IGHMBP2, IGHV1-12, IGHV1-14, IGHV1-17, IGHV1-18, IGHV1-2, IGHV1-24, IGHV1-3, IGHV1-45, IGHV1-46, IGHV1-58, IGHV1-67, IGHV1-68, IGHV1-69, IGHV1-8, IGHV1OR21-1, IGHV2-10, IGHV2-26, IGHV2-5, IGHV2-70, IGHV2OR16-5, IGHV3-11, IGHV3-13, IGHV3-15, IGHV3-16, IGHV3-19, IGHV3-20, IGHV3-21, IGHV3-22, IGHV3-23, IGHV3-25, IGHV3-29, IGHV3-30, IGHV3-30-2, IGHV3-32, IGHV3-33, IGHV3-33-2, IGHV3-35, IGHV3-36, IGHV3-37, IGHV3-38, IGHV3-41, IGHV3-42, IGHV3-43, IGHV3-47, IGHV3-48, IGHV3-49, IGHV3-50, IGHV3-52, IGHV3-53, IGHV3-54, IGHV3-57, IGHV3-6, IGHV3-60, IGHV3-62, IGHV3-63, IGHV3-64, IGHV3-65, IGHV3-66, IGHV3-7, IGHV3-71, IGHV3-72, IGHV3-73, IGHV3-74, IGHV3-75, IGHV3-76, IGHV3-79, IGHV3-9, IGHV3OR16-8, IGHV4-28, IGHV4-31, IGHV4-34, IGHV4-39, IGHV4-4, IGHV4-55, IGHV4-59, IGHV4-61, IGHV4-80, IGHV5-51, IGHV5-78, IGHV6-1, IGHV7-27, IGHV7-34-1, IGHV7-40, IGHV7-56, IGHV7-81, IGHVII-1-1, IGHVII-15-1, IGHVII-20-1, IGHVII-22-1, IGHVII-26-2, IGHVII-28-1, IGHVII-30-1, IGHVII-31-1, IGHVII-33-1, IGHVII-40-1, IGHVII-43-1, IGHVII-44-2, IGHVII-46-1, IGHVII-49-1, IGHVII-51-2, IGHVII-53-1, IGHVII-60-1, IGHVII-62-1, IGHVII-65-1, IGHVII-67-1, IGHVII-74-1, IGHVII-78-1, IGHVIII-11-1, IGHVIII-13-1, IGHVIII-16-1, IGHVIII-2-1, IGHVIII-22-2, IGHVIII-25-1, IGHVIII-26-1, IGHVIII-38-1, IGHVIII-44, IGHVIII-47-1, IGHVIII-5-1, IGHVIII-51-1, IGHVIII-5-2, IGHVIII-67-2, IGHVIII-67-3, IGHVIII-67-4, IGHVIII-76-1, IGHVIII-82 and IGHVIV-44-1.


For IGK, this included the following genes: IGKC, IGKJ1, IGKJ2, IGKJ3, IGKJ4, IGKJ5, IGKV1-12, IGKV1-13, IGKV1-16, IGKV1-17, IGKV1-22, IGKV1-27, IGKV1-32, IGKV1-33, IGKV1-35, IGKV1-37, IGKV1-39, IGKV1-5, IGKV1-6, IGKV1-8, IGKV1-9, IGKV1D-12, IGKV1D-13, IGKV1D-16, IGKV1D-17, IGKV1D-22, IGKV1D-27, IGKV1D-32, IGKV1D-33, IGKV1D-35, IGKV1D-37, IGKV1D-39, IGKV1D-42, IGKV1D-43, IGKV1D-8, IGKV1OR22-1, IGKV2-10, IGKV2-14, IGKV2-18, IGKV2-19, IGKV2-23, IGKV2-24, IGKV2-26, IGKV2-28, IGKV2-29, IGKV2-30, IGKV2-36, IGKV2-38, IGKV2-4, IGKV2-40, IGKV2D-10, IGKV2D-14, IGKV2D-18, IGKV2D-19, IGKV2D-23, IGKV2D-24, IGKV2D-26, IGKV2D-28, IGKV2D-29, IGKV2D-30, IGKV2D-36, IGKV2D-38, IGKV2D-40, IGKV2OR22-3, IGKV2OR22-4, IGKV3-11, IGKV3-15, IGKV3-20, IGKV3-25, IGKV3-31, IGKV3-34, IGKV3-7, IGKV3D-11, IGKV3D-15, IGKV3D-20, IGKV3D-25, IGKV3D-31, IGKV3D-34, IGKV3D-7, IGKV3OR22-2, IGKV4-1, IGKV5-2, IGKV6-21, IGKV6D-21, IGKV6D-41, and IGKV7-3.


For IGL, this included the following genes: IGLC1, IGLC2, IGLC3, IGLC4, IGLC5, IGLC6, IGLC7, IGLCOR22-1, IGLJ1, IGLJ2, IGLJ3, IGLJ4, IGLJ5, IGLJ6, IGLJ7, IGLL1, IGLL3, IGLON5, IGLV10-54, IGLV10-67, IGLV11-55, IGLV1-36, IGLV1-40, IGLV1-41, IGLV1-44, IGLV1-47, IGLV1-50, IGLV1-51, IGLV1-62, IGLV2-11, IGLV2-14, IGLV2-18, IGLV2-23, IGLV2-28, IGLV2-33, IGLV2-34, IGLV2-5, IGLV2-8, IGLV3-1, IGLV3-10, IGLV3-12, IGLV3-13, IGLV3-15, IGLV3-16, IGLV3-17, IGLV3-19, IGLV3-2, IGLV3-21, IGLV3-22, IGLV3-24, IGLV3-25, IGLV3-26, IGLV3-27, IGLV3-29, IGLV3-30, IGLV3-31, IGLV3-32, IGLV3-4, IGLV3-6, IGLV3-7, IGLV3-9, IGLV4-3, IGLV4-60, IGLV4-69, IGLV5-37, IGLV5-45, IGLV5-48, IGLV5-52, IGLV6-57, IGLV7-35, IGLV7-43, IGLV7-46, IGLV8-61, IGLV9-49, IGLVI-20, IGLVI-38, IGLVI-42, IGLVI-56, IGLVI-63, IGLVI-68, IGLVI-70, IGLVIV-53, IGLVIV-59, IGLVIV-64, IGLVIV-65, IGLVIV-66-1, IGLVV-58, IGLVV-66, IGLVVI-22-1, IGLVVI-25-1, and IGLVVII-41-1.


Patients with a best overall response of minimal response (MR) or better (i.e., MR, partial response [PR], very good partial response [VGPR] and complete response [CR]) were grouped into a responder category (N=27; abbreviated as “R”) and patients with a best overall response of stable disease (SD) or worse (i.e., SD and progressive disease [PD]) were grouped into a non-responder category (N=33; abbreviated as “NR”). Associations between response and expression of IGH, IGK and IGL loci were then tested with the Wilcoxon test, yielding the following results (Table 1):












TABLE 1







IG Locus
Wilcoxon P-Value



















IGH
0.003



IGK
0.49



IGL
0.05



Max (IGK, IGL)
0.13










Due to the strength of the association, further analysis focused on IGH expression (FIGS. 3A and 3B). Various IGH locus expression cutoffs were considered and it was ultimately determined that an FPKQ cutoff of 160,000 (defining “IGH-High”>=160,000 and “IGH-Low” <160,000) yielded the most clinically useful division the patients, predicting response in our training data with 55% sensitivity and 91% specificity. This same cutoff was then applied to time-to-progression (TTP) data for these patients, finding that IGH-High patients in this cohort have a 5.4-fold longer median TTP than IGH-Low patients (7.3 months vs. 1.3 months; log-rank P-value=0.003; FIG. 4).


To understand, how IG expression changes during treatment with proteasome inhibitors, the data was examined from one patient for which both a screening tumor sample and a sample collected on Cycle 1 Day 2 (C1D2) of treatment with single-agent carfilzomib were available. The results of this analysis are shown in Table 2.









TABLE 2







Expression












Gene
Screening
C1D2
Fold-Change







IGHA1
4.2E+05
1.3E+05
3.37



IGHV3-30
8.8E+04
2.4E+04
3.63



IGHV3-33
7.7E+04
2.1E+04
3.77



IGKC
5.3E+05
1.4E+05
3.88



ICKV3-20
3.9E+05
9.6E+04
4.11










As demonstrated in Table 2, IG expression is substantially (3- to 4-fold) lower in the sample collected one day after the first dose of carfilzomib. Similarly, RNA-Seq data from a multiple myeloma cell line (U266) continuously exposed to either bortezomib or carfilzomib for 24 hours was examined (FIG. 5). As in the patient samples above, expression of IG genes was substantially (3- to 4-fold) lower following exposure to either bortezomib or carfilzomib for 24 hours, which suggests that IG levels may be a proximal cause of death in cells exposed to proteasome inhibitors.


Looking more comprehensively, the inventors found a large set of genes (N=81) with expression that is significantly associated with response to carfilzomib (Wilcoxon P-value<10−3). The set includes an unexpectedly large number of genes encoding Ig folds that emerge as a small correlated cluster (FIG. 6), which implies that this class of proteins sensitizes cells to proteasome inhibition. This further suggests that high expression of immunoglobulin folds may be particularly proteotoxic to cells. A striking gene in this cluster is Fc gamma receptor 2B (FCGR2B), which is a receptor that normally binds Ig and down-modulates Ig production in B cells. Therefore, high FCGR2B expression may demarcate a tumor that is experiencing particularly high levels of proteotoxic stress from Ig production. Because of the close functional relationship between the IGH biomarker and FCGR2B, the inventors considered whether FCGR2B expression is an additional biomarker. The association between FCGR2B and response to carfilzomib (as defined above for IGH) is particularly strong (Wilcoxon P-value=4×10−4; FIGS. 7A, 7B). Unlike for IGH, for FCGR2B it is apparent that both high and low FPKQ cutoffs are valuable for predicting responder and non-responder categories, respectively. In this case we determined that a high FPKQ cutoff of 75 (defining “FCGR2B-High”>=75) and low FPKQ cutoff of 5 (defining “FCGR2B-Low”<5) yielded the most clinically useful division of our patients. The FCGR2B-High cutoff predicts responders in the training data with 37% sensitivity and 100% specificity, while the FCGR2B-Low cutoff predicts non-responders with 33% sensitivity and 96% specificity.


Combining the two FCGR2B cutoffs with a slightly lowered IGH-High cutoff of 140,000, we are able to achieve sensitivity of 70% and specificity of 94% for predicting responders in the training data. Next, these same cutoffs were applied to time-to-progression (TTP) data for these patients, finding that IGH+FCGR2B-Positive patients in this cohort have a 7.3-fold longer median TTP than IGH+FCGR2B-Negative patients (8.9 months vs. 1.2 months; log-rank P-value=3×10−5; FIG. 8).


Additional cutoffs for each individual biomarker (IGH and FCGR2B) and cutoffs for the combination of the two biomarkers were selected and the sensitivity and specificity for each of these additional cutoffs are listed in Tables 3A and 3B.












TABLE 3A






Biomarker




Biomarker
Cutoff
Sensitivity
Specificity


















FCGR2B
0
1
0


FCGR2B
1
0.962962963
0.151515152


FCGR2B
2
0.962962963
0.242424242


FCGR2B
3
0.962962963
0.272727273


FCGR2B
4
0.962962963
0.333333333


FCGR2B
7
0.962962963
0.363636364


FCGR2B
9
0.925925926
0.424242424


FCGR2B
10
0.851851852
0.424242424


FCGR2B
20
0.703703704
0.545454545


FCGR2B
30
0.592592593
0.666666667


FCGR2B
40
0.592592593
0.787878788


FCGR2B
50
0.481481481
0.818181818


FCGR2B
60
0.444444444
0.939393939


FCGR2B
70
0.37037037
0.96969697


FCGR2B
80
0.37037037
1


FCGR2B
100
0.259259259
1


FCGR2B
150
0.074074074
1


FCGR2B
170
0
1


IGH
0
1
0


IGH
10000
0.962962963
0.151515152


IGH
20000
0.925925926
0.212121212


IGH
30000
0.888888889
0.242424242


IGH
40000
0.888888889
0.242424242


IGH
50000
0.814814815
0.242424242


IGH
60000
0.814814815
0.272727273


IGH
70000
0.814814815
0.303030303


IGH
80000
0.814814815
0.363636364


IGH
90000
0.777777778
0.454545455


IGH
100000
0.777777778
0.484848485


IGH
110000
0.740740741
0.545454545


IGH
120000
0.703703704
0.606060606


IGH
130000
0.703703704
0.666666667


IGH
140000
0.703703704
0.696969697


IGH
150000
0.666666667
0.727272727


IGH
160000
0.666666667
0.727272727


IGH
200000
0.592592593
0.878787879


IGH
250000
0.333333333
0.909090909


IGH
300000
0.185185185
0.96969697


IGH
400000
0.074074074
0.96969697


IGH
450000
0.074074074
1


IGH
500000
0
1




















TABLE 3B





IGH_Cutoff
FCGR2B_low_Cutoff
FCGR2B_high_Cutoff
Sensitivity
Specificity



















100000
0
200
0
1


400000
100
200
0.074074074
1


1000000
0
110
0.148148148
1


200000
100
200
0.185185185
1


10000
100
200
0.222222222
1


1000000
3
110
0.259259259
1


150000
90
300
0.296296296
1


90000
90
500
0.333333333
1


1000000
2
90
0.37037037
1


90000
70
500
0.407407407
1


130000
50
110
0.444444444
0.96969697


200000
10
80
0.481481481
0.96969697


150000
40
90
0.518518519
0.96969697


110000
40
100
0.518518519
0.939393939


200000
10
200
0.518518519
0.909090909


200000
9
200
0.555555556
0.909090909


200000
8
500
0.592592593
0.909090909


200000
10
80
0.62962963
0.909090909


200000
9
90
0.666666667
0.909090909


200000
3
80
0.703703704
0.909090909


200000
2
80
0.703703704
0.878787879


200000
4
60
0.703703704
0.848484849


200000
0
60
0.703703704
0.818181818


140000
9
120
0.703703704
0.787878788


140000
5
80
0.777777778
0.787878788


130000
5
70
0.777777778
0.757575758


120000
2
80
0.777777778
0.727272727


200000
3
40
0.814814815
0.727272727


200000
0
40
0.814814815
0.696969697


200000
2
30
0.814814815
0.666666667


80000
5
70
0.814814815
0.636363636


80000
8
60
0.814814815
0.606060606


120000
5
40
0.851851852
0.606060606


100000
5
40
0.851851852
0.575757576


80000
4
40
0.888888889
0.575757576


80000
6
30
0.888888889
0.545454546


30000
6
110
0.888888889
0.515151515


10000
9
200
0.888888889
0.484848485


20000
5
100
0.925925926
0.484848485


40000
7
30
0.925925926
0.454545455


30000
3
40
0.925925926
0.424242424


10000
5
120
0.962962963
0.424242424


60000
5
10
0.962962963
0.393939394


140000
1
10
0.962962963
0.363636364


10000
2
60
0.962962963
0.333333333


100000
1
10
0.962962963
0.303030303


10000
1
100
0.962962963
0.272727273


70000
1
10
0.962962963
0.242424242


20000
0
100
0.962962963
0.212121212


20000
0
50
0.962962963
0.181818182


10000
0
200
0.962962963
0.151515152


40000
0
20
0.962962963
0.121212121


60000
0
10
1
0.090909091


90000
70
0
1
0









The % sensitivity and % specificity values shown in Tables 3A and 3B are datapoints of the response operating characteristic (ROC) curves shown in FIG. 9. FIG. 9 contains additional cutoffs (other than those in Tables 3A and 3B) as well as the corresponding % sensitivity and % specificity for each additional cutoff. The curve in FIG. 9 for IGH and FCGR2B (labeled as “IGH+FCGR2B”) allows for one to evaluate different combined cutoffs in terms of % sensitivity and % specificity. The different cutoffs thus provide for a multitude of diagnostic thresholds above which are determinative of a patient's treatment regimen. Cutoffs having the desired % sensitivity/% specificity values are then applied to TTP data.


Example 3: Additional Biomarkers

As discussed in Example 2, a large set of genes demonstrated expression that is significantly associated with patient response to carfilzomib. A table listing this set of genes is set forth herein as Table 4. Table 4 includes, for each gene: (i) the HUGO gene symbol, if available, (ii) the Ensembl Gene Name, (iii) gene expression level data, and (iv) statistical data: the P-value and Q-value.














TABLE 4








Δ gene




HUGO Gene


expression




Symbol
Ensembl Gene Name
log2(R/NR)
level
P-value
Q-value




















ABI2
ENSG00000138443
0.58949304
up
0.001738
0.1271


AC004381.6
ENSG00000005189
−1.204670581
down
0.000103
0.090927


AC005076.5
ENSG00000224046
−0.653150315
down
0.002953
0.154081


AC005104.3
ENSG00000223374
0.765035153
up
0.006398
0.199433


AC005943.5
ENSG00000267161
−1.004306556
down
0.003757
0.167074


AC006378.2
ENSG00000236861
0.923095301
up
0.00163
0.125963


AC007246.3
ENSG00000231312
0.507143559
up
0.001835
0.1271


AC007381.3
ENSG00000228590
1.794650455
up
0.001471
0.125963


AC007386.2
ENSG00000237638
1.314108591
up
0.001551
0.125963


AC009005.2
ENSG00000267751
−1.012595637
down
0.003809
0.167074


AC108488.3
ENSG00000234171
−0.530514717
down
0.005379
0.185044


ACAT2
ENSG00000120437
−0.691367312
down
0.001474
0.125963


ACOT7
ENSG00000097021
−1.127585567
down
0.006514
0.199433


ACYP1
ENSG00000119640
−0.679663744
down
0.001835
0.1271


ADCK1
ENSG00000063761
−0.429549728
down
0.004209
0.174124


ADIRF
ENSG00000148671
1.068032417
up
0.005074
0.182007


AGMAT
ENSG00000116771
−1.902702799
down
0.004209
0.174124


AKAP9
ENSG00000127914
0.578033255
up
0.005379
0.185044


ALCAM
ENSG00000170017
1.998126961
up
1.29E−05
0.067115


ALDH4A1
ENSG00000159423
−0.887790496
down
0.00488
0.180898


AMDHD2
ENSG00000162066
−0.533471154
down
0.001937
0.129464


AMH
ENSG00000104899
−1.185346863
down
0.005212
0.184691


ANKRD20A3
ENSG00000132498
2.350221687
up
0.000209
0.090927


ANKRD20A4
ENSG00000172014
2.863800069
up
0.004287
0.175935


ANKRD20A5P
ENSG00000186481
1.556593095
up
0.000788
0.112803


ANLN
ENSG00000011426
−1.397930353
down
0.000939
0.11973


ANTXR1
ENSG00000169604
2.166414183
up
0.004533
0.179635


APH1B
ENSG00000138613
0.516819563
up
0.006212
0.196883


APOBEC3H
ENSG00000100298
−1.351138348
down
0.000744
0.112803


ARHGAP11A
ENSG00000198826
−0.655219921
down
0.006212
0.196883


ARHGAP23
ENSG00000225485
0.522801324
up
0.004647
0.179635


ARHGAP31-
ENSG00000241155
0.652112201
up
0.000254
0.090927


AS1







ARL8B
ENSG00000134108
0.292126417
up
0.003809
0.167074


ARMC5
ENSG00000140691
−0.323867652
down
0.001474
0.125963


ASB1
ENSG00000065802
−0.417067595
down
9.32E−06
0.067115


ASF1B
ENSG00000105011
−0.792494369
down
0.003622
0.165504


ASPDH
ENSG00000204653
1.13291303
up
0.002752
0.149035


ASPH
ENSG00000198363
0.552936064
up
0.006514
0.199433


ATAD5
ENSG00000176208
−0.926161416
down
0.002804
0.149035


ATG4A
ENSG00000101844
0.454441032
up
0.001474
0.125963


ATP2C1
ENSG00000017260
0.414259932
up
0.002044
0.13169


ATXN7L2
ENSG00000162650
−0.602432936
down
0.001947
0.12962


AUNIP
ENSG00000127423
−1.420068922
down
0.002804
0.149035


B2M
ENSG00000166710
0.438515982
up
0.003272
0.160028


B4GALT4
ENSG00000121578
0.499817339
up
0.005922
0.194138


BAHD1
ENSG00000140320
0.521294292
up
0.001319
0.125963


BBX
ENSG00000114439
0.442989973
up
0.001835
0.1271


BCAR3
ENSG00000137936
1.528931715
up
0.001937
0.129464


BET1L
ENSG00000177951
0.544159583
up
0.004423
0.177259


BEX2
ENSG00000133134
1.389224675
up
0.001053
0.125345


BIK
ENSG00000100290
0.790115837
up
0.002156
0.135243


BIRC5
ENSG00000089685
−1.476698726
down
0.002526
0.144291


BMPR1A
ENSG00000107779
1.417631639
up
0.002526
0.144291


BRCA1
ENSG00000012048
−1.220603695
down
0.001179
0.125345


BTD
ENSG00000169814
0.7332119
up
0.001474
0.125963


BTLA
ENSG00000186265
1.270519458
up
0.002526
0.144291


C12orf23
ENSG00000151135
0.47094641
up
0.003809
0.167074


C14orf80
ENSG00000185347
−0.701479303
down
0.000552
0.1043


C16orf59
ENSG00000162062
−1.158684113
down
0.000622
0.106442


C17orf53
ENSG00000125319
−0.565512324
down
0.006514
0.199433


C19orf12
ENSG00000131943
0.739111952
up
0.000135
0.090927


C19orf40
ENSG00000131944
−0.636229388
down
0.002397
0.140035


C1orf112
ENSG00000000460
−0.903350952
down
0.001835
0.1271


C1orf132
ENSG00000203709
0.77824824
up
0.005124
0.182007


C22orf26
ENSG00000182257
−1.201787593
down
0.001295
0.125963


C3orf17
ENSG00000163608
0.355702903
up
0.001937
0.129464


C3orf37
ENSG00000183624
0.568954382
up
0.001319
0.125963


CACNB1
ENSG00000067191
−1.119138756
down
0.004647
0.179635


CAMK2B
ENSG00000058404
1.080919995
up
0.001079
0.125345


CAPN15
ENSG00000103326
−0.314834712
down
0.003272
0.160028


CAPN7
ENSG00000131375
0.429443198
up
0.000661
0.106442


CARHSP1
ENSG00000153048
−0.497561027
down
0.000661
0.106442


CASC4
ENSG00000166734
0.636516042
up
0.002044
0.13169


CBX7
ENSG00000100307
0.589142024
up
0.002804
0.149035


CCDC101
ENSG00000176476
−0.295016908
down
0.004005
0.170766


CCDC103
ENSG00000167131
−1.302299288
down
0.001137
0.125345


CCDC137
ENSG00000185298
−0.344295242
down
0.001646
0.125963


CCDC144CP
ENSG00000154898
1.94825361
up
0.001319
0.125963


CCDC18
ENSG00000122483
−0.661657101
down
0.001835
0.1271


CCDC34
ENSG00000109881
−1.207485494
down
0.001395
0.125963


CCHCR1
ENSG00000204536
−0.487115512
down
0.005713
0.1918


CCNA2
ENSG00000145386
−1.518792258
down
0.001937
0.129464


CCNF
ENSG00000162063
−0.797996486
down
0.000245
0.090927


CCPG1
ENSG00000260916
0.805364603
up
0.002044
0.13169


CD200
ENSG00000091972
2.296546672
up
0.000382
0.097757


CD276
ENSG00000103855
2.23215983
up
0.000202
0.090927


CD46
ENSG00000117335
0.535055566
up
0.005124
0.182007


CD47
ENSG00000196776
0.418206401
up
0.002397
0.140035


CDC25B
ENSG00000101224
−0.626962109
down
0.002397
0.140035


CDC45
ENSG00000093009
−1.488486593
down
0.00488
0.180898


CDC7
ENSG00000097046
−0.703380887
down
0.00488
0.180898


CDCA2
ENSG00000184661
−1.693705194
down
0.005124
0.182007


CDCA4
ENSG00000170779
−0.934167192
down
0.000279
0.092728


CDCA7L
ENSG00000164649
−0.90401625
down
0.003109
0.157059


CDCA8
ENSG00000134690
−1.131192478
down
0.001474
0.125963


CDKN3
ENSG00000100526
−0.866129394
down
0.003109
0.157059


CDT1
ENSG00000167513
−1.509518103
down
0.000788
0.112803


CENPC
ENSG00000145241
0.477584435
up
0.005379
0.185044


CENPH
ENSG00000153044
−1.059152551
down
0.006212
0.196883


CENPL
ENSG00000120334
−0.689597294
down
0.003622
0.165504


CENPW
ENSG00000203760
−1.336241011
down
8.35E−05
0.090927


CEP250
ENSG00000126001
−0.533656922
down
0.001646
0.125963


CEP55
ENSG00000138180
−1.336985816
down
0.004423
0.177259


CEP78
ENSG00000148019
−0.713457762
down
6.76E−05
0.086188


CFLAR-AS1
ENSG00000226312
0.787576615
up
0.001474
0.125963


CHAF1A
ENSG00000167670
−0.516246117
down
0.001646
0.125963


CHAF1B
ENSG00000159259
−1.117439741
down
0.000155
0.090927


CHTF18
ENSG00000127586
−0.923356052
down
6.21E−05
0.086188


CIT
ENSG00000122966
−1.008818794
down
0.001179
0.125345


CLIC2
ENSG00000155962
2.236011571
up
0.000245
0.090927


CLIC5
ENSG00000112782
1.869491174
up
0.001806
0.1271


CLPTM1
ENSG00000104853
0.466267313
up
0.000788
0.112803


CLSPN
ENSG00000092853
−1.383198126
down
0.000788
0.112803


CNTN1
ENSG00000018236
2.897399147
up
0.002156
0.135243


COPZ2
ENSG00000005243
2.563354592
up
0.000135
0.090927


CRBN
ENSG00000113851
0.479237667
up
0.003809
0.167074


CREB3L2
ENSG00000182158
1.297905196
up
9.60E−05
0.090927


CRELD2
ENSG00000184164
0.431307815
up
0.004209
0.174124


CRYBG3
ENSG00000080200
0.744498557
up
0.001319
0.125963


CRYBG3
ENSG00000233280
1.330532021
up
0.000519
0.103273


CSPG4P11
ENSG00000259726
1.650979674
up
0.000849
0.118158


CSPG4P12
ENSG00000259295
1.31746016
up
0.000744
0.112803


CTD-
ENSG00000244513
0.416073076
up
0.004647
0.179635


2013N24.2







CTD-
ENSG00000262526
1.864150843
up
0.001937
0.129464


2545G14.7







CTD-
ENSG00000247735
−1.395823366
down
0.003752
0.167074


2574D22.2







CTIF
ENSG00000134030
0.507094853
up
0.006212
0.196883


CTNNA1
ENSG00000044115
0.445702107
up
0.000177
0.090927


CXorf40B
ENSG00000197021
0.343593674
up
0.001558
0.125963


CXXC5
ENSG00000171604
0.520417324
up
0.005379
0.185044


CYFIP1
ENSG00000068793
0.603707882
up
0.001179
0.125345


CYP4F35P
ENSG00000265787
2.131597155
up
0.001551
0.125963


DAGLB
ENSG00000164535
−0.338981228
down
0.003443
0.164705


DBF4B
ENSG00000161692
−0.536907475
down
0.00488
0.180898


DBI
ENSG00000155368
0.437631696
up
0.003272
0.160028


DCK
ENSG00000156136
−0.710740223
down
0.002397
0.140035


DDX42
ENSG00000198231
0.208628409
up
0.002044
0.13169


DEPDC1
ENSG00000024526
−1.138548661
down
0.002953
0.154081


DERL1
ENSG00000136986
0.629070039
up
0.000622
0.106442


DERL3
ENSG00000099958
0.87425742
up
0.000552
0.1043


DGKI
ENSG00000157680
1.522742848
up
0.004528
0.179635


DHFR
ENSG00000228716
−0.596887056
down
0.001474
0.125963


DLG1
ENSG00000075711
0.720945313
up
0.003272
0.160028


DLGAP5
ENSG00000126787
−1.781943562
down
0.002526
0.144291


DNAJB9
ENSG00000128590
0.762742522
up
0.000215
0.090927


DNAJC1
ENSG00000136770
0.851198135
up
0.003622
0.165504


DNASE1L3
ENSG00000163687
1.150573975
up
0.003411
0.164705


DNMT1
ENSG00000130816
−0.401905618
down
0.001474
0.125963


DOT1L
ENSG00000104885
−0.440849834
down
0.001474
0.125963


DQX1
ENSG00000144045
−1.58805775
down
0.005822
0.194138


DST
ENSG00000151914
1.119680181
up
0.00488
0.180898


DTL
ENSG00000143476
−0.571087426
down
0.005458
0.187352


DTYMK
ENSG00000168393
−0.672857439
down
0.001835
0.1271


DVL3
ENSG00000161202
0.480840184
up
0.000622
0.106442


E2F1
ENSG00000101412
−0.713005902
down
0.003809
0.167074


E2F8
ENSG00000129173
−1.822880964
down
0.004647
0.179635


ECHDC2
ENSG00000121310
0.80675917
up
0.001319
0.125963


EFCAB11
ENSG00000140025
−0.83951759
down
0.000316
0.09315


EIF2AK4
ENSG00000128829
0.889621607
up
0.004005
0.170766


EIF2B5
ENSG00000145191
0.52014902
up
0.001179
0.125345


EMC7
ENSG00000134153
0.527652572
up
0.002156
0.135243


ENDOU
ENSG00000111405
0.996677877
up
0.000764
0.112803


EPDR1
ENSG00000086289
1.355276772
up
0.003109
0.157059


EPM2AIP1
ENSG00000178567
0.832768018
up
0.000622
0.106442


ERBB2
ENSG00000141736
−0.520928902
down
0.000939
0.11973


ERI1
ENSG00000104626
−0.601791404
down
0.000245
0.090927


ESCO2
ENSG00000171320
−1.5468643
down
0.006514
0.199433


ESPL1
ENSG00000135476
−0.932485167
down
0.003622
0.165504


ETV5
ENSG00000244405
0.880406053
up
0.001738
0.1271


FAM114A1
ENSG00000197712
0.986415082
up
0.00046
0.097757


FAM174A
ENSG00000174132
0.851426172
up
0.005124
0.182007


FAM219A
ENSG00000164970
0.431957782
up
0.001474
0.125963


FAM64A
ENSG00000129195
−1.675400995
down
0.000432
0.097757


FAM72B
ENSG00000188610
−0.874624959
down
0.000261
0.090927


FAM83D
ENSG00000101447
−0.996366514
down
0.000203
0.090927


FAM95B1
ENSG00000223839
1.618293895
up
0.004209
0.174124


FANCA
ENSG00000187741
−0.802052008
down
0.000406
0.097757


FANCB
ENSG00000181544
−0.733853593
down
0.001137
0.125345


FBLN2
ENSG00000163520
4.055675182
up
0.002318
0.139712


FBXL15
ENSG00000107872
0.347540494
up
0.00488
0.180898


FCF1
ENSG00000119616
−0.367208753
down
0.000432
0.097757


FCGR2B
ENSG00000072694
1.7069366
up
0.000552
0.1043


FCGR2C
ENSG00000244682
1.833667492
up
0.001937
0.129464


FCRL5
ENSG00000143297
1.284877883
up
0.001179
0.125345


FCRLA
ENSG00000132185
3.193292318
up
8.96E−05
0.090927


FGFR1OP
ENSG00000213066
−0.491542609
down
0.003272
0.160028


FOXM1
ENSG00000111206
−1.131740677
down
0.004332
0.176822


FUS
ENSG00000089280
−0.352688172
down
0.000261
0.090927


FXYD5
ENSG00000089327
0.875760601
up
0.001646
0.125963


FXYD7
ENSG00000221946
1.006169767
up
0.003585
0.165504


GALE
ENSG00000117308
−0.542602271
down
0.002953
0.154081


GBF1
ENSG00000107862
0.476441929
up
0.000279
0.092728


GINS2
ENSG00000131153
−0.805724953
down
0.003809
0.167074


GINS4
ENSG00000147536
−0.996858828
down
0.000661
0.106442


GLMN
ENSG00000174842
−0.690322855
down
0.001247
0.125963


GMEB1
ENSG00000162419
−0.25590121
down
0.004423
0.177259


GNG7
ENSG00000176533
0.927431553
up
0.004209
0.174124


GOLGA4
ENSG00000144674
0.386722654
up
0.004005
0.170766


GOLGA6L4
ENSG00000184206
0.812716533
up
0.000189
0.090927


GPBP1
ENSG00000062194
0.242730418
up
0.006514
0.199433


GPR176
ENSG00000166073
2.035062721
up
0.001114
0.125345


GPRASP1
ENSG00000198932
1.071771461
up
0.003443
0.164705


GPSM2
ENSG00000121957
−0.885489534
down
6.29E-05
0.086188


GRIP1
ENSG00000155974
1.231430795
up
0.004231
0.174124


GSG2
ENSG00000177602
−1.394544327
down
0.000406
0.097757


GTF2I
ENSG00000077809
0.40001011
up
0.000886
0.118955


HAUS4
ENSG00000092036
−0.602117697
down
0.002397
0.140035


HDAC9
ENSG00000048052
0.684068321
up
0.001646
0.125963


HELLS
ENSG00000119969
−0.623431519
down
0.001558
0.125963


HENMT1
ENSG00000162639
−0.624504013
down
0.001474
0.125963


HERPUD1
ENSG00000051108
0.778585363
up
0.001646
0.125963


HLA-DOB
ENSG00000241106
1.443362687
up
3.51E-05
0.067115


HMGB1P5
ENSG00000132967
−0.489741805
down
0.005645
0.190341


HMGN2
ENSG00000198830
−0.343343082
down
0.004647
0.179635


HNRNPCP1
ENSG00000258900
−0.953716452
down
0.004743
0.180898


HSDL2
ENSG00000119471
−0.610519971
down
0.006212
0.196883


ICAM5
ENSG00000105376
−1.044794924
down
0.004209
0.174124


IDUA
ENSG00000127415
0.955710969
up
0.001646
0.125963


IFT20
ENSG00000109083
0.302770683
up
0.003109
0.157059


ITPRIPL1
ENSG00000198885
−2.2280116
down
0.000586
0.106442


ITSN1
ENSG00000205726
0.905232008
up
0.002804
0.149035


KATNA1
ENSG00000186625
−0.391521115
down
0.001179
0.125345


KCNAB1
ENSG00000169282
1.001128869
up
0.000359
0.097757


KCNAB3
ENSG00000170049
−1.056336096
down
0.002273
0.138647


KIAA0226
ENSG00000145016
0.617682853
up
0.002953
0.154081


KIAA0586
ENSG00000100578
−0.431753645
down
0.006212
0.196883


KIAA1147
ENSG00000257093
0.482751184
up
0.006514
0.199433


KIAA1522
ENSG00000162522
1.035387539
up
0.005124
0.182007


KIAA1731
ENSG00000166004
−0.469427264
down
0.000788
0.112803


KIF14
ENSG00000118193
−1.705333985
down
0.001474
0.125963


KIF18A
ENSG00000121621
−0.692732773
down
0.001179
0.125345


KIF20B
ENSG00000138182
−0.628519438
down
0.001053
0.125345


KIF2C
ENSG00000142945
−1.365646338
down
0.000432
0.097757


KIF4A
ENSG00000090889
−1.904610649
down
0.003622
0.165504


KMT2C
ENSG00000055609
0.228004365
up
0.005645
0.190341


KPNB1
ENSG00000108424
−0.394700087
down
0.001835
0.1271


L2HGDH
ENSG00000087299
−0.921327169
down
0.000836
0.117391


LAMTOR5-
ENSG00000224699
−0.981944468
down
0.002047
0.13169


AS1







LINC00337
ENSG00000225077
−1.120082576
down
0.005676
0.19098


LINC00662
ENSG00000261824
0.773101729
up
0.005922
0.194138


LINC00883
ENSG00000243701
0.810773121
up
0.004005
0.170766


LMF1
ENSG00000103227
0.814937221
up
0.000622
0.106442


LPCAT3
ENSG00000111684
−0.559209562
down
0.004005
0.170766


LRRC4B
ENSG00000131409
0.793959329
up
0.004213
0.174124


LRRN2
ENSG00000170382
1.865843931
up
0.005922
0.194138


MAGED2
ENSG00000102316
0.720903224
up
0.001179
0.125345


MAP4
ENSG00000047849
0.724838313
up
0.002662
0.147631


MAP4K3
ENSG00000011566
0.589878912
up
0.004209
0.174124


MAPRE3
ENSG00000084764
0.784298761
up
0.000261
0.090927


MBD4
ENSG00000129071
0.276423792
up
0.000788
0.112803


MCM10
ENSG00000065328
−1.334114767
down
0.001674
0.1271


MCM2
ENSG00000073111
−1.247745579
down
0.001474
0.125963


MCM4
ENSG00000104738
−0.927747184
down
0.001646
0.125963


MEF2A
ENSG00000068305
0.506720645
up
0.000744
0.112803


MEI1
ENSG00000167077
1.179961962
up
0.000118
0.090927


MEMO1
ENSG00000162959
−0.281226311
down
0.004209
0.174124


MFN1
ENSG00000171109
0.332545316
up
0.006212
0.196883


MGAT1
ENSG00000131446
0.290644604
up
0.006514
0.199433


MGME1
ENSG00000125871
−0.433132545
down
0.003809
0.167074


MGST3
ENSG00000143198
0.659094058
up
0.005124
0.182007


MICA
ENSG00000204520
0.620119411
up
0.000939
0.11973


MID2
ENSG00000080561
0.756163973
up
0.005645
0.190341


MIR4435-1HG
ENSG00000172965
−1.062297175
down
0.00488
0.180898


MKX
ENSG00000150051
2.704795186
up
0.000382
0.097757


MLF1IP
ENSG00000151725
−0.868339353
down
0.005645
0.190341


MMRN2
ENSG00000173269
1.180392072
up
0.002662
0.147631


MOXD1
ENSG00000079931
1.559687372
up
0.004647
0.179635


MRAS
ENSG00000158186
1.52251306
up
0.00488
0.180898


MSH2
ENSG00000095002
−1.009515534
down
0.006514
0.199433


MTFR2
ENSG00000146410
−0.846036223
down
0.000489
0.099767


MXD4
ENSG00000123933
0.515542459
up
0.004423
0.177259


MYBL2
ENSG00000101057
−1.035211825
down
0.001247
0.125963


MYEF2
ENSG00000104177
1.112353906
up
0.005124
0.182007


MZB1
ENSG00000170476
0.965983178
up
0.002662
0.147631


NAA30
ENSG00000139977
−0.341182009
down
0.002662
0.147631


NBEA
ENSG00000172915
1.297503689
up
0.005124
0.182007


NCAPH
ENSG00000121152
−1.60849361
down
0.005379
0.185044


NDFIP1
ENSG00000131507
0.620791298
up
0.000202
0.090927


NDUFA3
ENSG00000170906
0.577079384
up
0.002662
0.147631


NEDD4L
ENSG00000049759
1.124026207
up
0.001247
0.125963


NOL12
ENSG00000256872
−0.5896443
down
0.00046
0.097757


NPRL3
ENSG00000103148
−0.392961317
down
0.00046
0.097757


NR1D2
ENSG00000174738
0.455919632
up
0.003622
0.165504


NR3C1
ENSG00000113580
0.731585314
up
0.005124
0.182007


NSUN5
ENSG00000130305
−0.478595971
down
0.00488
0.180898


NUDT1
ENSG00000106268
−0.852740182
down
0.000155
0.090927


NUF2
ENSG00000143228
−1.103956787
down
0.001558
0.125963


NXPE4
ENSG00000137634
4.468868564
up
0.003745
0.167074


OIP5
ENSG00000104147
−1.202560379
down
0.00488
0.180898


ORC1
ENSG00000085840
−1.065526966
down
0.001835
0.1271


P4HTM
ENSG00000178467
0.696412296
up
0.003272
0.160028


PAIP2B
ENSG00000124374
0.608423558
up
0.003443
0.164705


PAM
ENSG00000145730
0.736380555
up
0.001835
0.1271


PAQR4
ENSG00000162073
−1.249506432
down
0.000316
0.09315


PARL
ENSG00000175193
0.385732857
up
0.000519
0.103273


PARPBP
ENSG00000185480
−1.127390341
down
0.001319
0.125963


PATZ1
ENSG00000100105
−0.613716272
down
0.002044
0.13169


PC
ENSG00000173599
−2.196281547
down
0.001835
0.1271


PCBP3
ENSG00000183570
1.620969616
up
0.001558
0.125963


PCBP4
ENSG00000090097
1.027008279
up
1.76E−05
0.067115


PCDHB10
ENSG00000120324
1.367015984
up
0.002959
0.154081


PCDHB16
ENSG00000196963
2.272079546
up
0.001461
0.125963


PCDHB9
ENSG00000177839
1.659119974
up
0.000156
0.090927


PCDHGA10
ENSG00000253846
1.761871736
up
0.001852
0.12771


PCYT1A
ENSG00000161217
0.388019625
up
0.006514
0.199433


PDCD1LG2
ENSG00000197646
1.007150591
up
0.005645
0.190341


PDE6G
ENSG00000185527
−2.06520501
down
0.001474
0.125963


PDE8A
ENSG00000073417
0.898071797
up
0.000661
0.106442


PDIA3
ENSG00000167004
0.599823335
up
8.96E−05
0.090927


PDXDC2P
ENSG00000196696
0.390023823
up
0.002156
0.135243


PDZK1IP1
ENSG00000162366
2.059953642
up
0.005845
0.194138


PFN2
ENSG00000070087
1.726786978
up
0.002804
0.149035


PGP
ENSG00000184207
−0.394712174
down
0.001738
0.1271


PHF19
ENSG00000119403
−0.59874177
down
0.000432
0.097757


PKMYT1
ENSG00000127564
−1.403609452
down
0.001937
0.129464


PLCB4
ENSG00000101333
1.358514715
up
0.006212
0.196883


PLK1
ENSG00000166851
−0.980915772
down
0.002804
0.149035


PLK4
ENSG00000142731
−0.863342434
down
0.000215
0.090927


POLA2
ENSG00000014138
−0.356587586
down
0.005645
0.190341


POLD3
ENSG00000077514
−0.649754527
down
3.02E−05
0.067115


PON2
ENSG00000105854
1.064018977
up
3.25E−05
0.067115


POPDC2
ENSG00000121577
0.507108724
up
0.004647
0.179635


POU2F2
ENSG00000028277
0.829755089
up
0.001319
0.125963


PPFIBP2
ENSG00000166387
0.701578337
up
0.004209
0.174124


PPIB
ENSG00000166794
0.480516293
up
0.002804
0.149035


PPID
ENSG00000171497
−0.266755933
down
0.002804
0.149035


PPIP5K1
ENSG00000168781
0.808884722
up
0.000939
0.11973


PPP1R16B
ENSG00000101445
−1.201260806
down
0.003809
0.167074


PRIM1
ENSG00000198056
−0.995746296
down
0.000297
0.092779


PRKAA1
ENSG00000132356
0.401998563
up
0.001738
0.1271


PRKAR1B
ENSG00000188191
−0.660679976
down
0.002397
0.140035


PRKCA
ENSG00000154229
1.369576279
up
0.001474
0.125963


PSENEN
ENSG00000205155
0.368704282
up
0.002662
0.147631


PTHLH
ENSG00000087494
2.864363043
up
0.000427
0.097757


PTPRM
ENSG00000173482
1.799116837
up
0.003443
0.164705


RAB3B
ENSG00000169213
1.817774395
up
0.005124
0.182007


RABAC1
ENSG00000105404
0.682722137
up
0.004005
0.170766


RAC3
ENSG00000169750
−0.783424905
down
0.00488
0.180898


RAD18
ENSG00000070950
−0.388012916
down
0.005922
0.194138


RAD51B
ENSG00000182185
−0.647226192
down
0.004866
0.180898


RAPGEF3
ENSG00000079337
1.460778486
up
0.001835
0.1271


RASGRP3
ENSG00000152689
1.361593186
up
0.000886
0.118955


RBBP8
ENSG00000101773
−0.420670803
down
0.006514
0.199433


RBKS
ENSG00000171174
−0.607072797
down
0.002273
0.138647


RBL1
ENSG00000080839
−0.651514248
down
0.003109
0.157059


RECQL4
ENSG00000160957
−1.055176871
down
0.002273
0.138647


REEP5
ENSG00000129625
0.483198621
up
0.00046
0.097757


RFC3
ENSG00000133119
−0.845003493
down
0.005379
0.185044


RFC5
ENSG00000111445
−0.488787867
down
0.002804
0.149035


RHOQ
ENSG00000119729
0.981684777
up
0.001319
0.125963


RIC3
ENSG00000166405
2.061495289
up
0.000622
0.106442


RMDN3
ENSG00000137824
0.687762803
up
0.000661
0.106442


RMI2
ENSG00000175643
−1.105998037
down
0.000701
0.111769


RNASEH2A
ENSG00000104889
−0.588788676
down
0.003272
0.160028


RNF13
ENSG00000082996
0.430500699
up
0.000406
0.097757


RNF168
ENSG00000163961
0.789370661
up
0.001835
0.1271


RP11-110I1.12
ENSG00000255121
−1.089180897
down
0.001053
0.125345


RP11-
ENSG00000247679
0.494691737
up
0.005124
0.182007


1277A3.1







RP11-
ENSG00000261050
1
up
0.001558
0.125963


145F16.2







RP11-22P6.3
ENSG00000260442
−1.218957746
down
0.000305
0.09315


RP11-23N2.4
ENSG00000260618
0.599669898
up
0.006212
0.196883


RP11-295D4.1
ENSG00000262712
−0.543310808
down
0.004227
0.174124


RP11-382J12.1
ENSG00000246366
0.476637496
up
0.005645
0.190341


RP11-
ENSG00000258017
−0.732892348
down
0.002273
0.138647


386G11.10







RP11-
ENSG00000250116
0.874469118
up
0.002044
0.13169


417F21.1







RP11-
ENSG00000260872
0.813273679
up
0.001646
0.125963


680G24.5







RP11-690I21.1
ENSG00000237641
0.982445377
up
0.001045
0.125345


RP11-81H14.2
ENSG00000251301
1.154574974
up
0.002273
0.138647


RP1-290I10.6
ENSG00000229950
1.515905482
up
0.006057
0.196883


RP3-412A9.11
ENSG00000198832
1.332016018
up
5.86E−05
0.086188


RP4-742C19.8
ENSG00000233899
0.631077104
up
0.002526
0.144291


RPL7AP10
ENSG00000240522
−1.230279468
down
0.002679
0.148063


RPN1
ENSG00000163902
0.384107056
up
0.003622
0.165504


RPS11P5
ENSG00000232888
0.666696911
up
0.004645
0.179635


RSPH3
ENSG00000130363
−0.543360762
down
0.005922
0.194138


RTKN
ENSG00000114993
0.84231452
up
0.002273
0.138647


RTN4IP1
ENSG00000130347
−0.635965627
down
0.004005
0.170766


S100Z
ENSG00000171643
1.755116374
up
0.000586
0.106442


SAPCD1
ENSG00000228727
−0.731740287
down
0.006514
0.199433


SAPCD2
ENSG00000186193
−1.176135844
down
0.005922
0.194138


SASS6
ENSG00000156876
−0.232149316
down
0.005124
0.182007


SBNO1
ENSG00000139697
−0.315910334
down
0.003622
0.165504


SCAMP5
ENSG00000198794
0.861087469
up
0.000836
0.117391


SDHAP1
ENSG00000185485
0.572219514
up
0.000489
0.099767


SDHAP3
ENSG00000185986
−0.83746263
down
0.006514
0.199433


SEC13
ENSG00000157020
0.55661178
up
0.001053
0.125345


SEC22C
ENSG00000093183
0.355646837
up
0.001738
0.1271


SEC61A1
ENSG00000058262
0.610846513
up
0.000886
0.118955


SEC62
ENSG00000008952
0.788131385
up
0.00011
0.090927


SELK
ENSG00000113811
0.619756413
up
0.000432
0.097757


SENP2
ENSG00000163904
0.491242791
up
0.002273
0.138647


SERP1
ENSG00000120742
0.427941701
up
0.000886
0.118955


SERPINI1
ENSG00000163536
1.401734038
up
0.004423
0.177259


SGOL1
ENSG00000129810
−1.315415063
down
0.001179
0.125345


SGOL2
ENSG00000163535
−1.04754504
down
0.005379
0.185044


SH3BP5-AS1
ENSG00000224660
0.61664396
up
0.000489
0.099767


SH3PXD2A
ENSG00000107957
0.855689288
up
0.000441
0.097757


SHCBP1
ENSG00000171241
−0.653402279
down
0.005379
0.185044


SHMT1
ENSG00000176974
−0.916266061
down
0.005379
0.185044


SIT1
ENSG00000137078
1.491042511
up
0.003622
0.165504


SKA1
ENSG00000154839
−1.388698342
down
0.002156
0.135243


SKA3
ENSG00000165480
−1.49596618
down
0.001179
0.125345


SLAMF1
ENSG00000117090
2.637174504
up
0.000552
0.1043


SLC22A5
ENSG00000197375
0.364043198
up
0.005379
0.185044


SLC29A2
ENSG00000174669
−2.117487943
down
0.003109
0.157059


SLC35B1
ENSG00000121073
0.54289746
up
0.001558
0.125963


SLC35G2
ENSG00000168917
2.36121245
up
0.000417
0.097757


SLC51A
ENSG00000163959
0.619956943
up
0.003272
0.160028


SLIT1
ENSG00000187122
2.454001823
up
0.005971
0.195061


SLX4
ENSG00000188827
−0.309372634
down
0.005922
0.194138


SMPD1
ENSG00000166311
0.685554031
up
0.003622
0.165504


SOGA3
ENSG00000214338
2.141127737
up
0.004321
0.176822


SOWAHB
ENSG00000186212
2.27080955
up
0.001624
0.125963


SOX5
ENSG00000134532
1.658450619
up
0.003578
0.165504


SRGAP3
ENSG00000196220
1.029665782
up
0.000215
0.090927


SRI
ENSG00000075142
0.37152875
up
0.000744
0.112803


SSR3
ENSG00000114850
0.4896736
up
0.001319
0.125963


ST6GAL1
ENSG00000073849
0.766044282
up
0.000939
0.11973


ST8SIA4
ENSG00000113532
0.850904988
up
0.001247
0.125963


STARD4
ENSG00000164211
0.367759238
up
0.006212
0.196883


STARD5
ENSG00000172345
0.709779447
up
0.003272
0.160028


STIL
ENSG00000123473
−0.577961832
down
0.003622
0.165504


STMN1
ENSG00000117632
−0.958006821
down
0.005124
0.182007


TANC2
ENSG00000170921
2.470345102
up
0.004645
0.179635


TBC1D31
ENSG00000156787
−0.55473257
down
0.005379
0.185044


TBL3
ENSG00000183751
−0.371607611
down
0.00488
0.180898


TCEAL3
ENSG00000196507
1.039621308
up
0.000215
0.090927


TCF12
ENSG00000140262
0.573433117
up
0.003272
0.160028


TCHP
ENSG00000139437
−0.394692495
down
0.006212
0.196883


TEX19
ENSG00000182459
−1.232891533
down
0.005548
0.189988


TEX30
ENSG00000151287
−0.741242721
down
0.000997
0.125345


TFG
ENSG00000114354
0.52087412
up
0.004647
0.179635


THOC6
ENSG00000131652
−0.412866319
down
0.000261
0.090927


TICAM2
ENSG00000243414
0.757722382
up
0.002044
0.13169


TIMELESS
ENSG00000111602
−0.624056848
down
0.000661
0.106442


TK1
ENSG00000167900
−1.120895213
down
0.002804
0.149035


TMCO6
ENSG00000113119
−0.446075861
down
0.002662
0.147631


TMED7
ENSG00000134970
0.506103886
up
0.006514
0.199433


TMEM108
ENSG00000144868
1.262393672
up
0.005335
0.185044


TMEM115
ENSG00000126062
0.360490115
up
0.001179
0.125345


TMEM150A
ENSG00000168890
0.292194
up
0.006514
0.199433


TMEM243
ENSG00000135185
0.771028169
up
0.005379
0.185044


TMEM50B
ENSG00000142188
0.479584127
up
0.003443
0.164705


TMEM57
ENSG00000204178
0.382913174
up
0.005124
0.182007


TMEM63C
ENSG00000165548
2.463768871
up
0.002526
0.144291


TMOD2
ENSG00000128872
1.778485679
up
0.006212
0.196883


TMPO-AS1
ENSG00000257167
−0.726697259
down
0.003851
0.168443


TPX2
ENSG00000088325
−1.221950415
down
0.003809
0.167074


TRGV5
ENSG00000211697
0.933839652
up
0.004528
0.179635


TRIM52
ENSG00000183718
0.558151646
up
0.004209
0.174124


TROAP
ENSG00000135451
−1.70863633
down
0.000297
0.092779


TSKU
ENSG00000182704
1.51653736
up
0.005976
0.195061


TSPAN3
ENSG00000140391
0.841070593
up
0.002156
0.135243


TTC17
ENSG00000052841
0.342525973
up
0.003622
0.165504


TTK
ENSG00000112742
−0.988321296
down
0.003109
0.157059


TTLL5
ENSG00000119685
−0.327071817
down
0.000406
0.097757


TUB
ENSG00000166402
1.767676597
up
0.002317
0.139712


TUBA1B
ENSG00000123416
−0.538417101
down
0.001646
0.125963


TXNDC15
ENSG00000113621
0.575328234
up
0.003809
0.167074


TXNDC5
ENSG00000239264
0.756015355
up
0.003443
0.164705


TYMS
ENSG00000176890
−1.081232402
down
0.002397
0.140035


UBA7
ENSG00000182179
0.856044718
up
0.001395
0.125963


UBE2C
ENSG00000175063
−1.269467358
down
0.001179
0.125345


UBE2E2
ENSG00000182247
0.870140435
up
2.40E−05
0.067115


UBXN4
ENSG00000144224
0.38181031
up
0.006514
0.199433


UHRF1
ENSG00000034063
−1.469473531
down
0.001738
0.1271


USP32P3
ENSG00000189423
3.195455286
up
0.001473
0.125963


VAPB
ENSG00000124164
−0.257533585
down
0.004423
0.177259


VIMP
ENSG00000131871
0.687773457
up
0.003809
0.167074


VPS37D
ENSG00000176428
1.550120285
up
0.005095
0.182007


WASL
ENSG00000106299
0.492658686
up
0.004647
0.179635


WBP5
ENSG00000185222
1.50717312
up
0.000297
0.092779


WDHD1
ENSG00000198554
−1.152433926
down
0.001632
0.125963


WDR62
ENSG00000075702
−1.166051204
down
0.000245
0.090927


WEE1
ENSG00000166483
−1.043540913
down
0.000359
0.097757


XRCC2
ENSG00000196584
−0.775604531
down
0.005922
0.194138


XRCC3
ENSG00000126215
−0.655292946
down
0.000939
0.11973


YIPF2
ENSG00000130733
0.686984318
up
0.003109
0.157059


ZBTB4
ENSG00000174282
0.59662837
up
0.005124
0.182007


ZC2HC1A
ENSG00000104427
0.629482721
up
0.003622
0.165504


ZGLP1
ENSG00000220201
−0.463720366
down
0.005124
0.182007


ZHX1-
ENSG00000259305
0.431039293
up
0.001835
0.1271


C80RF76







ZMAT3
ENSG00000172667
0.647710648
up
0.002397
0.140035


ZNF101
ENSG00000181896
−0.605225083
down
0.001474
0.125963


ZNF14
ENSG00000105708
−0.800698079
down
0.002662
0.147631


ZNF204P
ENSG00000204789
3.260061269
up
0.002317
0.139712


ZNF358
ENSG00000198816
0.767920473
up
0.003109
0.157059


ZNF565
ENSG00000196357
0.615820899
up
0.005922
0.194138


ZNF609
ENSG00000180357
0.312449173
up
0.001114
0.125345


ZNF706
ENSG00000120963
0.465206779
up
0.004005
0.170766


ZNF738
ENSG00000172687
−1.270195442
down
0.006256
0.197853


ZNF827
ENSG00000151612
0.722894271
up
0.006212
0.196883


ZNF829
ENSG00000185869
−1.490325627
down
0.001114
0.125345


ZNF852
ENSG00000178917
0.526992277
up
0.00488
0.180898


ZNF880
ENSG00000221923
−2.04089136
down
0.004423
0.177259


ZNF93
ENSG00000184635
−1.691498564
down
0.003943
0.170766


Not available
ENSG00000233488
−1.126553625
down
1.76E−05
0.067115


Not available
ENSG00000259850
0.317164674
up
0.00488
0.180898


Not available
ENSG00000165406
0.486776476
up
0.000135
0.090927


Not available
ENSG00000233165
1.051483435
up
0.00488
0.180898


Not available
ENSG00000266348
1.615869627
up
0.002819
0.149305





Log2(R/NR), gene expression level of responders relative (R) to non-responders (NR). The terms “responder” and “non-responder” used in this example have the same meanings as those in Example 2.


Δ Gene expression level indicates the change in gene expression level of responders (R) relative to non-responders (NR); “up” means that the gene expression level was upregulated in R vs. NR and “down” means that the gene expression level was downregulated in R vs. NR.


Not available, HUGO gene name is not available.






Expression data was determined as essentially described in Example 2. Associations between response and expression of the indicated gene were tested with the Wilcoxon test yielding the P values indicated in Table 4. Q was determined as essentially described in Storey and Tibshirani, “Statistical significance for genome-wide experiments, Proceedings of the National Academy of Sciences 100:9440-9445 (2003).


Genes demonstrating strength of association between expression and response to carfilzomib are further analyzed by considering different expression cutoffs. An ROC curve for each gene is made so that each cutoff may be evaluated in terms of % specificity and % sensitivity. The cutoff is then applied to time-to-progression (TTP) data for the patients and clinical relevance is considered.


Example 4: Validation Studies

The analysis of bone marrow samples from carfilzomib (CFZ) Phase 2 clinical studies has shown that high IGH and FCGR2B [0] gene expression levels are predictive of response to the proteasome inhibitor carfilzomib in relapsed and refractory multiple myeloma patients [1]. Bone marrow samples from the carfilzomib Phase 3 clinical studies are analyzed to confirm these analysis results. Additionally, it is determined whether high IGH and FCGR2B gene expression levels are predictive of response to other therapeutic regimens such as corticosteroids plus optional cyclophosphamide (best supportive care; BSC) in patients with relapsed and refractory multiple myeloma and lenalidomide plus dexamethasone (Rd) in patients with multiple myeloma who have received one to three prior lines of therapy.


RNA sequencing data from CD138+ selected bone marrow samples from the carfilzomib Phase 3 clinical studies are analyzed with the goal of confirming that high IGH and/or FCGR2B gene expression levels are predictive of response to the proteasome inhibitor CFZ and not predictive of response to other therapeutic regimens, such as corticosteroids plus optional cyclophosphamide (BSC) and lenanlidomide plus dexamethasone (Rd). At the time of patient screening during the trials, bone marrow samples were collected and aspirated. These samples were used to obtain RNA sequencing data. RNA sequencing of these samples are performed at the Translational Genomics Research Institute (TGen).


The RNA samples are divided across the trial arms as follows: There were a total of 192 samples collected as part of the Phase 3 trial from patients treated either with CFZ or according to best supportive care (BSC) that were successfully sequenced. There were 424 samples collected as part of the Phase 3 trial from patients treated either with CFZ, lenalidomide, and dexamethasone (CRd) or with lenalidomide and dexamethasone (Rd) that were successfully sequenced.


Raw sequence reads are aligned and expression of genes and isoforms are quantified with a customized pipeline built in Array Studio v6.1. This pipeline [3] accepts Illumina adapter stripped, paired-end reads that are trimmed at the 5′ end if a base reaches PHRED quality score Q2 or lower. All reads are mapped to the transcriptome, as defined by the Ensembl annotation R.70 [4]. Reads mapped with mismatches and unmapped reads are subsequently aligned to the entire human genome, searching for novel exon junctions. Mappings of a particular read pairs to the genome and transcriptome are compared and the highest scoring mapping is kept, with transcriptome mappings preferred in the case of a tie. Reads that remain unmapped at this point are aligned against the newly identified exon junctions. Finally, all transcriptome mapping locations are translated to genomic coordinates to estimate the expected number of mappings using the EM algorithm [5]. The EM algorithm assigns reads with multiple mapping locations to a transcript isoform by calculating the conditional probability of a read mapping to a specific isoform, given all other mappings. Weighting the total number of mapped reads with this probability yields posterior expected read counts for the transcript. The EM counts are normalized by the length of the genes and number of reads in a library to yield FPKM values (Fragments Per Kilobase per Million reads). An additional normalization, referred to as quantile normalization, is applied to correct for biases introduced by the presence of one or two dominant transcripts (e.g., IGH, IGK, & IGL) in many of the samples. In the quantile normalization step each FPKM value in a sample is scaled by the 85th percentile FPKM value of that same sample to yield quantile normalized FPKM, referred to as FPKQ values.


IGH consists of many separately annotated genes in the ENSEMBL annotation. Therefore, estimates of IGH expression are calculated by summing the corresponding FPKQ of each gene of the IGH locus (cf. USPA No. 61/863,809 ‘Immunoglobulin Expression Levels as Biomarker for Proteasome Inhibitor’, 9/2013, for details). Expression cutoffs for IGH and FCGR2B determined as described above, are employed to quantify enrichment of responders, PFS, and OS on the four clinical trial arms in the biomarker positive versus negative subsets. For this analysis responders are defined as patients achieving a best overall response as determined by the PI of MR, PR, VGPR, or CR and non-responders as SD and PD.


The determined thresholds are then used to select the biomarker positive and negative samples for the biomarker subgroup analysis. In each group, comparison between treatments arms are performed and reported in tables accompanied by survival plots. A Cox regression model including the treatment group is fit and the hazard ratio for the CFZ arm and BSC (Rd) arm and their corresponding 95% confidence intervals are reported. A hazard ratio smaller than 1 implies that an extension of PFS in the CFZ arm compared to the BSC (Rd) arm was observed within the biomarker positive or negative subgroup of patients. The ratio of biomarker positive and biomarker negative hazard ratios are determined to fulfill the key Go criteria. A Wald interaction test is performed and a p-value reported as an indication whether the treatment effect varies according to biomarker status.


Example 5: qPCR

The RNA-Seq analysis of bone marrow samples from carfilzomib (CFZ) Phase 2 clinical studies has shown that high IGH and FCGR2B [0] gene expression levels are predictive of response to the proteasome inhibitor carfilzomib in relapsed and refractory multiple myeloma patients [1]. Bone marrow samples from the carfilzomib Phase 2 and Phase 3 clinical studies are analyzed using an alternative method to confirm these analysis results.


RNA from CD138+ selected bone marrow samples from the carfilzomib Phase 2 and Phase 3 clinical studies are analyzed with the goal of confirming that high IGH and/or FCGR2B gene expression levels can be measured using an RT-qPCR assay and can be used to determine predictiveness of response to the proteasome inhibitor CFZ and non predictiveness of response to other therapeutic regimens, such as corticosteroids plus optional cyclophosphamide (BSC) and lenanlidomide plus dexamethasone (Rd). At the time of patient screening during the trials, bone marrow samples were collected and aspirated.


There were a total of 75 samples collected as part of the Phase 2 study. There were a total of 192 samples collected as part of the Phase 3 trial from patients treated either with CFZ or according to best supportive care (BSC). There were 424 samples collected as part of the Phase 3 trial from patients treated either with CFZ, lenalidomide, and dexamethasone (CRd) or with lenalidomide and dexamethasone (Rd).


First, reverse transcription of RNA previously used for RNA-Seq data is performed. Then RT-qPCR (quantitative polymerase chain reaction) [2][3] is performed to determine the amount of Ig and/or FCGR2B in a sample by measuring the threshold cycle (Ct) or crossing point value. The Ct reflects the cycle at which the measured signal exceeds a defined background threshold. The flourescence signal is measured at the end of each amplification cycle and the Ct value results from the interpolation of the two signal measurements between which the threshold was crossed [2]. Negative specimens do not yield a Ct value. The quantitative Ct value is negatively associated with the (log) concentration of nucleic acids detected, i.e. the higher the Ct value the lower the input concentration.


Targeted primers are used to amplify the IgH locus and/or the FCGR2B gene of each sample. The expression levels of these two targets are normalized using house-keeping genes. A correlation between Ig and/or FCGR2B expression levels as measured using the RNA-Seq data (cf. Example 4) and as measured using the RT-qPCR method is established. RT-qPCR being representative of the amount of Ig and/or FCGR2B in a patient sample is validated. A pilot study performed on 19 samples from the Phase 3 trials has shown a good correlation (RA2=0.919) between RNA-Seq and RT-qPCR expression leves of the FCGR2B gene.


After this correlation is established for the Ig locus, an optimal cutoff to distinguish between responders and non-responders is determined by calculating sensitivity and specificity for all possible cut-off combinations within the range of the assay. An ROC analogous to the method described above is used to find an optimal cutoff to enrich for responders in the data.


The determined thresholds are then used to select the biomarker positive and negative samples for the biomarker subgroup analysis. In each group, comparisons between treatments arms are performed and reported in tables accompanied by survival plots. A Cox regression model including the treatment group is fitted and the hazard ratio for the CFZ arm and BSC (Rd) arm and their corresponding 95% confidence intervals are reported. A hazard ratio smaller than 1 implies that an extension of PFS in the CFZ arm compared to the BSC (Rd) arm was observed within the biomarker positive or negative subgroup of patients. A Wald interaction test is performed and a p-value reported as an indication whether the treatment effect varies according to biomarker status.


Example 6: Immunofluorescence Assay

An immunofluorescence (IF) based assay [2] is used to quantify the amount of Ig and/or FCGR2B protein in a sample. The IF technique comprises of two phases: (1) slide preparation (specimen fixation and permeabilization) and immunoreaction (in order: antigen retrieval, non-specific site block, primary antibody incubation, secondary incubation, couterstaining with a nuclear dye, and mounting the slide); (2) employment of systems of detection, interpretation and quantification of the obtained expression. As part of the clinical Phase 2 and 3 trials, cytospin slides of bone marrow aspirates were prepared from each sample. To visually confirm the presence of plasma cells, a CD138+ antibody was used to stain the sample. Specific antibodies against Ig and/or FCGR2B antigens that can also be visualized by staining are employed. The amount of staining is quantified using a flourescence microscope and standard image processing tools.


A correlation between Ig and/or FCGR2B expression levels as measured using the RNA-Seq data (cf. Example 4) and as measured using the IF method is establised. The IF assay being representative of the amount of Ig and/or FCGR2B in a patient sample is validated.


After this correlation is established, an optimal cutoff to distinguish between responders and non-responders is determined by calculating sensitivity and specificity for all possible cut-off combinations within the range of the assay. An ROC analogous to the method described above is used to find an optimal cutoff to enrich for responders in our data.


The determined thresholds are then used to select the biomarker positive and negative samples for the biomarker subgroup analysis. In each group, comparison between treatments arms are performed and reported in tables accompanied by survival plots. A Cox regression model including the treatment group is fitted and the hazard ratio for the CFZ arm and BSC (Rd) arm and their corresponding 95% confidence intervals are reported. A hazard ratio smaller than 1 implies that an extension of PFS in the CFZ arm compared to the BSC (Rd) arm was observed within the biomarker positive or negative subgroup of patients. A Wald interaction test is performed and a p-value reported as an indication whether the treatment effect varies according to biomarker status.


Example 7: Validation

This example demonstrates that sensitivity to carfilzomib correlates with level of expression of immunoglobulin.


Two hybridoma cell lines, Line A and Line B, were tested for immunoglobulin (Ig) gene expression levels by ELISA assay. Either hybridoma cell culture supernatant or hybridoma cell lysates were added to wells containing antibody specific for mouse IgG (Cat. No. E99-131; Lot No. E99-131-130419 Bethyl Laboratories Inc). As shown in FIG. 11A, hybridoma cells of Line A expressed Ig to a greater extent, relative to the Ig expression levels exhibited by Line B. Line A accordingly was considered as a high Ig-expressing (high Ig) hybridoma cell line, and Line B was considered a low Ig-expressing (low Ig) hybridoma cell lines.


The two hybridoma cell lines were tested for sensitivity to carfilzomib. Cells of each of Line A (high Ig) and Line B (low Ig) were treated with various doses (40 nM, 30 nM, 20 nM, 15 nM, 10 nM, 7.5 nM, 5 nM, 3.75 nM and 2.5 nM) of carfilzomib for 72 h. The viability of the cells were then measured by CellTiter-Glo® Luminescent Cell Viability Assay (Cat. No. G7570; Promega Corporation) and compared to no treatment control. As shown in FIG. 11B, cells of Line A were more sensitive to carfilzomib, relative to cells of Line B. The inhibitory concentration (IC50) of carfilzomib on the cells of Line A was 10.43 nM, whereas the IC50 of carfilzomib on the cells of Line B was 24.30.


The drug sensitivity assay performed on two additional pairs of hybridoma cell lines, each pair consisted of a high and a low Ig-expressing hybridoma cell line produced similar results to those achieved with Line A and Line B. For each pair, the cells of the high Ig-expressing hybridoma cell lines demonstrated a greater sensitivity to carfilzomib, relative to the cells of the low Ig-expressing lines.


Example 8: Quantitative RT-PCR

Taqman primers and probes for PCR amplification of FCGR2B was purchased from Life Technologies (Cat #4331182).


CD138 positive cells were collected from bone-marrow aspirates obtained from patients. Total RNA was then isolated from these cells using Trizol reagent (Cat. #15596-026; Life Technologies). The cDNAs were made using QuantiTect Reverse Transcription kit from Qiagen (Cat. #205310) and qRT-PCR assays were performed using Taqman assay reagents (Cat. #4440042; Life Technologies).


As shown in FIG. 12, the amplification and quantification of the expression of FCGR2B using the above primer pairs were successful. A strong correlation between RNA Seq data (FPKQ) and qRT-PCR data (R2=0.9563) indicate that the Taqman assay can be reliably used to determine FCGR2B transcript level from patient samples. Development of similar assays for IGH genes are in progress.


Once the assays are developed patient samples from a Phase II clinical trial will be subjected to the same quantitative RT-PCR measurements to establish a qRT-PCR based cut-off that will separate the responders from the non-responders.


Patient samples from a Phase III clinical trial will then be subjected to the same qRT-PCR assays and the cut-off established from Phase II study will be applied to parse responder from non-responder.


REFERENCES

References cited in Examples 1 and 2:

  • Dempster, A. P.; Laird, N. M., Rubin, D. B. (1977). Maximum Likelihood from Incomplete Data via the EM Algorithm. Journal of the Royal Statistical Society, Series B 39 (1): 1-38
  • Jun Hu, Huanying Ge, Matt Newman and Kejun Liu. OSA: a fast and accurate alignment tool for RNA-Seq. Bioinformatics (2012) 28 (14): 1933-1934.
  • Mulligan G, Mitsiades C, Bryant B, Zhan F, Chng W J, Roels S, Koenig E, Fergus A, Huang Y, Richardson P, Trepicchio W L, Broyl A, Sonneveld P, Shaughnessy J D Jr, Bergsagel P L, Schenkein D, Esseltine D L, Boral A, Anderson K C. Gene expression profiling and correlation with outcome in clinical trials of the proteasome inhibitor bortezomib. Blood. 2007 Apr. 15; 109(8):3177-88.
  • Rody A, Holtrich U, Pusztai L, Liedtke C, Gaetje R, Ruckhaeberle E, Solbach C, Hanker L, Ahr A, Metzler D, Engels K, Karn T, Kaufmann M. T-cell metagene predicts a favorable prognosis in estrogen receptor-negative and HER2-positive breast cancers. Breast Cancer Res. 2009; 11(2):R15.


REFERENCES CITED IN EXAMPLE 4



  • [0] Gray, K. A., Daugherty, L. C., Gordon, S. M., Seal, R. L., Wright, M. W., Bruford, E. A. ‘genenames.org: the HGNC resources in 2013’ Nucleic Acids Res. 2013 January; 41(Database issue):D545-52. doi: 10.1093/nar/gks1066. Epub 2012 Nov. 17 PMID:23161694

  • [1] Tuch, B. B., Löhr, A., Degenhardt, J. D., Kwei, K. A., Lowe, E., Keats, J. J., Kirk, C. J. USPA No. 61/863,809 ‘Immunoglobulin Expression Levels as Biomarker for Proteasome Inhibitor’, 9/2013

  • [2] Robert Hajek, Richard Vryce, Sunhee Ro, Barbara Klencke, Heinz Ludwig ‘Design and rationale of FOCUS(PX-171-011): A randomized, open-label, phase 3 study of carfolzomib versus best supportive care regimen in patients with relapsed and refractory multiple myeloma’ BMC Cancer 2012, 12:415

  • [3] Jun Hu, Huanying Ge, Matt Newman and Kejun Liu ‘OSA: a fast and accurate alignment tool for RNA-Seq’ Bioinformatics (2012) 28 (14): 1933-1934.

  • [4] Paul Flicek, Ikhlak Ahmed, M. Ridwan Amode, Daniel Barrell, Kathryn Beal, Simon Brent, Denise Carvalho-Silva, Peter Clapham, Guy Coates, Susan Fairley, Stephen Fitzgerald, Laurent Gil, Carlos Garcia-Girón, Leo Gordon, Thibaut Hourlier, Sarah Hunt, Thomas Juettemann, Andreas Kähäri, Stephen Keenan, Monika Komorowska, Eugene Kulesha, Ian Longden, Thomas Maurel, William McLaren, Mattieu Muffato, Rishi Nag, Bert Overduin, Miguel Pignatelli, Bethan Pritchard, Emily Pritchard, Harpreet Singh Riat, Graham R. S. Ritchie, Magali Ruffier, Michael Schuster, Daniel Sheppard, Daniel Sobral, Kieron Taylor, Anja Thormann, Stephen Trevanion, Simon White, Steven P. Wilder, Bronwen L. Aken, Ewan Birney, Fiona Cunningham, Ian Dunham, Jennifer Harrow, Javier Herrero, Tim J. P. Hubbard, Nathan Johnson, Rhoda Kinsella, Anne Parker, Giulietta Spudich, Andy Yates, Amonida Zadissa and Stephen M. J. Searle ‘Ensembl 2013’ Nucleic Acids Research 2013 41 Database issue:D48-D55

  • [5] Dempster, A. P.; Laird, N. M., Rubin, D. B. (1977) ‘Maximum Likelihood from Incomplete Data via the EM Algorithm’ Journal of the Royal Statistical Society, Series B 39 (1): 1-38

  • [6] Soreide, K. (2009) ‘Receiver-operating characteristic curve analysis in diagnostic, prognostic and predictive biomarker research’, Journal of Clinical Pathology 62:1-5

  • [7] Jiang, W., Freidlin, B., Simon, R. (2007) ‘Biomarker-adaptive threshold design: a procedure for evaluating treatment with possible biomarker-defined subset effect’ Journal of the National Cancer Institute 99(13):1036-43



REFERENCES CITED IN EXAMPLE 5



  • [0] Gray, K. A., Daugherty, L. C., Gordon, S. M., Seal, R. L., Wright, M. W., Bruford, E. A. ‘genenames.org: the HGNC resources in 2013’ Nucleic Acids Res. 2013 January; 41(Database issue):D545-52. doi: 10.1093/nar/gks1066. Epub 2012 Nov. 17 PMID:23161694

  • [1] Tuch, B. B., Löhr, A., Degenhardt, J. D., Kwei, K. A., Lowe, E., Keats, J. J., Kirk, C. J. USPA No. 61/863,809 ‘Immunoglobulin Expression Levels as Biomarker for Proteasome Inhibitor’, 9/2013

  • [2] Caraguel, C. G. B., Stryhn, H., Gagne, N., Dohoo, I. R., Hammell, K. L., “Selecting a Cutoff Value for Real-Time Polymerase Chain Reaction Results to Fit a Diasgnostic Purpose”, J VET Diagn Invest 2011, 32:2

  • [3] Mackay I M, Arden K E, Nitsche A: 2002, “Real-time PCR in virology”. Nucleic Acids Res 30:1292-1305.



REFERENCES CITED IN EXAMPLE 6



  • [0] Gray, K. A., Daugherty, L. C., Gordon, S. M., Seal, R. L., Wright, M. W., Bruford, E. A. ‘genenames.org: the HGNC resources in 2013’ Nucleic Acids Res. 2013 January; 41(Database issue):D545-52. doi: 10.1093/nar/gks1066. Epub 2012 Nov. 17 PMID:23161694

  • [1] Tuch, B. B., Löhr, A., Degenhardt, J. D., Kwei, K. A., Lowe, E., Keats, J. J., Kirk, C. J. USPA No. 61/863,809 ‘Immunoglobulin Expression Levels as Biomarker for Proteasome Inhibitor’, 9/2013

  • [2] Odell, I. D., Cook D., ‘Immunofluorescence Techniques’, J of Investig. Dermatology, 2013, 133 e4



A number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims.

Claims
  • 1.-20. (canceled)
  • 21. A method of treating a tumor from which a sample was obtained, wherein the level of expression of (i) immunoglobulin (Ig), (ii) Fc gamma receptor 2B (FCGR2B), or (iii) both Ig and FCGR2B of the sample was greater than a reference level, the method comprising the step of administering an effective amount of a proteasome inhibitor.
  • 22. The method of claim 21, wherein the proteasome inhibitor is carfilzomib, bortezomib, disulfiram, and oprozomib.
  • 23. The method of claim 22, wherein the proteasome inhibitor is carfilzomib.
  • 24. The method of claim 21, wherein the tumor is a hematological tumor.
  • 25. The method of claim 21, wherein the tumor is in a subject and the subject has multiple myeloma, optionally, relapsed or refractory multiple myeloma.
  • 26. The method of claim 25, wherein the subject has newly diagnosed multiple myeloma.
  • 27. The method of claim 25, wherein the subject received one to three prior lines of therapy.
  • 28. The method of claim 21, wherein the sample comprised CD138-positive tumor cells and the level of expression of (i) immunoglobulin (Ig), (ii) Fc gamma receptor 2B (FCGR2B), or (iii) both Ig and FCGR2B of the sample was greater than a reference level.
  • 29. The method of claim 21, wherein the reference level is a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
  • 30. The method of claim 29, wherein the ROC curve is based on (i) the distribution of Ig expression levels and/or FCGR2B expression levels of responders and (ii) the distribution of Ig expression levels and/or FCGR2B expression levels of non-responders.
  • 31. The method of claim 21, wherein the level of Ig expression and/or FCGR2B expression is at least 2-fold greater than the reference level.
  • 32. A method for treating a tumor in a subject, wherein the level of Ig expression and/or Fc gamma receptor 2B (FCGR2B) expression in the subject is greater than a reference level, said method comprising administering to the subject an effective amount of a proteasome inhibitor.
  • 33. A method for treating a tumor in a subject, said method comprising administering to the subject an effective amount of a proteasome inhibitor when the level of Ig expression and/or Fc gamma receptor 2B (FCGR2B) expression in the tumor is greater than a reference level.
  • 34. The method of claim 33, wherein: a. the proteasome inhibitor is carfilzomib, bortezomib, disulfiram, and oprozomib,b. the tumor is a hematological tumor,c. the tumor is in a subject and the subject has multiple myeloma, optionally, relapsed or refractory multiple myeloma, and/ord. the tumor comprises CD138-positive tumor cells and the level of expression of (i) immunoglobulin (Ig), (ii) Fc gamma receptor 2B (FCGR2B), or (iii) both Ig and FCGR2B of the tumor cells is greater than a reference level.
  • 35. The method of claim 34, wherein the proteasome inhibitor is carfilzomib.
  • 36. The method of claim 34, wherein the subject has newly diagnosed multiple myeloma.
  • 37. The method of claim 34, wherein the subject received one to three prior lines of therapy.
  • 38. The method of claim 33, wherein the reference level is a cutoff correlative with a % specificity of at least 50% and a % sensitivity of at least 50%, as determined by a receiver operating characteristic (ROC) curve.
  • 39. The method of claim 38, wherein the ROC curve is based on (i) the distribution of Ig expression levels and/or FCGR2B expression levels of responders and (ii) the distribution of Ig expression levels and/or FCGR2B expression levels of non-responders.
  • 40. The method of claim 33, wherein the level of Ig expression and/or FCGR2B expression is at least 2-fold greater than the reference level.
CROSS REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 17/129,573, filed Dec. 21, 2020, which is a division of U.S. patent application Ser. No. 14/910,668, filed Feb. 5, 2016, which is the U.S. national stage application of International Application No. PCT/US14/50333, filed Aug. 8, 2014, which claims the priority benefit of Provisional U.S. Patent Application No. 61/863,809, filed on Aug. 8, 2013, Provisional U.S. Patent Application No. 61/875,954, filed on Sep. 10, 2013, and Provisional U.S. Patent Application No. 62/005,904, filed on May 30, 2014, each of which is incorporated by reference in its entirety.

Provisional Applications (3)
Number Date Country
62005904 May 2014 US
61875954 Sep 2013 US
61863809 Aug 2013 US
Divisions (1)
Number Date Country
Parent 14910668 Feb 2016 US
Child 17129573 US
Continuations (1)
Number Date Country
Parent 17129573 Dec 2020 US
Child 17903700 US