US20210010091A1 - Classifier for the molecular classification of multiple myeloma - Google Patents

Classifier for the molecular classification of multiple myeloma Download PDF

Info

Publication number
US20210010091A1
US20210010091A1 US17/029,564 US202017029564A US2021010091A1 US 20210010091 A1 US20210010091 A1 US 20210010091A1 US 202017029564 A US202017029564 A US 202017029564A US 2021010091 A1 US2021010091 A1 US 2021010091A1
Authority
US
United States
Prior art keywords
gene
risk
patients
genes
multiple myeloma
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US17/029,564
Inventor
Rowan Kuiper
Pieter Sonneveld
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Erasmus University Medical Center
Original Assignee
Erasmus University Medical Center
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Erasmus University Medical Center filed Critical Erasmus University Medical Center
Priority to US17/029,564 priority Critical patent/US20210010091A1/en
Publication of US20210010091A1 publication Critical patent/US20210010091A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • C12Q1/6837Enzymatic or biochemical coupling of nucleic acids to a solid phase using probe arrays or probe chips
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • This application is in the field of molecular diagnostics and relates to a method for classifying samples obtained from patients diagnosed with multiple myeloma.
  • the application also relates to a method for determining the prognosis of an individual diagnosed with multiple myeloma as well as a method for the prediction of the response to treatment of an individual diagnosed with multiple myeloma.
  • MM Multiple myeloma
  • OS Median overall survival
  • ISS International Staging System
  • MM Based on cytogenetics, two classes of MM can be distinguished with implications for MM biology and prognosis.
  • Hyperdiploid MM ⁇ 60% of patients, characterized by trisomies of multiple odd chromosomes (3, 5, 7, 9, 11, 15, 19, and 21) has a relatively good prognosis.
  • Non-hyperdiploid MM ⁇ 40% of cases, is characterized by recurrent translocations involving the immunoglobulin heavy chain gene at 14q32, resulting in transcriptional activation of CCND1, CCND3, MAF, MAFB, or FGFR3/MMSET.
  • UAMS University of Arkansas for Medical Sciences
  • MM a number of classifications for myeloma consists of seven distinct gene expression clusters, including translocation clusters MS, MF, and CD-1/2, as well as a hyperdiploid cluster (HY), a cluster with proliferation-associated genes (PR), and a cluster characterized by low percentage of bone disease (LB).
  • HY hyperdiploid cluster
  • PR proliferation-associated genes
  • LB low percentage of bone disease
  • Gene expression is able to explain an even larger amount of variance in survival compared to ISS and cytogenetics.
  • One of the first survival signatures based on gene expression was the UAMS-70-gene classifier, and the further refined UAMS-17-gene classifier.
  • Other classifiers include the Millennium signature, the MRC-IX-6-gene signature, and the IFM classifier.
  • signatures were reported to predict plasma cell proliferation such as the recently published gene expression proliferation index (GPI).
  • the aim of this study was to develop a prognostic signature, based upon gene expression profiles (GEPs) of MM patients, treated with either standard induction treatment or bortezomib induction, followed in both cases by high-dose melphalan and maintenance.
  • GEPs gene expression profiles
  • a classifier comprising a 92-gene set capable of distinguishing between patients with a high risk and patients with a low risk.
  • the classifier yielded excellent results wherein the classification in the low risk group identified patients with a good overall survival, whereas the group identified as high risk showed significantly worse overall survival rates.
  • the disclosure therefore, relates to a method for determining the disease outcome or the prognosis of a patient diagnosed with multiple myeloma by classifying the patient into a high risk or a low risk category, the method comprising the steps of:
  • an EMC-92 score above a predetermined threshold indicates that the patient is to be classified in the high risk category and a score at or below the predetermined threshold indicates that the patient is to be classified in the low risk category.
  • FIGS. 1A-1D Performance of the EMC-92 classifier in predicting overall survival. High risk signature on four validation sets with a fixed cut-off value of 0.827.
  • FIG. 1A UAMS Total Therapy 2
  • FIG. 1B UAMS Total Therapy 3
  • FIG. 1C MRC-IX
  • FIG. 1D APEX.
  • FIG. 2 Relation between threshold and log-rank performance of the EMC-92 signature in the HOVON-65/GMMG-HD4 OS.
  • the model has an optimal performance for thresholds of at least 0.75.
  • a cut-off for high-risk was based on defining high-risk as having an OS of ⁇ 2 years within the training set, which corresponded to a threshold of 0.827.
  • a classifier comprising a 92-gene set capable of distinguishing between patients with a high risk and patients with a low risk is disclosed herein.
  • the classifier yielded excellent results wherein the classification in the low risk group identified patients with a good overall survival, whereas the group identified as high risk showed significantly worse overall survival rates.
  • the classifier was validated in an experimental setting wherein patients with poor overall survival (OS) were distinguished from patients with standard OS. Therefore, an SPCA model was built using the HOVON65/GMMG-HD4 data as a training set (see the experimental section below). A number of 1088 probe sets were found to be associated with progression-free survival (PFS) in a univariate Cox regression analysis (FDR ⁇ 10%). Based on these probe sets, a classifier with 92 probe sets was developed (Table 1). This classifier will be termed the EMC-92-gene signature.
  • a dichotomizing cut-off threshold was based on the clinically relevant definition of high-risk patients as those patients who have an overall survival of less than 2 years. This amounted to a proportion of 21.7% in the training set and a cut-off value of 0.827.
  • HOVON65/GMMG-HD4 Three datasets were available for this analysis: HOVON65/GMMG-HD4, APEX and MRC-IX.
  • Multivariate analysis on the HOVON65/GMMG-HD4 study demonstrates that together with the EMC-92-gene signature, del(17p), ⁇ 2 m[ ⁇ 3.5 mg/L] and allogenic transplantation were significantly related to shorter survival, whereas WHO status [0] was found to be significantly related to longer survival.
  • APEX albumin level
  • ISS and IgG isotype were found to be significantly related.
  • MRC-IX mainly ISS related covariants were found.
  • the EMC-92-gene classifier exclusively identified 31 patients correctly as high risk patients, which were missed by the UAMS-17-gene classifier (50% survival rate of 11, 24 and 51 months for the shared high risk group, intermediate high risk group and standard risk groups, respectively). Moreover, the UAMS-17-gene classifier exclusively identified 10 patients as high risk patients with a lower hazard ratio as compared to the 31 patients classified as high-risk by the EMC-92-gene classifier.
  • EMC-92-gene classifier The superiority of the EMC-92-gene classifier was even clearer in the APEX population.
  • 24 patients were exclusively identified as high risk in the EMC-92-gene classifier, which were missed in the UAMS-17-gene classifier.
  • These 24 patients formed a group whose overall survival after 20 months was 14% whereas the high risk population identified in both the EMC-92-gene and UAMS-17-gene classifier showed an overall survival of 25% after 20 months.
  • the UAMS-70-gene, MRC-IX-6-gene signature, GPI score, the Millennium and IFM signature were applied to the datasets.
  • the EMC-92-gene classifier had the highest hazard ratios and lowest p values of all classifiers.
  • the intersection of high-risk patients between the EMC-92-gene and UAMS-17-gene classifiers was ⁇ 8% of the total population. About 14% of patients were classified as high-risk by either one of these classifiers.
  • the 14% of patients uniquely classified as high-risk by either signature showed an intermediate hazard-ratio.
  • prognosis of MM patients is mainly based on ISS-stage and interphase fluorescence in situ hybridization (FISH).
  • FISH interphase fluorescence in situ hybridization
  • Previous classifiers include the UAMS-17/70-gene and MRC-IX-6-gene classifiers, both capable of predicting in independent datasets. [11, 14] In contrast, the Millennium and the IFM signatures demonstrate less solid performance in independent validation sets. [12, 13]
  • the EMC-92-gene expression signature presented herein is highly discriminative for patients with high-risk versus standard-risk MM across different (induction) regimes.
  • Validation in UAMS TT2 (thalidomide-based), [17] TT3 (bortezomib-based), [18] and MRC-IX trial (thalidomide maintenance in both young and elderly patients) [19, 20] showed high performance in these independent test environments. This is true for both the continuous fit of the model, which is a goodness-of-fit indicator, as well as the dichotomized output into high-risk/standard-risk, which is a requirement for practical use in a clinical setting.
  • the EMC-92-gene high-risk signature remains a strong predictor for early death. Still, there is strong evidence that ISS staging (serum albumin and ⁇ 2 m levels) turns out to be another major contributor for explaining survival-related variance in the presence of the signature. Therefore, incorporating ISS into the signature could potentially lead to an even better prediction of survival.
  • HY hyperdiploid patients with a generally favorable prognosis
  • MS and MF represent patients with translocations t(4;14) and t(14;16/20), which are usually thought to have an unfavorable prognosis.
  • PR represents the proliferation cluster, which was shown to be associated with poor prognosis.
  • chromosomal location of 1 q was highly enriched (Table 1) as was previously shown for the UAMS-17-gene signature.
  • probe sets located on chromosome 4 are enriched. These probe sets were found to be scattered over the entire chromosome and not only at the distal end of the p arm where MIVISET and FGFR3 are located. Chromosome 4 has previously not been considered a risk factor, but a low frequency of multiple gains and/or losses affecting this chromosome has been reported.
  • the EMC-92-gene signature was compared in a multivariable analysis to the UAMS-17/70-gene, MRC-IX-6-gene, GPI score, IFM and Millennium classifiers. Three pooled datasets were formed from publicly available MM datasets, allowing an independent comparison of the signatures that were not trained on those datasets (R. Kuiper et al., Leukemia 2012, 1-8 incorporated herein by reference). The outputs from the signatures were input into a Cox proportional hazards model, see Table 2. In all three comparisons, the EMC-92-gene signature obtained the most significant hazard ratio (HR) and, thus, is the most relevant prognostic factor of all signatures (including the UAMS-70 from Signal Genetics).
  • HR hazard ratio
  • the EMC-92-gene signature turns out to have the best dichotomized performance on its validation sets. Moreover, in comparison to other classifiers, the proportion of high-risk patients is higher. One would expect that differences between high-risk and standard-risk become less pronounced as the high-risk proportion increases. It should be mentioned that even at this high proportion, differences in survival time are larger for the EMC-92 as compared to other classifiers selecting smaller risk groups.
  • the EMC-92-gene signature had the strongest discriminative ability.
  • this study concerns the development of a robust high-risk signature, incorporates most known prognostic markers, clinical, cytogenetic and GEP based, and shows the developed EMC-92-gene signature to be the strongest independent prognostic marker for poor survival known.
  • This EMC-92-gene signature is able to select out a high-risk group of MM patients for whom in the future alternative, more intensive treatments should be sought.
  • the disclosure relates to a method for determining the disease outcome or the prognosis of a patient diagnosed with multiple myeloma by classifying the patient into a high risk or a low risk category, the method comprising the steps of:
  • an EMC-92 score above a predetermined threshold indicates that the patient is to be classified in the high-risk category and a score at or below the threshold indicates that the patient is to be classified in the low-risk category.
  • a preferred threshold value is at least 0.75, especially preferred is a threshold value of 0.827.
  • a signature must have both the ability to separate risk groups as clearly as possible and to predict stable groups of relevant size.
  • the EMC-92-gene signature meets both criteria.
  • a high-risk group of patients can be significantly determined and the proportion of high-risk patients is stable across the validation sets.
  • the validation sets represent different drug regimens, including thalidomide (MRC-IX, TT2) and bortezomib (APEX, TT3).
  • the signature is relevant to both transplant-eligible (for example, TT3) and non-transplant-eligible patients (subset of MRC-IX), as well as newly diagnosed (for example, TT2) and relapsed patients (APEX).
  • the predictions of the IFM-15 and MILLENNIUM-100 signatures in the validation sets fail to reach significance in independent data sets such as MRC-IX and TT3.
  • the HOVON65/GMMG-HD4 data was used as a training set. This multicenter trial compared the efficacy of bortezomib (PAD) to standard treatment (VAD) in newly diagnosed patients. Patients were randomized to induction treatment with three VAD or PAD cycles, [16] for a total of 290 patients, both follow-up and GEPs were available. [9]
  • TT2 and TT3 were derived from clinical trials performed in newly diagnosed patients, both treated with a complex regimen. The first was a randomized prospective treatment trial in which patients were randomly assigned to receive or not to receive thalidomide during all phases of treatment. [17] The latter was carried out by the same group according to the same regimen but with the addition of bortezomib to the thalidomide arm. [18] TT3 is a very small set with only 15 OS events but is included here for completeness.
  • the MRC-IX trial included both younger and older newly diagnosed patients. For younger patients, treatment consisted of induction with vincristine or no vincristine followed by transplantation. Older patients were treated initially with a thalidomide-vs. melphalan-based treatment. Maintenance for both young and old patients was a comparison of thalidomide vs. no thalidomide. [19, 20]
  • the trial and dataset denoted here as APEX consisted of the three trials APEX, SUMMIT and CREST. These trials aimed at testing the efficacy of bortezomib in relapse cases. [21-23]
  • Affymetrix GENECHIP® Human Genome U133 Plus 2.0 Array was used in the HOVON65/GMMG-HD4, TT2, TT3 and MRC-IX whereas Affymetrix HG U133 A/B chips was used in the APEX study.
  • a lower probe set expression boundary was set to the 5% lowest expression for the bioB hybridization controls in the HOVON65/GMMG-HD4 set. Probe sets with a lower expression in ⁇ 95% of the HOVON65/GMMG-HD4 patients were excluded. All data were MASS normalized, log 2 transformed and mean-variance scaled.
  • HOVON65/GMMG-HD4 molecular classification was performed previously. [9] To assign a cluster label to new validation samples, a Euclidean nearest neighbor algorithm was used with HOVON65/GMMG-HD4 being the reference set.
  • the HOVON65/GMMG-HD4 was used as a training set for building a GEP-based survival classifier.
  • the model was built using a Supervised Principal Component Analysis (SPCA) framework. All calculations were performed in the R statistical environment using the survival package for survival analysis.
  • SPCA Supervised Principal Component Analysis
  • All calculations were performed in the R statistical environment using the survival package for survival analysis.
  • the maxstat package was used to determine the optimal cut-off value for high-risk.
  • the performance of the EMC-92-gene signature in relation to available GEP-based prognostic signatures for OS in MM was evaluated. To this end, the following signatures were evaluated: UAMS-70, UAMS-17, UAMS-80, IFM-15, gene proliferation index (GPI-50), MRC-IX-6 and MILLENNIUM-100.
  • the performance of the EMC-92-gene signature was in line with the UAMS signatures, although they were derived from quite different patient populations.
  • the intersection of high-risk patients between the EMC-92 and UAMS-70 signatures was ⁇ 8% of the total population on the pooled data sets that were independent of both the training set and the UAMS-70 training set (that is, MRC-IX, TT3 and APEX; Supplemental Table S11 in reference 31). Approximately 13% of patients were classified as high-risk by either one of these signatures.
  • cytogenetic aberration frequencies in both populations were determined using an independent set for which cytogenetic variables were known, that is, MRC-IX ( FIG. 4 and Supplemental Table S13 in reference 31).
  • MRC-IX FIG. 4 and Supplemental Table S13 in reference 31.
  • poor prognostic cytogenetic aberrations 1 q gain, del(17p), t(4;14), t(14;16), t(14;20) and del(13q) were enriched in the high-risk populations ( FIG. 5 in reference 31), whereas the standard-risk cytogenetic aberrations such as t(11;14) were diminished in the high-risk populations.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Immunology (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Physics & Mathematics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Pathology (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

This disclosure is in the field of molecular diagnostics and relates to a method for classifying samples obtained from patients diagnosed with multiple myeloma into three newly defined clusters. The disclosure also relates to a method for determining the prognosis of an individual diagnosed with multiple myeloma as well as a method for the prediction of the response to treatment of an individual diagnosed with multiple myeloma. More in particular, the disclosure provides a method for determining the disease outcome or the prognosis of a patient diagnosed with multiple myeloma by classifying the patient into a high risk or a low risk category, based on a 92-gene classifier.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of co-pending U.S. patent application Ser. No. 15/959,703, filed Apr. 23, 2018, which is a continuation of U.S. patent application Ser. No. 14/232,176, filed Mar. 19, 2014, now U.S. Pat. No. 9,976,185, issued May 22, 2018, which is a national phase entry under 35 U.S.C. § 371 of International Patent Application PCT/EP2012/063722, filed Jul. 12, 2012, designating the United States of America and published in English as International Patent Publication WO 2013/007795 A1 on Jan. 17, 2013, which claims the benefit under Article 8 of the Patent Cooperation Treaty to European Patent Application Serial No. 11173971.0, filed Jul. 14, 2011.
  • TECHNICAL FIELD
  • This application is in the field of molecular diagnostics and relates to a method for classifying samples obtained from patients diagnosed with multiple myeloma. The application also relates to a method for determining the prognosis of an individual diagnosed with multiple myeloma as well as a method for the prediction of the response to treatment of an individual diagnosed with multiple myeloma.
  • BACKGROUND
  • Multiple myeloma (MM) is characterized by accumulation of malignant monoclonal plasma cells in the bone marrow. Median overall survival (OS) is 3 to 4 years but varies widely between patients. Currently, the International Staging System (ISS), based on serum β2m and albumin is clinically widely used to classify MM patients into three prognostic categories. [1]
  • Based on cytogenetics, two classes of MM can be distinguished with implications for MM biology and prognosis. Hyperdiploid MM, ˜60% of patients, characterized by trisomies of multiple odd chromosomes (3, 5, 7, 9, 11, 15, 19, and 21) has a relatively good prognosis. Non-hyperdiploid MM, ˜40% of cases, is characterized by recurrent translocations involving the immunoglobulin heavy chain gene at 14q32, resulting in transcriptional activation of CCND1, CCND3, MAF, MAFB, or FGFR3/MMSET.[2, 3] Translocation t(11;14), involving CCND1, confers a relatively favorable prognosis whereas translocation t(4;14), involving FGFR3 and MMSET, has poor prognosis.[4, 5] The translocations t(14;16) and t(14;20), involving the MAF oncogenes also confer a poor prognosis, although recently this has been debated.[6] In addition, del(17p), del(13q) and lq-gain detected with conventional karyotyping were reported to be associated with poor prognosis). [8]
  • Based on gene expression analysis, a number of classifications for MM have been published, which include the University of Arkansas for Medical Sciences (UAMS) classification and, more recently, a classification by our own group. The UAMS molecular classification of myeloma consists of seven distinct gene expression clusters, including translocation clusters MS, MF, and CD-1/2, as well as a hyperdiploid cluster (HY), a cluster with proliferation-associated genes (PR), and a cluster characterized by low percentage of bone disease (LB).[8] Our classification of MM resulted in three additional clusters: NFκB, CTA and PRL3.[9]
  • Gene expression is able to explain an even larger amount of variance in survival compared to ISS and cytogenetics. One of the first survival signatures based on gene expression was the UAMS-70-gene classifier, and the further refined UAMS-17-gene classifier.[10, 11] Other classifiers include the Millennium signature, the MRC-IX-6-gene signature, and the IFM classifier.[21-14] In addition, signatures were reported to predict plasma cell proliferation such as the recently published gene expression proliferation index (GPI).[15]
  • The aim of this study was to develop a prognostic signature, based upon gene expression profiles (GEPs) of MM patients, treated with either standard induction treatment or bortezomib induction, followed in both cases by high-dose melphalan and maintenance.
  • BRIEF SUMMARY
  • Presented herein is a classifier comprising a 92-gene set capable of distinguishing between patients with a high risk and patients with a low risk. In a survival analysis of newly diagnosed multiple myeloma (MM) patients, the classifier yielded excellent results wherein the classification in the low risk group identified patients with a good overall survival, whereas the group identified as high risk showed significantly worse overall survival rates.
  • The disclosure, therefore, relates to a method for determining the disease outcome or the prognosis of a patient diagnosed with multiple myeloma by classifying the patient into a high risk or a low risk category, the method comprising the steps of:
  • a) providing a gene chip comprising probes for the detection of at least the 92-gene set according to Table 1,
  • b) contacting the gene chip with a sample comprising mRNA from a patient,
  • c) determining the expression levels of the 92-gene set in the sample,
  • d) normalizing the expression levels using mean/variance normalization in order to obtain the normalized expression value,
  • e) multiplying the normalized expression value with the beta value according to Table 1 to obtain the calculated value for an individual probe,
  • f) determining an EMC-92 score by summation of the calculated values of the individual probes,
  • wherein an EMC-92 score above a predetermined threshold indicates that the patient is to be classified in the high risk category and a score at or below the predetermined threshold indicates that the patient is to be classified in the low risk category.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A-1D: Performance of the EMC-92 classifier in predicting overall survival. High risk signature on four validation sets with a fixed cut-off value of 0.827. FIG. 1A: UAMS Total Therapy 2; FIG. 1B: UAMS Total Therapy 3; FIG. 1C: MRC-IX; and FIG. 1D: APEX.
  • FIG. 2: Relation between threshold and log-rank performance of the EMC-92 signature in the HOVON-65/GMMG-HD4 OS. The model has an optimal performance for thresholds of at least 0.75. A cut-off for high-risk was based on defining high-risk as having an OS of <2 years within the training set, which corresponded to a threshold of 0.827.
  • DETAILED DESCRIPTION
  • A classifier comprising a 92-gene set capable of distinguishing between patients with a high risk and patients with a low risk is disclosed herein. In a survival analysis of newly diagnosed multiple myeloma (MM) patients, the classifier yielded excellent results wherein the classification in the low risk group identified patients with a good overall survival, whereas the group identified as high risk showed significantly worse overall survival rates.
  • The classifier was validated in an experimental setting wherein patients with poor overall survival (OS) were distinguished from patients with standard OS. Therefore, an SPCA model was built using the HOVON65/GMMG-HD4 data as a training set (see the experimental section below). A number of 1088 probe sets were found to be associated with progression-free survival (PFS) in a univariate Cox regression analysis (FDR <10%). Based on these probe sets, a classifier with 92 probe sets was developed (Table 1). This classifier will be termed the EMC-92-gene signature.
  • TABLE 1
    Chromo-
    # Probes Beta Gene some Band
    1 226217_at −0.0319 SLC30A7 1 p21.2
    2 208967_s_at 0.0113 AK2 1 p35.1
    3 202553_s_at 0.0054 SYF2 1 p36.11
    4 217728_at 0.0773 S100A6 1 q21.3
    5 223381_at −0.0070 NUF2 1 q23.3
    6 218365_s_at 0.0035 DARS2 1 q25.1
    7 211963_s_at 0.0303 ARPC5 1 q25.3
    8 222680_s_at 0.0205 DTL 1 q32.3
    9 221826_at 0.0200 ANGEL2 1 q32.3
    10 201795_at 0.0067 LBR 1 q42.12
    11 202813_at 0.0548 TARBP1 1 q42.2
    12 202322_s_at 0.0129 GGPS1 1 q42.3
    13 202728_s_at −0.1105 LTBP1 2 p22.3
    14 209683_at −0.0561 FAM49A 2 p24.2
    15 201930_at −0.0090 MCM6 2 q21.3
    16 228416_at −0.0778 ACVR2A 2 q22.3
    17 206204_at 0.0477 GRB14 2 q24.3
    18 215177_s_at −0.0768 ITGA6 2 q31.1
    19 224009_x_at −0.0520 DHRS9 2 q31.1
    20 AFFX-HUMISG 0.0525 STAT1 2 q32.2
    F3A/M97935_MA_at
    21 222154_s_at 0.0154 SPATS2L 2 q33.1
    22 207618_s_at 0.0746 BCS1L 2 q35
    23 239054_at −0.1088 SFMBT1 3 p21.1
    24 217852_s_at 0.0008 ARL8B 3 p26.1
    25 219510_at −0.0097 POLQ 3 q13.33
    26 202107_s_at 0.0225 MCM2 3 q21.3
    27 220351_at 0.0420 CCRL1 3 q22.1
    28 208942_s_at −0.0997 SEC62 3 q26.2
    29 233437_at 0.0446 GABRA4 4 p12
    30 225366_at 0.0140 PGM2 4 p14
    31 218662_s_at −0.0176 NCAPG 4 p15.31
    32 204379_s_at 0.0594 FGFR3 4 p16.3
    33 201307_at 0.0165 SEPT11 4 q21.1
    34 202542_s_at 0.0870 AIMP1 4 q24
    35 205046_at 0.0087 CENPE 4 q24
    36 226218_at −0.0644 IL7R 5 p13.2
    37 202532_s_at −0.0006 DHFR 5 q14.1
    38 226742_at −0.0345 SAR1B 5 q31.1
    39 231738_at 0.0686 PCDHB7 5 q31.3
    40 214150_x_at −0.0349 ATP6V0E1 5 q35.1
    41 201555_at −0.0052 MCM3 6 p12.2
    42 209026_x_at 0.0255 TUBB 6 p21.33
    43 211714_x_at 0.0221 TUBB 6 p21.33
    44 213002_at −0.0418 MARCKS 6 p22.2
    45 221041_s_at −0.0520 SLC17A5 6 q13
    46 217824_at −0.0041 NCUBE1 6 q15
    47 223811_s_at 0.0556 SUN1/GET4 7 p22.3
    48 202842_s_at −0.0626 DNAJB9 7 q31.1
    49 208232_x_at −0.0493 Unknown 8 p12
    50 208732_at −0.0618 RAB2A 8 q12.1
    51 201398_s_at −0.0254 TRAM1 8 q13.3
    52 233399_x_at −0.0184 ZNF252 8 q24.3
    53 200775_s_at 0.0163 HNRNPK 9 q21.32
    54 230034_x_at −0.0330 MRPL41 9 q34.3
    55 204026_s_at 0.0046 ZWINT 10 q21.1
    56 243018_at 0.0407 Unknown 11 p14.1
    57 222713_s_at 0.0278 FANCF 11 p14.3
    58 221755_at 0.0396 EHBP1L1 11 q13.1
    59 231210_at 0.0093 C11orf85 11 q13.1
    60 202884_s_at 0.0714 PPP2R1B 11 q23.1
    61 219550_at 0.0559 ROBO3 11 q24.2
    62 238780_s_at −0.0529 Unknown 11 q24.3
    63 208747_s_at −0.0874 C1S 12 p13.31
    64 38158_at 0.0423 ESPL1 12 q13.13
    65 217732_s_at −0.0252 ITM2B 13 q14.2
    66 214482_at 0.0861 ZBTB25 14 q23.3
    67 200701_at −0.0210 NPC2 14 q24.3
    68 238662_at 0.0490 ATPBD4 15 q14
    69 217548_at −0.0423 C15orf38 15 q26.1
    70 213007_at −0.0106 FANCI 15 q26.1
    71 231989_s_at 0.0730 SMG1 16 p12.3
    72 238116_at 0.0661 DYNLRB2 16 q23.2
    73 212282_at 0.0530 TMEM97 17 q11.2
    74 203145_at −0.0002 SPAG5 17 q11.2
    75 201292_at −0.0372 TOP2A 17 q21.2
    76 210334_x_at 0.0175 BIRC5 17 q25.3
    77 212055_at 0.0384 C18orf10 18 q12.2
    78 242180_at −0.0585 TSPAN16 19 p13.2
    79 208904_s_at −0.0334 RPS28 19 p13.2
    80 213350_at 0.0056 RPS11 19 q13.3
    81 200875_s_at 0.0437 NOP56 20 p13
    82 212788_x_at −0.0164 FTL 19 p13
    83 215181_at −0.0342 CDH22 20 q13.12
    84 221677_s_at 0.0126 DONSON 21 q22.11
    85 201102_s_at 0.0349 PFKL 21 q22.3
    86 208667_s_at −0.0390 ST13 22 q13.2
    87 216473_x_at −0.0576 DUX4 4/10 q35.2/
    q26.3
    88 200933_x_at −0.0323 RPS4X X q13.1
    89 218355_at 0.0116 KIF4A X q13.1
    90 221606_s_at 0.0208 HMGN5 X q21.1
    91 225601_at 0.0750 HMGB3 X q28
    92 214612_x_at 0.0496 MAGEA6 X q28
  • A dichotomizing cut-off threshold was based on the clinically relevant definition of high-risk patients as those patients who have an overall survival of less than 2 years. This amounted to a proportion of 21.7% in the training set and a cut-off value of 0.827. Within all four data sets TT2(n=351),[11] TT3(n=208),[11] MRC-IX(n=247)[14] and APEX(n=264),[12] the EMC-92-gene signature discriminated a high-risk group, which was significantly set apart from the standard-risk group (FIGS. 1A to 1D).
  • In datasets containing newly diagnosed patients, the EMC-92-gene signature selected a high-risk population of 17.7% on average, with a significantly shorter OS and hazard-ratios of 3.52 (p=2.5×10−8; TT2), 2.7 (p=0.07=2; TT3) and 2.38 (p=3.6×10−6=; MRC-IX). In the relapse setting, the EMC-92-gene signature also filtered out high-risk patients with a large hazard-ratio of 3.14 (p=5.3×10−9=; APEX). The proportion of high-risk patients in this latter study was lower compared to the MRC-IX and TT2 studies, but not significantly (15.9%, n=264 vs. 19.6%, n=; p=0.2) (Table 1.1).
  • In a multivariate covariant analysis, the EMC-92-gene signature was independent of most standard prognostic factors and clinical characteristics. Three datasets were available for this analysis: HOVON65/GMMG-HD4, APEX and MRC-IX. Multivariate analysis on the HOVON65/GMMG-HD4 study demonstrates that together with the EMC-92-gene signature, del(17p), β2m[≥3.5 mg/L] and allogenic transplantation were significantly related to shorter survival, whereas WHO status [0] was found to be significantly related to longer survival. In the APEX, albumin level, ISS and IgG isotype were found to be significantly related. For the MRC-IX, mainly ISS related covariants were found. WHO[2], 1q gain and IGH split showed a clear contribution. IGH split indicates all patients with cytogenetic aberration of the IGH locus. Age had a small but significant hazard-ratio here. In all three datasets, the EMC-92-gene signature remained a strong predictor for survival after correction for available variables.
  • The samples in all four validation sets were assigned a molecular cluster label by the nearest neighbor classification. Logistic regression for association between the molecular clusters and high-risk outcome revealed a significant relation between high-risk classification and the MF, MS, PR and HY clusters.
  • Comparing the UAMS-17-gene and EMC-92-gene set in independent datasets (for example, TT3, MRC-IX and APEX), a significantly higher proportion of patients was classified as high-risk by the EMC-92-gene signature (p=0.009). Moreover, the estimated hazard-ratios (high-risk/standard-risk) were higher in the EMC-92-gene classifier with the exception of the TT3 study.
  • In the MRCIX study population, the EMC-92-gene classifier exclusively identified 31 patients correctly as high risk patients, which were missed by the UAMS-17-gene classifier (50% survival rate of 11, 24 and 51 months for the shared high risk group, intermediate high risk group and standard risk groups, respectively). Moreover, the UAMS-17-gene classifier exclusively identified 10 patients as high risk patients with a lower hazard ratio as compared to the 31 patients classified as high-risk by the EMC-92-gene classifier.
  • The superiority of the EMC-92-gene classifier was even clearer in the APEX population. Here, 24 patients were exclusively identified as high risk in the EMC-92-gene classifier, which were missed in the UAMS-17-gene classifier. These 24 patients formed a group whose overall survival after 20 months was 14% whereas the high risk population identified in both the EMC-92-gene and UAMS-17-gene classifier showed an overall survival of 25% after 20 months.
  • In addition, the UAMS-70-gene, MRC-IX-6-gene signature, GPI score, the Millennium and IFM signature were applied to the datasets. In a pair-wise multivariate analysis based on the pooled independent datasets, including two classifiers at a time and correcting for study and age, the EMC-92-gene classifier had the highest hazard ratios and lowest p values of all classifiers.
  • The intersection of high-risk patients between the EMC-92-gene and UAMS-17-gene classifiers was ˜8% of the total population. About 14% of patients were classified as high-risk by either one of these classifiers. The intersecting high-risk group showed the largest differences compared to the intersecting standard-risk group as indicated by the hazard-ratios (HR=5.40; p=3.1×10−3; TT3), (HR=3.84; p=5×10−7; MRC-IX) and (HR=3.39; p=1.9×10−5; APEX). The 14% of patients uniquely classified as high-risk by either signature, showed an intermediate hazard-ratio. For the UAMS-17-gene high-risk group, this resulted in hazard-ratios of 4.08 (p=7.6×10−2), 1.92 (p=7.7×10−2) and 2.31 (p=2.3×10−2) for the TT3, MRX-IX and APEX. The EMC-92-gene high-risk group gave hazard-ratios of 0 (p=1.0 no events), 1.98 (p=2.9×1034) and 3.21 (p=1.6×10−6) for the TT3, MRX-IX and APEX.
  • In clinical practice, prognosis of MM patients is mainly based on ISS-stage and interphase fluorescence in situ hybridization (FISH). Several chromosomal aberrations detected by FISH have prognostic implications.[25] Del(17p) is considered the most important, associated with unfavorable outcome and present in 9% of patients.[26, 27] Still 60% of patients with this deletion do not display a specific poor outcome.[28] The combination of chromosomal aberration, t(4;14), del(17p) and ISS have further delineated patients with a poor prognosis.[29]
  • Previously, in the UAMS classification, the MS, MF and PR clusters showed lower PFS and OS, whereas clusters HY, LB, CD-1 and CD-2 were associated with longer PFS and OS.[8] Here, the variability in PFS and OS in the GEP-based clusters of the HOVON65 classification was evaluated. VAD-treated patients demonstrate significant differences in PFS and OS between clusters with a clearly reduced survival for the MF subgroup, whereas in bortezomib-(PAD-) treated patients, no significant differences were found.
  • Bortezomib-based treatment has been shown to overcome certain adverse prognostic markers such as del(13q), resulting in better PFS and OS in patients with poor prognostic markers such as ISS-3, del(17p), and t(4;14).[16] Both chromosomal markers and the HOVON65 GEP-based classification vary with treatment and are not applicable for diagnosing high-risk patients accurately. Therefore, a high-risk GEP signature was developed.
  • Previous classifiers include the UAMS-17/70-gene and MRC-IX-6-gene classifiers, both capable of predicting in independent datasets.[11, 14] In contrast, the Millennium and the IFM signatures demonstrate less solid performance in independent validation sets. [12, 13]
  • The EMC-92-gene expression signature presented herein, is highly discriminative for patients with high-risk versus standard-risk MM across different (induction) regimes. Validation in UAMS TT2 (thalidomide-based),[17] TT3 (bortezomib-based),[18] and MRC-IX trial (thalidomide maintenance in both young and elderly patients)[19, 20] showed high performance in these independent test environments. This is true for both the continuous fit of the model, which is a goodness-of-fit indicator, as well as the dichotomized output into high-risk/standard-risk, which is a requirement for practical use in a clinical setting.
  • In multivariate analyses, the EMC-92-gene high-risk signature remains a strong predictor for early death. Still, there is strong evidence that ISS staging (serum albumin and β2m levels) turns out to be another major contributor for explaining survival-related variance in the presence of the signature. Therefore, incorporating ISS into the signature could potentially lead to an even better prediction of survival.
  • Patients classified as high-risk are overrepresented within the molecular MF, MS and PR clusters and underrepresented within the HY cluster. This correlates well with previous data: HY represents hyperdiploid patients with a generally favorable prognosis; on the other hand, MS and MF represent patients with translocations t(4;14) and t(14;16/20), which are usually thought to have an unfavorable prognosis. Finally, PR represents the proliferation cluster, which was shown to be associated with poor prognosis.[8, 11, 15] In relation to this, pathway analysis of the EMC-92-gene signature demonstrated cell cycle regulation to be among the main functions found.
  • In the EMC-92-gene signature as well as the set of genes linked to survival in the univariate analysis, chromosomal location of 1 q was highly enriched (Table 1) as was previously shown for the UAMS-17-gene signature.[11] Also, probe sets located on chromosome 4 are enriched. These probe sets were found to be scattered over the entire chromosome and not only at the distal end of the p arm where MIVISET and FGFR3 are located. Chromosome 4 has previously not been considered a risk factor, but a low frequency of multiple gains and/or losses affecting this chromosome has been reported.[30]
  • The EMC-92-gene signature was compared in a multivariable analysis to the UAMS-17/70-gene, MRC-IX-6-gene, GPI score, IFM and Millennium classifiers. Three pooled datasets were formed from publicly available MM datasets, allowing an independent comparison of the signatures that were not trained on those datasets (R. Kuiper et al., Leukemia 2012, 1-8 incorporated herein by reference). The outputs from the signatures were input into a Cox proportional hazards model, see Table 2. In all three comparisons, the EMC-92-gene signature obtained the most significant hazard ratio (HR) and, thus, is the most relevant prognostic factor of all signatures (including the UAMS-70 from Signal Genetics).
  • TABLE 2
    Comparison of EMC 92 with conventional
    tests (HR = hazard ratio)
    Datasets pooled Signature HR P-value
    Comparison 1 MRCIX + SKY9211 1.75 4.60E−04
    APEX + TT3 UAMS1710 1.22 3.30E−01
    UAMS7010 1.80 1.10E−03
    IFM1512 1.25 9.10E−02
    Comparison 2 APEX + SKY92 2.53 3.70E−09
    TT2 + TT3 MRCIX613 1.50 4.10E−03
    IFM15 1.38 2.50E−02
    Comparison 3 MRCIX + SKY92 2.95 5.60E−12
    TT2 + TT3 Millennium10014 0.81 1.30E−01
    IFM15 1.13 4.00E−01
  • The EMC-92-gene signature turns out to have the best dichotomized performance on its validation sets. Moreover, in comparison to other classifiers, the proportion of high-risk patients is higher. One would expect that differences between high-risk and standard-risk become less pronounced as the high-risk proportion increases. It should be mentioned that even at this high proportion, differences in survival time are larger for the EMC-92 as compared to other classifiers selecting smaller risk groups.
  • In a multivariate analysis combining the signatures, the EMC-92-gene signature had the strongest discriminative ability.
  • In conclusion, a high-risk signature highly discriminative for patients with high-risk versus standard-risk MM, irrespective of treatment regime, age and relapse setting was developed. Use of this signature in the clinical setting may lead to a more informed treatment choice and potentially better outcome for the patient.
  • In conclusion, this study concerns the development of a robust high-risk signature, incorporates most known prognostic markers, clinical, cytogenetic and GEP based, and shows the developed EMC-92-gene signature to be the strongest independent prognostic marker for poor survival known. This EMC-92-gene signature is able to select out a high-risk group of MM patients for whom in the future alternative, more intensive treatments should be sought.
  • Hence, the disclosure relates to a method for determining the disease outcome or the prognosis of a patient diagnosed with multiple myeloma by classifying the patient into a high risk or a low risk category, the method comprising the steps of:
  • a) providing a gene chip comprising probes for the detection of at least the 92-gene set according to Table 1,
  • b) contacting the gene chip with a sample comprising mRNA from a patient,
  • c) determining the expression levels of the 92-gene set in the sample,
  • d) normalizing the expression levels using mean/variance normalization in order to obtain the normalized expression value,
  • e) multiply the normalized expression value with the beta value according to Table 1 to obtain the calculated value for an individual probe,
  • f) determine an EMC-92 score by summation of the calculated values of the individual probes,
  • wherein an EMC-92 score above a predetermined threshold indicates that the patient is to be classified in the high-risk category and a score at or below the threshold indicates that the patient is to be classified in the low-risk category.
  • As further detailed herein, a preferred threshold value is at least 0.75, especially preferred is a threshold value of 0.827.
  • In summary, the generation and validation of the EMC-92-gene signature, which was based on the HOVON65/GMMG-HD4 clinical trial, is reported herein. Conventional prognostic markers such as ISS stage and adverse cytogenetics have been augmented by signatures based on gene expression in order to increase accuracy in outcome prediction in MM. More accurate prognosis may lead to the development of treatment schedules that are specifically aimed at improving survival of high-risk MM patients.
  • For clinical relevance, a signature must have both the ability to separate risk groups as clearly as possible and to predict stable groups of relevant size. The EMC-92-gene signature meets both criteria. In all validation sets, a high-risk group of patients can be significantly determined and the proportion of high-risk patients is stable across the validation sets. The validation sets represent different drug regimens, including thalidomide (MRC-IX, TT2) and bortezomib (APEX, TT3). Also, the signature is relevant to both transplant-eligible (for example, TT3) and non-transplant-eligible patients (subset of MRC-IX), as well as newly diagnosed (for example, TT2) and relapsed patients (APEX). In contrast, the predictions of the IFM-15 and MILLENNIUM-100 signatures in the validation sets fail to reach significance in independent data sets such as MRC-IX and TT3.
  • In conclusion, a risk signature that is highly discriminative for patients with high-risk vs standard-risk MM, irrespective of treatment regime, age and relapse setting, has been developed. Use of this signature in the clinical setting may lead to a more informed treatment choice and potentially better outcome for the patient.
  • EXAMPLES Example 1 Patients
  • Five previously described datasets were used, of which both survival as well as GEPs of purified plasma cells obtained from bone marrow aspirates of myeloma patients, were available. These are HOVON65/GMMG-HD4 (N=320) (GSE19784),[9] Total Therapy 2 (TT2) (n=351),[11] TT3 (n=208) (GSE2658),[11] MRC-IX (n=247) (GSE15695),[14] and APEX (n=264) (GSE9782).[12]
  • The HOVON65/GMMG-HD4 data was used as a training set. This multicenter trial compared the efficacy of bortezomib (PAD) to standard treatment (VAD) in newly diagnosed patients. Patients were randomized to induction treatment with three VAD or PAD cycles,[16] for a total of 290 patients, both follow-up and GEPs were available.[9]
  • The other four independent datasets were used as validation. Two datasets, TT2 and TT3, were derived from clinical trials performed in newly diagnosed patients, both treated with a complex regimen. The first was a randomized prospective treatment trial in which patients were randomly assigned to receive or not to receive thalidomide during all phases of treatment.[17] The latter was carried out by the same group according to the same regimen but with the addition of bortezomib to the thalidomide arm.[18] TT3 is a very small set with only 15 OS events but is included here for completeness.
  • The MRC-IX trial included both younger and older newly diagnosed patients. For younger patients, treatment consisted of induction with vincristine or no vincristine followed by transplantation. Older patients were treated initially with a thalidomide-vs. melphalan-based treatment. Maintenance for both young and old patients was a comparison of thalidomide vs. no thalidomide.[19, 20] The trial and dataset denoted here as APEX consisted of the three trials APEX, SUMMIT and CREST. These trials aimed at testing the efficacy of bortezomib in relapse cases.[21-23]
  • The IFM dataset on which the IFM signature was based has not been evaluated due to an incompatible GEP platform.[13]
  • Example 2 Gene Expression Analysis
  • Two types of Affymetrix gene expression platforms were used. The Affymetrix GENECHIP® Human Genome U133 Plus 2.0 Array was used in the HOVON65/GMMG-HD4, TT2, TT3 and MRC-IX whereas Affymetrix HG U133 A/B chips was used in the APEX study. To allow for validation across different studies, only probe sets present on both platforms were included. A lower probe set expression boundary was set to the 5% lowest expression for the bioB hybridization controls in the HOVON65/GMMG-HD4 set. Probe sets with a lower expression in ≥95% of the HOVON65/GMMG-HD4 patients were excluded. All data were MASS normalized, log2 transformed and mean-variance scaled.
  • The HOVON65/GMMG-HD4 molecular classification was performed previously.[9] To assign a cluster label to new validation samples, a Euclidean nearest neighbor algorithm was used with HOVON65/GMMG-HD4 being the reference set.
  • The HOVON65/GMMG-HD4 was used as a training set for building a GEP-based survival classifier. The model was built using a Supervised Principal Component Analysis (SPCA) framework. All calculations were performed in the R statistical environment using the survival package for survival analysis. The maxstat package was used to determine the optimal cut-off value for high-risk.
  • Data were analyzed using Ingenuity Pathway Analysis (INGENUITY SYSTEMS®, on the World-Wide Web at Ingenuity.com). Both the gene set corresponding to the SPCA-based survival classifier as well as the gene set generated by the initial univariate ranking (FDR<10%) were analyzed. Probe sets present in both the HG U133 Plus 2.0 and A/B platforms were used as a reference. P-values were derived from right-tailed Fisher exact tests corrected for multiple testing using Benjamini Hochberg correction.
  • Example 3 Comparison with Published Gene Signatures
  • The performance of the EMC-92-gene signature in relation to available GEP-based prognostic signatures for OS in MM was evaluated. To this end, the following signatures were evaluated: UAMS-70, UAMS-17, UAMS-80, IFM-15, gene proliferation index (GPI-50), MRC-IX-6 and MILLENNIUM-100.
  • These signatures were evaluated as continuous variables as well as using the cut-off values as published (FIG. 2 and FIGS. 2A-2E in reference 31, and Supplemental Documents A and B in reference 31). Overall, the performance of the EMC-92-gene signature was found to be robust, consistent, which compares favorably with previously published signatures. Specifically, the EMC-92, UAMS, MRC-IX and GPI-50 signatures demonstrated significance in all validation sets tested both for the dichotomized and for the continuous values of the signatures. Significance was reached in three out of five studies for the IFM-15 signature using a dichotomized model, whereas the MILLENNIUM-100 signature had significant performance in the dichotomized model in one out of four independent studies. Thus, performance was less robust for the IFM-15 and MILLENNIUM-100 signatures. Although the proliferation index GPI-50 was found to be significant in all validation sets tested, the proportion of high-risk patients was much lower compared with the proportion found using either the EMC-92 or the UAMS-80 signatures. Ranked, weighted high-risk proportions are GPI: 10.0%, UAMS-17: 12.4%, UAMS-70: 13.0%, MRC-IX-6: 13.3%, EMC-92: 19.1% and UAMS-80: 23.4%. To determine which signature best explained the observed survival, pair-wise comparisons were performed. For every comparison, the EMC-92 was the strongest predictor for OS tested in an independent environment (FIG. 3 and Supplemental Table S9 in reference 31).
  • Example 4 Combined Risk Classifiers
  • The performance of the EMC-92-gene signature was in line with the UAMS signatures, although they were derived from quite different patient populations. The intersection of high-risk patients between the EMC-92 and UAMS-70 signatures was ˜8% of the total population on the pooled data sets that were independent of both the training set and the UAMS-70 training set (that is, MRC-IX, TT3 and APEX; Supplemental Table S11 in reference 31). Approximately 13% of patients were classified as high-risk by either one of these signatures. The intersecting high-risk group had the highest HR as compared with the intersecting standard-risk group (HR=3.87, 95% CI=2.76-5.42, P=3.6×10-15). Patients classified as high-risk by either signature showed an intermediate risk, that is, with a HR of 2.42, 95% CI=1.76-3.32, for the EMC-92-gene signature (P=5.1×10-8) and a HR of 2.22, 95% CI=1.20-4.11, for the UAMS-70 signature (P=1.1×10-2; Supplemental Table S12 in reference 31).
  • Example 5 EMC-92-Gene Signature and FISH
  • To compare the high-risk populations composition as defined by the EMC-92 and the UAMS-70 signatures, cytogenetic aberration frequencies in both populations were determined using an independent set for which cytogenetic variables were known, that is, MRC-IX (FIG. 4 and Supplemental Table S13 in reference 31). As expected, poor prognostic cytogenetic aberrations 1 q gain, del(17p), t(4;14), t(14;16), t(14;20) and del(13q) were enriched in the high-risk populations (FIG. 5 in reference 31), whereas the standard-risk cytogenetic aberrations such as t(11;14) were diminished in the high-risk populations. In contrast, only 15% (6 out of 39) of MRC-IX cases with high-risk status as determined by the EMC-92-gene signature showed absence of any poor prognostic cytogenetic aberrations, as opposed to 44% (74 out of 168) in standard-risk cases (P=1.8×10-3). Similarly, of the UAMS-70-defined high-risk patients 4% (1 out of 23) did not have any poor prognostic cytogenetics, whereas of the UAMS-70 defined standard risk patients, this proportion was 43% (79 out of 183) (P=5.3×10-3).
  • The following references are incorporated herein by reference. Their contents should be regarded as an integral part of this application.
  • REFERENCES
    • 1. Greipp P. R., J. San Miguel, B. G. Durie, J. J. Crowley, B. Barlogie, J. Blade, et al. International staging system for multiple myeloma. J. Clin. Oncol. 2005 May 20; 23(15):3412-20.
    • 2. Bergsagel P. L., W. M. Kuehl. Molecular Pathogenesis and a Consequent Classification of Multiple Myeloma. J. Clin. Oncol. 2005 Sepember 10; 23(26):6333-8.
    • 3. Fonseca R., C. S. Debes-Marun, E. B. Picken, G. W. Dewald, S. C. Bryant, J. M. Winkler et al. The recurrent IgH translocations are highly associated with nonhyperdiploid variant multiple myeloma. Blood 2003 October. 1; 102(7):2562-7.
    • 4. Fonseca R., J. D. Hoyer, P. Aguayo, S. M. Jalal, G. J. Ahmann, S. V. Rajkumar et al. Clinical significance of the translocation (11;14)(q13;q32) in multiple myeloma. Leuk. Lymphoma. 1999; 35(5-6):599-605.
    • 5. Keats J. J., T. Reiman, C. A. Maxwell, B. J. Taylor, L. M. Larratt, and M. J. Mant et al. In multiple myeloma, t(4;14)(p16;q32) is an adverse prognostic factor irrespective of FGFR3 expression. Blood 2003 February. 15; 101(4):1520-9.
    • 6. Avet-Loiseau H., F. Malard, L. Campion, F. Magrangeas, C. Sebban, and B. Lioure et al. Translocation t(14;16) and multiple myeloma: is it really an independent prognostic factor? Blood 2011 February 10; 117(6):2009-11.
    • 7. Cremer F. W., J. Bila, I. Buck, M. Kartal, D. Hose, C. Ittrich et al. Delineation of distinct subgroups of multiple myeloma and a model for clonal evolution based on interphase cytogenetics. Genes, Chromosomes & Cancer 2005 October; 44(2):194-203.
    • 8. Zhan F., Y. Huang, S. Colla, J.P. Stewart, I. Hanamura, S. Gupta et al. The molecular classification of multiple myeloma. Blood 2006 Sep. 15; 108(6):2020-8.
    • 9. Broyl A., D. Hose, H. Lokhorst, Y. de Knegt, J. Peeters, A. Jauch et al. Gene expression profiling for molecular classification of multiple myeloma in newly diagnosed patients. Blood 2010 Oct. 7; 116(14):2543-53.
    • 10. Chng W. J., W. M. Kuehl, P. L. Bergsagel, and R. Fonseca. Translocation t(4;14) retains prognostic significance even in the setting of high-risk molecular signature. Leukemia 2008 Feb.; 22(2):459-61.
    • 11. Shaughnessy J. D., Jr., F. Zhan, B. E. Burington, Y. Huang, S. Colla, I. Hanamura et al. A validated gene expression model of high-risk multiple myeloma is defined by deregulated expression of genes mapping to chromosome 1. Blood 2007 Mar. 15; 109(6):2276-84.
    • 12. Mulligan G., C. Mitsiades, B. Bryant, F. Zhan, W. J. Chng, S. Roels et al. Gene expression profiling and correlation with outcome in clinical trials of the proteasome inhibitor bortezomib. Blood 2007 Apr. 15; 109(8):3177-88.
    • 13. Decaux O., L. Lode, F. Magrangeas, C. Charbonnel, W. Gouraud, P. Jezequel, et al. Prediction of survival in multiple myeloma based on gene expression profiles reveals cell cycle and chromosomal instability signatures in high-risk patients and hyperdiploid signatures in low-risk patients: a study of the Intergroupe Francophone du Myelome. J. Clin. Oncol. 2008 Oct. 10; 26(29):4798-805.
    • 14. Dickens N. J., B. A. Walker, P. E. Leone, D. C. Johnson, J. L. Brito, A. Zeisig et al. Homozygous deletion mapping in myeloma samples identifies genes and an expression signature relevant to pathogenesis and outcome. Clin. Cancer Res. 2010 Mar. 15; 16(6):1856-64.
    • 15. Hose D., T. Reme, T. Hielscher, J. Moreaux, T. Messner, A. Seckinger et al. Proliferation is a central independent prognostic factor and target for personalized and risk-adapted treatment in multiple myeloma. Haematologica 2011 January; 96(1):87-95.
    • 16. Sonneveld P., I. Schmidt-Wolf, B. van der Holt, L. E. Jarari, U. Bertsch, H. Salwender et al. HOVON-65/GMMG-HD4 Randomized Phase III Trial Comparing Bortezomib, Doxorubicin, Dexamethasone (PAD) vs VAD Followed by High-Dose Melphalan (HDM) and Maintenance with Bortezomib or Thalidomide In Patients with Newly Diagnosed Multiple Myeloma (MM). Blood 2010 Nov. 19; 116(21):40.
    • 17. Barlogie B., M. Pineda-Roman, F. van Rhee, J. Haessler, E. Anaissie, K. Hollmig et al. Thalidomide arm of Total Therapy 2 improves complete remission duration and survival in myeloma patients with metaphase cytogenetic abnormalities. Blood 2008 Oct. 15; 112(8):3115-21.
    • 18. Pineda-Roman M., M. Zangari, J. Haessler, E. Anaissie, G. Tricot, F. van Rhee, et al. Sustained complete remissions in multiple myeloma linked to bortezomib in total therapy 3: comparison with total therapy 2. British journal of haematology 2008 March; 140(6):625-34.
    • 19. Morgan G. J., F. E. Davies, W. M. Gregory, S. E. Bell, A. J. Szubert, N. Navarro-Coy et al. Thalidomide Maintenance Significantly Improves Progression-Free Survival (PFS) and Overall Survival (OS) of Myeloma Patients When Effective Relapse Treatments Are Used: MRC Myeloma IX Results. Blood 2010 Nov. 19; 116(21):623.
    • 20. Morgan G. J., F. E. Davies, R. G. Owen, A. C. Rawstron, S. Bell, K. Cocks et al. Thalidomide Combinations Improve Response Rates; Results from the MRC IX Study. Blood 2007 Nov. 16; 110(11):3593-.
    • 21. Jagannath S., B. Barlogie, J. Berenson, D. Siegel, D. Irwin, P. G. Richardson et al. A phase 2 study of two doses of bortezomib in relapsed or refractory myeloma. British journal of haematology 2004 October; 127(2):165-72.
    • 22. Richardson P.G., B. Barlogie, J. Berenson, S. Singhal, S. Jagannath, D. Irwin et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. The New England journal of medicine, 2003 Jun. 26; 348(26):2609-17.
    • 23. Richardson P. G., P. Sonneveld, M. W. Schuster, D. Irwin, E. A. Stadtmauer, T. Facon et al. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. The New England journal of medicine 2005 Jun. 16; 352(24):2487-98.
    • 24. Bair E., T. Hastie, D. Paul, and R. Tibshirani. Prediction by Supervised Principal Components. J. Amer. Statistical Assoc. 2006 Mar. 1; 101(473):119-37.
    • 25. Avet-Loiseau H., F. Magrangeas, P. Moreau, M. Attal, T. Facon, K. Anderson et al. Molecular Heterogeneity of Multiple Myeloma: Pathogenesis, Prognosis, and Therapeutic Implications. J. Clin. Oncol. 2011 May 10; 29(14):1893-7.
    • 26. Avet-Loiseau H., M. Attal, P. Moreau, C. Charbonnel, F. Garban, C. Hulin et al. Genetic abnormalities and survival in multiple myeloma: the experience of the Intergroupe Francophone du Myelome. Blood 2007 Apr. 15; 109(8):3489-95.
    • 27. Fonseca R., P. L. Bergsagel, J. Drach, J. Shaughnessy, N. Gutierrez, A. K. Stewart et al. International Myeloma Working Group molecular classification of multiple myeloma: spotlight review. Leukemia 2009 Dec.; 23(12):2210-21.
    • 28. Avet-Loiseau H., X. Leleu, M. Roussel, P. Moreau, C. Guerin-Charbonnel, D. Caillot et al. Bortezomib plus dexamethasone induction improves outcome of patients with t(4;14) myeloma but not outcome of patients with del(17p). J. Clin. Oncol. 2010 Oct. 20; 28(30):4630-4.
    • 29. Neben K., A. Jauch, U. Bertsch, C. Heiss, T. Hielscher, and A. Seckinger et al. Combining information regarding chromosomal aberrations t(4; 14) and del(17p13) with the International Staging System classification allows stratification of myeloma patients undergoing autologous stem cell transplantation. Haematologica 2010 July; 95(7):1150-7.
    • 30. Carrasco D.R., G. Tonon, Y. Huang, Y. Zhang, R. Sinha, and B. Feng et al. High-resolution genomic profiles define distinct clinico-pathogenetic subgroups of multiple myeloma patients. Cancer Cell 2006; 9(4):313-25.
    • 31. Kuiper, R. et al. Leukemia 2012, 1-8 advance on line publication, 22 Jun. 2012; doi:10.1038/1eu.2012.127.

Claims (5)

What is claimed is:
1. A method for determining the prognosis of a subject diagnosed with multiple myeloma by classifying the patient into a high risk or a low risk category, the method comprising:
determining the expression level in a sample from the subject for each of the following 92 genes: SLC30A7, AK2, SYF2, S100A6, NUF2, DARS2, ARPC5, DTL, ANGEL2, LBR, TARBP1, GGPS1, LTBP1. FAM49A, MCM6, ACVR2A, GRB14, ITGA6, DHRS9, STAT1, SPATS2L, BCS1L, SFMBT1, ARL8B, POLQ, MCM2, CCRL1, SEC62, GABRA4, PGM2, NCAPG, FGFR3, SEPT11, AIMP1, CENPE, IL7R, DHFR, SAR1B, PCDHB7, ATP6V0E1, MCM3, TUBB, MARCKS, SLC17A5, NCUBE1, SUN1/GET4, DNAJB9, RAB2A, TRAM1, ZNF252, HNRNPK, MRPL41, ZWINT, FANCF, EHBP1L1, C15orf85, PPP2R1B, ROBO3, C1S, ESPL1, ITM2B, ZBTB25, NPC2, ATPBD4, C15orf38, FANCI, SMG1, DYNLRB2, TMEM97, SPAG5, TOP2A, BIRC5, C18orf10, TSPAN16, RPS28, RPS11, NOP56, FTL, CDH22, DONSON, PFKL, ST13, DUX4, RPS4X, KIF4A, HMGN5, HMGB3, MAGEA6, the gene at chromosome 8p12 detectable with probe 208232_x_at, the gene at chromosome 11p14.1 detectable with probe 243018_at, and the gene at chromosome 11q24.3 detectable with probe 238780_s_at; and
classifying the subject into a high risk or a low risk category based upon the gene expression levels from the 92 genes.
2. The method according to claim 1, wherein the sample comprises plasma cells.
3. The method according to claim 2, wherein determining the gene expression level comprises:
providing a probe set for the detection of each of the 92 genes;
contacting the probe set with a sample comprising mRNA from the subject; and
determining the expression level of each of the 92 genes.
4. The method according to claim 3, wherein the gene expression analysis is performed on a gene chip.
5. The method according to claim 4, wherein the method comprises
contacting a gene chip comprising probes for the detection of the genes with a sample comprising mRNA from the subject;
determining the expression level of the genes;
normalizing the expression levels using mean/variance normalization to obtain a normalized expression value for each gene;
multiplying the normalized expression value for each gene with the beta value for each gene to obtain the calculated value for each gene; and
determining a score by summation of the calculated values of the genes,
wherein a score above a predetermined threshold indicates that the subject is to be classified in the high risk category and a score at or below the predetermined threshold indicates that the subject is to be classified in the low risk category.
US17/029,564 2011-07-14 2020-09-23 Classifier for the molecular classification of multiple myeloma Abandoned US20210010091A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/029,564 US20210010091A1 (en) 2011-07-14 2020-09-23 Classifier for the molecular classification of multiple myeloma

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
EP11173971.0 2011-07-14
EP11173971A EP2546357A1 (en) 2011-07-14 2011-07-14 A new classifier for the molecular classification of multiple myeloma.
PCT/EP2012/063722 WO2013007795A1 (en) 2011-07-14 2012-07-12 A new classifier for the molecular classification of multiple myeloma
US201414232176A 2014-03-19 2014-03-19
US15/959,703 US10815532B2 (en) 2011-07-14 2018-04-23 Classifier for the molecular classification of multiple myeloma
US17/029,564 US20210010091A1 (en) 2011-07-14 2020-09-23 Classifier for the molecular classification of multiple myeloma

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/959,703 Continuation US10815532B2 (en) 2011-07-14 2018-04-23 Classifier for the molecular classification of multiple myeloma

Publications (1)

Publication Number Publication Date
US20210010091A1 true US20210010091A1 (en) 2021-01-14

Family

ID=46506441

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/232,176 Active 2032-12-01 US9976185B2 (en) 2011-07-14 2012-07-12 Classifier for the molecular classification of multiple myeloma
US15/959,703 Active 2032-07-15 US10815532B2 (en) 2011-07-14 2018-04-23 Classifier for the molecular classification of multiple myeloma
US17/029,564 Abandoned US20210010091A1 (en) 2011-07-14 2020-09-23 Classifier for the molecular classification of multiple myeloma

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US14/232,176 Active 2032-12-01 US9976185B2 (en) 2011-07-14 2012-07-12 Classifier for the molecular classification of multiple myeloma
US15/959,703 Active 2032-07-15 US10815532B2 (en) 2011-07-14 2018-04-23 Classifier for the molecular classification of multiple myeloma

Country Status (18)

Country Link
US (3) US9976185B2 (en)
EP (2) EP2546357A1 (en)
JP (1) JP6026529B2 (en)
KR (1) KR101933742B1 (en)
AU (1) AU2012282437B2 (en)
BR (1) BR112014000648B1 (en)
CA (1) CA2841536C (en)
CL (1) CL2014000103A1 (en)
DK (1) DK2732048T3 (en)
EA (1) EA032730B1 (en)
ES (1) ES2633165T3 (en)
IL (1) IL230164A (en)
MX (1) MX346314B (en)
NZ (1) NZ619896A (en)
PL (1) PL2732048T3 (en)
PT (1) PT2732048T (en)
WO (1) WO2013007795A1 (en)
ZA (1) ZA201400159B (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2546357A1 (en) 2011-07-14 2013-01-16 Erasmus University Medical Center Rotterdam A new classifier for the molecular classification of multiple myeloma.
WO2017138810A2 (en) * 2016-02-12 2017-08-17 Skylinedx B.V. Predicting response to immunomodulatory drugs (imids) in multiple myeloma patients
US20210166789A1 (en) 2017-04-04 2021-06-03 Skylinedx B.V. Method for identifying gene expression signatures
US10973820B2 (en) 2017-12-13 2021-04-13 Facio Intellectual Property B.V. Compounds for treatment of diseases related to DUX4 expression
US20210055301A1 (en) * 2018-04-28 2021-02-25 Beijing Normal University Molecular typing of multiple myeloma and application
CN110218789B (en) * 2019-04-22 2022-10-04 曾文炳 Gene probe composition and kit for predicting overall survival rate of multiple myeloma patients
TW202112368A (en) 2019-06-13 2021-04-01 荷蘭商法西歐知識產權股份有限公司 Inhibitor combinations for treatment of diseases related to dux4 expression
US20230271953A1 (en) 2019-11-29 2023-08-31 Facio Intellectual Property B.V. Novel compounds for treatment of diseases related to DUX4 expression
WO2021105474A1 (en) 2019-11-29 2021-06-03 Facio Intellectual Property B.V. New compounds for treatment of diseases related to dux4 expression

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070015146A1 (en) * 2001-05-22 2007-01-18 Gene Logic, Inc. Molecular nephrotoxicology modeling
AU2002359357A1 (en) * 2001-11-07 2003-07-09 The Board Of Trustees Of The University Of Arkansas Diagnosis prognosis and identification of potential therapeutic targets of multiple myeloma based on gene expression profiling
JP5531360B2 (en) * 2006-11-07 2014-06-25 ザ ボード オブ トラスティーズ オブ ザ ユニヴァーシティー オブ アーカンソー Identification and use of high-risk multiple myeloma genomic signatures based on gene expression profiling
US20080274911A1 (en) * 2006-11-07 2008-11-06 Burington Bart E Gene expression profiling based identification of genomic signature of high-risk multiple myeloma and uses thereof
EP1964930A1 (en) * 2007-02-28 2008-09-03 Institut National De La Sante Et De La Recherche Medicale (Inserm) Molecular classifier for prognosis in multiple myeloma
US20110301055A1 (en) * 2008-12-05 2011-12-08 Nicholas James Dickens Methods for determining a prognosis in multiple myeloma
WO2010151731A1 (en) * 2009-06-26 2010-12-29 University Of Utah Research Foundation Materials and methods for the identification of drug-resistant cancers and treatment of same
EP2546357A1 (en) 2011-07-14 2013-01-16 Erasmus University Medical Center Rotterdam A new classifier for the molecular classification of multiple myeloma.

Also Published As

Publication number Publication date
KR20140044364A (en) 2014-04-14
PT2732048T (en) 2017-07-18
BR112014000648A2 (en) 2017-02-14
AU2012282437B2 (en) 2016-04-21
EP2732048A1 (en) 2014-05-21
EA201400131A1 (en) 2014-04-30
NZ619896A (en) 2016-04-29
US20140221313A1 (en) 2014-08-07
EP2732048B1 (en) 2017-05-10
BR112014000648B1 (en) 2020-04-07
MX2014000440A (en) 2014-07-09
JP2014520540A (en) 2014-08-25
DK2732048T3 (en) 2017-08-07
JP6026529B2 (en) 2016-11-16
AU2012282437A1 (en) 2014-01-23
US9976185B2 (en) 2018-05-22
CA2841536A1 (en) 2013-01-17
IL230164A (en) 2017-12-31
US10815532B2 (en) 2020-10-27
MX346314B (en) 2017-03-15
EP2546357A1 (en) 2013-01-16
CA2841536C (en) 2020-09-22
ZA201400159B (en) 2017-09-27
CL2014000103A1 (en) 2014-07-04
PL2732048T3 (en) 2017-10-31
KR101933742B1 (en) 2018-12-28
EA032730B1 (en) 2019-07-31
WO2013007795A1 (en) 2013-01-17
US20180237866A1 (en) 2018-08-23
ES2633165T3 (en) 2017-09-19

Similar Documents

Publication Publication Date Title
US20210010091A1 (en) Classifier for the molecular classification of multiple myeloma
US20200291486A1 (en) Use of gene expression profiling to predict survival in cancer patient
US20200270691A1 (en) Diagnosis, prognosis and identification of potential therapeutic targets of multiple myeloma based on gene expression profiling
EP2087139B1 (en) Gene expression profiling based identification of genomic signatures of high-risk multiple myeloma
WO2014165753A1 (en) Methods and compositions for diagnosis of glioblastoma or a subtype thereof
US20190300956A1 (en) Method for identifying high-risk aml patients
US20210102260A1 (en) Patient classification and prognositic method
AU2017254645B2 (en) Evaluation of mantle cell lymphoma and methods related thereto
Rose et al. A robust molecular pattern for myelodysplastic syndromes in two independent cohorts investigated by next-generation sequencing can be revealed by comparative bioinformatic analyses.
EP3411517B1 (en) Method for identifying high-risk aml patients
WO2016008048A1 (en) Methods and devices for predicting anthracycline treatment efficacy
Dean et al. Genome-wide association study identifies variants at 16p13 associated with survival in multiple myeloma patients.
Walker et al. The varied distribution and impact of RAS codon and other key DNA alterations across the translocation cyclin D subgroups in multiple myeloma

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION