METHODS FOR PREDICTING MULTIPLE MYELOMA TREATMENT RESPONSE

Abstract
The present invention relates to a method of testing whether a patient suffering of multiple myeloma will respond or not to a DNA methyltransferase inhibitor (DNMTi) comprising: i. determining the expression level (ELi) of several genes G1-Gn selected from table A in a biological sample obtained from said patient ii. comparing the expression level (ELi) determined at step i) with a predetermined reference level (ELRi) iii. calculating the DMS score trough the following formula (I) wherein β1 represent the regression β coefficient reference value for the gene Gi and Ci=1 if the expression of the gene Gi (ELi) is higher than the predetermined reference level (ELRi) or Ci=−1 if the expression of the gene (ELi) is lower than or equal to the predetermined reference level (ELRi) comparing the score DMS determined at step iii) with a predetermined reference value DMSR and concluding that the patient will respond to the DNMTi when the DMS score is higher than the predetermined reference value DMSR or concluding that the patient will not respond to the DNMTi when the DMS score is lower than the predetermined reference value DMSR.
Description
FIELD OF THE INVENTION

The present invention relates to methods for predicting multiple myeloma treatment response.


BACKGROUND OF THE INVENTION

Malignant transformation requires oncogenic activation and inactivation of tumor suppressor genes, which help cancer cells overriding the normal mechanisms controlling cellular survival and proliferation (1,2). These molecular events are caused by genetic alterations (translocations, amplification, mutations) and also by epigenetic modifications (3). Epigenetic modifications include methylation of DNA cytosine residues and histone modifications and have been shown to be critical in the initiation and progression of cancers (4). DNA methyltransferase inhibitors or HDAC inhibitors are now being used in the treatment of several hematologic malignancies including MM (5-8).


Multiple myeloma (MM) is a plasma cell neoplasm characterized by the accumulation of malignant plasma cells, termed Multiple Myeloma Cells (MMCs) within the bone marrow (BM). Despite the recent introduction of therapies such as Lenalidomide and Bortezomib, MM remains an almost incurable disease. MM arises through the accumulation of multiple genetic changes that include an aberrant or overexpression of a D-type cyclin gene, cyclin D1 (CCND1) in the case of t(11;14) translocation or gain in 11q13, cyclin D3 (CCND3) in the case of the rare t(6;14) translocation, or cyclin D2 (CCND2) on the background of a translocation involving c-maf (414;16)) or MMSET/FGFR3 (t(4;14)) (9,10).


Recent studies have shown that epigenetic changes such as DNA methylation play a role by silencing various cancer-related genes in MM. Most of these studies have been performed on limited number of genes using methylation specific PCR (11-18). Among the genes identified with promoter hypermethylation in MM, cyclin-dependent kinase inhibitor 2A (CDKN2A) and transforming growth factor beta receptor 2 (TGFBR2) have been shown to be associated with a poor prognosis in MM patients with discrepant results for CDKN2A (12). Heller at al. have identified several epigenetic inactivated cancer related genes in 3 human myeloma cell lines (HMCLs) and validated the relevance of 10 of these genes in 6 additional HMCLs, premalignant PCs from 24 MGUS patients and MMCs from 111 patients with MM (19). Methylation of secreted protein acidic and rich in cysteine (SPARC) and Bcl2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3) promoters was associated with poor overall survival of MM patients (19). SOCS3 methylation was found to be associated with extramedullary manifestations, plasma cell leukemia and significant shortened survival in MM patients (20). More recently, Morgan et al have shown that the transition of normal plasma cell (PC) and MGUS stage to MM stage is associated with DNA hypomethylation, but the transition of intramedullary MM stage to plasma cell leukemia or HMCL stage is associated with DNA hypermethylation (21). They described 2 specific subgroups of hyperdiploid MM on the basis of their methylation profile, which had a significantly different overall survival (21).


DNMT inhibitors can be sub-divided into nucleoside analogue and non-nucleoside analogue families. 5-Azacytidine (azacytidine)/5-Aza-2′-deoxycytidine (decitabine) are both nucleoside analogues with Food and Drug Administration approval for use in myelodysplastic syndrome. Clinical trials in myeloma combining these demethylating agents with chemotherapy or other agents are underway (8).


An important objective for optimizing these clinical trials will be the identification of biomarkers predictive for sensitivity of MMCs to DNMTi. In the present invention, the inventors used gene expression profiling of Multiple Myeloma Cells (MMCs) to build a novel “DNA methylation gene expression score” that makes it possible identification of patients whose MMCs will be targeted by DNMT inhibition.


SUMMARY OF THE INVENTION

The present invention relates to a method of testing whether a patient suffering of multiple myeloma will respond or not to a DNA methyltransferase inhibitor (DNMTi).


DETAILED DESCRIPTION OF THE INVENTION

The multiple myeloma treatment response was investigated by the inventors using DNA methyltransferase inhibitors (DNMTi), human multiple myeloma cell lines and primary myeloma cells. The inventors analyzed gene expression profiles of 5 MM cell lines treated with decitabine using microarrays. The inventors identified 127 genes deregulated by decitabine. 47 out of 127 decitabine deregulated genes have prognostic value in inventor's cohort of 206 newly-diagnosed MM patients. The inventors summarized the prognostic information in a DNA methylation score (DM Score or DMS) built using the probe set signal value weighted by the beta coefficient of prognostic genes. The DM Score is predictive for DNMT inhibitor sensitivity of HMCLs and primary myeloma cells in vitro. The DM Score allows identification of myeloma patients that could benefit DNMT inhibitor treatment.


The inventors also demonstrated that the DM Score is also predictive of myeloma cells sensitivity for another DNMTi, 5-Azacytidine. HMCLs with a high DM Score exhibited significant 3-fold higher 5-Azacytidine sensitivity than HMCLs with a low DM Score and these results were validated in vitro with primary myeloma cells of patients.


Definitions

The term “patient” denotes a mammal. In a preferred embodiment of the invention, a patient refers to any patient (preferably human) afflicted with multiple myeloma. The term “multiple myeloma” refers to multiple myeloma such as revised in the World Health Organisation Classification C90.


The term “DNA methyltransferase inhibitors” or “DNMTi” has its general meaning in the art and refers to a multiple myeloma treatment. The term “DNA methyltransferase inhibitors” or “DNMTi” refers to DNA methyltransferase inhibitor that can be sub-divided into nucleoside analogue (5-Azacytidine (azacytidine), 5-Aza-2′-deoxycytidine (decitabine, 5-Aza-CdR), zebularine, 5-Fluoro-2′-deoxycytidine (5-F-CdR), 5,6-Dihydro-5-azacytidine (DHAC)) and non-nucleoside analogue families (Hydralazine, Procainamide, Procaine, EGCG ((−)-epigallocatechin-3-gallate), Psammaplin A, MG98, RG108) (8).


The term “biological sample” refers to multiple myeloma cells, bone marrow or medullary cell.


All the genes pertaining to the invention are known per se, and are listed in the below Table A.









TABLE A







Set of predictive genes.













Gene


β
Reference


Gene
Symbol
Gene name
Gene ID
coefficient
level (ELRi)















G1
COP1
Caspase-1 dominant-
1552701_a_at
0.669962539
25.72815534




negative inhibitor pseudo-




ICE


G2
DNAJB9
DnaJ (Hsp40) homolog;
1554462_a_at
−0.807267505
87.37864078




subfamily B; member 9


G3
INSIG1
insulin induced gene 1
201625_s_at
0.918185816
10.19417476


G4
SEL1L
sel-1 suppressor of lin-12-
202061_s_at
−0.683515307
86.40776699




like (C. elegans)


G5
FNDC3A
fibronectin type III domain
202304_at
−0.821182841
76.21359223




containing 3A


G6
IFI27
interferon, alpha-inducible
202411_at
0.87241554
60.19417476




protein 27


G7
STCH
stress 70 protein
202558_s_at
−0.924845543
27.66990291




chaperone; microsome-




associated; 60 kDa


G8
GALNT3
UDP-N-acetyl-alpha-D-
203397_s_at
0.838104028
30.58252427




galactosamine: polypeptide




N-




acetylgalactosaminyltransferase




3 (GalNAc-T3)


G9
TPST2
tyrosylprotein
204079_at
0.698525183
14.5631068




sulfotransferase 2


G10
KIF21B
kinesin family member
204411_at
1.057242019
26.21359223




21B


G11
G1P2
Interferon; alpha-inducible
205483_s_at
0.704117799
40.77669903




protein (clone IFI-15K)


G12
OAS1
2′,5′-oligoadenylate
205552_s_at
0.918506668
12.13592233




synthetase 1


G13
ITGB7
integrin, beta 7
205718_at
1.393658338
11.16504854


G14
SP110
SP110 nuclear body
208012_x_at
0.796781189
13.10679612




protein


G15
EIF2S2
eukaryotic translation
208725_at
−0.608613084
75.24271845




initiation factor 2, subunit




2 beta


G16
CORO1A
coronin, actin binding
209083_at
1.116949242
50.48543689




protein, 1A


G17
TUBA3
tubulin, alpha 1a
209118_s_at
0.794556227
15.04854369


G18
ADFP
Adipose differentiation-
209122_at
−0.742527707
73.30097087




related protein


G19
IFI35
interferon-induced protein 35
209417_s_at
0.608446212
29.12621359


G20
STAT1
signal transducer and
209969_s_at
0.865200608
20.87378641




activator of transcription 1


G21
HIST1H2BG
histone cluster 1, H2bc
210387_at
−0.713207906
78.15533981


G22
HLA-DPA1
major histocompatibility
211990_at
−0.708484927
79.12621359




complex, class II, DP




alpha 1


G23
RECQL
RecQ protein-like (DNA
213878_at
0.79702138
19.90291262




helicase Q1-like)


G24
CCPG1
cell cycle progression 1
214152_at
−0.686867998
48.05825243


G25
NFE2L1
nuclear factor (erythroid-
214179_s_at
0.694584139
12.13592233




derived 2)-like 1


G26
PARP12
poly (ADP-ribose)
218543_s_at
1.257650329
10.19417476




polymerase family,




member 12


G27
FKBP11
FK506 binding protein 11;
219118_at
−0.88381802
76.69902913




19 kDa


G28
EAF2
ELL associated factor 2
219551_at
−0.762323817
83.98058252


G29
C6orf48
chromosome 6 open
220755_s_at
−0.832289767
23.78640777




reading frame 48


G30
IL21R
interleukin 21 receptor
221658_s_at
0.612993436
24.75728155


G31
PGM3
Phosphoglucomutase 3
221788_at
−1.227167702
87.86407767


G32
EIF2C2
Eukaryotic translation
222294_s_at
−0.657954783
69.90291262




initiation factor 2C; 2


G33
TMEM39A
transmembrane protein
222690_s_at
0.787711031
64.5631068




39A


G34
SLAMF7
SLAM family member 7
222838_at
−0.601257629
64.0776699


G35
MYLIP
myosin regulatory light
223129_x_at
0.687458269
22.33009709




chain interacting protein


G36
PPAPDC1B
phosphatidic acid
223569_at
−0.860377506
83.49514563




phosphatase type 2 domain




containing 1B


G37
ARHGAP9
Rho GTPase activating
224451_x_at
−0.731399299
83.98058252




protein 9 /// Rho GTPase




activating protein 9


G38
RPL37
ribosomal protein L37
224763_at
−1.135732424
88.34951456


G39
GSK3B
Glycogen synthase kinase
226183_at
−0.772104529
83.98058252




3 beta


G40
ELL2
elongation factor; RNA
226982_at
−0.687762574
80.09708738




polymerase II; 2


G41
EST,
Expression sequence tag,
227755_at
−0.79716878
68.93203883



GenBank AA042983
GenBank AA042983


G42
SAMD9
sterile alpha motif domain
228531_at
0.813378182
46.60194175




containing 9


G43
GNG7
guanine nucleotide binding
228831_s_at
−0.641328827
80.09708738




protein (G protein);




gamma 7


G44
SEC63
SEC63 Homolog
229969_at
−1.216057852
85.9223301




(S. cerevisiae)


G45
EST,
Expression sequence tag,
230570_at
−0.659929394
82.52427184



GenBank AI702465
GenBank AI702465


G46
MGC15875
hypothetical protein
232488_at
−1.036621429
80.09708738




MGC15875


G47
SFN
stratifin
33322_i_at
0.721955343
12.62135922









Methods for Predicting Response

The present invention relates to a method of testing whether a patient suffering of multiple myeloma will respond or not to a DNA methyltransferase inhibitor (DNMTi) comprising:

    • i) determining the expression level (ELi) of several genes G1-Gn selected from table A in a biological sample obtained from said patient
    • ii) comparing the expression level (ELi) determined at step i) with a predetermined reference level (ELRi)
    • iii) calculating the DMS score trough the following formula






DMS
=




i
=
1

n



β





i
×
Ci






wherein βi represent the regression β coefficient reference value for the gene Gi and Ci=1 if the expression of the gene Gi (ELi) is higher than the predetermined reference level (ELRi) or Ci=−1 if the expression of the gene (ELi) is lower than or equal to the predetermined reference level (ELRi)

    • iv) comparing the score DMS determined at step with a predetermined reference value DMSR
    • v) and concluding that the patient will respond to the DNMTi when the DMS score is higher than the predetermined reference value DMSR or concluding that the patient will not respond to the DNMTi when the DMS score is lower than the predetermined reference value DMSR


In some embodiments, the levels of at least 25 genes from Table A are determined wherein said genes are:


202061_s_at SEL1L


202304_at FNDC3A


202558_s_at STCH


203397_s_at GALNT3


204079_at TPST2


205483_s_at G1P2


205552_s_at OAS1


205718_at ITGB7


208725_at EIF2S2


209083_at CORO1A


209118_s_at TUBA3


209122_at ADFP


209417_s_at IF135


211990_at HLA-DPA1


214152_at CCPG1


214179_s_at NFE2L1


219118_at FKBP11


221658_s_at IL21R


222690_s at TMEM39A


223129_x_at MYLIP


224451_x_at ARHGAP9


224763_at RPL37


227755_at EST, GenBank AA042983


228831_s_at GNG7


232488_at MGC15875


In some embodiment, the level of 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, or 47 genes from Table A are determined wherein every combinations of genes comprises a minimal set of 25 genes consisting of:


202061_s_at SEL1L


202304_at FNDC3A


202558_s_at STCH


203397_s_at GALNT3


204079_at TPST2


205483_s_at G1P2


205552_s_at OAS1


205718_at ITGB7


208725_at EIF2S2


209083_at CORO1A


209118_s_at TUBA3


209122_at ADFP


209417_s_at IF135


211990_at HLA-DPA1


214152_at CCPG1


214179_s_at NFE2L1


219118_at FKBP11


221658_s_at IL21R


222690_s_at TMEM39A


223129_x at MYLIP


22445 l_x_at ARHGAP9


224763_at RPL37


227755_at EST, GenBank AA042983


228831_s at GNG7


232488_at MGC15875


In some embodiments, the level of the 47 genes of Table A are determined


Determination of the expression level of the genes can be performed by a variety of techniques. Generally, the expression level as determined is a relative expression level. More preferably, the determination comprises contacting the biological sample with selective reagents such as probes, primers or ligands, and thereby detecting the presence, or measuring the amount, of polypeptide or nucleic acids of interest originally in the biological sample. Contacting may be performed in any suitable device, such as a plate, microtiter dish, test tube, well, glass, column, and so forth. In specific embodiments, the contacting is performed on a substrate coated with the reagent, such as a nucleic acid array or a specific ligand array. The substrate may be a solid or semi-solid substrate such as any suitable support comprising glass, plastic, nylon, paper, metal, polymers and the like. The substrate may be of various forms and sizes, such as a slide, a membrane, a bead, a column, a gel, etc. The contacting may be made under any condition suitable for a detectable complex, such as a nucleic acid hybrid or an antibody-antigen complex, to be formed between the reagent and the nucleic acids or polypeptides of the biological sample.


In a preferred embodiment, the expression level may be determined by determining the quantity of mRNA.


Methods for determining the quantity of mRNA are well known in the art. For example the nucleic acid contained in the biological sample is first extracted according to standard methods, for example using lytic enzymes or chemical solutions or extracted by nucleic-acid-binding resins following the manufacturer's instructions. The extracted mRNA is then detected by hybridization (e.g., Northern blot analysis) and/or amplification (e.g., RT-PCR). Preferably quantitative or semi-quantitative RT-PCR is preferred. Real-time quantitative or semi-quantitative RT-PCR is particularly advantageous.


Other methods of amplification include ligase chain reaction (LCR), transcription-mediated amplification (TMA), strand displacement amplification (SDA) and nucleic acid sequence based amplification (NASBA).


Nucleic acids having at least 10 nucleotides and exhibiting sequence complementarity or homology to the mRNA of interest herein find utility as hybridization probes or amplification primers. It is understood that such nucleic acids need not be identical, but are typically at least about 80% identical to the homologous region of comparable size, more preferably 85% identical and even more preferably 90-95% identical. In certain embodiments, it will be advantageous to use nucleic acids in combination with appropriate means, such as a detectable label, for detecting hybridization. A wide variety of appropriate indicators are known in the art including, fluorescent, radioactive, enzymatic or other ligands (e.g. avidin/biotin).


Probes typically comprise single-stranded nucleic acids of between 10 to 1000 nucleotides in length, for instance of between 10 and 800, more preferably of between 15 and 700, typically of between 20 and 500. Primers typically are shorter single-stranded nucleic acids, of between 10 to 25 nucleotides in length, designed to perfectly or almost perfectly match a nucleic acid of interest, to be amplified. The probes and primers are “specific” to the nucleic acids they hybridize to, i.e. they preferably hybridize under high stringency hybridization conditions (corresponding to the highest melting temperature Tm, e.g., 50% formamide, 5× or 6×SCC. SCC is a 0.15 M NaCl, 0.015 M Na-citrate).


The nucleic acid primers or probes used in the above amplification and detection method may be assembled as a kit. Such a kit includes consensus primers and molecular probes. A preferred kit also includes the components necessary to determine if amplification has occurred. The kit may also include, for example, PCR buffers and enzymes; positive control sequences, reaction control primers; and instructions for amplifying and detecting the specific sequences.


In a particular embodiment, the methods of the invention comprise the steps of providing total RNAs extracted from a biological samples and subjecting the RNAs to amplification and hybridization to specific probes, more particularly by means of a quantitative or semi-quantitative RT-PCR.


In another preferred embodiment, the expression level is determined by DNA chip analysis. Such DNA chip or nucleic acid microarray consists of different nucleic acid probes that are chemically attached to a substrate, which can be a microchip, a glass slide or a microsphere-sized bead. A microchip may be constituted of polymers, plastics, resins, polysaccharides, silica or silica-based materials, carbon, metals, inorganic glasses, or nitrocellulose. Probes comprise nucleic acids such as cDNAs or oligonucleotides that may be about 10 to about 60 base pairs. To determine the expression level, a biological sample from a test patient, optionally first subjected to a reverse transcription, is labelled and contacted with the microarray in hybridization conditions, leading to the formation of complexes between target nucleic acids that are complementary to probe sequences attached to the microarray surface. The labelled hybridized complexes are then detected and can be quantified or semi-quantified. Labelling may be achieved by various methods, e.g. by using radioactive or fluorescent labelling. Many variants of the microarray hybridization technology are available to the man skilled in the art (see e.g. the review by Hoheisel, Nature Reviews, Genetics, 2006, 7:200-210)


In this context, the invention further provides a DNA chip comprising a solid support which carries nucleic acids that are specific to the genes listed in table A.


Predetermined reference values ELRi or DMSR used for comparison may consist of “cut-off” values.


For example; each reference (“cut-off”) value ELRi for each gene may be determined by carrying out a method comprising the steps of:


a) providing a collection of samples from patients suffering of multiple myeloma;


b) determining the expression level of the relevant gene for each sample contained in the collection provided at step a);


c) ranking the samples according to said expression level


d) classifying said samples in pairs of subsets of increasing, respectively decreasing, number of members ranked according to their expression level,


e) providing, for each sample provided at step a), information relating to the actual clinical outcome for the corresponding cancer patient (i.e. the duration of the disease-free survival (DFS) or the overall survival (OS) or both);


f) for each pair of subsets of tumour tissue samples, obtaining a Kaplan Meier percentage of survival curve;


g) for each pair of subsets of tumour tissue samples calculating the statistical significance (p value) between both subsets


h) selecting as reference value ELR for the expression level, the value of expression level for which the p value is the smallest.


For example the expression level of a gene Gi has been assessed for 100 samples of 100 patients. The 100 samples are ranked according to the expression level of gene Gi. Sample 1 has the highest expression level and sample 100 has the lowest expression level. A first grouping provides two subsets: on one side sample Nr 1 and on the other side the 99 other samples. The next grouping provides on one side samples 1 and 2 and on the other side the 98 remaining samples etc., until the last grouping: on one side samples 1 to 99 and on the other side sample Nr 100. According to the information relating to the actual clinical outcome for the corresponding cancer patient, Kaplan Meier curves are prepared for each of the 99 groups of two subsets. Also for each of the 99 groups, the p value between both subsets was calculated. The reference value ELRi is then selected such as the discrimination based on the criterion of the minimum p value is the strongest. In other terms, the expression level corresponding to the boundary between both subsets for which the p value is minimum is considered as the reference value. It should be noted that according to the experiments made by the inventors, the reference value ELRi is not necessarily the median value of expression levels.


The man skilled in the art also understands that the same technique of assessment of the DMSR could be used for obtaining the reference value and thereafter for assessment of the response to DNMTi. However in one embodiment, the reference value DMSR is the median value of DMS.


In one embodiment, the reference value ELRi for the genes are described in table A (right column).


In one embodiment, the reference value DMSR is −19.7 for determining whether a patient suffering of multiple myeloma will respond to a DNMTi or −15.3 for predicting the survival time of patient suffering of multiple myeloma.


The regression β coefficient reference values may be easily determined by the skilled man in the art for each gene using a Cox model. The Cox model is based on a modeling approach to the analysis of survival data. The purpose of the model is to simultaneously explore the effects of several variables on survival. The Cox model is a well-recognised statistical technique for analysing survival data. When it is used to analyse the survival of patients in a clinical trial, the model allows us to isolate the effects of treatment from the effects of other variables. The logrank test cannot be used to explore (and adjust for) the effects of several variables, such as age and disease duration, known to affect survival. Adjustment for variables that are known to affect survival may improve the precision with which we can estimate the treatment effect. The regression method introduced by Cox is used to investigate several variables at a time. It is also known as proportional hazards regression analysis. Briefly, the procedure models or regresses the survival times (or more specifically, the so-called hazard function) on the explanatory variables. The hazard function is the probability that an individual will experience an event (for example, death) within a small time interval, given that the individual has survived up to the beginning of the interval. It can therefore be interpreted as the risk of dying at time t. The quantity h0 (t) is the baseline or underlying hazard function and corresponds to the probability of dying (or reaching an event) when all the explanatory variables are zero. The baseline hazard function is analogous to the intercept in ordinary regression (since exp0=1). The regression coefficient β gives the proportional change that can be expected in the hazard, related to changes in the explanatory variables. The coefficient β is estimated by a statistical method called maximum likelihood. In survival analysis, the hazard ratio (HR) (Hazard Ratio=exp(β)) is the ratio of the hazard rates corresponding to the conditions described by two sets of explanatory variables. For example, in a drug study, the treated population may die at twice the rate per unit time as the control population. The hazard ratio would be 2, indicating higher hazard of death from the treatment.


In one embodiment, the regression β coefficient reference values are described in table A.


The invention also relates to a kit for performing the methods as above described, wherein said kit comprises means for measuring the expression level of the genes listed in Table A. Typically the kit may include a primer, a set of primers, a probe, a set of probes as above described. In a particular embodiment, the probe or set of probes are labelled as above described. The kit may also contain other suitably packaged reagents and materials needed for the particular detection protocol, including solid-phase matrices, if applicable, and standards.


In a particular embodiment, the score may be generated by a computer program.


Methods of Treatment

The method of the invention allows to define a subgroup of patients who will be responsive (“responder”) or not (“non responder”) to the treatment with a DNA methyltransferase inhibitor.


A further object of the invention relates to a method for the treatment of multiple myeloma in a patient in need thereof


In the context of the invention, the term “treating” or “treatment”, as used herein, means reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.


In a particular embodiment, the method comprises the following steps


a) testing whether the patient will respond or not to a DNA methyltransferase inhibitor (DNMTi) by performing the method according to the invention


b) administering the DNA methyltransferase inhibitor, if said patient has as score higher than the reference value DMSR (i.e. the patient will respond to the DNA methyltransferase inhibitor).


A further object of the invention relates to a DNA methyltransferase inhibitor for use in the treatment of multiple myeloma in a patient in need thereof, wherein the patient was being classified as responder by the method as above described.


The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.





FIGURES


FIG. 1: DNA methylation Score in normal and malignant plasma cells


(A) DNA methylation Score in normal bone marrow plasma cells (N=7), in premalignant plasma cells of patients with monoclonal gammopathy of undetermined significance (MGUS, N=5), in multiple myeloma cells of patients with intramedullary MM (N=206) and in human myeloma cell lines (N=40). ** Indicate that the score value is significantly different with a P value <0.01. (B) DNA methylation Score in the 8 groups of the molecular classification of multiple myeloma. The DM Score was investigated in the 8 groups of the molecular classification of multiple myeloma in UAMS-TT2 cohort of patients. PR: proliferation, LB: low bone disease, MS: MMSET, HY: hyperdiploid, CD1: Cyclin D1, CD2: Cyclin D2, MF: MAF, MY: myeloid. * Indicate that the score value is significantly higher in the group compared to all the patients of the cohort (P<0.05). ** Indicate that the score value is significantly lower in the group compared to all the patients of the cohort (P<0.05).



FIG. 2: Prognostic value of DM Score in multiple myeloma.


Patients of HM cohort were ranked according to increased DM Score and a maximum difference in OS was obtained with DM Score=−15.3 splitting patients in a high risk (34.5%) and low risk (65.5%) groups. The prognostic value of DM Score was tested on an independent cohort of 345 patients from UAMS treated with TT2 therapy (UAMS- TT2 cohort). The parameters to compute the DM Score of patients of UAMS-TT2 cohort and the DM Score cut-off delineating the 2 prognostic groups were those defined with HM cohort.



FIG. 3: DM Score predicts for sensitivity of human myeloma cell lines to decitabine.


(A) HMCLs with high DM Score (N=5) exhibit significant higher DNMTi sensitivity compared to HMCLs with low DM Score (N=5). HMCLs were cultured for 4 days in 96-well flat-bottom microtiter plates in RPMI 1640 medium, 10% FCS, 2 ng/ml IL-6 culture medium (control), with graded decitabine concentrations. Data are mean values plus or minus standard deviation (SD) of 5 experiments determined on sextuplet culture wells.



FIG. 4: DM Score predicts for decitabine sensitivity of primary myeloma cells of patients.


Mononuclear cells from tumor samples of 12 patients with MM were cultured for 4 days in the presence of IL-6 (2 ng/ml) with or without graded decitabine concentrations. At day 4 of culture, the cell count and the viability were determined and the percentage of CD138+ viable plasma cells was determined by flow cytometry. Black color represents patients with high DM Score (N=6) and white represents patients with low DM Score values (N=6).



FIG. 5: DM Score predicts for sensitivity of human myeloma cell lines to 5-azacitidine.


HMCLs with a high DM Score (n=6) exhibit significant higher 5-azacitidine sensitivity compared to HMCLs with a low DM Score (n=6). HMCLs were cultured for 4 days in 96-well flat-bottom microtiter plates in RPMI 1640 medium, 10% FCS, 2 ng/ml IL-6 culture medium (control), and graded 5-azacitidine concentrations. Data are mean values plus or minus standard deviation (SD) of 5 experiments determined on sextuplet culture wells.



FIG. 6: DM Score predicts for sensitivity of primary myeloma cells of patients to 5-azacitidine.


Mononuclear cells from tumor samples of 14 patients with MM were cultured for 4 days in the presence of IL-6 (2 ng/ml) with or without graded 5-azacitidine concentrations. At day 4 of culture, the count of viable CD138′ MMCs was determined using flow cytometry. The black columns represent the mean±SD of primary myeloma cell counts (expressed as the percentage of the count without adding 5-azcytidine) of the 7 patients with a low DM Score and the white columns that of the 7 patients with a high DM Score.





EXAMPLE 1
Development of Gene Expression Based Score to Predict Sensitivity of Multiple Myeloma Cells to DNA Methylation Inhibitors

Material & Methods


Human Myeloma Cell Lines (HMCLs)


XG-1, XG-2, XG-3, XG-4, XG-5, XG-6, XG-7, XG-10, XG-11, XG-12, XG-13, XG-14, XG-16, XG-19, XG-20 and XG-21 human myeloma cell lines were obtained as previously described (22-26). JJN3 was kindly provided by Dr Van Riet (Bruxelles, Belgium), JIM3 by Dr MacLennan (Birmingham, UK) and MM1S by Dr S. Rosen (Chicago, USA). AMO-1, LP1, L363, U266, OPM2, and SKMM2 were from DSMZ (Germany) and RPMI8226 from ATTC (USA). All HMCLs derived in inventor's laboratory were cultured in the presence of recombinant IL-6. HMCLs microarray data have been deposited in the ArrayExpress public database under accession numbers E-TABM-937 and E-TABM-1088.


Primary Multiple Myeloma Cells


MMCs were purified from 206 patients with newly-diagnosed MM after written informed consent was given at the University hospitals of Heidelberg (Germany) or Montpellier (France) and agreement of the Center for Biological Resources of Montpellier University Hospital (N° DC-2008-417). The study was approved by the ethics boards of Heidelberg and Montpellier Universities. These 206 patients were treated with high dose Melphalan (HDM) and autologous stem cell transplantation (ASCT) (27) and were termed in the following Heidelberg-Montpellier (HM) series (Supplementary Table S1). The CEL files and MASS files have been deposited in the ArrayExpress public database (E-MTAB-372). The structural chromosomal aberrations including t(4;14)(p16.3;q32.3) and t(11;14)(q13;q32.3), as well as numerical aberrations including 17p13 and 1q21 gain, were assayed by fluorescence in situ hybridization (iFISH) (28). The inventors also used Affymetrix data of a cohort of 345 purified MMC from previously untreated patients from the University of Arkansas for Medical Sciences (UAMS, Little Rock, Ark.). The patients were treated with total therapy 2 including HDM and ASCT (29) and termed in the following UAMS-TT2 series. These data are publicly available via the online Gene Expression Omnibus (Gene Expression Profile of Multiple Myeloma, accession number GSE2658. http://www.ncbi.nlm nih.gov/geo/). As iFISH data were not available for UAMS-TT2 patients, t(4;14) translocation was evaluated using MMSET spike expression (30) and dell7p13 surrogated by TP53 probe set signal (31). After Ficoll-density gradient centrifugation, plasma cells were purified using anti-CD138 MACS microbeads (Miltenyi Biotech, Bergisch Gladbach, Germany).


Cell Culture and Treatment for Gene Expression Profiling


The human MM cell lines XG-5, XG-6, XG-7, XG-20 and LP1 were grown in RPMI 1640 supplemented with 10% fetal bovine serum and 2 ng/ml recombinant IL-6. Cells (2×105/mL) were treated either with 0.5 μmol/L 5-Aza-2′-deoxycytidine (decitabine) (Sigma, St. Louis, Mo.) for 7 days. Control cells were cultured in the same conditions without adding drug.


Growth Assay for Myeloma Cells


HMCLs were cultured for 4 days in 96-well flat-bottom microtiter plates in RPMI 1640 medium, 10% FCS, 2 ng/ml IL-6 culture medium (control), with graded decitabine concentrations. Cell growth was evaluated by quantifying intracellular ATP amount with a Cell Titer Glo Luminescent Assay (Promega, Madison, Wis.) with a Centro LB 960 luminometer (Berthold Technologies, Bad Wildbad, Germany).


Mononuclear Cell Culture


Mononuclear cells from tumor samples of 12 patients with MM (agreement of the Center for Biological Resources of Montpellier University Hospital (N° DC-2008-417)) were cultured for 4 days at 2×105 cells/ml in RPMI 1640 medium, 10% FCS, 2 ng/ml IL-6, with or without graded concentrations of decitabine. In each culture group, viability and cell counts were assayed and MMCs were stained with an anti-CD138-PE mAb (Immunotech, Marseille, France) as previously described (32).


Preparation of Complementary RNA (cRNA) and Microarray Hybridization


RNA was extracted using the RNeasy Kit (Qiagen, Hilden, Germany) as previously described (33,34). Biotinylated cRNA was amplified with a double in vitro transcription and hybridized to the human U133 2.0 plus GeneChips, according to the manufacturer's instructions (Affymetrix, Santa Clara, Calif.). Fluorescence intensities were quantified and analyzed using the GECOS software (Affymetrix).


Gene Expression Profiling and Statistical Analyses


Gene expression data were normalized with the MASS algorithm and analyzed with inventor's bioinformatics platforms—RAGE (http://rage.montp.inserm.fr/) (35) and Amazonia (http://amazonia.montp.inserm.fr/) (36)—or SAM (Significance Analysis of Microarrays) software (37). The statistical significance of differences in overall survival between groups of patients was calculated by the log-rank test. Multivariate analysis was performed using the Cox proportional hazards model. Survival curves were plotted using the Kaplan-Meier method. All these analyses have been done with R.2.10.1 (http://www.r-project.org/) and bioconductor version 2.5. Gene annotation and networks were generated through the use of Ingenuity Pathways Analysis (Ingenuity® Systems, Redwood City, Calif.) (38).


Results


Modulation of Gene Expression by Decitabine in HMCLs: Identification of Prognostic Genes


Five HMCLs were treated with 0.5 μM of decitabine for 7 days, a concentration which did not affect myeloma cell viability (Supplementary Table S2) (19). Using SAM supervised paired analysis, the expression of 48 genes was found to be significantly upregulated and that of 79 genes downregulated by decitabine treatment of 5 HMCLs (FDR <5%; Supplementary Table S3 and S4). Decitabine-regulated genes are significantly enriched in genes related to “Cancer” and “Cell death” pathways (FDR <5%; Ingenuity pathway analysis. Investigating the expression of these 127 decitabine-regulated genes in primary MMCs of a cohort of 206 newly-diagnosed patients (HM cohort), 22 genes had bad prognostic value and 25 a good one after Benjamini-Hochberg multiple testing correction (Supplementary Table S5). The prognostic information of decitabine regulated genes was gathered within an DNA methylation score (DM Score), which was the sum of the beta coefficients of the Cox model weighted by ±1 according to the patient MMC signal above or below the probe set maxstat value as previously described (38). The value of DM Score in normal, premalignant or malignant plasma cells is displayed in FIG. 1A. There is no significant difference of DM Score between cells from MGUS patients and normal BMPCs. MMCs of patients had a significantly higher DM Score than normal BMPCs or plasma cells from MGUS-patients (P<0.01), and HMCLs the highest score (P<0.001) (FIG. 1A). Investigating the DM Score in the 8 groups of the molecular classification of multiple myeloma (39), DM Score is significantly higher in the proliferation, t(4;14) and MAF subgroups (P<0.001) associated with a poor prognosis (39) and significantly lower in the low bone disease subgroup (P<0.001) (39) (FIG. 1B).


Prognostic value of DM score compared to usual prognostic factors


DM Score had prognostic value when used as a continuous variable (P≦10−4), or by splitting patients into two groups using Maxstat R function (38). A maximum difference in overall survival (OS) was obtained with DM Score=−15.3 splitting patients in a high-risk group of 34.5% patients (DM Score>−15.3) with a 42.1 months median OS and a low risk group of 65.5% patients (DM Score<−15.3) with not reached median survival (FIG. 2).


Using univariate Cox analysis, DM Score, UAMS-HRS, IFM-score and GPI had prognostic value as well as t(4;14), del17p, β2m, albumin and ISS using the HM patient cohort (Supplementary Table S6). When compared two by two, DM Score tested with ↑2m remained significant. When these parameters were tested together, DM Score, β2m and t(4;14) kept prognostic value. The DM Score is also prognostic in an independent cohort of 345 patients from UAMS treated with TT2 therapy (UAMS-TT2 cohort). For each patient of UAMS-TT2 cohort, DM Score was computed using parameters defined with HM patients' cohort. The median OS of patients within high score group (DM Score>−15.3) was 53.7 months and not reached for patients with low DM Score (P=0.0001) (FIG. 2). Using Cox univariate analysis, UAMS-HRS, IFM and GPI scores as well as t(4;14) and del17p had prognostic value. Comparing these prognostic factors two by two, DM Score remained significant compared to GPI, t(4;14), and dell7p in the UAMS-TT2 cohort (Supplementary Table S5). When these parameters were tested together, UAMS-HRS, t(4;14) and del17p kept prognostic value in UAMS-TT2 cohort.


DM Score is Predictive for Sensitivity of Human Myeloma Cell Lines or Patients' Primary MMCs to Decitabine In Vitro.


The inventors sought to determine whether DM Score could predict for the sensitivity of 10 HMCLs to DNMT inhibitor. Starting from a large cohort of 40 HMCLs (22), the 10 HMCLs with the highest or lowest DM Score were selected to assay decitabine sensitivity.


The 5 HMCLs with the highest DM Score exhibited a significant 11-fold higher decitabine sensitivity (median IC50=0.68 μM; range: 0.15 to 2.22 μM) than the 5 HMCLs with low DM Score (P=0.01; median IC50=7.94 μM; range: 2.92 to 60.81 μM) (FIG. 3). All HMCLs with the lowest DM Score and poorly sensitive to decitabine presented no ras mutations whereas 4 out of 5 HMCLs with the highest DM Score and higher decitabine sensitivity have ras mutations (Table 1).


Primary MMCs were cultured with their BM environment and recombinant IL-6 and graded concentrations of decitabine for 4 days. Primary MMCs of patients with a DM Score above median value (−19.7, FIG. 1) exhibited significant (P<0.01) 2.2-fold higher decitabine sensitivity than MMCs with DM Score below median (FIG. 4 and Table 2). The characteristics of patients with MM included in this study are described in Table 3.


Discussion


In this study, the inventors have identified a gene expression-based DNA methylation score (DM Score) which is predictive for patients' survival and for the in vitro sensitivity of human myeloma cell lines or patients' primary myeloma cells to a DNMT inhibitor, decitabine. Clinical trials in multiple myeloma combining these demethylating agents with chemotherapy or other agents are underway (8). The current identification of DM Score should be very useful to investigate whether the higher response to DNMTi is found in patients with highest DM Score and to speed up the investigation of the clinical efficacy of the novel agents.


Besides the potential utility of the current DM Score in selecting patients who could benefit from DNMTi therapies, the current study highlights pathways which could be involved in the emergence of multiple myeloma cells. Among the genes downregulated by decitabine treatment and associated with a poor prognosis, the inventors identified RECQ1 (ATP-dependent DNA helicase Q1) and KIF21B (kinesin family member 21B). RECQ helicase are a ubiquitous family of DNA unwinding enzymes involved in the maintenance of chromosome stability (40-42). Mutations in the genes of RECQ family members are linked with genetic disorders associated with genomic instability, cancer predisposition and features of premature ageing. Consistent with their ability to unwind DNA, several functions have been attributed to RECQ proteins, including roles in stabilization and repair of damaged DNA replication forks, telomere maintenance, homologous recombination, and DNA damage checkpoint signaling (40-42). Recent reports supported a cancer specific role for RECQ1 (43-45). RECQ1 silencing in cancer cells resulted in mitotic catastrophe and administration of siRNA targeting RECQ1 prevented tumor growth in murine models (43-45). More recently, it was demonstrated that RECQ1 is highly expressed in various types of solid tumors including colon carcinoma, thyroid cancer, lung cancer and brain glioblastoma tissues (46) In glioblastoma cell lines, depletion of RECQ1 by RNAi results in a significant reduction of cellular proliferation, perturbation of S-phase progression, spontaneous γ-H2AX foci formation and hypersensitivity to hydroxyurea and temozolomide treatments (46). KIF21B is a kinesin family member. Kinesins are a conserved class of microtubule-dependent molecular motor proteins that have adenosine triphosphatase activity and motion characteristics (47). Kinesins support several cellular functions, such as mitosis, meiosis and the transport of macromolecules. In mitosis of eukaryotic cells, kinesins participate in spindle formation, chromosome congression and alignment, and cytokinesis (48). Abnormal expression and function of kinesins are involved in the development or progression of several kinds of human cancers (49,50). Interestingly, KIF21B maps to chromosome 1q arm (1q32.1) whose amplification characterizes a significant fraction of high-risk MM tumors (51). More recently, KIF21B gene was found in a critical neighbor-gene model associated with a poor prognosis across independent data sets of respectively, 559, 247 and 264 myeloma patients (52). These data suggest that decitabine treatment could synergize with DNA-damaging agents, in MM, targeting genes involved in DNA-repair and maintenance of chromosome stability. These results demonstrated that the study of MMCs response to epigenetic-targeted treatments will extend our knowledge of MM development and progression and will lead to potential therapeutic advances. Epigenetic therapies could be combined with conventional therapies to develop personalized treatments in MM, render resistant tumors responsive to treatment.









TABLE 1







Characteristics of HMCLs5-aza sensitive and HMCLs5-aza resistant




















IL−6


Patient


t(14q32 or




HMCL


HMCL Name
dependence1
Origin2
Disease3
sample4
Gender
Isotype
22q11)
Target genes
Ras
TP53
CD45
classification





5-aza














Resistant


HMCLs


XG6
++
MN
MM
PB
F
Gl
t(16; 22)
c-Maf
wt
wt
+
CTA/MF


XG20
++
MN
PCL
PB
M
I
t(4; 14)
MMSET
wt
abn

MS


XG13
++
MN
PCL
PB
M
Gl
t(14; 16)
c-Maf
wt
abn
+
MF


SKMM2

CO
PCL
PB
M
Gk
t(11; 14)
CCND1
wt
abn

CD-1


LP1

CO
MM
PB
F
Gl
t(4; 14)
MMSET/FGFR3
wt
abn

MS


5-aza


Sensitive


HMCLs


XG12
++
MN
PCL
PB
F
I
t(14; 16)
c-Maf
mut
wt
+
CTA/MF


XG16
++
MN
PCL
PB
M
k
none
none
mut
abn
+
CTA/FRZB


XG19
++
MN
PCL
PB
F
Al
t(14; 16)
c-Maf
wt
wt
+
CTA/MF


JJN3

CO
MM
PE
F
Ak
t(14; 16)
c-Maf
mut
abn
+/−
MF


RPMI8226

CO
MM
PB
M
Gl
t(14; 16)
c-Maf
mut
abn

MF






1++ if growth is strictly dependent on adding exogenous IL-6, + if dependent on adding exogenous IL-6, − if not;




2Origin of the HMCL, MN Montpellier or Nantes, CO collected;




3Disease at diagnosis: MM multiple myeloma, PCL plasma cell leukemia, PCT plasmacytoma;




4Origin of the sample: AF ascitic fluid, BM bone marrow, PE pleural effusion, PB peripheral blood.














TABLE 2







Mononuclear cells from tumor samples of 12 patients with MM were


cultured for 4 days in the presence of IL-6 (2 ng/mL) with or without


increased doses of decitabine. At day 4 of culture, the cell count and


viability were determined and the percentage of CD138+


viable plasma cells was determined by flow cytometry.









Myeloma cell number/culture well














Patient

0.125 μM
0.5 μM
2 μM
8 μM



no.
Control
decitabine
decitabine
decitabine
decitabine

















Pa-
1
72168
43320
36738
20592
5800


tients
2
78182
49096
31086
22880
11600


with
3
60140
40432
28260
18304
10150


high
4
108252
63536
56520
29744
18850


DM
5
117273
69312
42390
32032
17400


Score
6
96224
60648
36738
20592
13050



Mean
88707
54391
38622
24024
12808


Pa-
1
24880
26100
27195
21028
18265


tients
2
57133
51984
45216
32032
21750


with
3
132308
77976
53694
22880
14500


low
4
53664
54336
51300
51230
53486


DM
5
1068
1066
870
1044
1039


Score
6
23716
51024
54726
55818
47440



Mean
48795
43748
38834
30672
26080
















TABLE 3







Characteristics of patients with a DM Score above (N = 6)


and under (N = 6) the median value.


















Durie and


Multiple myeloma





Monoclonal
Salmon
ISS
Serum β2-
molecular



Age
Sex
protein
stage
stage
microglobulin
classification










Patients with high DM Score














Patient 1
59
F
IgA Lambda
IIIA
III
6, 7
MF


Patient 2
48
M
Lambda
NA
NA
NA
CD1


Patient 3
63
M
BJ Lambda
NA
NA
NA
PR


Patient 4
69
M
IgG Lambda
IIIA
III
10.4
PR


Patient 5
70
F
IgA Lambda
IIIA
II
5
CD2


Patient 6
63
F
Asecret
III
III
13.5
CD1







Patients with low DM Score














Patient 1
72
M
IgG Lambda
IIIA
III
8.6
PR


Patient 2
54
M
IgA Lambda
IIIA
I
2.3
CD2


Patient 3
62
M
IgA Kappa
IIA
I
2.6
HY


Patient 4
55
M
BJ Kappa
IIIB
III
10
HY


Patient 5
69
F
IgG Kappa
IIIA
I
2.8
CD2


Patient 6
61
M
BJ Kappa
IIIA
II
4.6
HY
















SUPPLEMENTARY TABLE S1







Clinical patient data for age, serum-β2-microglobulin, and


plasma cell infiltration in the Heidelberg/Montpellier-group


(HM) and the Arkansas cohort. Median value and range are given.










HM cohort
Arkansas cohort


Characteristic
(n = 206)
(n = 345)





Age
58.5 [27-73]
57 [25-77]







Monoclonal protein









IgG
120
193


IgA
46
93


Bence Jones
35
47


Asecretory
4
6


IgD
1
3


NA
0
3







Myeloma in Durie and Salmon stage









I
22
NA


II
31
NA


III
153
NA







Myeloma in ISS stage









I
97
189


II
73
86


III
33
70


NA
3
0


Serum-β2-microglobulin
 2.99 [1.3-53.6]
  2.9 [1.0-38.7]


Plasma cells in bone marrow
  42 [1-100]
42 [4-98] 





NA, not available.


ISS, International Staging System.













SUPPLEMENTARY TABLE S2







Cell viability of HMCLs treated with either 0.5 μM decitabine


for 7 days. Date are the mean percentages ± SD of viable


cells evaluated by trypan blue exclusion (3 experiments).









Cell viability (%)










Day 3
Day 7












HMCLS
Day 0
Control
5-aza
Control
5-aza





XG-5
 70 ± 2
70 ± 1
65 ± 5
83 ± 5
69 ± 6


XG-6
 90 ± 2
90 ± 2
90 ± 1
93 ± 5
90 ± 1


XG-7
100 ± 0
90 ± 2
90 ± 2
93 ± 5
85 ± 5


XG-20
100 ± 0
91 ± 3
91 ± 3
95 ± 5
83 ± 5


LP1
100 ± 0
91 ± 2
91 ± 2
95 ± 5
86 ± 4
















SUPPLEMENTARY TABLE S3







Genes overexpressed in decitabine treated HMCLs. Five HMCLs were cultured


with or without 0.5 μM decitabine for 7 days and gene expression was profiled with


Affymetrix U133 plus 2.0. Genes significantly differentially expressed between control


and decitabine treated cells were identified using SAM supervised paired analysis with a


5% false discovery rate. When a gene was interrogated by several probe sets, we used the


probe set yielding to a maximum variance across control and decitabine treated cells.











Probeset
Gene
Ratio
Banding
Affymetrix description










Intercellular communication and membrane proteins











209122_at
ADFP
3.70
9p22.1
adipose differentiation-related protein


211990_at
HLA-DPA1
1.70
6p21.3
major histocompatibility complex; class II; DP






alpha 1


205718_at
ITGB7
2.11
12q13.13
integrin; beta 7


1569003_at
TMEM49
2.33
17q23.2
transmembrane protein 49


205483_s_at
G1P2
2.77
1p36.33
interferon; alpha-inducible protein (clone IFI-






15K)


200696_s_at
GSN
3.97
9q33
gelsolin (amyloidosis; Finnish type)







Signal transduction











203964_at
NMI
1.86
2p24.3-q21.3
N-myc (and STAT) interactor


205552_s_at
OAS1
1.65
12q24.1
2prime;5prime-oligoadenylate synthetase 1;






40/46 kDa


209969_s_at
STAT1
2.55
2q32.2
signal transducer and activator of transcription






1; 91 kDa


202693_s_at
STK17A
2.52
7p12-p14
serine/threonine kinase 17a (apoptosis-






inducing)







Cytoskeleton











223129_x_at
MYLIP
2.00
6p23-p22.3
myosin regulatory light chain interacting






protein


209083_at
CORO1A
2.38
16p11.2
coronin; actin binding protein; 1A


216323_x_at
H2-ALPHA
2.56
2q21.1
alpha-tubulin isotype H2-alpha


210527_x_at
TUBA2
2.50
13q11
tubulin; alpha 2


209118_s_at
TUBA3
2.23
12q12-12q14.3
tubulin; alpha 3


204141_at
TUBB2
5.07
6p25
tubulin; beta 2







Cancer testis antigens











235700_at
CT45-2
30.39
Xq26.3
cancer/testis antigen CT45-2


210437_at
MAGEA9
1.68
Xq28
melanoma antigen family A; 9


207847_s_at
MUC1
2.92
1q21
mucin 1; transmembrane







Protein binding











202411_at
IFI27
2.84
14q32
interferon; alpha-inducible protein 27


224917_at
MIRN21
1.99

microRNA 21


202814_s_at
HEXIM1
2.18
17q21.31
hexamethylene bis-acetamide inducible 1


211071_s_at
MLLT11
2.53
1q21
myeloid/lymphoid or mixed-lineage leukemia






(trithorax homolog; Drosophila); translocated






to 11


33322_i_at
SFN
4.34
1p36.11
stratifin







Metabolism











207761_s_at
DKFZP586A0522
2.97
12q13.12
DKFZP586A0522 protein/METTL7A






methyltransferase like 7A


201468_s_at
NQO1
2.15
16q22.1
NAD(P)H dehydrogenase; quinone 1


218543_s_at
PARP12
1.92
7q34
poly (ADP-ribose) polymerase family; member 12


214183_s_at
TKTL1
85.24
Xq28
transketolase-like 1


204079_at
TPST2
2.20
22q12.1
tyrosylprotein sulfotransferase 2


201243_s_at
ATP1B1
3.43
1q24
ATPase; Na+/K+ transporting; beta 1






polypeptide


210580_x_at
SULT1A3
2.52
16p11.2
sulfotransferase family; cytosolic; 1A; phenol-






preferring; member 3







Nuclear proteins and transcription factors











238825_at
ACRC
25.99
Xq13.1
acidic repeat containing


31845_at
ELF4
1.75
Xq26
E74-like factor 4 (ets domain transcription






factor)


208012_x_at
SP110
2.11
2q37.1
SP110 nuclear body protein


210387_at
HIST1H2BG
2.00
6p21.3
histone 1; H2bg


201565_s_at
ID2
2.76
2p25
inhibitor of DNA binding 2; dominant negative






helix-loop-helix protein


223484_at
NMES1
23.96
15q21.1
normal mucosa of esophagus specific 1







Apoptosis











1552701_a_at
COP1
2.34

caspase-1 dominant-negative inhibitor pseudo-






ICE


209417_s_at
IFI35
1.93
17q21
interferon-induced protein 35


202086_at
MX1
1.73
21q22.3
myxovirus (influenza virus) resistance 1;






interferon-inducible protein p78 (mouse)


219099_at
C12orf5
2.16
12p13.3
chromosome 12 open reading frame 5


201631_s_at
IER3
3.97
6p21.3
immediate early response 3







Others











227609_at
EPSTI1
1.99
13q13.3
epithelial stromal interaction 1 (breast)


217755_at
HN1
2.17
17q25.1
hematological and neurological expressed 1


215343_at
KIAA1509
2.21
14q32.12
KIAA1509


228531_at
SAMD9
2.06
7q21.2
sterile alpha motif domain containing 9


230000_at
C17orf27
2.41
17q25.3
chromosome 17 open reading frame 27


235964_x_at
C20orf118
2.12
20
Chromosome 20 open reading frame 118
















SUPPLEMENTARY TABLE S4







Genes underexpressed in decitabine treated HMCLs. Five HMCLs were cultured with or without


0.5 μM decitabine for 7 days and gene expression was profiled with Affymetrix U133


plus 2.0. Genes significantly differentially expressed between control and decitabine


treated cells were identified using SAM supervised paired analysis with a 5% false discovery


rate. When a gene was interrogated by several probe sets, we used the probe set yielding


to a maximum variance across control and decitabine treated cells.











Probeset
Gene
Ratio
Banding
Affymetrix description










Intercellular communication and membrane proteins











202304_at
FNDC3A
0.40
13q14.2
fibronectin type III domain containing 3A


209541_at
IGF1
0.43
12q22-q23
insulin-like growth factor 1 (somatomedin






C)


221658_s_at
IL21R
0.50
16p11
interleukin 21 receptor


210587_at
INHBE
0.55
12q13.3
inhibin; beta E


219702_at
PLAC1
0.42
Xq26
placenta-specific 1


209606_at
PSCDBP
0.48
2q11.2
pleckstrin homology; Sec7 and coiled-coil






domains; binding protein


202375_at
SEC24D
0.31
4q26
SEC24 related gene family; member D






(S. cerevisiae)


222838_at
SLAMF7
0.51
1q23.1-
SLAM family member 7





q24.1


222690_s_at
TMEM39A
0.46
3q13.33
transmembrane protein 39A







Signal transduction











224451_x_at
ARHGAP9
0.38
12q14
Rho GTPase activating protein 9 /// Rho






GTPase activating protein 9


212954_at
DYRK4
0.42
12p13.32
dual-specificity tyrosine-(Y)-






phosphorylation regulated kinase 4


228831_s_at
GNG7
0.33
19p13.3
guanine nucleotide binding protein (G






protein); gamma 7


209314_s_at
HBS1L
0.53
6q23-q24
HBS1-like (S. cerevisiae)


219188_s_at
LRP16
0.36
11q11
LRP16 protein


229549_at
OPN1SW
0.40
7q31.3-q32
Opsin 1 (cone pigments); short-wave-






sensitive (color blindness; tritan)







Cytoskeleton











213500_at
COPB2
0.58
3q23
Coatomer protein complex; subunit beta 2






(beta prime)


204411_at
KIF21B
0.53
1pter-q31.3
kinesin family member 21B


226438_at
SNTB1
0.36
8q23-q24
Syntrophin; beta 1 (dystrophin-associated






protein A1; 59 kDa; basic component 1)


226181_at
TUBE1
0.38
6q21
tubulin; epsilon 1







Cell cycle











214152_at
CCPG1
0.44
15q21.1
cell cycle progression 1


223569_at
PPAPDC1B
0.42
8p12
phosphatidic acid phosphatase type 2






domain containing 1B


223195_s_at
SESN2
0.47
1p35.3
sestrin 2







Metabolism











231202_at
ALDH1L2
0.45
12q23.3
aldehyde dehydrogenase 1 family; member






L2


219572_at
CADPS2
0.37

Ca2+-dependent activator protein for






secretion 2


212816_s_at
CBS
0.41
21q22.3
cystathionine-beta-synthase


218923_at
CTBS
0.42
1p22
chitobiase; di-N-acetyl-


201791_s_at
DHCR7
0.45
11q13.2-
7-dehydrocholesterol reductase





q13.5


204646_at
DPYD
0.48
1p22
dihydropyrimidine dehydrogenase


203397_s_at
GALNT3
0.65
2q24-q31
UDP-N-acetyl-alpha-D-






galactosamine: polypeptide N-






acetylgalactosaminyltransferase 3






(GalNAc-T3)


203157_s_at
GLS
0.42
2q32-q34
glutaminase


226183_at
GSK3B
0.49
3q13.3
Glycogen synthase kinase 3 beta


1555037_a_at
IDH1
0.53
2q33.3
isocitrate dehydrogenase 1 (NADP+);






soluble


201625_s_at
INSIG1
0.34
7q36
insulin induced gene 1


221760_at
MAN1A1
0.43
6q22
Mannosidase; alpha; class 1A; member 1


222805_at
MANEA
0.51
6q16.1
mannosidase; endo-alpha


232488_at
MGC15875
0.55
5q35.3
hypothetical protein MGC15875


225520_at
MTHFD1L
0.41
6q25.1
methylenetetrahydrofolate dehydrogenase






(NADP+ dependent) 1-like


202847_at
PCK2
0.47
14q11.2
phosphoenolpyruvate carboxykinase 2






(mitochondrial)


221788_at
PGM3
0.49
6q14.1-q15
Phosphoglucomutase 3


201397_at
PHGDH
0.36
1p12
phosphoglycerate dehydrogenase


205194_at
PSPH
0.38
7p15.2-
phosphoserine phosphatase





p15.1


200831_s_at
SCD
0.39
10q23-q24
stearoyl-CoA desaturase (delta-9-






desaturase)


209610_s_at
SLC1A4
0.54
2p15-p13
solute carrier family 1 (glutamate/neutral






amino acid transporter); member 4


208916_at
SLC1A5
0.40
19q13.3
solute carrier family 1 (neutral amino acid






transporter); member 5


209921_at
SLC7A11
0.41
4q28-q32
solute carrier family 7; (cationic amino






acid transporter; y+ system) member 11







Protein binding











215930_s_at
CTAGE5
0.30
14q13.3
CTAGE family; member 5


1554462_a_at
DNAJB9
0.42
7q31|14q24.2-
DnaJ (Hsp40) homolog; subfamily B;





q24.3
member 9


222294_s_at
EIF2C2
0.52
8q24
Eukaryotic translation initiation factor 2C;






2


235745_at
ERN1
0.35
17q24.2
endoplasmic reticulum to nucleus






signalling 1


219118_at
FKBP11
0.38
12q13.12
FK506 binding protein 11; 19 kDa


218361_at
GOLPH3L
0.45
1q21.2
golgi phosphoprotein 3-like


224763_at
RPL37
0.35
5p13
ribosomal protein L37


201915_at
SEC63
0.51
6q21
SEC63-like (S. cerevisiae)


202061_s_at
SEL1L
0.56
14q24.3-
sel-1 suppressor of lin-12-like (C. elegans)





q31


217790_s_at
SSR3
0.40
3q25.31
signal sequence receptor; gamma






(translocon-associated protein gamma)


202558_s_at
STCH
0.47
21q11.1|21q11
stress 70 protein chaperone; microsome-






associated; 60 kDa


222116_s_at
TBC1D16
0.70
17q25.3
TBC1 domain family; member 16


218145_at
TRIB3
0.41
20p13-
tribbles homolog 3 (Drosophila)





p12.2







Nuclear proteins and transcription factors











227558_at
CBX4
0.49
17q25.3
chromobox homolog 4 (Pc class homolog;







Drosophila)



219551_at
EAF2
0.46
3q13.33
ELL associated factor 2


226982_at
ELL2
0.45
5q15
elongation factor; RNA polymerase II; 2


202146_at
IFRD1
0.51
7q22-q31
interferon-related developmental regulator






1


214179_s_at
NFE2L1
0.59
17q21.3
nuclear factor (erythroid-derived 2)-like 1


213878_at
RECQL
0.44
12p12
RecQ protein-like (DNA helicase Q1-like)


208763_s_at
TSC22D3
0.53
Xq22.3
TSC22 domain family; member 3


225382_at
ZNF275
0.35
Xq28
zinc finger protein 275


227132_at
ZNF706
0.49
8q22.3
zinc finger protein 706







Others











227755_at

0.53

CDNA FLJ42435 fis; clone






BLADE2006849


208725_at
EIF2S2
0.46

eukaryotic translation initiation factor 2,






subunit 2 beta, Full-length cDNA clone






CS0DD001YD20 of Neuroblastoma Cot






50-normalized of Homo sapiens (human)


244623_at

0.39

Transcribed locus


226719_at

0.53

CDNA FLJ34899 fis; clone






NT2NE2018594


230570_at

0.37

Transcribed locus


229969_at

0.41

Transcribed locus; moderately similar to






XP_508230.1 PREDICTED: zinc finger






protein 195 [Pan troglodytes]


223136_at
AIG1
0.41
6q24.2
androgen-induced 1


222545_s_at
C10orf57
0.56
10q22.3
chromosome 10 open reading frame 57


220755_s_at
C6orf48
0.49
6p21.3
chromosome 6 open reading frame 48


219802_at
FLJ22028
0.44
12p12.1
hypothetical protein FLJ22028


212633_at
KIAA0776
0.49
6q16.1
KIAA0776


229090_at
LOC220930
0.45
10p11.23
hypothetical protein LOC220930
















SUPPLEMENTARY TABLE S5







Prognostic value of decitabine deregulated genes


in primary MMC of newly-diagnosed patients












Ajusted
Hazard


Probeset
NAME
P value
ratio










Bad prognostic genes










209417_s_at
IFI35
.03
1.84


221658_s_at
IL21R
.04
1.84


1552701_a_at
COP1
.02
1.95


223129_x_at
MYLIP
.01
1.99


214179_s_at
NFE2L1
.04
2.00


204079_at
TPST2
.03
2.01


205483_s_at
G1P2
.01
2.02


33322_i_at
SFN
.03
2.05


222690_s_at
TMEM39A
.02
2.20


209118_s_at
TUBA3
.01
2.21


208012_x_at
SP110
.01
2.22


213878_at
RECQL
.01
2.22


228531_at
SAMD9
.005
2.25


203397_s_at
GALNT3
.004
2.31


209969_s_at
STAT1
.003
2.37


202411_at
IFI27
.006
2.39


201625_s_at
INSIG1
.02
2.50


205552_s_at
OAS1
.01
2.50


204411_at
KIF21B
.0005
2.88


209083_at
CORO1A
.00008
3.05


218543_s_at
PARP12
.00005
3.52


205718_at
ITGB7
.00001
4.03







Good prognostic genes










221788_at
PGM3
.0003
.29


229969_at
SEC63
.0003
.29


224763_at
RPL37
.0006
.32


232488_at
MGC15875
.0007
.35


202558_s_at
STCH
.04
.39


219118_at
FKBP11
.03
.41


223569_at
PPAPDC1B
.01
.42


220755_s_at
C6orf48
.04
.43


202304_at
FNDC3A
.009
.44


1554462_a_at
DNAJB9
.02
.45


227755_at
EST, GenBank AA042983
.01
.45


226183_at
GSK3B
.02
.46


209122_at
ADFP
.01
.47


219551_at
EAF2
.03
.47


224451_x_at
ARHGAP9
.03
.48


210387_at
HIST1H2BG
.01
.49


211990_at
HLA-DPA1
.01
.49


202061_s_at
SEL1L
.04
.50


214152_at
CCPG1
.02
.50


226982_at
ELL2
.03
.50


222294_s_at
EIF2C2
.02
.52


228831_s_at
GNG7
.04
.52


230570_at
EST, GenBank AI702465
.04
.52


208725_at
EIF2S2
.04
.54


222838_at
SLAMF7
.04
.55
















SUPPLEMENTARY TABLE S6







Cox univariate and multivariate analysis


of OS in HM and TT2 patients' cohorts.










HM Cohort
TT2 Cohort



OAS
OAS













Pronostic
Proportional

Proportional




variable
hazard ratio
P-value
hazard ratio
P-value
















Univariate
DM Score
7.12
<.0001
2.06
<.0001


COX
β2m
1.1
<.0001
NA
NA


analysis -
ISS
1.73
.001
NA
NA


Overall
HRS
2.37
.01
4.67
<.0001


survival
IFM score
3.09
.0001
1.78
.004



t(4; 14)
2.14
.001
2.21
.001



del17p
3.44
.02
2.46
<.0001



GPI
2.21
.0001
1.75
<.0001


Multivariate
DM Score
5.91
<.0001
NA
NA


COX
ISS
1.40
NS
NA
NA


analysis -
DM Score
6.31
<.0001
NA
NA


Overall
β2m
1.1
.017
NA
NA


survival
DM Score
6.68
<.0001
1.46
NS



HRS
1.47
NS
4.02
<.0001



DM Score
6.80
<.0001
1.81
.006



IFM score
1.33
NS
1.37
NS



DM Score
6.28
<.0001
1.94
.001



t(4; 14)
1.88
NS
2.01
.003



DM Score
6.34
<.0001
2.00
<.0001



del17p
1.60
NS
2.32
.001



DM Score
5.97
<.0001
2.00
<.0001



GPI
1.47
NS
1.68
<.0001


Multivariate
DM Score
7.84
.005
1.40
NS


COX
β2m
1.1
NS
NA
NA


analysis -
ISS
1.37
NS
NA
NA


Overall
HRS
1.28
NS
3.66
<.0001


survival
IFM score
.62
NS
 .34
NS



t(4; 14)
2.58
.02
2.36
<.0001



del17p
1.72
NS
2.21
.002



GPI
1.61
NS
1.11
NS









The prognostic factors were tested as single variable or multi variables using Cox-model. P-values and the hazard ratios (HR) are shown. NS, Not significant at a 5% threshold; GPI, gene expression based proliferation index; ISS, International Staging System; HRS, high-risk score; IFM, Intergroupe Francophone du Myelome; NA, Not available.


EXAMPLE 2

In order to identify the minimal number of genes among the 47 genes used to calculate the DM score, the inventors used PAM (Prediction Analysis of Microarray) statistical technique used for class prediction from gene expression data using nearest shrunken centroids. 25 genes were identified and are depicted in Table B.












TABLE B







Probesets
Name









202061_s_at
SEL1L



202304_at
FNDC3A



202558_s_at
STCH



203397_s_at
GALNT3



204079_at
TPST2



205483_s_at
G1P2



205552_s_at
OAS1



205718_at
ITGB7



208725_at
EIF2S2



209083_at
CORO1A



209118_s_at
TUBA3



209122_at
ADFP



209417_s_at
IFI35



211990_at
HLA-DPA1



214152_at
CCPG1



214179_s_at
NFE2L1



219118_at
FKBP11



221658_s_at
IL21R



222690_s_at
TMEM39A



223129_x_at
MYLIP



224451_x_at
ARHGAP9



224763_at
RPL37



227755_at




228831_s_at
GNG7



232488_at
MGC15875










EXAMPLE 3
DNA Methylation Score is Predictive of Myeloma Cell Sensitivity to 5-Azacitidine

Epigenetics is characterized by a wide range of changes that are reversible and orchestrate gene expression. Recent studies of the epigenome have shown that epigenetic modifications play a role in cancer physiopathology including hematologic malignancies (Issa 2007, Issa, et at 2004, Oki, et al 2008, Smith, et al 2009). In MM, DNA hypomethylation was reported as the predominant early change during myelomagenesis that is gradually transformed to DNA hypermethylation in relapsed cases and during the progression of the disease (Heuck, et al 2013, Walker, et al 2010). Hypermethylation of GPX3, RBP1, SPARC and TGFBI genes was demonstrated to be associated with significantly shorter overall survival, independent of age, ISS score and adverse cytogenetics (Kaiser, et at 2013). DNA methylation is regulated by DNA methyltransferases (DNMT) (Hollenbach, et at 2010). Decitabine (5-aza-2′-deoxycytidine) or 5-azacytidine are both FDA (U.S. Food and Drug Administration)-approved DNMT inhibitors for the treatment of myelodysplastic syndrome (MDS) (Rodriguez-Paredes and Esteller 2011). 5-azacytidine is a ribonucleoside and Decitabine is a deoxyribonucleoside. Decitabine is incorporated only in DNA whereas 5-azacytidine incorporates both in DNA and RNA including rRNAs, tRNAs, mRNAs and miRNAs (Hollenbach, et al 2010). Once incorporated into DNA, 5-azacytidine and Decitabine will lead to DNMTs depletion, DNA hypomethylation and DNA damage induction (Flotho, et al 2009, Ghoshal, et al 2005, Hollenbach, et al 2010, Kiziltepe, et al 2007, Palii, et al 2008, Stresemann, et at 2006). Via incorporation into newly synthetized RNA, 5-azacytidine will alterate the processing of RNAs inhibiting protein synthesis (Hollenbach, et al 2010). The inventors have recently reported the building of a DNA methylation score (DM Score) predicting for the efficacy of decitabine to kill MM cells (MMCs). Given that 5-azacitidine can incorporate in both DNA and RNA and block DNA methylation and RNA traduction. The inventors have currently investigated whether DM Score could also predict for MMCs sensitivity to 5-azacitidine.


Material & Methods


Human Myeloma Cell Lines (HMCLs) and Primary Multiple Myeloma Cells of Patients.


Human myeloma cell lines XG-1, XG-2, XG-5, XG-6, XG-12, XG-13, XG-16, XG-19, XG-20, RPMI8226, LP1 and SKMM2 (HMCLs, N=12) were obtained as previously described (Gu, et al 2000, Moreaux, et al 2011, Rebouissou, et al 1998, Tarte, et al 1999, Zhang, et al 1994) or purchased from DSMZ and American Type Culture Collection. These HMCLs were extensively phenotypically and molecularly characterized as described (Moreaux, et al 2011). Microarray data are deposited in the ArrayExpress public database (accession numbers E-TABM-937 and E-TABM-1088). Bone marrow of patients presenting with previously untreated multiple myeloma (n=14) at the university hospital of Montpellier were obtained after patients' written informed consent in accordance with the Declaration of Helsinki and agreement of the Montpellier University Hospital Center for Biological Resources. MMCs were purified as previously published (Mahtouk, et al 2004) and whole genome gene expression profiling assayed with Affymetrix U133 2.0 plus microarrays (Affymetrix, Santa Clara, Calif., USA) (De Vos, et al 2002). Gene expression data were analyzed using the inventor's bioinformatics platforms (http://rage.montp.inserm.fr/ and http.//amazonia.montp.inserm.fr/) (Reme, et al 2008, Tanguy Le Carrour 2010) and computations performed using R 2.15.1 (http://www.r-project.org/) and bioconductor 2.0 (http://www.bioconductor.org).


Sensitivity of Myeloma Cell Lines and Primary Myeloma Cells to 5-Azacitidine.


HMCLs were cultured with graded 5-azacitidine (Sigma, St. Louis, Mo.) concentrations. HMCLs cell growth was quantified with a Cell Titer Glo Luminescent Assay (Promega, Madison, Wis.) and half inhibitory concentration (IC50) was determined using GraphPad Prism (http://www.graphpad.com/scientific-software/prism/).


Primary myeloma cells of 14 patients were cultured with or without graded concentrations of 5-azacitidine and MMC cytotoxicity evaluated using anti-CD138-PE mAb (Immunotech, Marseille, France) as described (Jourdan, et al 1998; Mahtouk, et al 2004; Moreaux, et al 2012).


Gene Set Enrichment Analysis (GSEA)


The inventors compared the gene expression levels from high DM Score versus low DM Score patients and picked up the genes which had significant different expression for Gene set enrichment analysis (GSEA). Gene set enrichment analysis was carried out by computing overlaps with canonical pathways and gene ontology gene sets obtained from the Broad Institute.


Results and Discussion


The efficacy of DM Score to predict MMCs sensitivity to 5-azacitidine was investigated on 12 HMCLs with high or low DM Score. The 6 HMCLs with high DM Score exhibited a 3 fold higher 5-azacitidine sensitivity (P=0.01; median IC50=2.43 μM; range: 1.12 to 8.25 μM) compared to the 6 ones with a low DM Score (median IC50=7.45 μM; range: 5.73 to 27.9) (FIG. 5). DM score could also predict for the ability of 5-azacytidine to kill patients' primary MMCs cultured together with their BM environment. The MMCs of patients with a high DM Score (N=7) exhibited a significant 1.6 fold higher 5-azacytidine sensitivity compared to low DM Score patients (N=7) (P<0.05, FIG. 6).


Thus, the DM Score, which was built using 47 genes whose expression is deregulated by decitabine in HMCLs and which have prognostic value for patients overall survival, can predict for the sensitivity of MM cell lines and primary MMCs to the two-clinical grade inhibitors of DNMT. These 47 genes include 22 genes associated with a bad prognosis and 25 with good one. Using GSEA analysis, MMCs of patients with a high DM Score show a significant enrichment in genes associated with proliferation (gene sets: REACTOME CELL CYCLE, CELL CYCLE G2 M and PLASMA CELLS VS PLASMABLAST DN, P<0.001, and supplementary Tables S7, S8 and S9). On the other hand, MMCs of patients with a low DM Score show a significant enrichment in genes coding for solute carrier group of membrane transport proteins (gene sets: REACTOME TRANSPORT OF INORGANIC CATIONS ANIONS AND AMINO ACIDS OLIGOPEPTIDES, P<0.001, and supplementary Tables S10).


Thus, the higher sensitivity of MMCs of patients with a high DM Score to DNMT inhibitors could be explained by the fact that these inhibitors are mainly active in cell cycling cells since incorporation into DNA is restricted to the S-phase (Hollenbach, et al 2010), and also by a reduced drug export in MMCs of these patients, resulting in higher intracellular drug accumulation.


Clinical trials evaluate the safety of DNMTi as monotherapy or in combination with lenalidomide or dexamethasone in MM (Maes, et al 2013; Toor, et al 2012) and investigate the link between HM Score and response of patients to DNMTi could be promising.


In conclusion, the DM Score allows identification of MM patients who could benefit from treatment with two clinical grade DNMT inhibitors and the development of personalized treatment.









TABLE S7







Genes set enrichment analysis revealed a significant overrepresentation of the REACTOME


CELL CYCLEset in high DM Score patients compared to low DM Score patients (P < .001).















RANK
RUNNING




GENE

METRIC
Enrichment
CORE


PROBE
SYMBOL
GENE_TITLE
SCORE
Score
ENRICHMENT















MYBL2
MYBL2
v-myb myeloblastosis
−0.167122021317482
−0.6420408
Yes




viral oncogene homolog




(avian)-like 2


CDKN2C
CDKN2C
cyclin-dependent kinase
−0.16787339746952057
−0.626822
Yes




inhibitor 2C (p18, inhibits




CDK4)


CDK6
CDK6
cyclin-dependent kinase 6
−0.19776728749275208
−0.6317126
Yes


LMNA
LMNA
lamin A/C
−0.20474471151828766
−0.61800563
Yes


CDKN2B
CDKN2B
cyclin-dependent kinase
−0.22542211413383484
−0.6069075
Yes




inhibitor 2B (p15, inhibits




CDK4)


TYMS
TYMS
thymidylate synthetase
−0.22692811489105225
−0.5853221
Yes


MCM10
MCM10
MCM10
−0.23174770176410675
−0.56442255
Yes




minichromosome




maintenance deficient 10




(S. cerevisiae)


BUB1B
BUB1B
BUB1 budding
−0.2420835942029953
−0.54413825
Yes




uninhibited by




benzimidazoles 1




homolog beta (yeast)


KIF23
KIF23
kinesin family member 23
−0.25110968947410583
−0.52252656
Yes


CHEK1
CHEK1
CHK1 checkpoint
−0.2538430988788605
−0.49904326
Yes




homolog (S. pombe)


HIST1H4H
HIST1H4H
histone cluster 1, H4h
−0.27717286348342896
−0.47906303
Yes


MCM2
MCM2
MCM2 minichromosome
−0.29232123494148254
−0.45348957
Yes




maintenance deficient 2,




mitotin (S. cerevisiae)


GINS1
GINS1
GINS complex subunit 1
−0.29381972551345825
−0.42524388
Yes




(Psf1 homolog)


KIF20A
KIF20A
kinesin family member
−0.30122432112693787
−0.3985847
Yes




20A


MCM4
MCM4
MCM4 minichromosome
−0.3015007972717285
−0.36983046
Yes




maintenance deficient 4




(S. cerevisiae)


OIP5
OIP5
Opa interacting protein 5
−0.30586564540863037
−0.3406566
Yes


CCND2
CCND2
cyclin D2
−0.31117963790893555
−0.31189123
Yes


RRM2
RRM2
ribonucleotide reductase
−0.3157253563404083
−0.28176954
Yes




M2 polypeptide


BIRC5
BIRC5
baculoviral IAP repeat-
−0.3474469482898712
−0.2515862
Yes




containing 5 (survivin)


AURKA
AURKA
aurora kinase A
−0.35163724422454834
−0.21778236
Yes


CENPA
CENPA
centromere protein A
−0.35916951298713684
−0.1837141
Yes


CCNB2
CCNB2
cyclin B2
−0.36041226983070374
−0.1490667
Yes


HIST1H4C
HIST1H4C
histone cluster 1, H4c
−0.37045058608055115
−0.11368413
Yes


CCNB1
CCNB1
cyclin B1
−0.38353657722473145
−0.077273406
Yes


CDCA8
CDCA8
cell division cycle
−0.39406412839889526
−0.039620794
Yes




associated 8


NEK2
NEK2
NIMA (never in mitosis
−0.4169292151927948
2.2989404E−4
Yes




gene a)-related kinase 2
















SUPPLEMENTARY TABLE S8







Genes set enrichment analysis revealed a significant overrepresentation of the CELL


CYCLE G2/M set in high DM Score patients compared to low DM Score patients (P < .001).















RANK
RUNNING




GENE

METRIC
Enrichment
CORE


PROBE
SYMBOL
GENE_TITLE
SCORE
Score
ENRICHMENT















CENPE
CENPE
centromere protein E,
−0.15664561092853546
−0.6155994
Yes




312 kDa


BMP2
BMP2
bone morphogenetic
−0.16032224893569946
−0.6001657
Yes




protein 2


TACC3
TACC3
transforming, acidic
−0.18774476647377014
−0.6039366
Yes




coiled-coil containing




protein 3


PRR5
PRR5
proline rich 5 (renal)
−0.18954475224018097
−0.582728
Yes


CENPF
CENPF
centromere protein F,
−0.19975358247756958
−0.5653478
Yes




350/400ka (mitosin)


LMNA
LMNA
lamin A/C
−0.20474471151828766
−0.5455384
Yes


TPX2
TPX2
TPX2, microtubule-
−0.22465506196022034
−0.52863073
Yes




associated, homolog




(Xenopus laevis)


SHCBP1
SHCBP1
SHC SH2-domain
−0.23987287282943726
−0.5050944
Yes




binding protein 1


KIF14
KIF14
kinesin family member
−0.2565326690673828
−0.48026097
Yes




14


DEPDC1B
DEPDC1B
DEP domain containing
−0.2718331515789032
−0.4513114
Yes




1B


HMMR
HMMR
hyaluronan-mediated
−0.2835068106651306
−0.42005372
Yes




motility receptor




(RHAMM)


DEPDC1
DEPDC1
DEP domain containing
−0.28364425897598267
−0.3862573
Yes




1


ANLN
ANLN
anillin, actin binding
−0.286072313785553
−0.35217157
Yes




protein


HMGB3
HMGB3
high-mobility group
−0.28622567653656006
−0.31806755
Yes




box 3


MCM4
MCM4
MCM4
−0.3015007972717285
−0.286271
Yes




minichromosome




maintenance deficient 4




(S. cerevisiae)


FOXM1
FOXM1
forkhead box M1
−0.3144078850746155
−0.25064352
Yes


CEP55
CEP55
centrosomal protein
−0.3161003887653351
−0.21320921
Yes




55 kDa


BIRC5
BIRC5
baculoviral IAP repeat-
−0.3474469482898712
−0.17479162
Yes




containing 5 (survivin)


AURKA
AURKA
aurora kinase A
−0.35163724422454834
−0.13289377
Yes


CENPA
CENPA
centromere protein A
−0.35916951298713684
−0.09055706
Yes


CCNB2
CCNB2
cyclin B2
−0.36041226983070374
−0.047613665
Yes


NEK2
NEK2
NIMA (never in mitosis
−0.4169292151927948
2.2932523E−4
Yes




gene a)-related kinase 2
















SUPPLEMENTARY TABLE S9







Genes set enrichment analysis revealed a significant overrepresentation of the PLASMA CELLS


VS PLASMABLAST set in high DM Score patients compared to low DM Score patients (P < .001).















RANK
RUNNING




GENE

METRIC
Enrichment
CORE


PROBE
SYMBOL
GENE_TITLE
SCORE
Score
ENRICHMENT















CLEC2B
CLEC2B
C-type lectin domain family 2,
−0.1298338621854782
−0.6410949
Yes




member B


PLK4
PLK4
polo-like kinase 4 (Drosophila)
−0.13095927238464355
−0.62984425
Yes


RNASE6
RNASE6
ribonuclease, RNase A family,
−0.13925980031490326
−0.62673587
Yes




k6


IFI30
IFI30
interferon, gamma-inducible
−0.1412345916032791
−0.61493564
Yes




protein 30


GLIPR1
GLIPR1
GLI pathogenesis-related 1
−0.141678586602211
−0.6014841
Yes




(glioma)


CENPE
CENPE
centromere protein E, 312 kDa
−0.15664561092853546
−0.60218084
Yes


CD52
CD52
CD52 molecule
−0.15694235265254974
−0.58677125
Yes


ITGB1
ITGB1
integrin, beta 1 (fibronectin
−0.1667313575744629
−0.5811952
Yes




receptor, beta polypeptide,




antigen CD29 includes MDF2,




MSK12)


PAPOLA
PAPOLA
poly(A) polymerase alpha
−0.19660720229148865
−0.5871552
Yes


GALNT3
GALNT3
UDP-N-acetyl-alpha-D-
−0.19789130985736847
−0.56841403
Yes




galactosamine: polypeptide N-




acetylgalactosaminyltransferase




3 (GalNAc-T3)


CD58
CD58
CD58 molecule
−0.21272079646587372
−0.5571741
Yes


GLDC
GLDC
glycine dehydrogenase
−0.22228969633579254
−0.5404011
Yes




(decarboxylating)


TYMS
TYMS
thymidylate synthetase
−0.22692811489105225
−0.5197276
Yes


GZMB
GZMB
granzyme B (granzyme 2,
−0.2282218039035797
−0.49754903
Yes




cytotoxic T-lymphocyte-




associated serine esterase 1)


ITGB7
ITGB7
integrin, beta 7
−0.24833402037620544
−0.47867823
Yes


KIF23
KIF23
kinesin family member 23
−0.25110968947410583
−0.45517108
Yes


KIF14
KIF14
kinesin family member 14
−0.2565326690673828
−0.43227983
Yes


PFKP
PFKP
phosphofructokinase, platelet
−0.2628379166126251
−0.40693212
Yes


HMMR
HMMR
hyaluronan-mediated motility
−0.2835068106651306
−0.38460782
Yes




receptor (RHAMM)


GINS1
GINS1
GINS complex subunit 1 (Psf1
−0.29381972551345825
−0.35759616
Yes




homolog)


SELL
SELL
selectin L (lymphocyte adhesion
−0.2999555468559265
−0.32998204
Yes




molecule 1)


TOP2A
TOP2A
topoisomerase (DNA) II alpha
−0.30873504281044006
−0.3015059
Yes




170 kDa


CDKN3
CDKN3
cyclin-dependent kinase
−0.30915674567222595
−0.27138063
Yes




inhibitor 3 (CDK2-associated




dual specificity phosphatase)


CCND2
CCND2
cyclin D2
−0.31117963790893555
−0.24082708
Yes


FOXM1
FOXM1
forkhead box M1
−0.3144078850746155
−0.20995656
Yes


RRM2
RRM2
ribonucleotide reductase M2
−0.3157253563404083
−0.17895667
Yes




polypeptide


CASP6
CASP6
caspase 6, apoptosis-related
−0.34289273619651794
−0.14781573
Yes




cysteine peptidase


CENPA
CENPA
centromere protein A
−0.35916951298713684
−0.11392827
Yes


S100A10
S100A10
S100 calcium binding protein
−0.37857523560523987
−0.077676095
Yes




A10


CCNB1
CCNB1
cyclin B1
−0.38353657722473145
−0.040018078
Yes


NEK2
NEK2
NIMA (never in mitosis gene
−0.4169292151927948
2.296113E−4
Yes




a)-related kinase 2
















SUPPLEMENTARY TABLE S10







Genes set enrichment analysis revealed a significant overrepresentation of the


REACTOME TRANSPORT OF INORGANIC CATIONS ANIONS AND AMINO ACID OLIGOPEPTIDES set


in low DM Score patients compared to high DM Score patients (P < .001).















RANK
RUNNING




GENE

METRIC
Enrichment
CORE


PROBE
SYMBOL
GENE_TITLE
SCORE
Score
ENRICHMENT















SLC24A3
SLC24A3
solute carrier family 24
0.18617968261241913
0.03475809
Yes




(sodium/potassium/calcium




exchanger), member 3


SLC17A8
SLC17A8
solute carrier family 17
0.17720697820186615
0.09601015
Yes




(sodium-dependent inorganic




phosphate cotransporter),




member 8


SLC7A8
SLC7A8
solute carrier family 7 (cationic
0.17013469338417053
0.15928961
Yes




amino acid transporter, y+




system), member 8


SLC1A1
SLC1A1
solute carrier family 1
0.15042848885059357
0.19263873
Yes




(neuronal/epithelial high




affinity glutamate transporter,




system Xag), member 1


SLC4A1
SLC4A1
solute carrier family 4, anion
0.134476438164711
0.21448465
Yes




exchanger, member 1




(erythrocyte membrane protein




band 3, Diego blood group)


SLC12A5
SLC12A5
solute carrier family 12,
0.13315615057945251
0.26718774
Yes




(potassium-chloride transporter)




member 5


SLC9A3
SLC9A3
solute carrier family 9
0.1316232681274414
0.3169685
Yes




(sodium/hydrogen exchanger),




member 3


SLC15A2
SLC15A2
solute carrier family 15
0.11438794434070587
0.3207876
Yes




(H+/peptide transporter),




member 2


SLC4A4
SLC4A4
solute carrier family 4, sodium
0.10706888139247894
0.34565148
Yes




bicarbonate cotransporter,




member 4


SLC12A1
SLC12A1
solute carrier family 12
0.106844961643219
0.3904528
Yes




(sodium/potassium/chloride




transporters), member 1


SLC24A5
SLC24A5
solute carrier family 24,
0.09523425251245499
0.39576042
Yes




member 5


SLC8A1
SLC8A1
solute carrier family 8
0.09496445208787918
0.43509954
Yes




(sodium/calcium exchanger),




member 1


SLC1A3
SLC1A3
solute carrier family 1 (glial
0.09355431795120239
0.47225094
Yes




high affinity glutamate




transporter), member 3


SLC8A3
SLC8A3
solute carrier family 8 (sodium-
0.08295272290706635
0.47784585
Yes




calcium exchanger), member 3









REFERENCES

Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.


1. Hahn W C, Weinberg R A. Rules for making human tumor cells. N Engl J Med. 2002; 347(20):1593-1603.


2. Vogelstein B, Kinzler K W. Cancer genes and the pathways they control. Nat Med. 2004; 10(8):789-799.


3. Baylin S B. DNA methylation and gene silencing in cancer. Nat Clin Pract Oncol. 2005; 2 Suppl 1:S4-11.


4. Kondo Y. Epigenetic cross-talk between DNA methylation and histone modifications in human cancers. Yonsei Med J. 2009; 50(4):455-463.


5. Issa J P. DNA methylation as a therapeutic target in cancer. Clin Cancer Res. 2007; 13(6): 1634-1637.


6. Issa J P, Garcia-Manero G, Giles F J, et al. Phase 1 study of low-dose prolonged exposure schedules of the hypomethylating agent 5-aza-2′-deoxycytidine (decitabine) in hematopoietic malignancies. Blood. 2004; 103(5):1635-1640.


7. Oki Y, Jelinek J, Shen L, Kantarjian H M, Issa J P. Induction of hypomethylation and molecular response after decitabine therapy in patients with chronic myelomonocytic leukemia. Blood. 2008; 111(4):2382-2384.


8. Smith E M, Boyd K, Davies F E. The potential role of epigenetic therapy in multiple myeloma. Br J Haematol. 2009.


9. Bergsagel P L, Kuehl W M. Molecular pathogenesis and a consequent classification of multiple myeloma. J Clin Oncol. 2005; 23(26):6333-6338.


10. Hideshima T, Bergsagel P L, Kuehl W M, Anderson K C. Advances in biology of multiple myeloma: clinical applications. Blood. 2004; 104(3):607-618.


11. Chen G, Wang Y, Huang H, et al. Combination of DNA methylation inhibitor 5-azacytidine and arsenic trioxide has synergistic activity in myeloma. Eur J Haematol. 2009; 82(3): 176-183.


12. de Carvalho F, Colleoni G W, Almeida M S, Carvalho A L, Vettore A L. TGFbetaR2 aberrant methylation is a potential prognostic marker and therapeutic target in multiple myeloma. Int J Cancer. 2009; 125(8):1985-1991.


13. Galm O, Yoshikawa H, Esteller M, Osieka R, Herman J G. SOCS-1, a negative regulator of cytokine signaling, is frequently silenced by methylation in multiple myeloma. Blood. 2003; 101(7):2784-2788.


14. Hatzimichael E, Dranitsaris G, Dasoula A, et al. Von Hippel-Lindau methylation status in patients with multiple myeloma: a potential predictive factor for the development of bone disease. Clin Lymphoma Myeloma. 2009; 9(3):239-242.


15. Hodge D R, Peng B, Cherry J C, et al. Interleukin 6 supports the maintenance of p53 tumor suppressor gene promoter methylation. Cancer Res. 2005; 65(11):4673-4682.


16. Ng M H, Chung Y F, Lo K W, Wickham N W, Lee J C, Huang D P. Frequent hypermethylation of p16 and p15 genes in multiple myeloma. Blood. 1997; 89(7):2500-2506.


17. Seidl S, Ackermann J, Kaufmann H, et al. DNA-methylation analysis identifies the E-cadherin gene as a potential marker of disease progression in patients with monoclonal gammopathies. Cancer. 2004; 100(12):2598-2606.


18. Tshuikina M, Jernberg-Wiklund H, Nilsson K, Oberg F. Epigenetic silencing of the interferon regulatory factor ICSBP/IRF8 in human multiple myeloma. Exp Hematol. 2008; 36(12):1673-1681.


19. Heller G, Schmidt W M, Ziegler B, et al. Genome-wide transcriptional response to 5-aza-2′-deoxycytidine and trichostatin a in multiple myeloma cells. Cancer Res. 2008; 68(1):44-54.


20. Wilop S, van Gemmeren T B, Lentjes M H, et al. Methylation-associated dysregulation of the suppressor of cytokine signaling-3 gene in multiple myeloma. Epigenetics. 2011; 6(8):1047-1052.


21. Walker B A, Wardell C P, Chiecchio L, et al. Aberrant global methylation patterns affect the molecular pathogenesis and prognosis of multiple myeloma. Blood. 2010.


22. Moreaux J, Klein B, Bataille R, et al. A high-risk signature for patients with multiple myeloma established from the molecular classification of human myeloma cell lines. Haematologica. 2011; 96(4):574-582.


23. Zhang X G, Gaillard J P, Robillard N, et al. Reproducible obtaining of human myeloma cell lines as a model for tumor stem cell study in human multiple myeloma. Blood. 1994; 83(12):3654-3663.


24. Rebouissou C, Wijdenes J, Autissier P, et al. A gp130 interleukin-6 transducer-dependent SCID model of human multiple myeloma. Blood. 1998;91(12):4727-4737.


25. Tarte K, Zhang X G, Legouffe E, et al. Induced expression of B7-1 on myeloma cells following retroviral gene transfer results in tumor-specific recognition by cytotoxic T cells. J Immunol. 1999; 163(1):514-524.


26. Gu Z J, Vos J D, Rebouissou C, et al. Agonist anti-gp130 transducer monoclonal antibodies are human myeloma cell survival and growth factors. Leukemia. 2000; 14(1):188-197.


27. Goldschmidt H, Sonneveld P, Cremer F W, et al. Joint HOVON-50/GMMG-HD3 randomized trial on the effect of thalidomide as part of a high-dose therapy regimen and as maintenance treatment for newly diagnosed myeloma patients. Ann Hematol. 2003; 82(10):654-659.


28. Cremer F W, Bila J, Buck I, et al. Delineation of distinct subgroups of multiple myeloma and a model for clonal evolution based on interphase cytogenetics. Genes Chromosomes Cancer. 2005; 44(2):194-203.


29. Barlogie B, Tricot G, Rasmussen E, et al. Total therapy 2 without thalidomide in comparison with total therapy 1: role of intensified induction and posttransplantation consolidation therapies. Blood. 2006; 107(7):2633-2638.


30. Sprynski A C, Hose D, Caillot L, et al. The role of IGF-1 as a major growth factor for myeloma cell lines and the prognostic relevance of the expression of its receptor. Blood. 2009; 113(19):4614-4626.


31. Xiong W, Wu X, Starnes S, et al. An analysis of the clinical and biologic significance of TP53 loss and the identification of potential novel transcriptional targets of TP53 in multiple myeloma. Blood. 2008; 112(10):4235-4246.


32. Moreaux J, Legouffe E, Jourdan E, et al. BAFF and APRIL protect myeloma cells from apoptosis induced by interleukin 6 deprivation and dexamethasone. Blood. 2004; 103(8):3148-3157.


33. Hose D, Reme T, Meissner T, et al Inhibition of aurora kinases for tailored risk-adapted treatment of multiple myeloma. Blood. 2009; 113(18):4331-4340.


34. Moreaux J, Cremer F W, Reme T, et al. The level of TACI gene expression in myeloma cells is associated with a signature of microenvironment dependence versus a plasmablastic signature. Blood. 2005; 106(3):1021-1030.


35. Reme T, Hose D, De Vos J, et al. A new method for class prediction based on signed-rank algorithms applied to Affymetrix microarray experiments. BMC Bioinformatics. 2008; 9:16.


36. Tanguy Le Carrour S A, Sylvie Tondeur, Ludovic Lhermitte, Ned Lamb, Thierry Reme, Veronique Pantesco, Samir Hamamah, Bernard Klein, John De Vos. Amazonia!: An Online Resource to Google and Visualize Public Human whole Genome Expression Data. The Open Bioinformatics Journal. 2010; 4:5-10.


37. Cui X, Churchill G A. Statistical tests for differential expression in cDNA microarray experiments. Genome Biol. 2003; 4(4):210.


38. Kassambara A, Hose D, Moreaux J, et al. Genes with a spike expression are clustered in chromosome (sub)bands and spike (sub)bands have a powerful prognostic value in patients with multiple myeloma. Haematologica. 2011.


39. Zhan F, Huang Y, Colla S, et al. The molecular classification of multiple myeloma. Blood. 2006; 108(6):2020-2028.


40. Chu W K, Hickson I D. RecQ helicases: multifunctional genome caretakers. Nat Rev Cancer. 2009; 9(9):644-654.


41. Harrigan J A, Bohr V A. Human diseases deficient in RecQ helicases. Biochimie. 2003; 85(11): 1185-1193.


42. Hickson I D. RecQ helicases: caretakers of the genome. Nat Rev Cancer. 2003; 3(3):169-178.


43. Arai A, Chano T, Futami K, et al. RECQL1 and WRN proteins are potential therapeutic targets in head and neck squamous cell carcinoma. Cancer Res. 2011; 71(13):4598-4607.


44. Futami K, Kumagai E, Makino H, et al. Induction of mitotic cell death in cancer cells by small interference RNA suppressing the expression of RecQL1 helicase. Cancer Sci. 2008; 99(1):71-80.


45. Futami K, Kumagai E, Makino H, et al. Anticancer activity of RecQL1 helicase siRNA in mouse xenograft models. Cancer Sci. 2008; 99(6):1227-1236.


46. Mendoza-Maldonado R, Faoro V, Bajpai S, et al. The human RECQ1 helicase is highly expressed in glioblastoma and plays an important role in tumor cell proliferation. Mol Cancer. 2011; 10:83.


47. Miki H, Okada Y, Hirokawa N. Analysis of the kinesin superfamily: insights into structure and function. Trends Cell Biol. 2005; 15(9):467-476.


48. Hirokawa N. Kinesin and dynein superfamily proteins and the mechanism of organelle transport. Science. 1998; 279(5350):519-526.


49. Yu Y, Feng Y M. The role of kinesin family proteins in tumorigenesis and progression: potential biomarkers and molecular targets for cancer therapy. Cancer. 2010; 116(22):5150-5160.


50. Zhu C, Zhao J, Bibikova M, et al. Functional analysis of human microtubule-based motor proteins, the kinesins and dyneins, in mitosis/cytokinesis using RNA interference. Mol Biol Cell. 2005; 16(7):3187-3199.


51. Avet-Loiseau H, Li C, Magrangeas F, et al. Prognostic significance of copy-number alterations in multiple myeloma. J Clin Oncol. 2009; 27(27):4585-4590.


52. Agnelli L, Forcato M, Ferrari F, et al. The reconstruction of transcriptional networks reveals critical genes with implications for clinical outcome of multiple myeloma. Clin Cancer Res. 2011; 17(23):7402-7412.


53. De Vos, J., Thykjaer, T., Tarte, K., Ensslen, M., Raynaud, P., Requirand, G., Pellet, F., Pantesco, V., Reme, T., Jourdan, M., Rossi, J. F., Orntoft, T. & Klein, B. (2002) Comparison of gene expression profiling between malignant and normal plasma cells with oligonucleotide arrays. Oncogene, 21, 6848-6857.


54. Flotho, C., Claus, R., Batz, C., Schneider, M., Sandrock, I., Ihde, S., Plass, C., Niemeyer, C. M. & Lubbert, M. (2009) The DNA methyltransferase inhibitors azacitidine, decitabine and zebularine exert differential effects on cancer gene expression in acute myeloid leukemia cells. Leukemia, 23, 1019-1028.


55. Ghoshal, K., Datta, J., Majumder, S., Bai, S., Kutay, H., Motiwala, T. & Jacob, S. T. (2005) 5-Aza-deoxycytidine induces selective degradation of DNA methyltransferase 1 by a proteasomal pathway that requires the KEN box, bromo-adjacent homology domain, and nuclear localization signal. Mol Cell Biol, 25, 4727-4741.


56. Heuck, C. J., Mehta, J., Bhagat, T., Gundabolu, K., Yu, Y., Khan, S., Chrysofakis, G., Schipke, C., Tariman, J., Vickrey, E., Pulliam, N., Nischal, S., Zhou, L., Bhattacharyya, S., Meagher, R., Hu, C., Maqbool, S., Suzuki, M., Parekh, S., Reu, F., Steidl, U., Greally, J., Verma, A. & Singhal, S. B. (2013) Myeloma is characterized by stage-specific alterations in DNA methylation that occur early during myelomagenesis. J Immunol, 190, 2966-2975.


57. Hollenbach, P. W., Nguyen, A. N., Brady, H., Williams, M., Ning, Y., Richard, N., Krushel, L., Aukerman, S. L., Heise, C. & MacBeth, K. J. (2010) A comparison of azacitidine and decitabine activities in acute myeloid leukemia cell lines. PLoS One, 5, e9001.


58. Jourdan, M., Ferlin, M., Legouffe, E., Horvathova, M., Liautard, J., Rossi, J. F., Wijdenes, J., Brochier, J. & Klein, B. (1998) The myeloma cell antigen syndecan-1 is lost by apoptotic myeloma cells. Br J Haematol, 100, 637-646.


59. Kaiser, M. F., Johnson, D. C., Wu, P., Walker, B. A., Brioli, A., Mirabella, F., Wardell, C. P., Melchor, L., Davies, F. E. & Morgan, G. J. (2013) Global methylation analysis identifies prognostically important epigenetically inactivated tumour suppressor genes in multiple myeloma. Blood.


60. Kiziltepe, T., Hideshima, T., Catley, L., Raje, N., Yasui, H., Shiraishi, N., Okawa, Y., Ikeda, H., Vallet, S., Pozzi, S., Ishitsuka, K., Ocio, E. M., Chauhan, D. & Anderson, K. C. (2007) 5-Azacytidine, a DNA methyltransferase inhibitor, induces ATR-mediated DNA double-strand break responses, apoptosis, and synergistic cytotoxicity with doxorubicin and bortezomib against multiple myeloma cells. Mol Cancer Ther, 6, 1718-1727.


61. Maes, K., Menu, E., Van Valckenborgh, E., Van Riet, I., Vanderkerken, K. & De Bruyne, E. (2013) Epigenetic Modulating Agents as a New Therapeutic Approach in Multiple Myeloma. Cancers, 5, 430-461.


62. Mahtouk, K., Jourdan, M., De Vos, J., Hertogh, C., Fiol, G., Jourdan, E., Rossi, J. F. & Klein, B. (2004) An inhibitor of the EGF receptor family blocks myeloma cell growth factor activity of HB-EGF and potentiates dexamethasone or anti-IL-6 antibody-induced apoptosis. Blood, 103, 1829-1837.


63. Moreaux, J., Reme, T., Leonard, W., Veyrune, J. L., Requirand, G., Goldschmidt, H., Hose, D. & Klein, B. (2012) Development of gene expression-based score to predict sensitivity of multiple myeloma cells to DNA methylation inhibitors. Mol Cancer Ther, 11, 2685-2692.


64. Palii, S. S., Van Emburgh, B. O., Sankpal, U. T., Brown, K. D. & Robertson, K. D. (2008) DNA methylation inhibitor 5-Aza-2′-deoxycytidine induces reversible genome-wide DNA damage that is distinctly influenced by DNA methyltransferases 1 and 3 B. Mol Cell Biol, 28, 752-771. 65. Rodriguez-Paredes, M. & Esteller, M. (2011) Cancer epigenetics reaches mainstream oncology. Nat Med, 17, 330-339.


66. Stresemann, C., Brueckner, B., Musch, T., Stopper, H. & Lyko, F. (2006) Functional diversity of DNA methyltransferase inhibitors in human cancer cell lines. Cancer Res, 66, 2794-2800. 67. Tanguy Le Carrour, S. A., Sylvie Tondeur, Ludovic Lhermitte, Ned Lamb, Thierry Reme, Veronique Pantesco, Samir Hamamah, Bernard Klein, John De Vos (2010) Amazonia!: An Online Resource to Google and Visualize Public Human whole Genome Expression Data. The Open Bioinformatics Journal, 4, 5-10.


68. Toor, A. A., Payne, K. K., Chung, H. M., Sabo, R. T., Hazlett, A. F., Kmieciak, M., Sanford, K., Williams, D. C., Clark, W. B., Roberts, C. H., McCarty, J. M. & Manjili, M. H. (2012) Epigenetic induction of adaptive immune response in multiple myeloma: sequential azacitidine and lenalidomide generate cancer testis antigen-specific cellular immunity. Br J Haematol, 158, 700-711.


69. Walker, B. A., Wardell, C. P., Chiecchio, L., Smith, E. M., Boyd, K. D., Neri, A., Davies, F. E., Ross, F. M. & Morgan, G. J. (2010) Aberrant global methylation patterns affect the molecular pathogenesis and prognosis of multiple myeloma. Blood.


70. Zhang, X. G., Gaillard, J. P., Robillard, N., Lu, Z. Y., Gu, Z. J., Jourdan, M., Boiron, J. M., Bataille, R. & Klein, B. (1994) Reproducible obtaining of human myeloma cell lines as a model for tumor stem cell study in human multiple myeloma. Blood, 83, 3654-3663.

Claims
  • 1. A method of testing whether a patient suffering of from multiple myeloma will respond or not to a DNA methyltransferase inhibitor (DNMTi) comprising: i.determining the expression level (ELi) of a plurality of genes G1-Gn selected from table A in a biological sample obtained from said patientii. comparing the expression level (ELi) determined at step i) with a predetermined reference level (ELRi)iii. calculating the DMS score trough the following formula
  • 2. A method for the treatment of multiple myeloma in a patient in need thereof comprising the steps of: a) testing whether the patient will respond or not to a DNA methyltransferase inhibitor (DNMTi) by performing the method according to claim 1b) administering the DNA methyltransferase inhibitor, if said patient has as a DMS score higher than the reference value DMSR.
  • 3. The method of claim 1, wherein at least 25 genes from Table A are determined, and wherein said 25 genes are: 202061_s_at SEL1L,202304_at FNDC3A,202558_s_at STCH,203397_s_at GALNT3,204079_at TPST2,205483_s_at G1P2,205552_s_at OAS1,205718_at ITGB7,208725_at EIF2S2,209083_at CORO1A,209118_s_at TUBA3,209122_at ADFP,209417_s_at IF135,211990_at HLA-DPA1,214152_at CCPG1,214179_s_at NFE2L1,219118_at FKBP11,221658_s_at IL21R,222690_s_at TMEM39A,223129_x_at MYLIP,224451_x_at ARHGAP9,224763_at RPL37,227755_at EST, GenBank AA042983,228831_s_at GNG7 and232488_at MGC15875.
  • 4. The method of claim 2, wherein at least 25 genes from Table A are determined, and wherein said 25 genes are: 202061_s_at SEL1L,202304_at FNDC3A,202558_s_at STCH,203397_s_at GALNT3,204079_at TPST2,205483_s_at G1P2,205552_s_at OAS1,205718_at ITGB7,208725_at EIF2S2,209083_at CORO1A,209118_s_at TUBA3,209122_at ADFP,209417_s_at IF135,211990_at HLA-DPA1,214152_at CCPG1,214179_s_at NFE2L1,219118_at FKBP11,221658_s_at IL21R,222690_s_at TMEM39A,223129_x_at MYLIP,224451_x_at ARHGAP9,224763_at RPL37,227755_at EST, GenBank AA042983,228831_s_at GNG7 and232488_at MGC15875.
Priority Claims (1)
Number Date Country Kind
12306141.8 Sep 2012 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2013/069736 9/23/2013 WO 00