METHODS FOR PREDICTING MULTIPLE MYELOMA TREATMENT RESPONSE

Abstract
The present invention relates to a method of testing whether a patient suffering of myeloma will respond or not to a histone deacetylase inhibitor (HDACi) comprising: determining the expression level (ELi) of several genes G1-Gn selected from table A in a biological sample obtained from said patient comparing the expression level (ELi) determined at step i) with a predetermined reference level (ELRi) iii) calculating the HAS score trough the following formula 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) comparing the score HAS determined at step iii) with a predetermined reference value HASR v) and concluding that the patient will respond to the HDACi when the HAS score is higher than the predetermined reference value HASR or concluding that the patient will not respond to the HDACi when the HAS score is lower than the predeference value HASR
Description
FIELD OF THE INVENTION

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


BACKGROUND OF THE INVENTION

The molecular events governing the onset and progression of malignant transformation involve 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 triggered 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 many cancers (4). DNA methyltransferase inhibitors or HDAC inhibitors are now being used in the treatment of some hematologic malignancies including multiple myeloma (MM) and myelodysplastic syndromes (5-8). 18 different HDACs were identified and divided into four classes based on cellular localization and function (9). Class I includes HDACs 1, 2, 3 and 8, which are restrictively nuclear. Class II HDACs includes HDACs 4, 5, 7 and 9 (class IIa) shuttling back and forth between the nucleus and the cytoplasm and HDACs 6 and 10 (class IIb), with their distinctive two zinc-dependent catalytic sites, are expressed only in the cytoplasm. Class III contains the NAD+ dependent sirtuin family, which does not act primarily on histones and class IV includes HDAC11 (9,10). Based on their chemical structure, HDACi can be grouped in four classes: hydroxamates (panobinostat, trichostatin-A (TSA), vorinostat, belinostat (PXD101), NVP-LAQ824 and givinostat (ITF2357)), cyclic peptide (romidepsin (depsipeptide)), aliphatic acids (valproic acid and sodium phenylbutyrate) and benzamides (MS-275, MGCD0103) (10). HDACi are characterized as class I-specific HDACs inhibitors (MGCD0103, romidepsin and MS-275) or as pan-HDAC inhibitors, denoting activity against both classes I and II HDACs (TSA, panobinostat, vorinostat and belinostat) (10). Multiple myeloma is a plasma cell neoplasm characterized by the accumulation of malignant plasma cells (PCs), termed Multiple Myeloma Cells (MMCs) within the bone marrow (BM). Despite the recent introduction of new 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 (t(14; 16)) or MMSET/FGFR3 (t(4; 14)) (11,12). HDACi have already been evaluated in MM including Trichostatin A (TSA) (13), vorinostat (14,15), NVP-LAQ824 (16), depsipeptide (17), KD5170 (18), valproic acid (19, 20) and panobinostat (10). In MM, HDACi induce G1 cell cycle arrest by enhancing expression of p21, p53 and dephosphorylation of Rb (13, 15, 20), induce apoptosis by dowregulation of Bcl-2 family members (15,17) and overcome drug resistance mediated by the bone marrow environment (15). Clinical trials were designed to analyze the activity of HDACi as single agents in Phase I/II trials in relapsed/refractory MM patients. When used as single agent, HDACi had modest activity (21,22), but in combination with other anti-MM treatments, they can induce durable responses (23,24).


The identification of biomarkers predictive for sensitivity of MMCs to HDACi is an important objective for optimizing these clinical trials. In the present invention, the inventors used gene expression profiling of Multiple Myeloma Cells (MMCs) to build a novel “histone acetylation gene expression score” that makes it possible identification of patients whose MMCs will be targeted by HDAC 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 histone deacetylase inhibitor (HDACi).


DETAILED DESCRIPTION OF THE INVENTION

The multiple myeloma treatment response was investigated by the inventors using histone deacetylase inhibitor (HDACi) and human multiple myeloma cell lines. The inventors analyzed gene expression profiles of 5 MM cells lines treated with trichostatin A (TSA). 95 genes were deregulated by TSA and 37 out of 95 TSA deregulated genes have prognostic value in a cohort of 206 newly-diagnosed MM patients. The inventors also built a histone acetylation scores (HA Score or HAS) using the probe set signal value weighted by the beta coefficient of prognostic genes. The HA Score is predictive for myeloma cells HDACi sensitivity of HMCL and primary myeloma cells in vitro. The HA Score allows identification of myeloma patients that could benefit HDAC inhibitor treatment.


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 “histone deacetylase inhibitor” or “HDACi” has its general meaning in the art and refers to a multiple myeloma treatment. The term “histone deacetylase inhibitor” or “HDACi” refers to histone deacetylase inhibitor that can be grouped in four classes: hydroxamates (panobinostat (LBH-589), trichostatin-A (TSA), vorinostat (SAHA), belinostat (PXD101), NVP-LAQ824 and givinostat (ITF2357)), cyclic peptide (romidepsin (depsipeptide)), aliphatic acids (valproic acid (VPA) and sodium phenylbutyrate) and benzamides (MS-275, MGCD0103) (10). HDACi are characterized as class I-specific HDACs inhibitors (MGCD0103, romidepsin and MS-275) or as pan-HDAC inhibitors, denoting activity against both classes I and II HDACs (TSA, panobinostat, vorinostat and belinostat) (10).


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



Reference


Gene
Symbol
Gene name
Gene ID
β coefficient
level (ELRi)















G1
SCN3A
sodium channel,
210432_s_at
0.996958090200582
64.0776699029126




voltage-gated, type







III, alpha subunit





G2
ANK3
ankyrin 3
206385_s_at
−0.913919887371127
81.0679611650485


G3
APLP2
amyloid beta (A4)
214875_x_at
0.784702839669874
24.7572815533981




precursor-like







protein 2





G4
QKI
quaking homolog,
212636_at
−0.804915153416405
85.4368932038835




KH domain RNA







binding





G5
SYT11
synaptotagmin XI
209198_s_at
−0.71983191352392
38.8349514563107


G6
KIAA1324L
KIAA1324-like
235301_at
−0.921569046866849
87.864077669903


G7
DHRS2
dehydrogenase/reductase
214079_at
1.13644894957913
10.6796116504854




(SDR family)







member 2





G8
DFNA5
deafness, autosomal
203695_s_at
−0.601888717627172
48.5436893203884




dominant 5





G9
STAT1
signal transducer and
209969_s_at
0.865200607771322
20.873786407767




activator of







transcription 1





G10
SERPINI1
serpin peptidase
205352_at
0.708779268580762
55.3398058252427




inhibitor, clade I







(neuroserpin),







member 1





G11
BBS9 or
Bardet-Biedl
209958_s_at
0.854129984250074
28.1553398058252



PTHB1
syndrome 9 or







parathyroid







hormone-responsive







B1





G12
RGS1
regulator of G-
216834_at
−0.604322500555556
65.5339805825243




protein signaling 1





G13
HLA-
major
211990_at
−0.708484927244178
79.126213592233



DPA1
histocompatibility







complex, class II,







DP alpha 1





G14
FN1
fibronectin 1
212464_s_at
−0.988700830206777
90.2912621359223


G15
KLHL24
kelch-like 24
226158_at
1.23690256697662
78.1553398058252


G16
HLA-
major
208894_at
−1.02237505806332
33.9805825242718



DRA
histocompatibility







complex, class II,







DR alpha





G17
PTPRG
protein tyrosine
204944_at
0.749927151477102
16.9902912621359




phosphatase,







receptor type, G





G18
RASGEF1B
RasGEF domain
230233_at
−0.855660771294511
89.3203883495146




family; member 1B





G19
OAS1
2′,5′-oligoadenylate
205552_s_at
0.918506668310864
69.9029126213592




synthetase 1





G20
TRIM38
tripartite motif-
203567_s_at
0.673917187650873
15.5339805825243




containing 38





G21
SERPINB1
Serpin peptidase
228726_at
−0.96828128571625
87.378640776699




inhibitor; clade B







(ovalbumin);







member 1





G22
TRPS1
trichorhinophalangeal
222651_s_at
0.776522209484358
13.1067961165049




syndrome I





G23
CFHR1
complement factor
215388_s_at
−0.644744853140614
77.1844660194175



or
H-related 1






CFHL1






G24
PHLDA1
pleckstrin
225842_at
−1.14097033702181
89.8058252427184




homology-like







domain, family A,







member 1





G25
HLA-
major
212998_x_at
−0.741541655889151
76.2135922330097



DQB1
histocompatibility







complex, class II,







DQ beta 1





G26
SELL
selectin L
204563_at
0.665456120458623
75.7281553398058


G27
HLA-
major
215193_x_at
−0.835746441816605
88.8349514563107



DRB1
histocompatibility







complex; class II;







DR beta 1





G28
NFKBIZ
nuclear factor of
223218_s_at
−0.7451541815469
79.126213592233




kappa light







polypeptide gene







enhancer in B-cells







inhibitor, zeta





G29
PARP14
poly (ADP-ribose)
224701_at
1.08062305938478
10.1941747572816




polymerase family,







member 14





G30
CFI
complement factor I
203854_at
0.85259982594094
10.6796116504854


G31
MAN1C1
mannosidase, alpha,
218918_at
−0.709223593142427
33.495145631068




class 1C, member 1





G32
BASP1
brain abundant,
202391_at
−0.9921976055676
83.495145631068




membrane attached







signal protein 1





G33
GDAP1
ganglioside-induced
226269_at
0.988539786187137
62.621359223301




differentiation-







associated protein 1





G34
EFHC1
EF-hand domain (C-
219833_s_at
−0.587820758510972
51.9417475728155




terminal) containing 1





G35
ANXA1
annexin A1
201012_at
0.699836394397964
25.7281553398058


G36
RTN2
reticulon 2
34408_at
−1.25994066884418
36.4077669902913


G37
DDX60L
DEAD (Asp-Glu-
228152_s_at
−0.629835803277543
54.8543689320388



or
Ala-Asp) box






FLJ31033
polypeptide 60-like









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 histone deacetylase inhibitor (HDACi) 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 HAS score trough the following formula






HAS
=




i
=
1

n







β





i
×
Ci








    • wherein βi represent the regression 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 HAS determined at step iii) with a predetermined reference value HASR

    • v) and concluding that the patient will respond to the HDACi when the HAS score is higher than the predetermined reference value HASR or concluding that the patient will not respond to the HDACi when the HAS score is lower than the predetermined reference value HASR





In some embodiments, the levels of at least 34 genes from Table A are determined wherein said genes are: SCN3A, ANK3, APLP2, QKI, SYT11, KIAA1324L, DHRS2, DFNA5, STAT1, SERPINI1, BBS9 (or PTHB1), RGS1, HLA-DPA1, FN1, KLHL24, HLA-DRA, PTPRG, RASGEF1B, OAS1, TRIM38, SERPINB1, TRPS1, CFHR1 (or CFHL1), PHLDA1, HLA-DQB1, SELL, HLA-DRB1, NFKBIZ, PARP14, CFI, MAN1C1, BASP1, GDAP1 and EFHC1.


In some embodiment, the level of 34, 35, 36, or 37 genes from Table A are determined wherein every combinations of genes comprises a minimal set of 34 genes consisting of: SCN3A, ANK3, APLP2, QKI, SYT11, KIAA1324L, DHRS2, DFNA5, STAT1, SERPINI1, BBS9 (or PTHB1), RGS1, HLA-DPA1, FN1, KLHL24, HLA-DRA, PTPRG, RASGEF1B, OAS1, TRIM38, SERPINB1, TRPS1, CFHR1 (or CFHL1), PHLDA1, HLA-DQB1, SELL, HLA-DRB1, NFKBIZ, PARP14, CFI, MAN1C1, BASP1, GDAP1 and EFHC1.


In some embodiment, the level of 35 genes from Table A are determined wherein said genes are:


SCN3A, ANK3, APLP2, QKI, SYT11, KIAA1324L, DHRS2, DFNA5, STAT1, SERPINI1, BBS9 (or PTHB1), RGS1, HLA-DPA1, FN1, KLHL24, HLA-DRA, PTPRG, RASGEF1B, OAS1, TRIM38, SERPINB1, TRPS1, CFHR1 (or CFHL1), PHLDA1, HLA-DQB1, SELL, HLA-DRB1, NFKBIZ, PARP14, CFI, MAN1C1, BASP1, GDAP1, EFHC1, and ANXA1, or,


SCN3A, ANK3, APLP2, QKI, SYT11, KIAA1324L, DHRS2, DFNA5, STAT1, SERPINI1, BBS9 (or PTHB1), RGS1, HLA-DPA1, FN1, KLHL24, HLA-DRA, PTPRG, RASGEF1B, OAS1, TRIM38, SERPINB1, TRPS1, CFHR1 (or CFHL1), PHLDA1, HLA-DQB1, SELL, HLA-DRB1, NFKBIZ, PARP14, CFI, MAN1C1, BASP1, GDAP1, EFHC1, and RTN2, or,


SCN3A, ANK3, APLP2, QKI, SYT11, KIAA1324L, DHRS2, DFNA5, STAT1, SERPINI1, BBS9 (or PTHB1), RGS1, HLA-DPA1, FN1, KLHL24, HLA-DRA, PTPRG, RASGEF1B, OAS1, TRIM38, SERPINB1, TRPS1, CFHR1 (or CFHL1), PHLDA1, HLA-DQB1, SELL, HLA-DRB1, NFKBIZ, PARP14, CFI, MAN1C1, BASP1, GDAP1, EFHC1, and DDX6OL (FLJ31033).


In some embodiment, the level of 36 genes from Table A are determined wherein said genes are:


SCN3A, ANK3, APLP2, QKI, SYT11, KIAA1324L, DHRS2, DFNA5, STAT1, SERPINI1, BBS9 (or PTHB1), RGS1, HLA-DPA1, FN1, KLHL24, HLA-DRA, PTPRG, RASGEF1B, OAS1, TRIM38, SERPINB1, TRPS1, CFHR1 (or CFHL1), PHLDA1, HLA-DQB1, SELL, HLA-DRB1, NFKBIZ, PARP14, CFI, MAN1C1, BASP1, GDAP1, EFHC1, ANXA1, and RTN2, or,


SCN3A, ANK3, APLP2, QKI, SYT11, KIAA1324L, DHRS2, DFNA5, STAT1, SERPINI1, BBS9 (or PTHB1), RGS1, HLA-DPA1, FN1, KLHL24, HLA-DRA, PTPRG, RASGEF1B, OAS1, TRIM38, SERPINB1, TRPS1, CFHR1 (or CFHL1), PHLDA1, HLA-DQB1, SELL, HLA-DRB1, NFKBIZ, PARP14, CFI, MAN1C1, BASP1, GDAP1, EFHC1, ANXA1, and DDX6OL (F1131033), or,


SCN3A, ANK3, APLP2, QKI, SYT11, KIAA1324L, DHRS2, DFNA5, STAT1, SERPINI1, BBS9 (or PTHB1), RGS1, HLA-DPA1, FN1, KLHL24, HLA-DRA, PTPRG, RASGEF1B, OAS1, TRIM38, SERPINB1, TRPS1, CFHR1 (or CFHL1), PHLDA1, HLA-DQB1, SELL, HLA-DRB1, NFKBIZ, PARP14, CFI, MAN1C1, BASP1, GDAP1, EFHC1, RTN2, and DDX6OL (FLJ31033).


In some embodiments, the level of the 37 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 HASR 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 HASR could be used for obtaining the reference value and thereafter for assessment of the response to HDACi. However in one embodiment, the reference value HASR is the median value of HAS.


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


Typically, the reference value HASR is −11.3 for determining whether a patient suffering of multiple myeloma will respond to an HDACi and for predicting the survival time of patient suffering of multiple myeloma.


Typically, the reference value HASR is −12.3 for determining whether a patient suffering of multiple myeloma will respond to an HDACi.


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 histone deacetylase 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 histone deacetylase inhibitor (HDACi) by performing the method according to the invention


b) administering the histone deacetylase inhibitor, if said patient has as score higher than the reference value HASR (i.e. the patient will respond to the histone deacetylase inhibitor).


A further object of the invention relates to a histone deacetylase 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: Histone acetylation Score in normal and malignant plasma cells


Histone acetylation 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.



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


Patients of HM cohort were ranked according to increased HA Score and a maximum difference in OS was obtained with HA Score=−11.3 splitting patients in a high risk (42.7%) and low risk (57.3%) groups. The prognostic value of HA Score was tested on an independent cohort of 345 patients from UAMS treated with TT2 therapy (UAMS-TT2 cohort). The parameters to compute the HA Score of patients of UAMS-TT2 cohort and the proportions delineating the 2 prognostic groups were those defined with HM cohort.



FIG. 3: HA Score predicts for sensitivity of human myeloma cell lines to trichostatin A.


(A) HMCLs with high HA Score (N=5) exhibit significant higher HDACi sensitivity compared to HMCLs with low HA 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 TSA concentrations. Data are mean values plus or minus standard deviation (SD) of 5 experiments determined on sextuplet culture wells.



FIG. 4: HA Score predicts for trichostatin A sensitivity of primary myeloma cells of patients. Mononuclear cells from tumor samples of 13 patients with MM were cultured for 4 days in the presence of IL-6 (2 ng/ml) with or without graded TSA 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 HA Score (N=8) and white represents patients with low HA Score values (N=5).



FIG. 5: HA Score predicts for sensitivity of human myeloma cell lines to HDACi in clinical development in MM.


HMCLs with high HA Score (N=5) exhibit significant higher HDACi sensitivity compared to HMCLs with low HA 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 Panobinostat (A), VPA (VPA) or SAHA (C) concentrations. Data are mean values plus or minus standard deviation (SD) of 5 experiments determined on sextuplet culture wells.





EXAMPLES
Example 1
Gene Expression-Based Prediction of Myeloma Cell Sensitivity to Histone Deacetylase Inhibitors (Moreaux et al., BJC, 2013).

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, XG-21, XG-22, XG-23 and XG-24 human myeloma cell lines were obtained as previously described (25-29). JJN3 was kindly provided by Dr Van Riet (Bruxelles, Belgium), JIM3 by Dr MacLennan (Birmingham, UK) and MM1S by Dr Rosen (Chicago, USA). AMO-1, LP1, L363, U266, OPM2, and SKMM2 were from DSMZ (Germany) and RPMI8226 from ATTC (USA). All HMCLs derived in our 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). 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) (30) 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 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 (31) 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/). After ficoll-density gradient centrifugation, plasma cells were purified using anti-CD138 MACS microbeads (Miltenyi Biotech, Bergisch Gladbach, Germany). The t(4; 14) translocation results in aberrant FGFR3 expression in 70% of patients and MMSET spiked expression in 100% of patients (32), and spiked MMSET expression has been taken as surrogate for the presence of t(4; 14) as previously described (33, 34).


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 with 0.33 μmol/L TSA (Sigma, St Louis, Mo.) for 24 h. Control cells were cultured in the same conditions without TSA.


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 TSA 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 13 patients with MM 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 TSA, valproic acid (VPA), vorinostat (SAHA) or panobinostat (LBH-589). 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 (35).


Preparation of Complementary RNA (cRNA) and Microarray Hybridization


RNA was extracted using the RNeasy Kit (Qiagen, Hilden, Germany) as previously described (36-37). 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 our bioinformatics platforms—RAGE (http://rage.montp.inserm.fr/) (38) and Amazonia (http://amazonia.montp.inserm.fr/) (39)—or SAM (Significance Analysis of Microarrays) software (40). Hierarchical clustering was performed with the Cluster and Treeview softwares from Eisen (41). The event free or overall survival of subgroups of patients was compared with the log-rank test and survival curves computed with the Kaplan-Meier method. The prognostic values of parameters were compared with univariate or multivariate Cox analysis. Statistical comparisons were done with Mann-Whitney, Chi-square, or Student t-tests. Statistical tests were performed with the software package SPSS 12.0 (SPSS, Chicago, Ill.). Biological pathways were analyzed with Ingenuity Pathways Analysis (Ingenuity® Systems, www.ingenuity.com).


Results


Identification of Prognostic Genes Whose Expression is Upregulated by Trichostatin A Treatment of Multiple Myeloma Cells.


Five HMCLs were treated with 0.33 μM TSA for 24 h, a concentration which did not affect myeloma cell viability (Supplementary Table S2) (42). Using SAM supervised paired analysis, the expression of 95 genes was found to be significantly upregulated by TSA treatment of these 5 HMCLs (FDR<5%; Supplementary Table S3). TSA-regulated genes are significantly enriched in genes related to “Immunological disease and Inflammatory disease” pathway (P<0.05; Ingenuity pathway analysis). Looking for the expression of these 95 TSA-regulated genes in primary MMCs of a cohort of 206 newly-diagnosed patients (HM cohort), 16 genes had a bad prognostic value and 21 a good one after Benjamini Hochberg multiple testing correction (Supplementary Table S4). The prognostic information of HDACi regulated genes was gathered within an histone acetylation score (HA 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 described (34). The value of HA Score in healthy, premalignant or malignant plasma cells is displayed in FIG. 1. Cells from MGUS patients had a significant higher HA Score than normal BMPCs (P<0.001), MMCs of patients a significantly higher HA Score than normal BMPCs or plasma cells from MGUS-patients (P<0.001), and HMCLs the highest score (P<0.001) (FIG. 1).


Prognostic Value of HA Score Compared to Usual Prognostic Factors.


HA Score had prognostic value when used as a continuous variable (P≦10−4), or by splitting patients into two groups using Maxstat R function (34). A maximum difference in overall survival (OS) was obtained with HA Score=−11.3 splitting patients in a high-risk group of 42.7% patients (HA Score>−11.3) with a 43.5 months median OS and a low risk group of 57.3% patients (HA Score≦−11.3) with not reached median survival (FIG. 2). Using univariate Cox analysis, HA Score, UAMS-HRS, IFM-score and GPI had prognostic value as well as t(4; 14), del17p, β32m, albumin and ISS using the HM patient cohort (Supplementary Table S5). When compared two by two, HA Score tested with β2m and t(4; 14) remained significant. When these parameters were tested together, HA Score, β2m, t(4; 14) and GPI kept prognostic value. The HA 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, HA Score was computed using parameters defined with HM patients' cohort. The median OS of patients within high score group was 71.4 months and not reached for patients with low HA 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, HA Score remained significant compared to UAMS-HRS, IFM, GPI, t(4; 14), and del17p in the UAMS-TT2 cohort (Supplementary Table S5). When these parameters were tested together, HA Score, UAMS-HRS, t(4; 14) and del17p kept prognostic value in UAMS-TT2 cohort.


HA Score is Predictive for Sensitivity of Human Myeloma Cell Lines or Patients' Primary MMCs to Trichostatin A In Vitro.


The inventors sought to determine whether HA Score could predict for the sensitivity of 10 HMCLs to HDAC inhibitor. Starting from a large cohort of 40 HMCLs (25), the 10 HMCLs with the highest or lowest HA Score were selected to assay TSA sensitivity. The 5 HMCLs with the highest HA Score exhibited a significant 5-fold higher TSA sensitivity (median IC50=10.97 nM ; range: 6.32 to 17.4 nM) than the 5 HMCLs with low HA Score (P=0.0004; median IC50=52.33 nM; range: 29.49 to 57.74 nM) (FIG. 3). No difference in recurrent genetic abnormalities were found between HMCLs with the highest or lowest HA Score (Table 1).


HA Score is Predictive for Sensitivity of Human Myeloma Cells to Other Clinical Grade HDACi In Vitro


The inventors sought to determine whether HA Score could predict for the sensitivity of myeloma cells to clinical grade HDAC inhibitors {Neri, 2012 #3317}. The 5 HMCLs with the highest HA Score exhibited a significant higher Panobinostat, VPA and Vorinostat sensitivity (median IC50=1.16 nM, 0.28 μM and 528 nM respectively) than the 5 HMCLs with low HA score (P=0.007, P=0.009 and P=0.02; median IC50=3.16 nM, 0.43 μM and 897 nM respectively) (FIGS. 5A-B&C).


Discussion


In this study, the inventors have identified a gene expression-based histone acetylation score (HA 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 pan-HDAC inhibitor, trichostatin A. Several trials have looked for the efficacy of various HDACi in patients with MM, when used alone (10,21,22) or in combination with usual anti-MMC drugs such as Dexamethasone, Lenalidomide or Bortezomib (10,23,24). These trials indicate a partial response, which could be explained by patients' heterogeneity. The current identification of HA Score should be very useful to investigate whether the higher response to HDACi is found in patients with highest HA Score and to speed up the investigation of the clinical efficacy of the novel agents.


Besides the tility of the current HA Score in selecting patients who could benefit from HDACi therapies, the current study highlights pathways which could be involved in the emergence of multiple myeloma cells. Among the genes upregulated by TSA treatment and associated with a favorable prognosis, the inventors identified NFKBIZ (nuclear factor of kappa light polypeptide gene enhancer in B-cell inhibitor zeta), BASP1 (Brain acid-soluble protein 1) and QKI (Quaking). NFKBIZ is a member of IκB family (43). NFKBIZ protein is localized in the nucleus where it interacts with and regulates nuclear NF-κB activity. Suppression of endogenous NFKBIZ renders cells more resistant to apoptosis, whereas its overexpression induces cell death (43,44). More recently, it was demonstrated that NFKBIZ inhibits the transcriptional activity of STAT3 leading to cell growth inhibition and apoptosis induction mediated by down-regulation of a known STAT3 target, Mcl-1 (45). This is of interest because the inventors previously demonstrated that Mcl-1 is the major antiapoptotic protein involved in IL-6-mediated survival of MMCs (46). BASP1 is a Myc oncogene target that is specifically repressed in Myc-transformed cells and conversely, has a strong potential to inhibit cell transformation induced by Myc (47). The inhibition of Myc induced fibroblast cell transformation by BASP1 also prevents the transcriptional activation or repression of known Myc target genes. BASP1 appears to be a potential tumor suppressor in cancer (47). In MM, malignant features includes activation of Myc and of NF-κB pathway (11,48,49). HDAC inhibitors appear useful to target NF-κB and Myc activation in MMC through upregulation of NFKBIBZ and BASP1 expression. RNA binding protein QKI belongs to the evolutionarily conserved signal transduction and activator of RNA family. It has been demonstrated that overexpression of QKI induced the G1 cell cycle arrest in oligodendrocyte progenitor cells (50). Furthermore, QKI inhibits colon cancer cell growth, acting as a tumor suppressor (51). It was demonstrated that QKI protein is directly transcribed by E2F1, which in turn negatively regulates the cell cycle by targeting multiple cell cycle regulators including p27, cyclin D1 and c-fos (52). These results demonstrated that a better understanding of the cellular response to epigenetic-targeted treatments will increase our knowledge of MM development and progression and will provide potential therapeutic advances. Epigenetic therapies could be combined with conventional therapies to develop personalized treatments in MM and render resistant tumors responsive to treatment. These advances may limit the side effects of treatment, improving compliance with dosing regimens and overall quality of life. Our methodology could be extended to other anti-MM treatments.









TABLE 1







Characteristics of HMCLsTSA sensitive and HMCLsTSA resistant



















HMCL
IL-6


Patient


t(14q32 or




HMCL


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










TSA Resistant HMCLs



















XG7
+
MN
MM
PB
F
Ak
 t(4; 14)
MMSET
mut
wt
+/−
MS


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

MS


AMO1

CO
PCT
AF
F
Ak
t(12; 14)
unknown
wt
wt
+
CD-2L


JJN3

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


LP1

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

MS







TSA Sensitive HMCLs



















XG5
++
MN
MM
PB
F
I
t(11; 14)
CCND1
wt
abn

CD-1


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


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


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


XG21
++
MN
MM
PE
M
I
t(11; 14)
CCND1
wt
wt
+
CD-1









Primary MMCs were cultured with their BM environment and recombinant IL-6 and graded concentrations of TSA for 4 days. Primary MMCs of patients with a HA Score above median value (−11.3, FIG. 1) exhibited significant (P<0.05) 2.4-fold higher TSA sensitivity than MMCs with HA Score below median (FIG. 4 and Table 2). The characteristics of patients with MM included in this study are described in Table 3.









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 TSA. 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
















5.1
10.3
20.7
41.3



Patient

nM
nM
nM
nM



no.
Control
TSA
TSA
TSA
TSA

















Patients
1
66154
43320
31086
22880
7250


with
2
114266
115520
45216
20592
29000


high
3
114266
72200
50868
38896
18850


HA
4
96224
83752
25434
20592
11600


Score
5
33540
19950
12170
7650
1736



6
72168
37544
33912
18304
7250



7
70092
48249
38249
29064
5238



8
100440
78844
41860
32752
29790



Mean
83394
62422
34849
23841
13839


Patients
1
75175
72200
64998
54912
33350


with
2
69161
60648
62172
41184
26100


low
3
15072
14750
18486
14328
12765


HA
4
272272
306768
254478
243978
235040


Score
5
21450
19100
19458
18168
18876



Mean
90626
94693
83918
74514
65226
















TABLE 3







Characteristics of patients with a HA Score above (N =


8) and under (N = 5) the median value.





















Multiple






Durie

Serum
myeloma





Mono-
and

β2-
molecular





clonal
Salmon
ISS
micro-
classifi-



Age
Sex
protein
stage
stage
globulin
cation











Patients with high HA Score














Patient 1
69
F
IgG
IIIA
I
2.8
CD2





Kappa


Patient 2
48
M
Lambda
NA
NA
NA
CD1


Patient 3
55
M
BJ
IIIB
III
10
HY





Kappa


Patient 4
69
M
IgG
IIIA
III
10.4
PR





Lambda


Patient 5
70
F
IgA
IIIA
II
5
CD2





Lambda


Patient 6
63
F
Asecret
III
III
13.5
CD1


Patient 7
54
M
IgA
IIIA
I
2.3
CD2





Lambda


Patient 8
72
M
IgG
IIIA
III
8.6
PR





Lambda







Patients with low HA Score














Patient 1
63
M
BJ
NA
NA
NA
PR





Lambda


Patient 2
62
M
IgA
IIA
I
2.6
HP





Kappa


Patient 3
83
M
IgG
IIIA
III
8.3
HY





Kappa


Patient 4
59
F
IgA
IIIA
III
6.7
MF





Lambda


Patient 5
47
M
IgA
IIIB
III
24.4
PR





Kappa
















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
42 [1-100]
42 [4-98] 



marrow





NA, not available.


ISS, International Staging System.













SUPPLEMENTARY TABLE S2







Cell viability of HMCLs treated with 0.33 μM TSA for 24 h.


Date are the mean percentages ± SD of viable cells evaluated


by trypan blue exclusion (3 experiments).









Cell viability (%)










Day 1













HMCLS
Day 0
Control
TSA






XG-5
 70 ± 2
 70 ± 2
 70 ± 3



XG-6
100 ± 0
100 ± 0
100 ± 1



XG-7
100 ± 0
100 ± 0
100 ± 0



XG-20
100 ± 0
100 ± 0
100 ± 1



LP1
100 ± 0
100 ± 0
100 ± 0
















SUPPLEMENTARY TABLE S3







Genes overexpressed in TSA treated HMCLs. Five HMCLs were cultured


with or without 0.33 μM TSA for 1 day and gene expression was


profiled with Affymetrix U133 plus 2.0. Genes significantly differentially


expressed between control and TSA 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 TSA treated cells.











Probeset
Gene
Ratio
Banding
Affymetrix description










Intercellular communication and membrane proteins











209462_at
APLP1
3.37
19q13.1
amyloid beta (A4) precursor-like






protein 1


214875_x_at
APLP2
1.62
11q23-q25|
amyloid beta (A4) precursor-like





11q24
protein 2


209906_at
C3AR1
7.86
12p13.31
complement component 3a receptor 1


1557905_s_at
CD44
2.69
11p13
CD44 antigen (homing function and






Indian blood group system)


219505_at
CECR1
1.86
22q11.2
cat eye syndrome chromosome






region; candidate 1


215388_s_at
CFH
1.94
1q32
complement factor H


209732_at
CLEC2B
1.84
12p13-p12
C-type lectin domain family 2;






member B


226281_at
DNER
15.70
2q36.3
delta-notch-like EGF repeat-






containing transmembrane


212464_s_at
FN1
5.28
2q34
fibronectin 1


216041_x_at
GRN
2.58
17q21.32
granulin


200696_s_at
GSN
5.81
9q33
gelsolin (amyloidosis; Finnish type)


211990_at
HLA-DPA1
2.02
6p21.3
major histocompatibility complex;






class II; DP alpha 1


201137_s_at
HLA-DPB1
1.51
6p21.3
major histocompatibility complex;






class II; DP beta 1


212998_x_at
HLA-DQB1
1.51
6p21.3
major histocompatibility complex;






class II; DQ beta 1


208894_at
HLA-DRA
1.58
6p21.3
major histocompatibility complex;






class II; DR alpha


215193_x_at
HLA-DRB1
1.64
6p21.3
major histocompatibility complex;






class II; DR beta 1


216331_at
ITGA7
2.18
12q13
integrin; alpha 7


214020_x_at
ITGB5
2.73
3q21.2
Integrin; beta 5


203413_at
NELL2
6.16
12q13.11-q13.12
NEL-like 2 (chicken)


204563_at
SELL
4.02
1q23-q25
selectin L (lymphocyte adhesion






molecule 1)


228726_at
SERPINB1
2.97
6p25
Serpin peptidase inhibitor; clade B






(ovalbumin); member 1


205352_at
SERPINI1
4.22
3q26.1
serpin peptidase inhibitor; clade I






(neuroserpin); member 1


209848_s_at
SILV
26.38
12q13-q14
silver homolog (mouse)


1569003_at
TMEM49
2.57
17q23.2
transmembrane protein 49







Signal transduction











221718_s_at
AKAP13
2.18
15q24-q25
A kinase (PRKA) anchor protein 13


218501_at
ARHGEF3
2.01
3p21-p13
Rho guanine nucleotide exchange






factor (GEF) 3


219546_at
BMP2K
2.43
4q21.21
BMP2 inducible kinase


208891_at
DUSP6
1.99
12q22-q23
dual specificity phosphatase 6


226269_at
GDAP1
1.91
8q21.11
Ganglioside-induced differentiation-






associated protein 1


223218_s_at
NFKBIZ
2.70
3p12-q12
nuclear factor of kappa light






polypeptide gene enhancer in B-cells






inhibitor; zeta


203355_s_at
PSD3
4.19
8pter-p23.3
pleckstrin and Sec7 domain






containing 3


204944_at
PTPRG
1.80
3p21-p14
protein tyrosine phosphatase; receptor






type; G


230233_at
RASGEF1B
4.44
4q21.3
RasGEF domain family; member 1B


226436_at
RASSF4
1.32
10q11.21
Ras association (RalGDS/AF-6)






domain family 4


216834_at
RGS1
2.68
1q31
regulator of G-protein signalling 1


34408_at
RTN2
3.75
19q13.32
reticulon 2


209969_s_at
STAT1
3.72
2q32.2
signal transducer and activator of






transcription 1; 91 kDa







Cytoskeleton











200965_s_at
ABLIM1
1.98
10q25
actin binding LIM protein 1


206385_s_at
ANK3
2.57
10q21
ankyrin 3; node of Ranvier (ankyrin






G)


225481_at
FRMD6
1.23
14q22.1
FERM domain containing 6


203854_at
IF
3.96
4q25
I factor (complement)


224823_at
MYLK
1.71
3q21
myosin; light polypeptide kinase


218678_at
NES
1.50
1q23.1
nestin


209958_s_at
PTHB1
2.03
7p14
parathyroid hormone-responsive B1







Cell cycle











209304_x_at
GADD45B
1.51
19p13.3
growth arrest and DNA-damage-






inducible; beta







Metabolism











213106_at
ATP8A1
2.60
4p14-p12
ATPase; aminophospholipid






transporter (APLT); Class I; type 8A;






member 1


213317_at
CLIC5
4.98
6p12.1-21.1
Chloride intracellular channel 5


214079_at
DHRS2
8.51
14q11.2
dehydrogenase/reductase (SDR






family) member 2


201431_s_at
DPYSL3
2.55
5q32
dihydropyrimidinase-like 3


219833_s_at
EFHC1
3.22
6p12.3
EF-hand domain (C-terminal)






containing 1


210299_s_at
FHL1
1.34
Xq26
four and a half LIM domains 1


202838_at
FUCA1
3.52
1p34
fucosidase; alpha-L-1; tissue


218918_at
MAN1C1
2.10
1p35
mannosidase; alpha; class 1C;






member 1


211685_s_at
NCALD
1.80
8q22-q23
neurocalcin delta


201468_s_at
NQO1
2.29
16q22.1
NAD(P)H dehydrogenase; quinone 1


1555426_a_at
OTUD5
2.50
Xp11.23
OTU domain containing 5


207808_s_at
PROS1
3.03
3q11.2
protein S (alpha)


210432_s_at
SCN3A
1.37
2q24
sodium channel; voltage-gated; type






III; alpha


224818_at
SORT1
1.97
1p21.3-p13.1|
sortilin 1





1p21.3-p13.1



206310_at
SPINK2
7.86
4q12
serine peptidase inhibitor; Kazal type






2 (acrosin-trypsin inhibitor)







Protein binding











202391_at
BASP1
1.30
5p15.1-p14
brain abundant; membrane attached






signal protein 1


208791_at
CLU
2.55
8p21-p12
clusterin (complement lysis inhibitor;






SP-40; 40; sulfated glycoprotein 2;






testosterone-repressed prostate






message 2; apolipoprotein J)


203695_s_at
DFNA5
7.46
7p15
deafness; autosomal dominant 5


226158_at
KLHL24
2.32
3q27.1
kelch-like 24 (Drosophila)


204745_x_at
MT1G
2.61
16q13
metallothionein 1G


212185_x_at
MT2A
2.54
16q13
metallothionein 2A


202073_at
OPTN
2.93
10p13
optineurin


209198_s_at
SYT11
6.99
1q21.2
synaptotagmin XI


213361_at
TDRD7
2.36
9q22.33
tudor domain containing 7


201009_s_at
TXNIP
2.52
1q21.1
thioredoxin interacting protein







Nuclear proteins and transcription factors











205249_at
EGR2
6.47
10q21.1
early growth response 2 (Krox-20






homolog; Drosophila)


228260_at
ELAVL2
2.97
9p21
ELAV (embryonic lethal; abnormal






vision; Drosophila)-like 2 (Hu antigen






B)


219209_at
IFIH1
2.33
2p24.3-q24.3
interferon induced with helicase C






domain 1


238430_x_at
MGC19764
3.16
17q12
likely ortholog of mouse schlafen 5


223484_at
NMES1
7.17
15q21.1
normal mucosa of esophagus specific 1


205552_s_at
OAS1
2.79
12q24.1
2prime; 5prime-oligoadenylate






synthetase 1; 40/46 kDa


224701_at
PARP14
2.73
3q21.1
poly (ADP-ribose) polymerase






family; member 14


212636_at
QKI
3.04
6q26-27
quaking homolog; KH domain RNA






binding (mouse)


225123_at
SESN3
5.02
11q21
Sestrin 3


222651_s_at
TRPS1
2.08
8q24.12
trichorhinophalangeal syndrome I







Apoptosis











201012_at
ANXA1
7.51
9q12-q21.2|
annexin A1





9q12-q21.2



210538_s_at
BIRC3
4.09
11q22
baculoviral IAP repeat-containing 3


226530_at
BMF
2.19
15q14
Bcl2 modifying factor


204415_at
G1P3
2.81
1p35
interferon; alpha-inducible protein






(clone IFI-6-16)







Others











236099_at

1.78

Transcribed locus


215079_at

1.45

LOC441207


226725_at

3.75

Transcribed locus


225842_at

2.10

CDNA clone IMAGE: 5531727


231040_at

1.78

CDNA FLJ43172 fis; clone






FCBBF3007242


219637_at
ARMC9
4.55
2q37.1
armadillo repeat containing 9


229973_at
C1orf173
8.67
1p31.1
chromosome 1 open reading frame






173


228152_s_at
FLJ31033
4.08
4q32.3
hypothetical protein FLJ31033


235301_at
KIAA1324L
2.26
7q21.12
KIAA1324-like


225688_s_at
PHLDB2
6.58
3q13.2
pleckstrin homology-like domain;






family B; member 2


203567_s_at
TRIM38
2.83
6p21.3
tripartite motif-containing 38
















SUPPLEMENTARY TABLE S4







Prognostic value of TSA deregulated genes in primary MMC of


newly-diagnosed patients.












Ajusted P value





(Benjamini hochberg





multiple testing



Probeset
NAME
correction)
Hazard ratio










Bad prognostic genes










204563_at
SELL
.04
1.94


203567_s_at
TRIM38
.04
1.96


201012_at
ANXA1
.02
2.01


205352_at
SERPINI1
.04
2.03


204944_at
PTPRG
.01
2.12


222651_s_at
TRPS1
.03
2.17


214875_x_at
APLP2
.01
2.19


203854_at
IF
.03
2.34


209958_s_at
PTHB1
.01
2.35


209969_s_at
STAT1
.009
2.37


205552_s_at
OAS1
.01
2.50


226269_at
GDAP1
.008
2.69


210432_s_at
SCN3A
.007
2.71


224701_at
PARP14
.01
2.94


214079_at
DHRS2
4.76e−05
3.11


226158_at
KLHL24
.01
3.44







Good prognostic genes










34408_at
RTN2
2.42e−05
.28


225842_at

9.96e−05
.32


208894_at
HLA-DRA
.01
.36


212464_s_at
FN1
.01
.37


202391_at
BASP1
7.01e−05
.37


228726_at
SERPINB1
.009
.38


235301_at
KIAA1324L
.01
.39


206385_s_at
ANK3
.007
.40


230233_at
RASGEF1B
.04
.42


215193_x_at
HLA-DRB1
.01
.43


212636_at
QKI
.02
.44


212998_x_at
HLA-DQB1
.01
.47


223218_s_at
NFKBIZ
.03
.47


209198_s_at
SYT11
.03
.48


211990_at
HLA-DPA1
.02
.49


218918_at
MAN1C1
.04
.49


215388_s_at
CFH ///
.04
.52



CFHL1




228152_s_at
FLJ31033
.03
.53


216834_at
RGS1
.04
.54


203695_s_at
DFNA5
.04
.54


219833_s_at
EFHC1
.04
.55
















SUPPLEMENTARY TABLES S5







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
HA Score
18.07
<.0001
1.95
<.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
HA Score
15.71
<.0001
NA
NA


COX
ISS
1.44
NS
NA
NA


analysis -
HA Score
16.49
<.0001
NA
NA


Overall
β2m
1.1
.008
NA
NA


survival
HA Score
17.39
<.0001
1.57
.02



HRS
1.88
NS
4.11
<.0001



HA Score
17.34
<.0001
1.83
.002



IFM score
1.44
NS
1.61
0.02



HA Score
16.70
<.0001
1.91
.001



t(4; 14)
2.38
.01
2.15
.001



HA Score
14.41
<.0001
1.90
.001



del17p
1.63
NS
2.37
.001



HA Score
15.56
<.0001
1.68
.009



GPI
1.65
NS
1.55
.005


Multivariate
HA Score
10.85
<.0001
1.50
.03


COX
β2m
1.1
.02
NA
NA


analysis -
ISS
1.13
NS
NA
NA


Overall
HRS
1.70
NS
3.92
<.0001


survival
IFM score
.45
NS
.89
NS



t(4; 14)
3.89
.003
2.32
.001



del17p
1.42
NS
2.35
.001



GPI
2.11
.03
1.19
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 Myélome; NA, Not available.


Example 2

In order to identify the minimal number of genes among the 37 genes used to calculate the HA score, the inventors used PAM (Prediction Analysis of Microarray) statistical technique used for class prediction from gene expression data using nearest shrunken centroids. 34 genes were identified and are depicted in Table B.









TABLE B







the minimal number of genes among the 37 genes used


to calculate the HA score.











Gene





Symbol
Gene name
Gene ID






SCN3A
sodium channel,
210432_s_at




voltage-gated, type





III, alpha subunit




ANK3
ankyrin 3
206385_s_at



APLP2
amyloid beta (A4)
214875_x_at




precursor-like protein 2




QKI
quaking homolog, KH
212636_at




domain RNA binding




SYT11
synaptotagmin XI
209198_s_at



KIAA1324L
KIAA1324-like
235301_at



DHRS2
dehydrogenase/reductase
214079_at




(SDR family)





member 2




DFNA5
deafness, autosomal
203695_s_at




dominant 5




STAT1
signal transducer and
209969_s_at




activator of





transcription 1




SERPINI1
serpin peptidase
205352_at




inhibitor, clade I





(neuroserpin),





member 1




BBS9 or
Bardet-Biedl
209958_s_at



PTHB1
syndrome 9 or





parathyroid hormone-





responsive B1




RGS1
regulator of G-protein
216834_at




signaling 1




HLA-
major
211990_at



DPA1
histocompatibility





complex, class II, DP





alpha 1




FN1
fibronectin 1
212464_s_at



KLHL24
kelch-like 24
226158_at



HLA-
major
208894_at



DRA
histocompatibility





complex, class II, DR





alpha




PTPRG
protein tyrosine
204944_at




phosphatase, receptor





type, G




RASGEF1B
RasGEF domain
230233_at




family; member 1B




OAS1
2′,5′-oligoadenylate
205552_s_at




synthetase 1




TRIM38
tripartite motif-
203567_s_at




containing 38




SERPINB1
Serpin peptidase
228726_at




inhibitor; clade B





(ovalbumin); member 1




TRPS1
trichorhinophalangeal
222651_s_at




syndrome I




CFHR1
complement factor H-
215388_s_at



or
related 1




CFHL1





PHLDA1
pleckstrin homology-
225842_at




like domain, family A,





member 1




HLA-
major
212998_x_at



DQB1
histocompatibility





complex, class II, DQ





beta 1




SELL
selectin L
204563_at



HLA-
major
215193_x_at



DRB1
histocompatibility





complex; class II; DR





beta 1




NFKBIZ
nuclear factor of
223218_s_at




kappa light





polypeptide gene





enhancer in B-cells





inhibitor, zeta




PARP14
poly (ADP-ribose)
224701_at




polymerase family,





member 14




CFI
complement factor I
203854_at



MAN1C1
mannosidase, alpha,
218918_at




class 1C, member 1




BASP1
brain abundant,
202391_at




membrane attached





signal protein 1




GDAP1
ganglioside-induced
226269_at




differentiation-





associated protein 1




EFHC1
EF-hand domain (C-
219833_s_at




terminal) containing 1









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. Nov. 14, 2002; 347(20):1593-1603.


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


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


4. Kondo Y. Epigenetic cross-talk between DNA methylation and histone modifications in human cancers. Yonsei Med J. Aug. 31, 2009; 50(4):455-463.


5. Issa J P. DNA methylation as a therapeutic target in cancer. Clin Cancer Res. Mar. 15, 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. Mar. 1, 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. Feb. 15, 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. Nov. 13, 2009.


9. Lane A A, Chabner B A. Histone deacetylase inhibitors in cancer therapy. J Clin Oncol. Nov. 10, 2009; 27(32):5459-5468.


10. Neri P, Bahlis N J, Lonial S. Panobinostat for the treatment of multiple myeloma. Expert Opin Investig Drugs. Mar. 12, 2012.


11. Bergsagel P L, Kuehl W M. Molecular pathogenesis and a consequent classification of multiple myeloma. J Clin Oncol. Sep. 10, 2005; 23(26):6333-6338.


12. Hideshima T, Bergsagel P L, Kuehl W M, Anderson K C. Advances in biology of multiple myeloma: clinical applications. Blood. Aug. 1, 2004; 104(3):607-618.


13. Lavelle D, Chen Y H, Hankewych M, DeSimone J. Histone deacetylase inhibitors increase p21(WAF1) and induce apoptosis of human myeloma cell lines independent of decreased IL-6 receptor expression. Am J Hematol. November 2001; 68(3):170-178.


14. Mitsiades C S, Mitsiades N S, McMullan C J, et al. Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc Natl Acad Sci USA. Jan. 13, 2004; 101(2):540-545.


15. Mitsiades N, Mitsiades C S, Richardson P G, et al. Molecular sequelae of histone deacetylase inhibition in human malignant B cells. Blood. 2003; 16:16.


16. Catley L, Weisberg E, Tai Y T, et al. NVP-LAQ824 is a potent novel histone deacetylase inhibitor with significant activity against multiple myeloma. Blood. Oct. 1, 2003; 102(7):2615-2622.


17. Khan S B, Maududi T, Barton K, Ayers J, Alkan S. Analysis of histone deacetylase inhibitor, depsipeptide (FR901228), effect on multiple myeloma. Br J Haematol. April 2004; 125(2):156-161.


18. Feng R, Ma H, Hassig C A, et al. KD5170, a novel mercaptoketone-based histone deacetylase inhibitor, exerts antimyeloma effects by DNA damage and mitochondrial signaling. Mol Cancer Ther. June 2008; 7(6):1494-1505.


19. Kaiser M, Zavrski I, Sterz J, et al. The effects of the histone deacetylase inhibitor valproic acid on cell cycle, growth suppression and apoptosis in multiple myeloma. Haematologica. February 2006; 91(2):248-251.


20. Neri P, Tagliaferri P, Di Martino M T, et al. In vivo anti-myeloma activity and modulation of gene expression profile induced by valproic acid, a histone deacetylase inhibitor. Br J Haematol. November 2008; 143(4):520-531.


21. Richardson P, Mitsiades C, Colson K, et al. Phase I trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) in patients with advanced multiple myeloma. Leuk Lymphoma. March 2008; 49(3):502-507.


22. Niesvizky R, Ely S, Mark T, et al. Phase 2 trial of the histone deacetylase inhibitor romidepsin for the treatment of refractory multiple myeloma. Cancer. Jan. 15, 2011; 117(2):336-342.


23. Badros A, Burger A M, Philip S, et al. Phase I study of vorinostat in combination with bortezomib for relapsed and refractory multiple myeloma. Clin Cancer Res. Aug. 15, 2009; 15(16):5250-5257.


24. Harrison S J, Quach H, Link E, et al. A high rate of durable responses with romidepsin, bortezomib, and dexamethasone in relapsed or refractory multiple myeloma. Blood. Dec. 8, 2011; 118(24):6274-6283.


25. 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. April 2011; 96(4):574-582.


26. 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.


27. 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.


28. 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.


29. 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.


30. 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. October 2003; 82(10):654-659.


31. 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. Apr. 1, 2006; 107(7):2633-2638.


32. Santra M, Zhan F, Tian E, Barlogie B, Shaughnessy J, Jr. A subset of multiple myeloma harboring the t(4; 14)(p16; q32) translocation lacks FGFR3 expression but maintains an IGH/MMSET fusion transcript. Blood. Mar. 15, 2003; 101(6):2374-2376.


33. 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. May 7, 2009; 113(19):4614-4626.


34. 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. Nov. 18, 2011.


35. Moreaux J, Legouffe E, Jourdan E, et al. BAFF and APRIL protect myeloma cells from apoptosis induced by interleukin 6 deprivation and dexamethasone. Blood. Apr. 15, 2004; 103(8):3148-3157.


36. Hose D, Reme T, Meissner T, et al. Inhibition of aurora kinases for tailored risk-adapted treatment of multiple myeloma. Blood. Apr. 30, 2009; 113(18):4331-4340.


37. 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. Aug. 1, 2005; 106(3):1021-1030.


38. 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.


39. 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.


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


41. Eisen M B, Spellman P T, Brown P O, Botstein D. Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci USA. Dec. 8, 1998; 95(25):14863-14868.


42. 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. Jan. 1, 2008; 68(1):44-54.


43. Totzke G, Essmann F, Pohlmann S, Lindenblatt C, Janicke R U, Schulze-Osthoff K. A novel member of the IkappaB family, human IkappaB-zeta, inhibits transactivation of p65 and its DNA binding. J Biol Chem. May 5, 2006; 281(18):12645-12654.


44. Yamazaki S, Muta T, Takeshige K. A novel IkappaB protein, IkappaB-zeta, induced by proinflammatory stimuli, negatively regulates nuclear factor-kappaB in the nuclei. J Biol Chem. Jul. 20, 2001; 276(29):27657-27662.


45. Wu Z, Zhang X, Yang J, et al. Nuclear protein IkappaB-zeta inhibits the activity of STAT3. Biochem Biophys Res Commun. Sep. 18, 2009; 387(2):348-352.


46. Jourdan M, Veyrune J L, Vos J D, Redal N, Couderc G, Klein B. A major role for Mcl-1 antiapoptotic protein in the IL-6-induced survival of human myeloma cells. Oncogene. 2003; 22(19):2950-2959.


47. Hartl M, Nist A, Khan M I, Valovka T, Bister K. Inhibition of Myc-induced cell transformation by brain acid-soluble protein 1 (BASP1). Proc Natl Acad Sci USA. Apr. 7, 2009; 106(14):5604-5609.


48. Annunziata C M, Davis R E, Demchenko Y, et al. Frequent engagement of the classical and alternative NF-kappaB pathways by diverse genetic abnormalities in multiple myeloma. Cancer Cell. August 2007; 12(2):115-130.


49. Keats J J, Fonseca R, Chesi M, et al. Promiscuous mutations activate the noncanonical NF-kappaB pathway in multiple myeloma. Cancer Cell. August 2007; 12(2):131-144.


50. Larocque D, Galarneau A, Liu H N, Scott M, Almazan G, Richard S. Protection of p27(Kip1) mRNA by quaking RNA binding proteins promotes oligodendrocyte differentiation. Nat Neurosci. January 2005; 8(1):27-33.


51. Yang G, Fu H, Zhang J, et al. RNA-binding protein quaking, a critical regulator of colon epithelial differentiation and a suppressor of colon cancer. Gastroenterology. January 2010; 138(1):231-240 e231-235.


52. Yang G, Lu X, Wang L, et al. E2F1 and RNA binding protein QKI comprise a negative feedback in the cell cycle regulation. Cell Cycle. Aug 15 2011; 10(16):2703-2713.


53. Moreaux J, Reme T, Leonard W, Veyrune J L, Requirand G, Goldschmidt H, Hose D, Klein B. Gene expression-based prediction of myeloma cell sensitivity to histone deacetylase inhibitors. Br J Cancer. 2013 Aug. 6; 109(3):676-85.

Claims
  • 1. A method of testing whether a patient suffering of multiple myeloma will respond or not to a histone deacetylase inhibitor (HDACi) 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 HAS score trough the following formula
  • 2. The method of testing whether a patient suffering of multiple myeloma will respond or not to a histone deacetylase inhibitor (HDACi) according to claim 1 wherein the step i) comprises determining the expression level (ELi) of a combination of 34, 35, 36, or 37 genes from Table A wherein said combination of genes comprises a minimal set of 34 genes consisting of: SCN3A, ANK3, APLP2, QKI, SYT11, KIAA1324L, DHRS2, DFNA5, STAT1, SERPINI1, BBS9 (or PTHB1), RGS1, HLA-DPA1, FN1, KLHL24, HLA-DRA, PTPRG, RASGEF1B, OAS1, TRIM38, SERPINB1, TRPS1, CFHR1 (or CFHL1), PHLDA1, HLA-DQB1, SELL, HLA-DRB1, NFKBIZ, PARP14, CFI, MAN1C1, BASP1, GDAP1 and EFHC1.
  • 3. 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 histone deacetylase inhibitor (HDACi) by performing the method according to claim 1, andb) administering the histone deacetylase inhibitor, if said patient has as score higher than the reference value HASR.
  • 4. The method of claim 3, wherein said step i) comprises determining the expression level (ELi) of a combination of 34, 35, 36, or 37 genes from Table A wherein said combination of genes comprises a minimal set of 34 genes consisting of: SCN3A, ANK3, APLP2, QKI, SYT11, KIAA1324L, DHRS2, DFNA5, STAT1, SERPINI1, BBS9 (or PTHB1), RGS1, HLA-DPA1, FN1, KLHL24, HLA-DRA, PTPRG, RASGEF1B, OAS1, TRIM38, SERPINB1, TRPS1, CFHR1 (or CFHL1), PHLDA1, HLA-DQB1, SELL, HLA-DRB1, NFKBIZ, PARP14, CFI, MAN1C1, BASP1, GDAP1 and EFHC1.
Priority Claims (1)
Number Date Country Kind
12306225.9 Oct 2012 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2013/070964 10/8/2013 WO 00