Genetic marker for early breast cancer prognosis prediction and diagnosis, and use thereof

Information

  • Patent Grant
  • 10655187
  • Patent Number
    10,655,187
  • Date Filed
    Tuesday, October 9, 2018
    5 years ago
  • Date Issued
    Tuesday, May 19, 2020
    4 years ago
Abstract
The present invention relates to a gene for predicting or diagnosing the prognosis of early-stage breast cancer and to a use thereof, and more specifically relates to a genetic marker for predicting or diagnosing the prognosis of breast cancer, including TRBC1 (T cell receptor beta constant 1), BTN3A2 (butyrophilin, subfamily 3, member A2) or HLA-DPA1 (major histocompatibility complex, class II, DP alpha 1) for providing information necessary for predicting or diagnosing the prognosis of a breast cancer patient. The genetic marker of the present invention allows the prediction or diagnosis of the prognosis of a breast cancer patient, and can therefore advantageously be used for the purpose of providing a direction as to the future course of breast cancer treatment, including a decision on whether anticancer therapy is necessary.
Description
BACKGROUND
1. Field

The present invention relates to a gene for early-stage breast cancer prognosis prediction and diagnosis and a use thereof, and more specifically, to a genetic marker for early-stage breast cancer prognosis prediction and diagnosis, of TRBC1 (T cell receptor beta constant 1), BTN3A2 (butyrophilin, subfamily 3, member A2), or HLA-DPA1 (major histocompatibility complex, class II, DP alpha 1), for providing information necessary for the prognosis prediction and diagnosis of a breast cancer patient, and to a use thereof.


2. Discussion of the Background

As human genome information has been actively utilized, cancer research has focused on the establishment of mechanisms at the genome level. Particularly, cancer cell characteristics can be identified in a macroscopic view, based on information about expression patterns of tens of thousands of genes or on the increase or decrease in the number of genes using microarrays. This analysis of the genome-level information is very innovative in understanding organic and complicated life phenomena, and will be more commonly used. Specifically, in cases of complex diseases such as cancer, the analysis of a small number of particular genes is likely to obtain narrow results, and it is important to capture large behavior patterns with respect to the occurrence and development of cancer, and thus genome information analysis is absolutely necessary. As described above, most of the genome information that is a basis for cancer research is created using genome chips such as a microarray, and technologies that can obtain a lot of information at once are evolving day by day. In spite of the disadvantages of high costs, research using microarrays is being actively developed, so the amount of related information is explosively increasing. Since the mid-2000s, such genome information has started to be collected and made into a database, and secondary and tertiary analysis using the information thus obtained is becoming a focal point for the research of life phenomena.


Tens of thousands of probes indicating approximately 20,000 to 30,000 genes are embedded in general expression gene chips, and more than one million probes are often embedded in microarrays that measure precise information, such as SNP. Methods using these microarrays are very efficient since they are relatively simple and standardized and a large amount of information can be obtained at once in a short time, but analyzing the obtained results is a key point as well as being a difficult bottleneck. Comprehensive analysis for tens of thousands of genes incomparable to existing analysis for a small number of genes must be supported by a broad knowledge of the genome as well as statistical analysis techniques, so useful information can be eventually obtained. Besides, high-performance computing equipment capable of storing and analyzing large amounts of information are needed, and the related computational techniques are also needed. Meanwhile, it is difficult to perform for the researchers who are familiar with conventional biological research ranges and experimental methods, and thus, the methods cannot be favorably utilized even though genome information increases at an extraordinary rate in Korea. Considering the domestic situation with respect to capital and technology research that are insufficient compared with North America and Europe, actively utilizing known genome information should be at the head of bioinformatics. Genome analysis has been actively introduced in the research of, particularly, cancers, and a considerable amount of related-information has been accumulated.


Breast cancer is frequently detected in the early stage since self-diagnosis is possible and the importance of self-diagnosis is highly publicized. It was difficult to determine whether early-stage breast cancer patients should be allowed to receive anticancer treatments after surgery. It is possible to roughly predict a prognosis through pathological observation, but the observation result is difficult to normalize and quantify, and the reliability on prognosis prediction is low, and thus, most of early-stage breast cancer patients are recommended to receive anticancer treatments in actual clinical practice. Due to the nature of anticancer treatments, they are very expensive, while the patients suffer from very serious pains. It is estimated that more than half of early-stage breast cancer patients do not need to receive anticancer treatments. Therefore, if unnecessary anticancer treatments are reduced by analyzing the characteristics of the early-stage breast cancer to predict prognosis of patient, it may be a great help to increase the quality of life of the patients. As the information about tens of thousands of breast cancer gene expression patterns is obtained at once using microarrays, research for classifying breast cancer types at the molecular level and establishing mechanisms of cancer occurrence and development are being actively conducted. It is important to predict the prognosis of the early-stage breast cancer patients in clinical practice. The work of identifying genes for prognosis prediction using microarrays has already started in the early 2000s. Although research that uses microarrays is expensive, a significant number of breast cancer tissue expression profiles have been produced and available to researchers. Starting from the identifying of 70 prognosis prediction genes by analyzing the early-stage breast cancer tissues and survival data of 78 patients followed up for 10 years in 2002, a dozen genes for prognosis prediction genes were then published, and among these genes, several genes have already been commercialized and used in clinical practice (Chang, H. Y., et al., Gene expression signature of fibroblast serum response predicts human cancer progression: similarities between tumors and wounds. PLoSBiol 2(2): p. E7(2004); van de Vijver, M. J., et al., A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 347(25):1999-2009(2002); van 't Veer, L. J., et al., Gene expression profiling predicts clinical outcome of breast cancer. Nature 415(6871): 530-536(2002); Wang, Y., et al., Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet 365(9460): 671-679(2005); Buyse, M., et al., Validation and clinical utility of a 70-gene prognostic signature for women with node-negative breast cancer. J Natl Cancer Inst, 98(17):1183-92(2006); Paik, S., Development and clinical utility of a 21-gene recurrence score prognostic assay in patients with early-stage breast cancer treated with tamoxifen. Oncologist 12(6):631-635(2007); Paik, S., et al., A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med 351(27):2817-2826(2004); Sotiriou, C., et al., Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst 98(4):262-72(2006); Pawitan, Y., et al., Gene expression profiling spares early-stage breast cancer patients from adjuvant therapy: derived and validated in two population-based cohorts. Breast Cancer Res 7(6):R953-964(2005); Miller, L. D., et al., An expression signature for p53 status in human breast cancer predicts mutation status, transcriptional effects, and patient survival. Proc Natl Acad Sci USA, 102(38):13550-13555(2005); Bild, A. H., et al., Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature 439(7074):353-357(2006); Teschendorff, A. E., et al., A consensus prognostic gene expression classifier for ER positive breast cancer. Genome Biol 7(10):R101(2006); Desmedt, C., et al., Strong time dependence of the 76-gene prognostic signature for node-negative breast cancer patients in the TRANSBIG multicenter independent validation series. Clin Cancer Res 13(11): 3207-3214(2007)). Representative examples thereof are MammaPrint (Agendia) and Oncotype DX (Genomic Health), which are being currently used in clinical practice. However, they have been used as one of the references for prognosis (van de Vijver, M. J., et al., A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 347(25):1999-2009(2002); Paik, S., et al., A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med 351(27):2817-2826(2004)).


Throughout the entire specification, many research papers and patent documents are referenced and their citations are disclosed. The disclosure of cited papers and patent documents are entirely incorporated by reference into the present specification, and the level of the technical field within which the present invention falls, and details of the present invention are explained more clearly.


SUMMARY

The present inventors have endeavored to develop a gene diagnosis system capable of predicting the prognosis of the early-stage breast cancer patient and determining whether anticancer treatment is performed on the early-stage breast cancer patient by using an FFPE sample of the tissue containing cancer cells of the patient. As a result, the present inventors have identified genes associated with prognosis prediction by collecting and analyzing microarray data and clinical information, which are obtained from the early-stage breast cancer tissue; selected genes and sets thereof, which are suitable for the application to the FFPE sample, among the identified genes; and validated utility of the selected genes and gene sets, thereby completing the present invention.


Therefore, an aspect of the present invention is to provide a genetic marker for predicting or diagnosing the prognosis of a breast cancer patient, and a use thereof.


Another aspect of the present invention is to provide a novel method for predicting or diagnosing the prognosis of a breast cancer patient.


Still another aspect of the present invention is to provide a kit for predicting or diagnosing the prognosis of a breast cancer patient.


Still another aspect of the present invention is to provide a method for calculating a breast cancer prognosis predictive value in order to provide information necessary for predicting or diagnosing the prognosis of a breast cancer patient, the method comprising isolating mRNA from a patient sample, measuring the gene expression level, normalizing the gene expression level, and calculating a predictive value.


In accordance with an aspect of the present invention, there is provided a genetic marker for predicting or diagnosing the prognosis of a breast cancer patient and a use thereof.


In accordance with another aspect of the present invention, there is provided a novel method for predicting or diagnosing the prognosis of a breast cancer patient.


In accordance with still another aspect of the present invention, there is provided a kit for predicting or diagnosing the prognosis of a breast cancer patient.


In accordance with still another aspect of the present invention, there is provided a method for calculating a predictive value of the prognosis of breast cancer to provide information necessary for predicting or diagnosing the prognosis of a breast cancer patient, the method comprising isolating mRNA from a patient sample, measuring a gene expression level, normalizing the gene expression level, and calculating a predictive value.


In accordance with still another aspect of the present invention, there is provided a primer pair for at least one gene selected from the group consisting of T cell receptor beta constant 1 (TRBC1), butyrophilin, subfamily 3, member A2 (BTN3A2), and major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1), wherein the primer pair is capable of amplifying a target gene through PCR amplification.


In accordance with still another aspect of the present invention, there is provided a use of a primer pair for preparing an agent for predicting the prognosis of breast cancer, wherein the primer pair is for at least one gene selected from the group consisting of TRBC1, BTN3A2, and HLA-DPA1, and wherein the primer pair is capable of amplifying a target gene through PCR amplification.


In accordance with further aspect of the present invention, there is provided a method for diagnosing a prognosis of breast cancer and treating breast cancer in a breast cancer patient, the method comprising the steps of:


collecting a sample from the breast cancer patient;


isolating mRNA from the sample of the breast cancer patient;


measuring a first mRNA expression level for the mRNA of at least one i-gene selected from the group consisting of BTN3A2 (butyrophilin, subfamily 3, member A2), T cell receptor beta constant 1 (TRBC1), and major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1), and a second mRNA expression level for the mRNA of at least one p-gene selected from the group consisting of the p-genes of the following Table 1;


normalizing the first and second mRNA expression levels to determine a normalized value;


diagnosing the prognosis of breast cancer patient by using the determined normalized value of the first and second mRNA expression levels, wherein an overexpression of the i-gene indicates a good prognosis of breast cancer, while an overexpression of the p-gene indicates a poor prognosis of breast cancer; and


treating the diagnosed breast cancer patient by administering at least one of an anti-cancer agent, a surgery and a radiation therapy.









TABLE 1







List of p-genes









No.
Gene ID
Gene Name












1
APITD1
apoptosis-inducing, TAF9-like domain 1


2
BID
BH3 interacting domain death agonist


3
BUB1B
BUB1 mitotic checkpoint serine/threonine




kinase B


4
BUB1
BUB1 mitotic checkpoint serine/threonine kinase


5
CKS1B
CDC28 protein kinase regulatory subunit 1B


6
CKS2
CDC28 protein kinase regulatory subunit 2


7
DLGAP5
discs, large (Drosophila) homolog-associated




protein 5


8
POLA1
polymerase (DNA directed), alpha 1, catalytic




subunit


9
POLA2
polymerase (DNA directed), alpha 2, accessory




subunit


10
DSCC1
DNA replication and sister chromatid cohesion 1


11
DNA2
DNA replication helicase/nuclease 2


12
E2F8
E2F transcription factor 8


13
ERCC6L
excision repair cross-complementation




group 6-like


14
FBXO5
F-box protein 5


15
FANCI
Fanconi anemia, complementation group I


16
GADD45GIP1
growth arrest and DNA-damage-inducible,




gamma interacting protein 1


17
GINS1
GINS complex subunit 1 (Psf1 homolog)


18
GINS2
GINS complex subunit 2 (Psf2 homolog)


19
MAD2L1
MAD2 mitotic arrest deficient-like 1 (yeast)


20
MAD2L1BP
MAD2L1 binding protein


21
MIS18A
MIS18 kinetochore protein A


22
MYBL2
v-myb avian myeloblastosis viral oncogene




homolog-like 2


23
NAA50
N(alpha)-acetyltransferase 50, NatE catalytic




subunit


24
NEK2
NIMA-related kinase 2


25
NSL1
NSL1, MIS12 kinetochore complex component


26
PBK
PDZ binding kinase


27
RAB11A
RAB11A, member RAS oncogene family


28
RAD51C
RAD51 paralog C


29
RAD54B
RAD54 homolog B (S. cerevisiae)


30
RANBP1
RAN binding protein 1


31
RALA
v-ral simian leukemia viral oncogene




homolog A (ras related)


32
RACGAP1
Rac GTPase activating protein 1


33
SSNA1
Sjogren syndrome nuclear autoantigen 1


34
STAMBP
STAM binding protein


35
SSSCA1
Sjogren syndrome/scleroderma autoantigen 1


36
TAF2
TAF2 RNA polymerase II, TATA box binding




protein (TBP)-associated factor, 150 kDa


37
TIPIN
TIMELESS interacting protein


38
TIPRL
TOR signaling pathway regulator


39
TRIAP1
TP53 regulated inhibitor of apoptosis 1


40
TTK
TTK protein kinase


41
ZWINT
ZW10 interacting kinetochore protein


42
ASPM
asp (abnormal spindle) homolog, microcephaly




associated (Drosophila)


43
AURKA
aurora kinase A


44
AURKB
aurora kinase B


45
BRD7
bromodomain containing 7


46
CSNK2A1
casein kinase 2, alpha 1 polypeptide


47
CDC20
cell division cycle 20


48
CDC25C
cell division cycle 25C


49
CENPA
centromere protein A


50
CENPE
centromere protein E, 312 kDa


51
CENPF
centromere protein F, 350/400 kDa


52
CENPI
centromere protein I


53
CENPM
centromere protein M


54
CENPN
centromere protein N


55
CENPU
centromere protein U


56
CEP55
centrosomal protein 55 kDa


57
CHEK1
checkpoint kinase 1


58
CDT1
chromatin licensing and DNA replication factor 1


59
C11orf80
chromosome 11 open reading frame 80


60
CCNA2
cyclin A2


61
CCNB1
cyclin B1


62
CCNB2
cyclin B2


63
CCNE2
cyclin E2


64
CDK1
cyclin-dependent kinase 1


65
CDKN3
cyclin-dependent kinase inhibitor 3


66
CKAP5
cytoskeleton associated protein 5


67
DTL
denticleless E3 ubiquitin protein ligase




homolog (Drosophila)


68
DCTN2
dynactin 2 (p50)


69
DYNLT1
dynein, light chain, Tctex-type 1


70
ECD
ecdysoneless homolog (Drosophila)


71
ECT2
epithelial cell transforming 2


72
EIF4G1
eukaryotic translation initiation factor 4 gamma, 1


73
EIF4EBP1
eukaryotic translation initiation factor 4E




binding protein 1


74
EZR
ezrin


75
FEN1
flap structure-specific endonuclease 1


76
FOXM1
forkhead box M1


77
GSK3B
glycogen synthase kinase 3 beta


78
HMGN5
high mobility group nucleosome binding domain 5


79
INTS7
integrator complex subunit 7


80
KIF11
kinesin family member 11


81
KIF14
kinesin family member 14


82
KIF20A
kinesin family member 20A


83
KIF23
kinesin family member 23


84
KIF2C
kinesin family member 2C


85
KIF4A
kinesin family member 4A


86
KIFC1
kinesin family member C1


87
MIF
macrophage migration inhibitory factor




(glycosylation-inhibiting factor)


88
MELK
maternal embryonic leucine zipper kinase


89
MED1
mediator complex subunit 1


90
MCM10
minichromosome maintenance complex




component 10


91
MCM2
minichromosome maintenance complex




component 2


92
MCM6
minichromosome maintenance complex




component 6


93
MAP2K1
mitogen-activated protein kinase kinase 1


94
MSH6
mutS homolog 6


95
MLF1
myeloid leukemia factor 1


96
NCAPG
non-SMC condensin I complex, subunit G


97
NUSAP1
nucleolar and spindle associated protein 1


98
NUP155
nucleoporin 155 kDa


99
NUP93
nucleoporin 93 kDa


100
ORC4
origin recognition complex, subunit 4


101
ORC5
origin recognition complex, subunit 5


102
PIN1
peptidylprolyl cis/trans isomerase,




NIMA-interacting 1


103
PIK3R4
phosphoinositide-3-kinase, regulatory subunit 4


104
PTTG1
pituitary tumor-transforming 1


105
PTTG3P
pituitary tumor-transforming 3, pseudogene


106
PLK1
polo-like kinase 1


107
PLK4
polo-like kinase 4


108
PRIM1
primase, DNA, polypeptide 1 (49 kDa)


109
PA2G4
proliferation-associated 2G4, 38 kDa


110
LEPREL4
leprecan-like 4


111
PSMC3
proteasome (prosome, macropain) 26S subunit,




ATPase, 3


112
PSMC6
proteasome (prosome, macropain) 26S subunit,




ATPase, 6


113
PSMD10
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 10


114
PSMD12
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 12


115
PSMD14
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 14


116
PSMD2
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 2


117
PSMD3
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 3


118
PSMD4
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 4


119
PSMD6
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 6


120
PSMD7
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 7


121
PSMA1
proteasome (prosome, macropain) subunit,




alpha type, 1


122
PSMA2
proteasome (prosome, macropain) subunit,




alpha type, 2


123
PSMA3
proteasome (prosome, macropain) subunit,




alpha type, 3


124
PSMA4
proteasome (prosome, macropain) subunit,




alpha type, 4


125
PSMA6
proteasome (prosome, macropain) subunit,




alpha type, 6


126
PSMA7
proteasome (prosome, macropain) subunit,




alpha type, 7


127
PSMB3
proteasome (prosome, macropain) subunit,




beta type, 3


128
PSMB5
proteasome (prosome, macropain) subunit,




beta type, 5


129
PSMB7
proteasome (prosome, macropain) subunit,




beta type, 7


130
PRMT1
protein arginine methyltransferase 1


131
PPP2R3B
protein phosphatase 2, regulatory subunit B″, beta


132
PPP3CA
protein phosphatase 3, catalytic subunit, alpha




isozyme


133
PRC1
protein regulator of cytokinesis 1


134
RRM2
ribonucleotide reductase M2


135
RPS6KB1
ribosomal protein S6 kinase, 70 kDa,




polypeptide 1


136
SPAG5
sperm associated antigen 5


137
SKA1
spindle and kinetochore associated complex




subunit 1


138
STMN1
stathmin 1


139
SLBP
stem-loop binding protein


140
SMC2
structural maintenance of chromosomes 2


141
SMC4
structural maintenance of chromosomes 4


142
SMC5
structural maintenance of chromosomes 5


143
TERF1
telomeric repeat binding factor




(NIMA-interacting) 1


144
TXNL4A
thioredoxin-like 4A


145
TRIP13
thyroid hormone receptor interactor 13


146
TOP2A
topoisomerase (DNA) II alpha 170 kDa


147
TFDP2
transcription factor Dp-2 (E2F dimerization




partner 2)


148
TACC3
transforming, acidic coiled-coil containing




protein 3


149
TUBB3
tubulin, beta 3 class III


150
TUBB4B
tubulin, beta 4B class IVb


151
TUBB
tubulin, beta class I


152
TSG101
tumor susceptibility 101


153
UBE2C
ubiquitin-conjugating enzyme E2C


154
UBE2L3
ubiquitin-conjugating enzyme E2L 3


155
UBE2S
ubiquitin-conjugating enzyme E2S


156
USP9X
ubiquitin specific peptidase 9, X-linked


157
VRK1
vaccinia related kinase 1


158
ZFHX3
zinc finger homeobox 3


159
ZWILCH
zwilch kinetochore protein


160
MMP11
Matrix Metallopeptidase 11









In another aspect of the present invention, there is provided the above method wherein the i-gene is BTN3A2 (butyrophilin, subfamily 3, member A2).


In still another aspect of the present invention, there is provided the above method wherein the p-gene is selected from the group consisting of AURKA (Aurora Kinase A), CCNB2 (Cyclin B2), FOXM1 (Forkhead box protein M1), MMP11 (Matrix Metallopeptidase 11), PTTG1 (Pituitary Tumor-Transforming 1), RACGAP1 (Rac GTPase Activating Protein 1), RRM2 (Ribonucleotide Reductase M2), TOP2A (Topoisomerase II Alpha) and UBE2C (Ubiquitin-Conjugating Enzyme E2C).


In another aspect of the present invention, there is provided the above method wherein the step of measuring the expression levels is conducted through PCR amplification of a target gene.


In still another aspect of the present invention, there is provided the above method wherein the sample is a formalin-fixed paraffin-embedded (FFPE) sample of tissue containing cancer cells of the breast cancer patient.


In still further aspect of the present invention, there is provided the above method wherein the step of normalizing is conducted by calculating a ratio of a mean expression level of the gene with a mean expression level of at least one standard gene selected from the group consisting of CTBP1 (C-terminal-binding protein 1), TBP (TATA-binding protein), HMBS (hydroxymethylbilane synthase), CUL1 (cullin 1), and UBQLN1 (ubiquilin-1).


In accordance with further aspect of the present invention, there is provided a method for determining a predictive value of the prognosis of breast cancer to provide information necessary for predicting or diagnosing the prognosis of a breast cancer patient, the method comprising the steps of:


isolating mRNA from a sample of the breast cancer patient;


measuring a first mRNA expression level for the mRNA of at least one i-gene selected from the group consisting of BTN3A2 (butyrophilin, subfamily 3, member A2), T cell receptor beta constant 1 (TRBC1), and major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1), and a second mRNA expression level for the mRNA of at least one p-gene selected from the group consisting of the p-genes of the Table 1;


normalizing the first and second mRNA expression levels to obtain normalized values;


inserting the normalized values into a pre-determined calculation formula to obtain a numerical predictive value; and


determining the prognosis of breast cancer as being good or poor depending on the numerical predictive value.


In another aspect of the present invention, there is provided the above method wherein the i-gene is BTN3A2 (butyrophilin, subfamily 3, member A2).


In still another aspect of the present invention, there is provided the above method wherein the p-gene is selected from the group consisting of AURKA (Aurora Kinase A), CCNB2 (Cyclin B2), FOXM1 (Forkhead box protein M1), MMP11 (Matrix Metallopeptidase 11), PTTG1 (Pituitary Tumor-Transforming 1), RACGAP1 (Rac GTPase Activating Protein 1), RRM2 (Ribonucleotide Reductase M2), TOP2A (Topoisomerase II Alpha) and UBE2C (Ubiquitin-Conjugating Enzyme E2C).


In further another aspect of the present invention, there is provided the above method wherein an overexpression of the i-gene indicates a good prognosis of breast cancer, while an overexpression of the p-gene indicates a poor prognosis of breast cancer.


In still further aspect of the present invention, there is provided the above method wherein the step of measuring the expression levels is conducted through PCR amplification of a target gene.


In further aspect of the present invention, there is provided the above method wherein the sample is a formalin-fixed paraffin-embedded (FFPE) sample of a tissue containing cancer cells of a patient.


In another aspect of the present invention, there is provided the above method wherein the step of normalizing is conducted by calculating a ratio of a mean expression level of the gene with a mean expression level of at least one standard gene selected from the group consisting of CTBP1 (C-terminal-binding protein 1), TBP (TATA-binding protein), HMBS (hydroxymethylbilane synthase), CUL1 (cullin 1), and UBQLN1 (ubiquilin-1).


In still another aspect of the present invention, there is provided the above method wherein the breast cancer patient determined as poor prognosis is treated by administering at least one of an anti-cancer agent, a surgery and a radiation therapy.


In accordance with further aspect of the present invention, there is provided a method for predicting or diagnosing the prognosis of breast cancer in a breast cancer patient, the method comprising a step of using a plurality of primer pairs, wherein the plurality of the primer pairs comprises a primer pair for at least one i-gene selected from the group consisting of BTN3A2 (butyrophilin, subfamily 3, member A2), T cell receptor beta constant 1 (TRBC1), and major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1), and a primer pair for at least one p-gene selected from the group consisting of the p-genes of the Table 1, wherein the primer pairs are selected to amplify the at least one i-gene and the at least one p-gene through PCR amplification.


In another aspect of the present invention, there is provided the above method wherein the i-gene is BTN3A2 (butyrophilin, subfamily 3, member A2).


In still another aspect of the present invention, there is provided the above method wherein the p-gene is selected from the group consisting of AURKA (Aurora Kinase A), CCNB2 (Cyclin B2), FOXM1 (Forkhead box protein M1), MMP11 (Matrix Metallopeptidase 11), PTTG1 (Pituitary Tumor-Transforming 1), RACGAP1 (Rac GTPase Activating Protein 1), RRM2 (Ribonucleotide Reductase M2), TOP2A (Topoisomerase II Alpha) and UBE2C (Ubiquitin-Conjugating Enzyme E2C).


In still further aspect of the present invention, there is provided the above method wherein the breast cancer patient diagnosed as poor prognosis is treated by administering at least one of an anti-cancer agent, a surgery and a radiation therapy.


Unless defined otherwise, all technical and scientific terms used herein have the same meanings as are commonly understood by a person skilled in the art. The following reference documents provide one of skills that have general definitions of many terms used herein: Singleton et al., DICTIONARY OF MICROBIOLOGY AND MOLECULAR BIOLOTY (2ded. 1994); THE CAMBRIDGE DICTIONARY OF SCIENCE AND TECHNOLOGY (Walkered., 1988); and Hale & Marham, THE HARPER COLLINS DICTIONARY OF BIOLOGY





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A schematically shows a normalization procedure by curation and pre-processing of microarray data of breast cancer tissue. FIG. 1B schematically shows an identification procedure of prognostic genes in the discovery dataset.



FIGS. 2A-2D show validation results of the prognostic model (frozen samples) in the discovery dataset. FIG. 2A shows that prognostic indexes of all patients using the prognostic model (frozen samples) were divided into four and classified into four prognostic groups, and the separation of the observed survival probability of each prognostic group was verified. The observed survival probability was compared with the predicted survival probability. FIG. 2B shows that observed survival probabilities were compared with predicted survival probabilities using the prognostic models in overall patients. FIG. 2C shows that overall patients were divided into four groups with respect to the most influential p.mean, and then the concurrence between the observed survival probability and the predicted survival probability by the prognostic model (frozen samples) in each group was verified. FIG. 2D shows that the concurrence between the observed survival probability and the predicted survival probability with respect to 5-year survival probability was verified.



FIGS. 3A-3C show validation results of the prognostic model (frozen samples) in validation set 1. The validation method was the same as that in the discovery dataset. FIG. 3A shows validation results on the determination, and FIG. 3B shows validation results on the calibration of the overall time period of observation. FIG. 3C shows validation results on the calibration of 5-year survival probability.



FIGS. 4A-4C show validation results of the prognostic model (frozen samples) in validation set 2. The validation method was the same as that in the discovery dataset. FIG. 4A shows validation results on determination, and FIG. 4B shows validation results on calibration of the overall time period of observation. FIG. 4C shows validation results on the calibration of 5-year survival probability.



FIG. 5 shows validation results of the prognostic model (frozen samples) in validation set 3. The validation method was the same as that in the discovery dataset.



FIGS. 6A-6I show correlation measurement results between FFPE sample (siemens, vertical axis)/frozen sample (horizontal axis) with respect to selected p-gene, and gene names and correlation values (cor) are shown, respectively.



FIGS. 7A-7F show correlation measurement results between FFPE sample (siemens, vertical axis)/frozen sample (horizontal axis) with respect to selected i-gene, and gene names and correlation values (cor) are shown, respectively.



FIG. 8 shows a Kaplan-Meier plot of DIMES (Distant Metastasis Free Survival) over the course of 15 years for ER+ breast cancer patients who had been treated with tamoxifen for 5 years after surgery.



FIG. 9 shows the C-indices of BTN3A2, RRM2, and the combination of BTN3A2 and RRM2, respectively.





DETAILED DESCRIPTION OF THE ILLUSTRATED EMBODIMENTS

The present invention will be described in more detail.


The present invention provides a genetic marker for predicting or diagnosing the prognosis of a breast cancer patient and a use thereof. More specifically, the present invention provides a genetic marker of T cell receptor beta constant 1 (TRBC1), butyrophilin, subfamily 3, member A2 (BTN3A2), or major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1), for predicting or diagnosing the prognosis of breast cancer, especially, early-stage breast cancer. In addition, the present invention provides a method for predicting the prognosis of breast cancer, the method comprising: (a) isolating mRNA from a sample; (b) measuring the mRNA expression level of at least one gene selected from the group consisting of T cell receptor beta constant 1 (TRBC1), butyrophilin, subfamily 3, member A2 (BTN3A2), and major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1); and (c) normalizing the mRNA expression level of the gene, wherein the overexpression of the gene is determined as indicating good prognosis.


Herein, T cell receptor beta constant 1 (TRBC1), butyrophilin, subfamily 3, member A2 (BTN3A2), or major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1) may serve as a genetic marker in the present invention. They may be used for predicting or diagnosing the prognosis of early-stage breast cancer by being independently selected, or by a combination of two or three genes. Each gene may be a sequence thereof known in the art or a sequence of the synonym thereof, and preferably a sequence thereof derived from a human being. More preferably, TRBC1 may be Genbank Accession No. BC030533.1; BTN3A2 may be Genbank Accession No. NM_007047.3; and HLA-DPA1 may be Genbank Accession No. NM_001242524.1, NM_001242525.1, or NM_033554.3. The synonym and the sequence of each gene may be searched in Genbank or Swissprot.


Herein, the breast cancer may be an invasive breast cancer, or breast cancer stage I, II, or III. Herein, the breast cancer may be estrogen receptor positive (ER+).


As used herein, the term “prognosis” refers to symptoms in the future or prospects of progress determined by disease diagnose. The prognosis in cancer patients generally means whether or not the cancer is metastatic within a certain period or the survival period after the occurrence of cancer or the surgical procedure. The prediction of prognosis (or the diagnosis of prognosis) presents clues for future direction of breast cancer treatment, especially, including whether chemotherapy treatment is performed on early-stage breast cancer patients, and thus is a very important clinical challenge. The prediction of prognosis includes predictions of the response of patients to disease therapeutic agents and therapeutic progress.


Herein, the sample may be a breast cancer tissue of a breast cancer patient. The breast cancer tissue may contain some normal cells, and may preferably be a formalin-fixed paraffin-embedded (FFPE) sample of the breast cancer tissue including cancer cells of the patient.


The marker for predicting or diagnosing the prognosis of breast cancer according to the present invention may be detected through polymerase chain reaction (PCR) amplification of a target gene. The detection of the target gene according to the present invention is preferably a detection of the expression level of the target gene, and more preferably a quantitative detection of the expression level of the target gene. For the detection of the expression level, the isolation of mRNA in the sample tissue and the synthesis of cDNA from the mRNA may be needed. For the isolation of mRNA, the isolation methods of mRNA from a sample, which are known in the art, may be employed, and since the sample is preferably an FFPE sample, the isolation methods of mRNA, which are appropriate for the FFPE sample, may be employed. For the synthesis of cDNA, the cDNA synthesis methods using mRNA as a template, which are known in the art, may be employed. Preferably, the detection of the marker for predicting or diagnosing the prognosis of breast cancer according to the present invention is a quantitative detection of the mRNA expression in the FFPE sample, and thus may be the detection by the isolation method of mRNA and the reverse transcription quantitative polymerase chain reaction (RT-qPCR) method with respect to the FFPE sample.


Herein, the detection may be a measurement of the mRNA expression level. The measurement of the expression level may be conducted using the methods known in the art, but may be conducted by an optical quantitative analysis system using a probe labeled with a reporter fluorescent dye and/or a quencher fluorescent dye. The measurement may be conducted using a commercially available system, for example, ABI PRISM 7700™ Sequence Detection System™, Roche Molecular BiochemicalsLightcycler, and an operating system affiliated therewith, such as software. The measurement data may be expressed as a measurement value or a threshold cycle (Ct or Cp). The point in which the measured fluorescent value is first recorded as being statistically significant is defined as the threshold cycle. The threshold cycle is inversely proportional to the value at the beginning in which targets of detection are present as a template of PCR, and thus the lower threshold cycle value indicates the presence of the quantitatively increased targets of detection.


Meanwhile, the present invention provides a composition for predicting or diagnosing the prognosis of breast cancer, the composition comprising a primer pair as an active ingredient, wherein the primer pair is for at least one gene selected from the group consisting of TRBC1, BTN3A2, and HLA-DPA1, and wherein the primer pair is capable of amplifying a target gene through PCR amplification.


As used herein, the term “primer” refers to an oligonucleotide, and the primer may act as an initial point of synthesis in the condition where the synthesis of the primer extension products that are complementary to a nucleic acid chain (template) is induced, that is, the presence of nucleotides and polymerases such as DNA polymerases, and appropriate temperature and pH. Preferably, the primer is deoxyribonucleotide, and has a single chain. The primer used herein may include naturally occurring dNMPs (that is, dAMP, dGMP, dCMP, and dTMP), modified nucleotides, or non-naturally occurring nucleotides. Also, the primer may include ribonucleotides.


The primer of the present invention may be an extension primer that is annealed to a target nucleic acid to form a sequence complementary to the target nucleic acid by template-dependent nucleic acid polymerase. The extension primer is extended to a site at which an immobilized probe is annealed, and thus occupies the site at which the probe is annealed.


The extension primer used herein includes a hybrid nucleotide sequence complementary to the first site of the target nucleic acid. The term “complementary” refers to being sufficiently complementary such that primers or probes are selectively hybridized with the target nucleic acid sequence under predetermined annealing or hybridizing conditions, encompassing the terms “substantially complementary” and “perfectly complementary”, and means preferably “perfectly complementary”. As used herein, the term “substantially complementary sequence” in conjunction with the primer sequence, means including the sequence that is partially un-identical to the sequence of the comparative target within the range in which the sequence is annealed to a particular sequence to serve as a primer, as well as the perfectly identical sequence.


The primer needs to be long enough to prime the synthesis of extension products in the presence of polymerases. The appropriate length of the primer varies depending on several factors, such as temperature, field of application, and primer source, but the primer generally has 15˜30 nucleotides. Short primer molecules generally require lower temperatures in order to form sufficiently stable hybrid complexes together with templates. The term “annealing” or “priming” refers to the apposition of oligodeoxynucleotide or nucleic acid to the template nucleic acid, and the apposition enables the polymerase to polymerize nucleotides to form a nucleic acid molecule, which is complementary to the template nucleic acid or a portion thereof.


The sequences of primers do not need to have a perfectly complementary sequence to some sequences of templates. The primers are good enough so long as they have sufficient complementarity within the scope in which they can perform their inherent actions through hybridization with the template. Therefore, the primer of the present invention does not need to have a perfectly complementary sequence to the foregoing nucleotide sequence as a template. The primers are good enough so long as they have sufficient complementarity within the scope in which they can perform their actions through hybridization with the gene sequence. This design of the primers may be easily carried out by a person skilled in the art with reference to the foregoing nucleotide sequences. For instance, the design of the primers may be carried out using computer programs for primer design (e.g., PRIMER 3 program).


The present invention provides a kit for predicting or diagnosing the prognosis of breast cancer, the kit comprising the primer pair. The kit of the present invention may further comprise tools and/or reagents known in the art which are used for PCR, RNA separation in samples, and cDNA synthesis, in addition to primer pairs allowing PCR amplification of TRBC1, BTN3A2, and/or HLA-DPA1. The kit of the present invention may further comprise, if necessary, tubes which are to be used to mix respective components, well plates, instruction manuals describing how to use, or the like.


The present invention provides a method for calculating a predictive value of the prognosis of breast cancer to provide information necessary for predicting or diagnosing the prognosis of a breast cancer patient, the method comprising:


(a) isolating mRNA from a sample;


(b) measuring the mRNA expression level of at least one gene selected from the group consisting of T cell receptor beta constant 1 (TRBC1), butyrophilin, subfamily 3, member A2 (BTN3A2), and major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1);


(c) normalizing the mRNA expression level of the gene;


(d) inserting the normalized value into a predetermined calculation formula to calculate a numerical value; and


(e) determining the breast cancer prognosis as being good(favorable) or poor(unfavorable) depending on the numerical value.


The present invention also provides a method for calculating a predictive value of the prognosis of breast cancer to provide information necessary for predicting or diagnosing the prognosis of a breast cancer patient, the method comprising:


(a) isolating mRNA from a sample of the breast cancer patient;


(b) measuring the mRNA expression level of at least one gene selected from the i-gene group consisting of TRBC1 (T cell receptor beta constant 1), BTN3A2 (butyrophilin, subfamily 3, member A2), and HLA-DPA1 (major histocompatibility complex, class II, DP alpha 1), and the mRNA expression level of at least one gene selected from the p-gene group consisting of AURKA (Aurora Kinase A), CCNB2 (Cyclin B2), FOXM1 (Forkhead box protein M1), MMP11 (Matrix Metallopeptidase 11), PTTG1 (Pituitary Tumor-Transforming 1), RACGAP1 (Rac GTPase Activating Protein 1), RRM2 (Ribonucleotide Reductase M2), TOP2A (Topoisomerase II Alpha) and UBE2C (Ubiquitin-Conjugating Enzyme E2C);


(c) normalizing the mRNA expression level of the gene; and


(d) calculating a predictive value of the prognosis of breast cancer by combining the normalized value of the gene,


wherein the predictive value indicates the prognosis of breast cancer as being good or poor.


Herein, the expression level of the target gene for detection needs to be normalized since the overall gene expression level may vary depending on the target patient or sample. The normalization is made through the difference in the expression of the gene, which can indicate the difference from the basic expression level, and preferably, the normalization may be carried out by measuring the expression levels of one to five genes selected from C-terminal-binding protein 1 (CTBP1), TATA-binding protein (TBP), hydroxymethylbilane synthase (HMBS), cullin 1 (CUL1), and ubiquilin-1 (UBQLN1) (or the mean of expression levels of selected multiple genes), and calculating the ratio thereof.


Meanwhile, the present invention provides a method for breast cancer prognosis prediction and diagnosis, the method comprising a step of using a primer pair to measure the mRNA expression level of a selected gene from a sample of a breast cancer patient,


wherein the primer pair is for at least one gene selected from the group consisting of TRBC1, BTN3A2, and HLA-DPA1 and


wherein the primer pair is capable of amplifying a target gene through PCR amplification.


Furthermore, the present invention provides a primer pair for at least one gene selected from the group consisting of TRBC1, BTN3A2, and HLA-DPA1, wherein the primer pair is capable of amplifying a target gene through PCR amplification.


Still furthermore, the present invention provides a use of a primer pair for preparing a preparation for predicting the prognosis of breast cancer, wherein the primer pair is for at least one gene selected from the group consisting of TRBC1, BTN3A2, and HLA-DPA1, and wherein the primer pair is capable of amplifying a target gene through PCR amplification.


Some embodiments of the present invention provide a method for diagnosing a prognosis of breast cancer and treating breast cancer in a breast cancer patient, the method comprising the steps of:


collecting a sample from the breast cancer patient;


isolating mRNA from the sample of the breast cancer patient;


measuring a first mRNA expression level for the mRNA of at least one i-gene selected from the group consisting of BTN3A2 (butyrophilin, subfamily 3, member A2), T cell receptor beta constant 1 (TRBC1), and major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1), and a second mRNA expression level for the mRNA of at least one p-gene selected from the group consisting of the p-genes of the following Table 1;


normalizing the first and second mRNA expression levels to determine a normalized value;


diagnosing the prognosis of breast cancer patient by using the determined normalized value of the first and second mRNA expression levels, wherein an overexpression of the i-gene indicates a good prognosis of breast cancer, while an overexpression of the p-gene indicates a poor prognosis of breast cancer; and


treating the diagnosed breast cancer patient by administering at least one of an anti-cancer agent, a surgery and a radiation therapy.









TABLE 1







List of p-genes









No.
Gene ID
Gene Name












1
APITD1
apoptosis-inducing, TAF9-like domain 1


2
BID
BH3 interacting domain death agonist


3
BUB1B
BUB1 mitotic checkpoint serine/threonine




kinase B


4
BUB1
BUB1 mitotic checkpoint serine/threonine kinase


5
CKS1B
CDC28 protein kinase regulatory subunit 1B


6
CKS2
CDC28 protein kinase regulatory subunit 2


7
DLGAP5
discs, large (Drosophila) homolog-associated




protein 5


8
POLA1
polymerase (DNA directed), alpha 1,




catalytic subunit


9
POLA2
polymerase (DNA directed), alpha 2,




accessory subunit


10
DSCC1
DNA replication and sister chromatid cohesion 1


11
DNA2
DNA replication helicase/nuclease 2


12
E2F8
E2F transcription factor 8


13
ERCC6L
excision repair cross-complementation




group 6-like


14
FBXO5
F-box protein 5


15
FANCI
Fanconi anemia, complementation group I


16
GADD45GIP1
growth arrest and DNA-damage-inducible,




gamma interacting protein 1


17
GINS1
GINS complex subunit 1 (Psf1 homolog)


18
GINS2
GINS complex subunit 2 (Psf2 homolog)


19
MAD2L1
MAD2 mitotic arrest deficient-like 1 (yeast)


20
MAD2L1BP
MAD2L1 binding protein


21
MIS18A
MIS18 kinetochore protein A


22
MYBL2
v-myb avian myeloblastosis viral oncogene




homolog-like 2


23
NAA50
N(alpha)-acetyltransferase 50, NatE catalytic




subunit


24
NEK2
NIMA-related kinase 2


25
NSL1
NSL1, MIS12 kinetochore complex component


26
PBK
PDZ binding kinase


27
RAB11A
RAB11A, member RAS oncogene family


28
RAD51C
RAD51 paralog C


29
RAD54B
RAD54 homolog B (S. cerevisiae)


30
RANBP1
RAN binding protein 1


31
RALA
v-ral simian leukemia viral oncogene




homolog A (ras related)


32
RACGAP1
Rac GTPase activating protein 1


33
SSNA1
Sjogren syndrome nuclear autoantigen 1


34
STAMBP
STAM binding protein


35
SSSCA1
Sjogren syndrome/scleroderma autoantigen 1


36
TAF2
TAF2 RNA polymerase II, TATA box binding




protein (TBP)-associated factor, 150 kDa


37
TIPIN
TIMELESS interacting protein


38
TIPRL
TOR signaling pathway regulator


39
TRIAP1
TP53 regulated inhibitor of apoptosis 1


40
TTK
TTK protein kinase


41
ZWINT
ZW10 interacting kinetochore protein


42
ASPM
asp (abnormal spindle) homolog,




microcephaly associated (Drosophila)


43
AURKA
aurora kinase A


44
AURKB
aurora kinase B


45
BRD7
bromodomain containing 7


46
CSNK2A1
casein kinase 2, alpha 1 polypeptide


47
CDC20
cell division cycle 20


48
CDC25C
cell division cycle 25C


49
CENPA
centromere protein A


50
CENPE
centromere protein E, 312 kDa


51
CENPF
centromere protein F, 350/400 kDa


52
CENPI
centromere protein I


53
CENPM
centromere protein M


54
CENPN
centromere protein N


55
CENPU
centromere protein U


56
CEP55
centrosomal protein 55 kDa


57
CHEK1
checkpoint kinase 1


58
CDT1
chromatin licensing and DNA replication factor 1


59
C11orf80
chromosome 11 open reading frame 80


60
CCNA2
cyclin A2


61
CCNB1
cyclin B1


62
CCNB2
cyclin B2


63
CCNE2
cyclin E2


64
CDK1
cyclin-dependent kinase 1


65
CDKN3
cyclin-dependent kinase inhibitor 3


66
CKAP5
cytoskeleton associated protein 5


67
DTL
denticleless E3 ubiquitin protein ligase




homolog (Drosophila)


68
DCTN2
dynactin 2 (p50)


69
DYNLT1
dynein, light chain, Tctex-type 1


70
ECD
ecdysoneless homolog (Drosophila)


71
ECT2
epithelial cell transforming 2


72
EIF4G1
eukaryotic translation initiation factor 4 gamma, 1


73
EIF4EBP1
eukaryotic translation initiation factor 4E




binding protein 1


74
EZR
ezrin


75
FEN1
flap structure-specific endonuclease 1


76
FOXM1
forkhead box M1


77
GSK3B
glycogen synthase kinase 3 beta


78
HMGN5
high mobility group nucleosome binding domain 5


79
INTS7
integrator complex subunit 7


80
KIF11
kinesin family member 11


81
KIF14
kinesin family member 14


82
KIF20A
kinesin family member 20A


83
KIF23
kinesin family member 23


84
KIF2C
kinesin family member 2C


85
KIF4A
kinesin family member 4A


86
KIFC1
kinesin family member C1


87
MIF
macrophage migration inhibitory factor




(glycosylation-inhibiting factor)


88
MELK
maternal embryonic leucine zipper kinase


89
MED1
mediator complex subunit 1


90
MCM10
minichromosome maintenance complex




component 10


91
MCM2
minichromosome maintenance complex




component 2


92
MCM6
minichromosome maintenance complex




component 6


93
MAP2K1
mitogen-activated protein kinase kinase 1


94
MSH6
mutS homolog 6


95
MLF1
myeloid leukemia factor 1


96
NCAPG
non-SMC condensin I complex, subunit G


97
NUSAP1
nucleolar and spindle associated protein 1


98
NUP155
nucleoporin 155 kDa


99
NUP93
nucleoporin 93 kDa


100
ORC4
origin recognition complex, subunit 4


101
ORC5
origin recognition complex, subunit 5


102
PIN1
peptidylprolyl cis/trans isomerase,




NIMA-interacting 1


103
PIK3R4
phosphoinositide-3-kinase, regulatory subunit 4


104
PTTG1
pituitary tumor-transforming 1


105
PTTG3P
pituitary tumor-transforming 3, pseudogene


106
PLK1
polo-like kinase 1


107
PLK4
polo-like kinase 4


108
PRIM1
primase, DNA, polypeptide 1 (49 kDa)


109
PA2G4
proliferation-associated 2G4, 38 kDa


110
LEPREL4
leprecan-like 4


111
PSMC3
proteasome (prosome, macropain) 26S subunit,




ATPase, 3


112
PSMC6
proteasome (prosome, macropain) 26S subunit,




ATPase, 6


113
PSMD10
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 10


114
PSMD12
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 12


115
PSMD14
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 14


116
PSMD2
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 2


117
PSMD3
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 3


118
PSMD4
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 4


119
PSMD6
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 6


120
PSMD7
proteasome (prosome, macropain) 26S subunit,




non-ATPase, 7


121
PSMA1
proteasome (prosome, macropain) subunit,




alpha type, 1


122
PSMA2
proteasome (prosome, macropain) subunit,




alpha type, 2


123
PSMA3
proteasome (prosome, macropain) subunit,




alpha type, 3


124
PSMA4
proteasome (prosome, macropain) subunit,




alpha type, 4


125
PSMA6
proteasome (prosome, macropain) subunit,




alpha type, 6


126
PSMA7
proteasome (prosome, macropain) subunit,




alpha type, 7


127
PSMB3
proteasome (prosome, macropain) subunit,




beta type, 3


128
PSMB5
proteasome (prosome, macropain) subunit,




beta type, 5


129
PSMB7
proteasome (prosome, macropain) subunit,




beta type, 7


130
PRMT1
protein arginine methyltransferase 1


131
PPP2R3B
protein phosphatase 2, regulatory subunit B″, beta


132
PPP3CA
protein phosphatase 3, catalytic subunit,




alpha isozyme


133
PRC1
protein regulator of cytokinesis 1


134
RRM2
ribonucleotide reductase M2


135
RPS6KB1
ribosomal protein S6 kinase, 70 kDa,




polypeptide 1


136
SPAG5
sperm associated antigen 5


137
SKA1
spindle and kinetochore associated complex




subunit 1


138
STMN1
stathmin 1


139
SLBP
stem-loop binding protein


140
SMC2
structural maintenance of chromosomes 2


141
SMC4
structural maintenance of chromosomes 4


142
SMC5
structural maintenance of chromosomes 5


143
TERF1
telomeric repeat binding factor (NIMA-




interacting) 1


144
TXNL4A
thioredoxin-like 4A


145
TRIP13
thyroid hormone receptor interactor 13


146
TOP2A
topoisomerase (DNA) II alpha 170 kDa


147
TFDP2
transcription factor Dp-2 (E2F dimerization




partner 2)


148
TACC3
transforming, acidic coiled-coil containing




protein 3


149
TUBB3
tubulin, beta 3 class III


150
TUBB4B
tubulin, beta 4B class IVb


151
TUBB
tubulin, beta class I


152
TSG101
tumor susceptibility 101


153
UBE2C
ubiquitin-conjugating enzyme E2C


154
UBE2L3
ubiquitin-conjugating enzyme E2L 3


155
UBE2S
ubiquitin-conjugating enzyme E2S


156
USP9X
ubiquitin specific peptidase 9, X-linked


157
VRK1
vaccinia related kinase 1


158
ZFHX3
zinc finger homeobox 3


159
ZWILCH
zwilch kinetochore protein


160
MMP11
Matrix Metallopeptidase 11









In some exemplary embodiments of the present invention, the i-gene may be BTN3A2 (butyrophilin, subfamily 3, member A2).


In another exemplary embodiments of the present invention, the p-gene may be selected from the group consisting of AURKA (Aurora Kinase A), CCNB2 (Cyclin B2), FOXM1 (Forkhead box protein M1), MMP11 (Matrix Metallopeptidase 11), PTTG1 (Pituitary Tumor-Transforming 1), RACGAP1 (Rac GTPase Activating Protein 1), RRM2 (Ribonucleotide Reductase M2), TOP2A (Topoisomerase II Alpha) and UBE2C (Ubiquitin-Conjugating Enzyme E2C).


The steps of measuring mRNA expression levels of target genes and normalizing the measured mRNA expression levels are described above and well known in the art, while the step of treating the diagnosed breast cancer patient may be conducted by administering at least one of an anti-cancer agent, a surgery and a radiation therapy which is considered appropriate by one skilled in a cancer therapy.


The following references are made in the above-mentioned nucleotide and protein works (Maniatis et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. (1982); Sambrook et al., Molecular Cloning: A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory Press(1989); Deutscher, M., Guide to Protein Purification Methods Enzymology, vol. 182. Academic Press. Inc., San Diego, Calif. (1990); Ausubel et al., Current Protocols of Molecular Biology, John Wiley and Sons (1997); Rupp and Locker, Lab Invest. 56: A67 (1987); De Andres et al., BioTechniques 18: 42044 (1995); Held et al., Genome Research 6:986-994 (1996); T. E. Godfrey et al. J. Molec. Diagnostics 2: 84-91 (2000); K. Specht et al., Am. J. Pathol. 158: 419-29 (2001)).


Accordingly, the present invention provides a genetic marker for predicting or diagnosing the prognosis of early-stage breast cancer. The genetic marker of the present invention enables the prediction or diagnosis of the prognosis of a breast cancer patient, and thus can be favorably used to present clues for the future direction of breast cancer treatment, including the determination on whether anticancer treatment is needed.


Hereinafter, the present invention will be described in detail with reference to the following examples.


However, the following examples are merely for illustrating the present invention and are not intended to limit the scope of the present invention.


For examples in the present specification, the disclosures of Korean Patent Application Publication No. 10-2012-0079295 and PCT Publication No. WO2012093821A2 are entirely incorporated into the present specification by reference, and the level of the technical field to which the present pertain and the details of the present invention are explained more clearly.


Methods


Collection of Expression Profile in Early-Stage Breast Cancer Tissue


Expression profiles and clinical information obtained using frozen cancer tissues of early-stage breast cancer patients were collected from open database GEO (http://www.ncbi.nlm.nih.gov/geo). Each of a total nine independent expression profile sets was a relatively big dataset composed of 100 or more samples, and was made in order to conduct researches on prognosis of early-stage breast cancer patients (2, 4, 9, 10, 13, 25, 32, 33). Of these, eight datasets were made using microarray platform, Affymetrix U133A, and the other one dataset was manufactured using Agilent Hu25K. In most cases, important clinical information (age, sex, cancer size, cancer metastasis state, and degree of cancer differentiation) and survival information of patients were collected together. Of the eight datasets made by Affymetrix U133A, six datasets included survival information about distant metastasis (distant metastasis free survival), and the other two datasets included survival information about overall survival. Agilent data included survival information about distant metastasis. Survival analysis on the base of whether distant metastasis occurred or not was performed based on the facts that distant metastasis is the most decisive event in deciding prognosis, the distant metastasis is determined by unique characteristics of cancer, and the largest number of patients had information on the distant metastasis in the collected data. Through comparision of the collected information of all patients, the expression profiles of duplicated 186 cases were removed, and a total of 1,861 unique cases were researched. With respect to seven datasets made by the same platform (Affymetrix U133A), raw files (.CEL) of the expression profiles of corresponding patients were combined together, and then normalized. The normalization was conducted by the rma method (background correction: rma, normalization: quantile, summarization: medianpolish). For the normalization, custom CDF (http://brainarray.mbni.med.umich.edu/Brainarray/) ENTREZG version 13 developed by Manhong Dai et al., was used (34). After the normalization, the expression level of each probe was converted from the one-color expression level into, for example, the two-color expression level by subtracting the mean value of each probe in the discovery dataset. Of the total eight normalized dataset, five datasets were combined together, and then used as the discovery dataset, while two datasets were separately combined, and then used as validation dataset 1, and the other dataset was used as validation dataset 2. Agilent dataset was also used as validation dataset 3.


Prognosis of Patients and Definition of ER Status


In order to identify genes associated with prognosis of patients, the collected patients were classified into a good (favorable) prognostic group and a poor (unfavorable) prognostic group. In general clinical practice, five-year survival or metastasis information is used for such a classification. In other words, the prognosis is poor if metastasis or death occurs within five years, and the prognosis is good if metastasis or death does not occur within five years. The distribution of survival times of metastatic patients was investigated using patient information in the discovery dataset. About 73% or more of the metastatic patients had metastasis within 5 years, and less than 7% of them had metastasis after 10 years. Based on this fact, among patients in the discovery dataset, 217 patients who had metastasis within five years were classified as the “poor prognostic group”, while 281 patients who had no metastasis for ten years or longer were classified as the “good prognostic group”. As a result of classification, the median survival time of the good prognostic group was 2.4 years, while the median survival time of the poor prognostic group was 12.9 years. Error due to unreliable survival information could be minimized by clearly dividing the poor prognostic group and the good prognostic group. The expression or not of the estrogen receptor (ER) is the most commonly used criterion when breast cancer patients are classified by subtypes. In clinical practice, the breast cancer patients are classified into ER+ and ER− through ER immunohistochemistry (IHC) readout results by pathologists. Considering that about 200 patients had no ER IHC information in the collected discovery dataset, and the determination of ER IHC was made for each of five datasets constituting the discovery dataset, independently, the ER status was determined using the mRNA expression level of ESR1 gene in the expression profile for each patient. For the patients having ER IHC information, region of convergence (ROC) analysis was conducted using the ER IHC information and the ERS1 mRNA expression level. Through a comparison between the ER IHC results and the ERS1 mRNA expression level, the cutoff for ER status was determined at the point where the accuracy was highest (accuracy=0.88). The cases showing higher expression levels than the cutoff were classified as ER+, and the cases showing lower expression levels than the cutoff were classified as ER−. In the discovery dataset, 864 patients were designated as ER+ and 240 patients as ER−.


Selection of Prognostic Genes


The good prognosis group was defined as ER+, and the poor prognosis group was defined as ER− in the discovery dataset. 275 patients had good prognosis, while 218 patients had poor prognosis. Through significant analysis of microarray (SAM), genes showing a difference in the expression level among the prognostic groups were investigated. By using the q-value of the SAM analysis results, overexpressed genes in the good prognosis group and the poor prognosis group were selected. As a result of combining the selected genes, a total of 302 gene sets that were not duplicated have been made, and a cluster analysis for identifying expression patterns of these genes was conducted using the principal component analysis (PCA) method. Two principal components were selected, and in order to investigate biological functions related to each principal component, GO function analysis was performed for each cluster.


As a result of GO analysis, principal component 1 was shown to concentrate on the proliferation, whereas principal component 2 was shown to concentrate on the immune response. From genes belonging to two principal components involved in the proliferation and the immune response, genes showing the highest expression level between two prognosis groups were selected, respectively. The genes of the respective gene sets are named as p-gene in view of representing expression patterns of the proliferation and i-gene in view of representing expression patterns of the immune response, respectively.


Configuration of Prognostic Model Using Parametric Survival Analysis


Regression analysis in which expression levels of p-gene and i-gene were covariates was performed using the accelerated failure time (AFT) model of parametric survival models. Four p-genes were converted into p.mean by calculating the mean value for each patient, and five i-genes were also converted into i.mean by calculating the mean value of each patient before application. AFT model is specified as:

Ti=T0 exp(β1χ12χ2+ . . . +βqχqi  Equation 1


wherein, Ti is the survival time of the i-th object; T0 is the baseline survival time; χi is the vector of the covariates (j=1, 2, . . . , q); β is the coefficient of corresponding covariates; and ε is the error. In this model, the covariates synergistically influence the baseline survival time, and thus this model is called the accelerated failure time (AFT) model in the industries in which this model is frequently used. The synergistic effect on the survival time, Φ=β1χ1χ12χ2+ . . . +βqχq is called the acceleration factor. Where the natural logarithm in Equation (1) is calculated,

log Ti=log T01χ12χ2+ . . . +βqχq+ε*  Equation 2


Thus, the AFT model has the same form as the general linear regression model. However, since the dependent variable log T does not normally distribute and the survival analysis data always includes censored cases, which are not permitted in the linear regression model, Equation (2) cannot be processed like in the linear regression model. Since the distribution of ε* of Equation (2) may vary for different datasets, unlike where the normal distribution is assumed in the linear regression model, the practical statistical processing is inconvenient. In order to overcome this, log T0 and ε* are modified, and expressed as follows:

log Ti=log T01χi2χ2+ . . . +βqχq+σW  Equation 3


wherein W follows the distribution of log T, and the variance thereof is fixed as the value of the normalized distribution. Here, σ is the constant as the scale parameter, and the value thereof is determined depending on the dataset.


Various candidate prognostic models were fit to Weibull, loglogistic, and lognormal distributions, using AFT model, and then the most appropriate model was selected. For the risk distribution for AFT model, the hazard function, which can be obtained by creating the generation life table of survival information in the discovery dataset, was used. Since the hazard function obtained by the generation life table is shown to have a unimodal form, it was predicted that the Weibull, loglogistic, and lognormal distributions would be well appropriate. The final model was selected in consideration of Akaike's information criterion (AIC) and the R square (R2).


Validation of Prognostic Model


The performances of the selected model were assessed according to their “calibration” and “discrimination” aspects. “Calibration” is the degree of concurrence between the predicted survival probability produced by the prognostic model and the actually observed survival probability, and “Discrimination” is the ability of the prognostic model to correctly separate the given patient group into different prognostic groups. Herein, the actually observed survival probability refers to the value obtained by Kaplan-Meier method. The AFT-based prognostic model was used to obtain survival probabilities of all time zones for each patient. The survival probability predicted by the model was compared with the survival probability by Kaplan-Meier method. In order to obtain the predicted survival probabilities for the overall time period like in Kaplan-Meier, survival probability curves of all patients were obtained by calculating the mean survival probability for each time zone from 0 to 25 years by the 0.1 year unit. Together with the comparison of survival probability for the overall survival time, the 5-year survival probability was also compared. The 5-year predicted survival probability of patients produced by using the prognostic model in the given dataset was compared with, as the observed value, the 5-year survival probability calculated by using Hare which is the hazard regression analysis.


For “discrimination”, the prognostic indexes of all patients in the given dataset were divided into four sections, and survival probabilities of patients for the respective sections were compared by the KM graph. The prognostic index is the dependent variable in the survival model. The more clearly the KM graphs of the four prognostic groups are separated, the better the discrimination power of the model.


Both “calibration” and “determination” for the discovery dataset and three independent validation datasets were investigated.


Important R packages used in statistical analysis are as below:


affy: pre-processing of .CEL files using rma algorithm


samr: identifying genes showing the difference in the expression level between prognostic groups


GOstats: investigating function associated with selected gene set


KMsurv: creating a life table using survival data of the discovery dataset


rma: estimating coefficients of prognostic model using AFT model and conducting calibration on the model.


Selection of Gene Sets Applicable to FFPE Sample


RNA extracted from formalin-fixed paraffin embedded (FFPE) tissues and samples has not been accepted in the expression analysis due to many procedures causing the obstruction to RNA stability, such as immobilization causing cross-linking between tissues in the treatment process of tissues or samples. Herein, in order to develop a method for breast cancer prognosis prediction, suitable for FFPE samples, based on the actual breast cancer treatment procedure, gene sets were selected by placing a higher priority on genes having higher interqurtile ranges (IQR) and genes having higher mean expression levels in the order of higher degree of contribution, from p-genes (proliferation-related gene set) representing the expression pattern of proliferation and i-genes (immune response-related gene set) representing the expression pattern of the immune response, which were obtained according to the principal component analysis and function analysis with respect to the principal component analysis (GO function analysis or GO analysis).


As used herein, several genes having the same pattern in p-gene and i-gene were selected since the microarray data has a limitation in measuring accurate expression levels and the mean expression level for several genes rather than the expression level of one gene representing the pattern can represent the actual expression pattern better.


Measurement of Gene Correlation Between FFPE Sample and Frozen Sample and Selection of Genes


27 types of FFPE and frozen samples collected from the same patients were secured, and RNA was extracted from the FFPE or frozen samples by the RNA extraction method. The expression levels of 32 types of genes selected were measured by using the extracted RNA as a template.


The gene expression levels need to be normalized due to the difference thereof between individuals. Therefore, the expression levels of five genes selected as a normalization gene, that is, of C-terminal-binding protein 1 (CTBP1), TATA-binding protein (TBP), hydroxymethylbilane synthase (HMBS), cullin 1 (CUL1), and ubiquilin-1 (UBQLN1), were normalized, and then the gene expression correlation between FFPE sample and frozen sample was measured.


Based on the correlation measurement results and the expression level results for each gene, the genes that have a high correlation rate and various gene expression distributions between samples were finally selected as highly reliable genes for early-stage breast cancer prognosis prediction.


Results


Selection of Prognostic Genes for Prognostic Model


Five independent breast cancer datasets composed of expression profiles of early-stage breast cancer tissue were pooled into a discovery dataset of 1,104 samples. All patients did not receive chemotherapy, while most of them have little metastasis to axillary lymph nodes (N0 or N−), or have early-stage breast cancer (1st stage or 2nd stage). Among them, 1072 patients with information about distant metastasis were subjected to statistical analysis. In order to find genes associated with prognosis, expression profiles of the good prognostic group (having no metastasis for more than 10 years) and the poor prognostic group (having metastasis within 5 years) were compared. 182 genes showing high expression levels were selected from the good prognostic group, and 120 genes showing high expression levels were selected from the poor prognostic group (results not shown, FDR<0.001).


Principal component analysis was performed on the expression levels of the selected 302 genes. GO function analysis was performed on principal component 1 and principal component 2. Principal component 1 was definitely associated with the proliferation, while principal component 2 was strongly associated with the immune response. Based on this result, genes belonging to principal components 1 and 2 were selected, respective, and thus the prognostic model was allowed to reflect the two expression patterns.


Nine genes that were not only associated with the prognosis but also had the largest expression difference among the groups were selected. The gene selected from principal component 1 representing the proliferation is named p-gene, and the gene selected from principal 2 representing the immune response is named i-gene.


Comparison between ER+ Breast Cancer and ER− Breast Cancer


It is known that the expression or not of the estrogen receptor (ER) is closely associated with the occurrence and development of breast cancer. Two functions shown in the genes selected in association with prognosis, that is, proliferation and immune response are interestingly noticed in the mechanism of cancer. ER− breast cancer was compared with ER+ breast cancer using the selected 16 genes (p-genes and i-genes). In order to display the intensity of each function, the p-genes and i-genes were stratified into three stages (p1, p2, p3 or i1, i2, i3) according to the expression level. Here, p1 was a p-gene group showing the lowest expression level, and was assumed to make the slowest proliferation; p3 was a p-gene group showing the highest expression level, and was assumed to make the highest proliferation; and p2 was a p-gene group showing a moderate expression level, and was assumed to make a moderate level of proliferation. As used herein, it was an i-gene group showing the lowest expression level, and was assumed to make the weakest immune response; i3 was an i-gene group showing the highest expression level, and was assumed to make the strongest immune response; and i2 was considered to show a moderate expression level, and was assumed to do a moderate level of activity.


1,072 cases in the discovery dataset were classified according to the expression levels of p-gene and i-gene, and the intensity of each function according to the ER status was investigated. The proportion of p3 type indicating very active proliferation was higher in ER− breast cancer than in ER+ breast cancer. About 62% of ER− breast cancer cases showed very high p-gene expression levels (p3), while only 18% of ER+ breast cancer cases showed high p-gene expression levels, which supported the previously reported fact that ER− breast cancer tends to be more aggressive than ER+ breast cancer. About 35% of ER+ breast cancer cases showed weak p-genes (p1), while the proportion of p1 was only 9% in ER− breast cancer cases. The active immune response function is another characteristic of ER− breast cancer, and 38% or more of ER− breast cancer cases showed very high i-gene expression levels (i3). Whereas, only 21% of ER+ breast cancer cases showed high i-gene expression levels. It was observed that more active proliferation led to more active immune response in both ER+ and ER−, while ER− breast cancer showed a much more active immune response.


Besides, it was observed that the differentiation grade of the breast cancer was also closely related with the proliferation. Generally, poorly differentiated breast cancer (G3) showed a fast proliferation, whereas well-differentiated breast cancer (G1) showed a slow proliferation. It was observed that the prognosis of patients was also correlated with the proliferation. It was observed that more poor prognostic patients having metastasis within five years were concentrated in the group with a fast proliferation.


In summary, it was found that ER− breast cancer was more active than ER+ breast cancer in view of both the proliferation and the immune response, and it was supposed that the expression level of ER influences the mechanisms of occurrence and development of breast cancer.


Establishment of Prognostic Model


The AFT prognostic model of early-stage breast cancer patient metastasis was created using survival information of the discovery dataset and the selected p-genes and i-genes.


The generation life table on a yearly basis was created using the survival information of the discovery dataset, and the degree of hazard was roughly calculated.


Since the probability of death obtained by the generation life table is shown to have a unimodal form, it was predicted that the Weibull, loglogistic, and lognormal distributions would be well appropriate. The covariates to be included in the prognostic model are p.mean and i.mean. p.mean is the mean in the p-genes, and i.mean is the mean in the i-genes.


As a result of applying three models to the Weibull, loglogistic and lognormal distributions, the lognormal distribution was most appropriate. The final model following the lognormal distribution was selected using Akaikes information criterion (AIC).

log(T)=−0.689×p.mean+0.274×i.mean+3.219  Equation 4


According to the above estimated model, the p.mean, that is, the proliferation has a negative correlation (−0.689, p value=2.47×e−17) with the survival time (T), and thus, more active proliferation shortens the survival time. On the contrary, the i.mean has a positive correlation (0.274, p value=3.69×e−11) with the survival time, which indicates that a more active immune response lengthens the survival time. It could be concluded from the above estimated factors that proliferation plays a decisive role in the breast cancer prognosis, and more active proliferation leads to poor prognosis, while the immune response acts as a defense mechanism against fast proliferation.


Validation of Prognostic Model


The performances of the prognostic model, which was created using expression profiles for 1,072 early-stage breast cancer patients in the discovery dataset, were validated according to “calibration” and “discrimination” aspects. Here, the “calibration” is the degree of concurrence between the predicted survival probability produced through the model and the actually observed survival probability. The actually observed survival probability refers to the survival probability obtained using the Kaplan-Meier method. In addition, the “discrimination” is the ability of the model to correctly separate the patients into different prognostic groups. Validations for the two performances were performed on the discovery dataset developing the model and three independent validation datasets.


In the discovery dataset developing the prognostic model, the prognostic indexes (PI) were divided into four and classified into four prognostic groups. The survival probabilities observed in the four prognostic groups classified by the prognostic index were compared using the KM graph. As a result, it could be seen that four prognostic groups were very well classified, and the predicted survival probability corresponded well with the observed survival probability for each prognostic group.


The KM survival probability and the predicted survival probability produced by the prognostic model were compared with each other using the graph. The survival probabilities for all time zones of all patients were obtained in the prognostic model, and thus, in order to obtain the survival probability curve for the overall time period like the survival curve of KM, survival probability curves were drawn using the mean survival probability for each time zone (from 0 to 25 year, 0.1-year interval) of each patient. The predicted survival probability was slightly higher than the KM survival probability, but they are similar as a whole. In addition to the comparison of survival probability for the overall survival time, the comparison of 5-year survival probability was also conducted. The 5-year survival probability predicted by the model was similar to the actually observed 5-year survival probability. Particularly, as the predicted survival probability was higher, the concurrence between the predicted probability and the observed survival probability was stronger.


For more objective validation, the prognostic model was validated using three independent validation datasets. The first validation dataset was obtained by combining two datasets generated by Affymetrix U133A platform. The second validation dataset was the dataset generated by Affymetric U133A platform, and all were ER+ patients taking tamoxifen for five years. The third validation dataset was the dataset that was used to identify and validate 70 prognostic genes (currently, commercialized as MammaPrint) and generated by Agilent Hu25K platform. Validation datasets 1 and 2 were generated by the same platform as in the discovery dataset, Affymetrix U133A, and the expression levels thereof were normalized together with the discovery dataset. Validation datasets 1 and 2 were assessed in terms of calibration and discrimination aspects, while validation datasets 3 was assessed in terms of only the discrimination aspect due to the normalization of expression levels.


Selection of Gene Sets Applicable to FFPE Sample


32 types of genes were selected by placing a higher priority on genes having higher interqurtile ranges (IQR) and genes having higher mean expression levels in the order of higher degree of contribution, from 182 genes showing high expression levels in the good prognostic group and 120 genes showing high expression levels in the poor prognostic group in the discovery dataset of 1,104 samples.


Measurement of Gene Correlation Between FFPE Sample and Frozen Sample and Selection of Genes


For the measurement of correlation between FFPE sample and frozen sample, samples that secure both FFPE sample and frozen sample from patients or their cancer tissues are needed. With respect to 27 pairs of FFPE and frozen samples thus obtained, the expression levels of 32 types of the selected genes and the correlation between FFPE and frozen samples were measured. Based on the measurement results, genes that have a high correlation rate and various gene expression distributions among samples were found 12 types in p-genes and 15 types in i-genes, which were selected as highly reliable genes for early-stage breast cancer prognosis prediction.


Among the selected genes above, 9 types of p-genes and 6 types of i-genes were selected as genes to be included in the kit for prognosis prediction.


The correlation measurement results for the respective genes are shown in FIGS. 6A-6I and 7A-7F. In each drawing, the solid line represents the equivalent value of the horizontal axis and the vertical axis (slope: 1, that is, the line that connects values of which the horizontal axis value is identical to the vertical axis value).


Meanwhile, among the respective genes, the expression levels of TRBC1, BTN3A2, and HLA-DPA1 were significantly different between the good prognostic group and the poor prognostic group. Through the above analysis, it could be concluded that the expression of TRBC1, BTN3A2, and HLA-DPA1 was significantly increased in the good prognostic group, verifying that their increased expression (overexpression) indicated good breast cancer prognosis.


Improvement in Predicting Prognosis of Breast Cancer Through Gene Combinations


The prognosis of each breast cancer patient is different among breast cancer patients, and the accurate prediction of the prognosis helps the patient to decide an appropriate treatment if needed. The purpose of this study was to investigate the gene expression in tissues of breast cancer patients after surgery so as to find a method for predicting a more accurate prognosis. For this purpose, the gene expression data collected from cancer tissues of early breast cancer patients (a total of 298 patients) was obtained from a public database GEO (http://www.ncbi.nlm.nih.gov/geo). The gene expression dataset as used in this experiment was obtained through microarray from 298 patients with ER+ breast cancer who had been treated with tamoxifen for 5 years after surgery.


For this purpose, RNA was extracted from cancer tissues of each patient and microarray experiment was performed according to the protocol of oligonucleotide microarray (U133A GeneChip; Affymetrix). The raw intensity values of the measured genes were normalized using MAS 5.0 (R/Bioconductor, www.bioconductor.org) and transformed into log 2 values. This dataset derived from this procedures was used to select a combination of genes which is capable of increasing a predictive performance of breast cancer prognosis through the combination of one of the i-genes, BTN3A2 gene and at least another prognostic genes.


Since the distant metastasis of breast cancer has a significant effect on the selection of the type of cancer therapy in comparison with its local or regional recurrence, Distant Metastasis Free Survival (DMFS) was designated as a primary endpoint in this analysis. As used herein, DMFS (Distant Metastasis Free Survival) means an interval between the surgery of a breast cancer patient and the diagnosis of a distant metastasis or death from any cause.


As shown in FIG. 8, it was found that the overall probability rate of DMFS (Distant Metastasis Free Survival) as determined by Kaplan-Meier estimation was 85.1% (81.0%-89.4%) within 5 years and 74.0% (68.5%-79.9%) within 10 years, respectively.


The C-index (concordance index) is an index for evaluating the accuracy of prediction in which the C-index of being closer to 1 is defined as being more accurate in prediction. The predictive accuracy of BTN3A2 gene for distant metastasis was found to be 0.55 through survival analysis.


Another prognosis-predictive indicator is a hazard ratio (HR) calculated via Cox's proportional hazard model. As for the risk of distant metastasis, when the hazard ratio is greater than 1, the risk of distant metastasis increases as the expression level of a target gene increases. On the contrary, when the hazard ratio is less than 1, the risk of distant metastasis decreases as the expression level of a target gene increases. With regard to BTN3A2 gene, its hazard ratio was found to be 0.82 (0.67-1.00), indicating that an increased expression of BTN3A2 gene is associated with a good prognosis.









TABLE 2







Prognostic Values of BTN3A2 gene













Hazard Ratio (HR)












Gene Name
C-Index
HR (95% C.I.)
p-value







BTN3A2
0.55
0.82 (0.67-1.00)
0.046










Since BTN3A2 is known to be an immune-related gene, BTN3A2 gene was named ‘i-gene’ in this analysis. As shown in Table 2, these results suggest that BTN3A2 gene is a prognostic gene which can provide a statistically significant prognostic value.


Although BTN3A2 gene alone may provide a statistically significant prognostic value, the present inventors had sought to find a combination of BT3A2 gene and one or more another genes which leads to a more accurate prognosis prediction.


To determine the prognostic significance of another genes in combination with BTN3A2 gene, bivariate Cox analysis was conducted on BTN3A2 and a total of 13,268 another genes for predicting the probability of distant metastasis, respectively. Bivariate Cox analysis is used to determine whether each of two prognostic factors is an independent prognostic factor when the two prognostic factors are combined. Therefore, when each is confirmed as a significant (p-value<0.05) prognostic factor, the combination of the two factors is considered as a combination which may increase a prediction accuracy.


As a result of such bivariate Cox analysis, a total of 1,673 genes were found to be statistically significant (p-value<0.05) for predicting the probability of distant metastasis when used together with BTN3A2 gene. As used herein, those statistically significant genes are named as ‘prognostic gene’ as described above.


Further, among the prognostic 1,673 genes, a total of 1,635 genes as indicated in the following Table 3 were found to have a higher c-index (i.e., higher prediction accuracy) when used in combination with BTN3A2, in comparison with their single use.


Out of the 1,635 genes which have a higher c-index when used in combination with BTN3A2, a total of 848 genes were showed to be associated with a poor prognosis as their hazard ratio was greater than 1 in bivariate Cox analysis. Conversely, the hazard ratio of the remaining 786 genes was smaller than 1, respectively, indicating that they are associated with a good prognosis. For all of its combinations with each of the 1,635 genes, the hazard ratio of BTN3A2 was smaller than 1, indicating that BTN3A2 is associated with a good prognosis.









TABLE 3







1,635 prognostic genes for use in combination with BTN3A2 gene











prognostic



BTN3A2
gene















Gene
GENE


p-

p-


No.
Symbol
ID
Gene Name
HR
value
HR
value

















1
A2M
2
alpha-2-macroglobulin
0.81
0.044
0.56
0.003


2
AAMDC
28971
adipogenesis associated, Mth938
0.79
0.025
1.33
0.014





domain containing


3
AATF
26574
apoptosis antagonizing transcription
0.82
0.047
1.80
0.021





factor


4
ABAT
18
4-aminobutyrate aminotransferase
0.77
0.007
0.58
0.000


5
ABCA1
19
ATP-binding cassette, sub-family A
0.79
0.015
1.89
0.005





(ABC1), member 1


6
ABCA7
10347
ATP-binding cassette, sub-family A
0.79
0.026
0.63
0.028





(ABC1), member 7


7
ABCB1
5243
ATP-binding cassette, sub-family B
0.78
0.014
0.53
0.000





(MDR/TAP), member 1


8
ABCC1
4363
ATP-binding cassette, sub-family C
0.80
0.031
0.47
0.006





(CFTR/MRP), member 1


9
ABCD4
5826
ATP-binding cassette, sub-family D
0.81
0.039
0.35
0.000





(ALD), member 4


10
ABCE1
6059
ATP-binding cassette, sub-family E
0.77
0.012
2.41
0.002





(OABP), member 1


11
ABCG1
9619
ATP-binding cassette, sub-family G
0.80
0.022
1.50
0.043





(WHITE), member 1


12
ACACB
32
acetyl-CoA carboxylase beta
0.78
0.017
0.60
0.005


13
ACADVL
37
acyl-CoA dehydrogenase, very long
0.77
0.014
0.51
0.004





chain


14
ACAT2
39
acetyl-CoA acetyltransferase 2
0.78
0.011
2.01
0.004


15
ACBD3
64746
acyl-CoA binding domain containing 3
0.78
0.010
1.94
0.004


16
ACBD4
79777
acyl-CoA binding domain containing 4
0.78
0.006
0.52
0.001


17
ACO1
48
aconitase 1, soluble
0.80
0.020
0.39
0.000


18
ACOT13
55856
acyl-CoA thioesterase 13
0.76
0.009
2.62
0.002


19
ACOX1
51
acyl-CoA oxidase 1, palmitoyl
0.77
0.009
1.66
0.022


20
ACTB
60
actin, beta
0.77
0.011
0.52
0.010


21
ACTG1
71
actin, gamma 1
0.74
0.004
0.50
0.006


22
ACTL6A
86
actin-like 6A
0.81
0.042
2.28
0.003


23
ACTR10
55860
actin-related protein 10 homolog (S.
0.78
0.019
1.87
0.041






cerevisiae)



24
ADAM2
2515
ADAM metallopeptidase domain 2
0.79
0.023
1.22
0.021


25
ADAMTS5
11096
ADAM metallopeptidase with
0.81
0.036
0.74
0.044





thrombospondin type 1 motif, 5


26
ADCY9
115
adenylate cyclase 9
0.81
0.020
0.51
0.001


27
ADIPOR1
51094
adiponectin receptor 1
0.81
0.040
2.14
0.002


28
ADRA2A
150
adrenoceptor alpha 2A
0.80
0.011
0.50
0.000


29
ADSL
158
adenylosuccinate lyase
0.77
0.011
2.67
0.004


30
AGAP10
119016
ArfGAP with GTPase domain, ankyrin
0.76
0.010
0.60
0.030





repeat and PH domain 10


31
AGAP11
119016
ankyrin repeat and GTPase domain Arf
0.76
0.007
0.56
0.006





GTPase activating protein 11


32
AGBL2
79841
ATP/GTP binding protein-like 2
0.79
0.015
0.59
0.020


33
AGL
178
amylo-alpha-1, 6-glucosidase, 4-alpha-
0.79
0.022
1.28
0.035





glucanotransferase


34
AHNAK
79026
AHNAK nucleoprotein
0.76
0.005
0.60
0.009


35
AHNAK2
113146
AHNAK nucleoprotein 2
0.79
0.022
0.77
0.047


36
AHSA2
130872
AHA1, activator of heat shock 90 kDa
0.78
0.016
0.74
0.010





protein ATPase homolog 2 (yeast)


37
AHSG
197
alpha-2-HS-glycoprotein
0.77
0.012
1.50
0.001


38
AK4
205
adenylate kinase 4
0.80
0.028
0.67
0.016


39
AK5
26289
adenylate kinase 5
0.78
0.014
0.75
0.001


40
AKAP11
11215
A kinase (PRKA) anchor protein 11
0.81
0.043
0.56
0.010


41
AKR7A2
8574
aldo-keto reductase family 7, member
0.81
0.023
0.48
0.004





A2 (aflatoxin aldehyde reductase)


42
ALDH18A1
5832
aldehyde dehydrogenase 18 family,
0.78
0.018
1.61
0.046





member A1


43
ALDH1A2
8854
aldehyde dehydrogenase 1 family,
0.79
0.020
0.80
0.010





member A2


44
ALDH6A1
4329
aldehyde dehydrogenase 6 family,
0.80
0.021
0.50
0.019





member A1


45
ALMS1
7840
Alstrom syndrome 1
0.78
0.011
0.72
0.048


46
AMOTL2
51421
angiomotin like 2
0.79
0.018
0.55
0.018


47
AMT
275
aminomethyltransferase
0.78
0.013
0.71
0.031


48
ANGEL1
23357
angel homolog 1 (Drosophila)
0.77
0.016
0.59
0.039


49
ANGPTL7
10218
angiopoietin-like 7
0.78
0.017
0.78
0.038


50
ANKRD10-IT1
100505494
ANKRD10 intronic transcript 1 (non-
0.76
0.007
0.62
0.008





protein coding)


51
ANKRD40
91369
ankyrin repeat domain 40
0.77
0.014
1.26
0.048


52
ANXA2
302
annexin A2
0.77
0.012
1.93
0.018


53
ANXA8
653145
annexin A8
0.75
0.009
0.67
0.022


54
AP1B1
162
adaptor-related protein complex 1, beta
0.79
0.021
1.63
0.045





1 subunit


55
AP1M2
10053
adaptor-related protein complex 1, mu
0.82
0.048
1.57
0.024





2 subunit


56
AP5M1
55745
adaptor-related protein complex 5, mu
0.78
0.011
1.69
0.020





1 subunit


57
APITD1
378708
apoptosis-inducing, TAF9-like domain
0.81
0.041
1.86
0.027





1


58
APLP2
334
amyloid beta (A4) precursor-like
0.81
0.024
0.67
0.033





protein 2


59
APOC1
341
apolipoprotein C-I
0.76
0.008
1.34
0.026


60
APOC3
345
apolipoprotein C-III
0.77
0.015
1.62
0.000


61
APOO
79135
apolipoprotein O
0.74
0.007
2.27
0.000


62
AQP1
358
aquaporin 1 (Colton blood group)
0.81
0.041
0.63
0.004


63
AQP9
366
aquaporin 9
0.77
0.007
1.33
0.001


64
ARF1
375
ADP-ribosylation factor 1
0.78
0.016
1.88
0.025


65
ARGLU1
55082
arginine and glutamate rich 1
0.79
0.026
0.58
0.001


66
ARHGEF4
50649
Rho guanine nucleotide exchange
0.77
0.014
0.59
0.020





factor (GEF) 4


67
ARHGEF5
7984
Rho guanine nucleotide exchange
0.82
0.049
0.76
0.040





factor (GEF) 5


68
ARID4B
23029
AT rich interactive domain 4B (RBP1-
0.78
0.011
1.57
0.029





like)


69
ARIH2
10425
ariadne RBR E3 ubiquitin protein
0.78
0.014
0.52
0.048





ligase 2


70
ARMC1
55156
armadillo repeat containing 1
0.81
0.043
1.75
0.001


71
ARMC2-AS1
9551
ARMC2 antisense RNA 1
0.80
0.026
1.83
0.043


72
ARPC3
10094
actin related protein 2/3 complex,
0.78
0.012
1.60
0.039





subunit 3, 21 kDa


73
ARPC4
10093
actin related protein 2/3 complex,
0.78
0.014
0.68
0.003





subunit 4, 20 kDa


74
ARPC5L
81873
actin related protein 2/3 complex,
0.81
0.021
2.21
0.002





subunit 5-like


75
ARPIN
348110
actin-related protein 2/3 complex
0.82
0.036
0.73
0.035





inhibitor


76
ASAH1
427
N-acylsphingosine amidohydrolase
0.79
0.025
0.65
0.017





(acid ceramidase) 1


77
ASAP1
50807
ArfGAP with SH3 domain, ankyrin
0.80
0.020
2.14
0.002





repeat and PH domain 1


78
ASCC3
10973
activating signal cointegrator 1
0.75
0.007
1.53
0.021





complex subunit 3


79
ASH2L
9070
ash2 (absent, small, or homeotic)-like
0.81
0.037
1.80
0.003





(Drosophila)


80
ASIC2
40
acid-sensing (proton-gated) ion
0.82
0.046
1.24
0.045





channel 2


81
ASIC4
55515
acid-sensing (proton-gated) ion
0.81
0.035
1.64
0.001





channel family member 4


82
ASL
435
argininosuccinate lyase
0.80
0.024
1.67
0.030


83
ASPM
259266
asp (abnormal spindle) homolog,
0.79
0.011
1.32
0.002





microcephaly associated (Drosophila)


84
ASRGL1
80150
asparaginase like 1
0.82
0.048
1.25
0.042


85
ATAD2
29028
ATPase family, AAA domain
0.80
0.022
1.39
0.004





containing 2


86
ATF6
22926
activating transcription factor 6
0.80
0.022
1.76
0.018


87
ATG101
60673
autophagy related 101
0.80
0.038
1.84
0.035


88
ATG5
9474
autophagy related 5
0.74
0.003
2.16
0.002


89
ATM
472
ATM serine/threonine kinase
0.79
0.032
0.41
0.000


90
ATOH1
474
atonal homolog 1 (Drosophila)
0.82
0.045
1.34
0.026


91
ATP2A2
488
ATPase, Ca++ transporting, cardiac
0.82
0.050
1.99
0.008





muscle, slow twitch 2


92
ATP5A1
498
ATP synthase, H+ transporting,
0.81
0.034
0.46
0.008





mitochondrial F1 complex, alpha





subunit 1, cardiac muscle


93
ATP5C1
509
ATP synthase, H+ transporting,
0.79
0.016
1.94
0.014





mitochondrial F1 complex, gamma





polypeptide 1


94
ATP5H
10476
ATP synthase, H+ transporting,
0.79
0.026
1.62
0.034





mitochondrial Fo complex, subunit d


95
ATP6V0B
533
ATPase, H+ transporting, lysosomal
0.81
0.047
1.52
0.028





21 kDa, V0 subunit b


96
ATP6V1E1
529
ATPase, H+ transporting, lysosomal
0.78
0.014
1.58
0.022





31 kDa, V1 subunit E1


97
ATP7B
540
ATPase, Cu++ transporting, beta
0.77
0.012
0.68
0.030





polypeptide


98
ATP8B1
5205
ATPase, aminophospholipid
0.78
0.013
0.72
0.009





transporter, class I, type 8B, member 1


99
AURKA
6790
aurora kinase A
0.80
0.024
1.59
0.002


100
AURKB
9212
aurora kinase B
0.81
0.029
1.26
0.044


101
AZGP1
563
alpha-2-glycoprotein 1, zinc-binding
0.79
0.020
0.80
0.031


102
AZGP1
563
alpha-2-glycoprotein 1, zinc-binding
0.78
0.018
0.81
0.040


103
AZIN1
51582
antizyme inhibitor 1
0.79
0.024
1.86
0.000


104
B3GALT4
8705
UDP-Gal:betaGlcNAc beta 1,3-
0.81
0.035
1.50
0.035





galactosyltransferase, polypeptide 4


105
BANP
54971
BTG3 associated nuclear protein
0.78
0.024
0.39
0.006


106
BAP1
8314
BRCA1 associated protein-1 (ubiquitin
0.77
0.010
0.50
0.010





carboxy-terminal hydrolase)


107
BARX2
8538
BARX homeobox 2
0.78
0.019
0.74
0.004


108
BBOX1
8424
butyrobetaine (gamma), 2-oxoglutarate
0.81
0.043
0.86
0.029





dioxygenase (gamma-butyrobetaine





hydroxylase) 1


109
BBS1
582
Bardet-Biedl syndrome 1
0.81
0.018
0.51
0.026


110
BCAM
4059
basal cell adhesion molecule (Lutheran
0.77
0.008
0.64
0.013





blood group)


111
BCAP29
55973
B-cell receptor-associated protein 29
0.81
0.030
1.60
0.013


112
BCAS2
10286
breast carcinoma amplified sequence 2
0.79
0.017
1.64
0.034


113
BCCIP
56647
BRCA2 and CDKN1A interacting
0.77
0.013
1.37
0.017





protein


114
BCHE
590
butyrylcholinesterase
0.79
0.016
0.84
0.043


115
BET1
10282
Bet1 golgi vesicular membrane
0.80
0.026
1.66
0.047





trafficking protein


116
BICC1
80114
BicC family RNA binding protein 1
0.80
0.026
0.52
0.025


117
BID
637
BH3 interacting domain death agonist
0.77
0.007
1.72
0.011


118
BIK
638
BCL2-interacting killer (apoptosis-
0.79
0.027
1.26
0.018





inducing)


119
BIN3
55909
bridging integrator 3
0.77
0.012
0.44
0.001


120
BIN3-IT1
80094
BIN3 intronic transcript 1 (non-protein
0.81
0.031
0.67
0.001





coding)


121
RDH5
100528022
retinol dehydrogenase 5 (11-cis/9-cis)
0.80
0.033
0.62
0.000


122
BLZF1
8548
basic leucine zipper nuclear factor 1
0.79
0.022
1.52
0.028


123
BNC1
646
basonuclin 1
0.77
0.012
0.83
0.041


124
BOLA2
552900
bolA family member 2
0.81
0.037
1.48
0.017


125
BRD7
29117
bromodomain containing 7
0.76
0.007
1.54
0.021


126
BRIX1
55299
BRX1, biogenesis of ribosomes,
0.80
0.028
1.45
0.044





homolog (S. cerevisiae)


127
BSDC1
55108
BSD domain containing 1
0.80
0.027
0.46
0.029


128
BTF3
689
basic transcription factor 3
0.78
0.014
0.46
0.003


129
BTN1A1
696
butyrophilin, subfamily 1, member A1
0.78
0.012
0.83
0.018


130
BUB1
699
BUB1 mitotic checkpoint
0.78
0.014
1.73
0.005





serine/threonine kinase


131
BUB1B
701
BUB1 mitotic checkpoint
0.80
0.031
1.71
0.002





serine/threonine kinase B


132
BUD31
8896
BUD31 homolog (S. cerevisiae)
0.80
0.018
1.88
0.037


133
BYSL
705
bystin-like
0.76
0.009
1.51
0.017


134
BZW1
9689
basic leucine zipper and W2 domains 1
0.82
0.042
1.59
0.048


135
BZW2
28969
basic leucine zipper and W2 domains 2
0.82
0.048
1.58
0.006


136
C11orf48
79081
chromosome 11 open reading frame 48
0.77
0.010
1.56
0.018


137
C11orf57
55216
chromosome 11 open reading frame 57
0.77
0.013
0.44
0.037


138
C11orf58
10944
chromosome 11 open reading frame 58
0.79
0.023
1.86
0.036


139
C11orf63
79864
chromosome 11 open reading frame 63
0.79
0.019
0.74
0.001


140
C11orf80
79703
chromosome 11 open reading frame 80
0.82
0.039
1.31
0.004


141
C12orf29
91298
chromosome 12 open reading frame 29
0.81
0.033
1.42
0.026


142
C14orf132
56967
chromosome 14 open reading frame
0.80
0.024
0.74
0.040





132


143
C16orf59
80178
chromosome 16 open reading frame 59
0.81
0.034
1.29
0.021


144
C16orf80
29105
chromosome 16 open reading frame 80
0.76
0.008
1.58
0.037


145
C17orf75
64149
chromosome 17 open reading frame 75
0.79
0.024
1.33
0.020


146
C18orf32
6139
chromosome 18 open reading frame 32
0.81
0.044
0.58
0.015


147
C19orf53
28974
chromosome 19 open reading frame 53
0.78
0.017
1.72
0.025


148
C19orf54
284325
chromosome 19 open reading frame 54
0.80
0.029
0.54
0.000


149
C1GALT1
56913
core 1 synthase, glycoprotein-N-
0.81
0.030
1.63
0.016





acetylgalactosamine 3-beta-





galactosyltransferase 1


150
C1orf21
81563
chromosome 1 open reading frame 21
0.83
0.048
0.85
0.039


151
C20orf24
55969
chromosome 20 open reading frame 24
0.80
0.027
1.85
0.004


152
C2orf43
60526
chromosome 2 open reading frame 43
0.80
0.026
0.49
0.048


153
TOMM7
201725
translocase of outer mitochondrial
0.80
0.025
0.44
0.002





membrane 7 homolog (yeast)


154
C5AR1
728
complement component 5a receptor 1
0.80
0.023
1.44
0.032


155
C7orf63
79846
chromosome 7 open reading frame 63
0.79
0.024
0.82
0.018


156
C9orf114
51490
chromosome 9 open reading frame 114
0.80
0.023
1.72
0.029


157
CA4
762
carbonic anhydrase IV
0.78
0.013
0.68
0.012


158
CACNA1C
775
calcium channel, voltage-dependent, L
0.82
0.047
0.76
0.020





type, alpha 1C subunit


159
CACNA1D
776
calcium channel, voltage-dependent, L
0.79
0.013
0.59
0.001





type, alpha 1D subunit


160
CACNB2
783
calcium channel, voltage-dependent,
0.81
0.030
0.72
0.028





beta 2 subunit


161
CACNG1
786
calcium channel, voltage-dependent,
0.83
0.043
1.45
0.000





gamma subunit 1


162
CACYBP
27101
calcyclin binding protein
0.82
0.045
1.73
0.008


163
CADM3-AS1
100131825
CADM3 antisense RNA 1
0.78
0.011
0.68
0.031


164
CADPS2
93664
Ca++-dependent secretion activator 2
0.80
0.032
0.66
0.031


165
CALCOCO1
57658
calcium binding and coiled-coil
0.77
0.007
0.60
0.013





domain 1


166
CALML3
810
calmodulin-like 3
0.73
0.006
0.65
0.003


167
CAMKMT
79823
calmodulin-lysine N-methyltransferase
0.81
0.036
0.73
0.037


168
CAMKV
79012
CaM kinase-like vesicle-associated
0.76
0.010
0.69
0.004


169
CAMSAP1
157922
calmodulin regulated spectrin-
0.82
0.036
2.34
0.031





associated protein 1


170
CAMSAP2
23271
calmodulin regulated spectrin-
0.81
0.040
1.67
0.021





associated protein family, member 2


171
CAMTA2
23125
calmodulin binding transcription
0.80
0.021
0.37
0.003





activator 2


172
CAND1
55832
cullin-associated and neddylation-
0.80
0.022
1.71
0.014





dissociated 1


173
CAPN10
11132
calpain 10
0.81
0.028
0.67
0.000


174
CAPN15
6650
calpain 15
0.80
0.038
0.59
0.026


175
CAPRIN2
65981
caprin family member 2
0.81
0.036
0.52
0.009


176
CARD10
29775
caspase recruitment domain family,
0.79
0.018
0.64
0.004





member 10


177
CASC1
55259
cancer susceptibility candidate 1
0.80
0.024
0.75
0.003


178
CASK
8573
calcium/calmodulin-dependent serine
0.82
0.044
1.54
0.034





protein kinase (MAGUK family)


179
CASP2
835
caspase 2, apoptosis-related cysteine
0.76
0.009
0.60
0.032





peptidase


180
CASP3
836
caspase 3, apoptosis-related cysteine
0.77
0.008
2.03
0.016





peptidase


181
CASP5
838
caspase 5, apoptosis-related cysteine
0.80
0.038
1.26
0.044





peptidase


182
CAST
831
calpastatin
0.81
0.046
0.59
0.022


183
CATSPER2
117155
cation channel, sperm associated 2
0.79
0.017
0.72
0.003


184
CBX7
23492
chromobox homolog 7
0.81
0.040
0.74
0.014


185
CCBL1
883
cysteine conjugate-beta lyase,
0.76
0.007
0.61
0.027





cytoplasmic


186
CCDC101
112869
coiled-coil domain containing 101
0.81
0.034
0.49
0.035


187
CCDC132
55610
coiled-coil domain containing 132
0.80
0.024
0.79
0.031


188
CCDC176
80127
coiled-coil domain containing 176
0.76
0.011
0.48
0.002


189
CCDC59
29080
coiled-coil domain containing 59
0.77
0.008
2.26
0.002


190
CCNA2
890
cyclin A2
0.82
0.043
1.70
0.006


191
CCNB1
891
cyclin B1
0.79
0.021
1.58
0.004


192
CCNB2
9133
cyclin B2
0.77
0.007
1.67
0.001


193
CCNC
892
cyclin C
0.80
0.021
1.68
0.043


194
CCNE2
9134
cyclin E2
0.80
0.027
2.07
0.000


195
CCNO
10309
cyclin O
0.80
0.026
0.80
0.027


196
CCR6
1235
chemokine (C-C motif) receptor 6
0.81
0.042
0.75
0.011


197
CCT3
7203
chaperonin containing TCP1, subunit 3
0.76
0.010
2.35
0.001





(gamma)


198
CCT5
22948
chaperonin containing TCP1, subunit 5
0.79
0.015
1.73
0.008





(epsilon)


199
CCT6A
908
chaperonin containing TCP1, subunit
0.78
0.010
2.45
0.000





6A (zeta 1)


200
CD55
1604
CD55 molecule, decay accelerating
0.78
0.027
1.43
0.039





factor for complement (Cromer blood





group)


201
CDC14B
8555
cell division cycle 14B
0.74
0.006
0.48
0.004


202
CDC20
991
cell division cycle 20
0.78
0.011
1.32
0.019


203
CDC25C
995
cell division cycle 25C
0.80
0.038
1.70
0.011


204
CDIPT
10423
CDP-diacylglycerol--inositol 3-
0.79
0.028
0.63
0.012





phosphatidyltransferase


205
CDK1
983
cyclin-dependent kinase 1
0.76
0.005
1.56
0.001


206
CDK2AP2
10263
cyclin-dependent kinase 2 associated
0.78
0.014
1.43
0.019





protein 2


207
CDK5RAP3
80279
CDK5 regulatory subunit associated
0.78
0.015
0.63
0.023





protein 3


208
CDKN1C
1028
cyclin-dependent kinase inhibitor 1C
0.83
0.044
0.51
0.001





(p57, Kip2)


209
CDKN3
1033
cyclin-dependent kinase inhibitor 3
0.81
0.037
2.26
0.000


210
CDO1
1036
cysteine dioxygenase type 1
0.81
0.035
0.66
0.006


211
CDT1
81620
chromatin licensing and DNA
0.78
0.015
1.32
0.040





replication factor 1


212
CELA2A
51032
chymotrypsin-like elastase family,
0.81
0.031
1.27
0.038





member 2A


213
CELSR2
1952
cadherin, EGF LAG seven-pass G-type
0.78
0.014
0.68
0.013





receptor 2


214
CENPA
1058
centromere protein A
0.73
0.002
2.00
0.000


215
CENPBD1P1
65996
CENPBD1 pseudogene 1
0.78
0.016
1.33
0.047


216
CENPE
1062
centromere protein E, 312 kDa
0.78
0.012
1.33
0.001


217
CENPF
1063
centromere protein F, 350/400 kDa
0.79
0.016
1.64
0.002


218
CENPI
2491
centromere protein I
0.81
0.030
1.50
0.040


219
CENPM
79019
centromere protein M
0.75
0.005
1.39
0.001


220
CENPN
55839
centromere protein N
0.78
0.006
1.50
0.001


221
CENPU
79682
centromere protein U
0.79
0.025
1.77
0.000


222
CEP164
22897
centrosomal protein 164 kDa
0.74
0.003
0.51
0.002


223
CEP55
55165
centrosomal protein 55 kDa
0.75
0.007
1.48
0.001


224
CFHR4
10877
complement factor H-related 4
0.79
0.020
1.40
0.014


225
CFHR5
81494
complement factor H-related 5
0.79
0.024
0.71
0.041


226
CH25H
9023
cholesterol 25-hydroxylase
0.81
0.042
0.77
0.027


227
CHAC1
79094
ChaC, cation transport regulator
0.81
0.043
1.45
0.033





homolog 1 (E. coli)


228
CHD3
1107
chromodomain helicase DNA binding
0.73
0.003
0.39
0.001





protein 3


229
CHEK1
1111
checkpoint kinase 1
0.80
0.024
1.75
0.016


230
CHL1
10752
cell adhesion molecule L1-like
0.81
0.041
0.86
0.040


231
CHN1
1123
chimerin 1
0.78
0.010
1.44
0.010


232
CHRNB2
1141
cholinergic receptor, nicotinic, beta 2
0.78
0.022
1.39
0.017





(neuronal)


233
CHRNB4
1143
cholinergic receptor, nicotinic, beta 4
0.79
0.016
0.83
0.032





(neuronal)


234
CIC
23152
capicua transcriptional repressor
0.78
0.015
0.65
0.023


235
CIDEC
63924
cell death-inducing DFFA-like effector
0.82
0.037
0.61
0.009





c


236
CILP
8483
cartilage intermediate layer protein,
0.81
0.042
0.80
0.026





nucleotide pyrophosphohydrolase


237
CIRBP
1153
cold inducible RNA binding protein
0.75
0.004
0.47
0.000


238
CKAP5
9793
cytoskeleton associated protein 5
0.78
0.020
2.12
0.022


239
CKS1B
1163
CDC28 protein kinase regulatory
0.78
0.013
1.52
0.027





subunit 1B


240
CKS2
1164
CDC28 protein kinase regulatory
0.79
0.023
1.55
0.002





subunit 2


241
CLCN7
1186
chloride channel, voltage-sensitive 7
0.78
0.014
0.62
0.029


242
CLDN8
9073
claudin 8
0.82
0.038
0.64
0.000


243
CLIP2
7461
CAP-GLY domain containing linker
0.82
0.039
0.68
0.016





protein 2


244
CLK4
57396
CDC-like kinase 4
0.80
0.023
0.56
0.005


245
CLPB
81570
ClpB caseinolytic peptidase B
0.78
0.016
1.19
0.048





homolog (E. coli)


246
CLPP
8192
caseinolytic mitochondrial matrix
0.77
0.013
1.48
0.015





peptidase proteolytic subunit


247
CLPS
1208
colipase, pancreatic
0.80
0.035
1.28
0.048


248
CLUAP1
23059
clusterin associated protein 1
0.82
0.027
0.43
0.010


249
CLUHP3
100132341
clustered mitochondria (cluA/CLU1)
0.82
0.046
0.61
0.000





homolog pseudogene 3


250
CMC2
56942
C-x(9)-C motif containing 2
0.68
0.000
2.37
0.000


251
CNIH4
29097
cornichon family AMPA receptor
0.77
0.011
1.73
0.002





auxiliary protein 4


252
CNKSR1
10256
connector enhancer of kinase
0.80
0.035
0.45
0.001





suppressor of Ras 1


253
CNN3
1266
calponin 3, acidic
0.83
0.045
0.64
0.002


254
CNPY2
10330
canopy FGF signaling regulator 2
0.75
0.007
1.73
0.025


255
CNR1
1268
cannabinoid receptor 1 (brain)
0.75
0.008
0.63
0.005


256
COL16A1
1307
collagen, type XVI, alpha 1
0.82
0.039
0.62
0.002


257
COL17A1
1308
collagen, type XVII, alpha 1
0.81
0.031
0.73
0.005


258
COL19A1
1310
collagen, type XIX, alpha 1
0.77
0.014
1.28
0.038


259
COL4A6
1288
collagen, type IV, alpha 6
0.78
0.014
0.61
0.002


260
COL7A1
1294
collagen, type VII, alpha 1
0.81
0.035
0.76
0.048


261
COMMD8
54951
COMM domain containing 8
0.79
0.014
1.53
0.037


262
COPB1
1315
coatomer protein complex, subunit beta
0.80
0.024
2.57
0.005





1


263
COPB2
9276
coatomer protein complex, subunit beta
0.76
0.004
1.96
0.001





2 (beta prime)


264
COPS3
8533
COP9 signalosome subunit 3
0.80
0.025
1.63
0.034


265
COPS4
51138
COP9 signalosome subunit 4
0.79
0.035
2.32
0.009


266
COQ2
27235
coenzyme Q2 4-hydroxybenzoate
0.79
0.013
1.67
0.013





polyprenyltransferase


267
COQ3
51805
coenzyme Q3 methyltransferase
0.81
0.026
3.03
0.001


268
CORO1C
23603
coronin, actin binding protein, 1C
0.74
0.003
1.78
0.003


269
COX17
10063
COX17 cytochrome c oxidase copper
0.78
0.017
1.75
0.001





chaperone


270
COX6B1
1340
cytochrome c oxidase subunit VIb
0.77
0.010
2.22
0.002





polypeptide 1 (ubiquitous)


271
COX7B
1349
cytochrome c oxidase subunit VIIb
0.78
0.013
1.81
0.014


272
CPEB1
64506
cytoplasmic polyadenylation element
0.81
0.039
0.64
0.025





binding protein 1


273
CPNE3
8895
copine III
0.80
0.031
2.00
0.000


274
CPSF6
11052
cleavage and polyadenylation specific
0.77
0.010
2.50
0.008





factor 6, 68 kDa


275
CREB3L2
64764
cAMP responsive element binding
0.82
0.045
0.54
0.032





protein 3-like 2


276
CRELD1
78987
cysteine-rich with EGF-like domains 1
0.82
0.044
0.54
0.042


277
CRIM1
51232
cysteine rich transmembrane BMP
0.80
0.025
0.73
0.028





regulator 1 (chordin-like)


278
CRIPT
9419
cysteine-rich PDZ-binding protein
0.78
0.016
1.42
0.024


279
CRISP3
10321
cysteine-rich secretory protein 3
0.79
0.019
1.08
0.043


280
CRNKL1
51340
crooked neck pre-mRNA splicing
0.80
0.033
1.50
0.041





factor 1


281
CROT
54677
carnitine O-octanoyltransferase
0.79
0.024
0.60
0.002


282
CRTAP
10491
cartilage associated protein
0.78
0.012
0.39
0.001


283
CRTC3
64784
CREB regulated transcription
0.80
0.030
0.52
0.042





coactivator 3


284
CRYBA2
1412
crystallin, beta A2
0.79
0.018
1.51
0.008


285
CSAD
51380
cysteine sulfinic acid decarboxylase
0.77
0.009
0.76
0.021


286
CSH1
1442
chorionic somatomammotropin
0.80
0.037
1.45
0.007





hormone 1 (placental lactogen)


287
CSNK1G2
1455
casein kinase 1, gamma 2
0.77
0.008
0.47
0.009


288
CSNK2A1
1457
casein kinase 2, alpha 1 polypeptide
0.75
0.004
2.13
0.002


289
CSNK2B
1460
casein kinase 2, beta polypeptide
0.78
0.016
1.62
0.044


290
CSRP1
1465
cysteine and glycine-rich protein 1
0.80
0.021
0.60
0.011


291
CSTF1
1477
cleavage stimulation factor, 3′ pre-
0.76
0.007
1.63
0.005





RNA, subunit 1, 50 kDa


292
CTDSP1
58190
CTD (carboxy-terminal domain, RNA
0.78
0.013
0.49
0.006





polymerase II, polypeptide A) small





phosphatase 1


293
CTDSPL
10217
CTD (carboxy-terminal domain, RNA
0.80
0.014
0.54
0.005





polymerase II, polypeptide A) small





phosphatase-like


294
CTNNB1
1499
catenin (cadherin-associated protein),
0.80
0.016
0.64
0.028





beta 1, 88 kDa


295
CTNNBL1
56259
catenin, beta like 1
0.78
0.013
1.64
0.021


296
CTNND1
1500
catenin (cadherin-associated protein),
0.81
0.024
0.47
0.008





delta 1


297
CTNS
1497
cystinosin, lysosomal cystine
0.81
0.041
0.35
0.003





transporter


298
CTSA
5476
cathepsin A
0.79
0.022
1.63
0.030


299
CTSG
1511
cathepsin G
0.79
0.024
0.78
0.028


300
CTSV
1515
cathepsin V
0.82
0.042
1.35
0.047


301
CXCL10
3627
chemokine (C-X-C motif) ligand 10
0.74
0.004
1.29
0.027


302
CXCL11
6373
chemokine (C-X-C motif) ligand 11
0.72
0.001
1.42
0.003


303
CXCL9
4283
chemokine (C-X-C motif) ligand 9
0.75
0.004
1.20
0.036


304
CXorf40A
91966
chromosome X open reading frame
0.80
0.031
1.44
0.022





40A


305
CXorf40A
91966
chromosome X open reading frame
0.80
0.030
1.64
0.005





40A


306
CXorf40B
541578
chromosome X open reading frame
0.80
0.029
1.42
0.033





40B


307
CXXC1
30827
CXXC finger protein 1
0.76
0.006
0.52
0.007


308
CYCS
54205
cytochrome c, somatic
0.82
0.032
1.80
0.005


309
CYP4A11
1579
cytochrome P450, family 4, subfamily
0.77
0.010
0.76
0.014





A, polypeptide 11


310
CYP4F8
11283
cytochrome P450, family 4, subfamily
0.78
0.025
0.89
0.019





F, polypeptide 8


311
CYR61
3491
cysteine-rich, angiogenic inducer, 61
0.82
0.041
0.79
0.026


312
CYSLTR2
57105
cysteinyl leukotriene receptor 2
0.77
0.010
0.80
0.024


313
DAAM1
23002
dishevelled associated activator of
0.81
0.036
1.90
0.002





morphogenesis 1


314
DAD1
1603
defender against cell death 1
0.80
0.038
1.77
0.037


315
DARS2
55157
aspartyl-tRNA synthetase 2,
0.82
0.050
1.75
0.024





mitochondrial


316
DBT
1629
dihydrolipoamide branched chain
0.76
0.008
0.69
0.016





transacylase E2


317
DCK
1633
deoxycytidine kinase
0.77
0.007
1.70
0.004


318
DCTN2
10540
dynactin 2 (p50)
0.77
0.012
1.85
0.015


319
DCUN1D1
54165
DCN1, defective in cullin neddylation
0.77
0.010
2.50
0.003





1, domain containing 1


320
DDX17
10521
DEAD (Asp-Glu-Ala-Asp) box
0.77
0.009
0.55
0.013





helicase 17


321
DDX23
9416
DEAD (Asp-Glu-Ala-Asp) box
0.76
0.009
1.70
0.006





polypeptide 23


322
DDX39A
10212
DEAD (Asp-Glu-Ala-Asp) box
0.80
0.017
1.92
0.005





polypeptide 39A


323
DDX49
54555
DEAD (Asp-Glu-Ala-Asp) box
0.78
0.015
1.76
0.032





polypeptide 49


324
DDX5
1655
DEAD (Asp-Glu-Ala-Asp) box
0.79
0.022
0.59
0.030





helicase 5


325
DERL1
79139
derlin 1
0.74
0.003
1.89
0.000


326
DGAT1
8694
diacylglycerol O-acyltransferase 1
0.81
0.033
1.33
0.040


327
DGKA
1606
diacylglycerol kinase, alpha 80 kDa
0.79
0.029
0.51
0.010


328
DHCR24
1718
24-dehydrocholesterol reductase
0.79
0.015
0.66
0.013


329
DHCR7
1717
7-dehydrocholesterol reductase
0.80
0.030
1.40
0.011


330
DHPS
1725
deoxyhypusine synthase
0.78
0.018
1.47
0.017


331
DHX9
1660
DEAH (Asp-Glu-Ala-His) box helicase
0.80
0.028
2.07
0.025





9


332
DIEXF
27042
digestive organ expansion factor
0.82
0.049
2.16
0.013





homolog (zebrafish)


333
DKC1
1736
dyskeratosis congenita 1, dyskerin
0.81
0.040
1.78
0.026


334
OBSCN
84033
obscurin, cytoskeletal calmodulin and
0.77
0.010
0.76
0.005





titin-interacting RhoGEF


335
DLGAP5
9787
discs, large (Drosophila) homolog-
0.75
0.004
1.76
0.000





associated protein 5


336
DNA2
1763
DNA replication helicase/nuclease 2
0.81
0.034
1.49
0.047


337
DNAAF1
123872
dynein, axonemal, assembly factor 1
0.81
0.026
0.77
0.021


338
DNAJB14
79982
DnaJ (Hsp40) homolog, subfamily B,
0.80
0.019
1.37
0.036





member 14


339
DNAJC9
23234
DnaJ (Hsp40) homolog, subfamily C,
0.79
0.017
2.17
0.001





member 9


340
DNM1
1759
dynamin 1
0.80
0.028
0.77
0.043


341
DOCK3
1795
dedicator of cytokinesis 3
0.79
0.023
1.38
0.010


342
DPP3
10072
dipeptidyl-peptidase 3
0.77
0.015
1.35
0.017


343
DPY19L4
286148
dpy-19-like 4 (C. elegans)
0.81
0.047
1.83
0.003


344
LRRC37A2
474170
leucine rich repeat containing 37,
0.81
0.028
0.65
0.049





member A2


345
DRG1
4733
developmentally regulated GTP
0.78
0.018
2.02
0.007





binding protein 1


346
DSC3
1825
desmocollin 3
0.77
0.017
0.68
0.019


347
DSCC1
79075
DNA replication and sister chromatid
0.80
0.021
1.93
0.000





cohesion 1


348
DSERG1
751816
Down syndrome encephalopathy
0.79
0.018
0.77
0.040





related protein 1


349
DSG1
1828
desmoglein 1
0.76
0.014
0.82
0.027


350
DST
667
dystonin
0.80
0.036
0.64
0.016


351
DTL
51514
denticleless E3 ubiquitin protein ligase
0.80
0.023
1.47
0.003





homolog (Drosophila)


352
DTX2
113878
deltex 2, E3 ubiquitin ligase
0.80
0.030
0.66
0.003


353
DUSP6
1848
dual specificity phosphatase 6
0.81
0.040
0.71
0.031


354
DYNC2H1
79659
dynein, cytoplasmic 2, heavy chain 1
0.82
0.041
0.52
0.004


355
DYNLRBl
83658
dynein, light chain, roadblock-type 1
0.81
0.035
1.62
0.017


356
DYNLT1
6993
dynein, light chain, Tctex-type 1
0.77
0.010
2.21
0.003


357
DYRK2
8445
dual-specificity tyrosine-(Y)-
0.78
0.009
1.91
0.008





phosphorylation regulated kinase 2


358
E2F8
79733
E2F transcription factor 8
0.81
0.025
1.46
0.004


359
EAF2
55840
ELL associated factor 2
0.79
0.013
1.30
0.036


360
EBAG9
9166
estrogen receptor binding site
0.79
0.022
1.48
0.008





associated, antigen, 9


361
ECD
11319
ecdysoneless homolog (Drosophila)
0.75
0.007
1.70
0.036


362
ECHDC2
55268
enoyl CoA hydratase domain
0.76
0.007
0.49
0.000





containing 2


363
ECT2
1894
epithelial cell transforming 2
0.79
0.022
1.69
0.001


364
EEF1A1
1915
eukaryotic translation elongation factor
0.74
0.005
0.47
0.003





1 alpha 1


365
EEF1E1
9521
eukaryotic translation elongation factor
0.78
0.015
2.06
0.004





1 epsilon 1


366
EEF1G
1937
eukaryotic translation elongation factor
0.79
0.019
0.48
0.002





1 gamma


367
EEF2
1938
eukaryotic translation elongation factor
0.74
0.006
0.41
0.000





2


368
EFCAB14
9813
EF-hand calcium binding domain 14
0.79
0.023
0.46
0.026


369
EFNA5
1946
ephrin-A5
0.75
0.006
0.59
0.002


370
EFR3A
23167
EFR3 homolog A (S. cerevisiae)
0.80
0.033
1.59
0.037


371
EFR3B
22979
EFR3 homolog B (S. cerevisiae)
0.79
0.018
0.68
0.015


372
EGF
1950
epidermal growth factor
0.80
0.029
0.82
0.032


373
EGFL8
9374
EGF-like-domain, multiple 8
0.79
0.020
0.63
0.019


374
EGFR
1956
epidermal growth factor receptor
0.81
0.050
0.56
0.005


375
EGR1
1958
early growth response 1
0.79
0.019
0.67
0.001


376
EGR3
1960
early growth response 3
0.79
0.016
0.54
0.000


377
EI24
9538
etoposide induced 2.4
0.77
0.010
0.59
0.026


378
EIF2S2
8894
eukaryotic translation initiation factor
0.76
0.008
1.54
0.018





2, subunit 2 beta, 38 kDa


379
EIF3J
8669
eukaryotic translation initiation factor
0.79
0.022
1.70
0.028





3, subunit J


380
EIF3L
51386
eukaryotic translation initiation factor
0.81
0.035
0.62
0.026





3, subunit L


381
EIF4B
1975
eukaryotic translation initiation factor
0.80
0.021
0.54
0.012





4B


382
EIF4E2
9470
eukaryotic translation initiation factor
0.76
0.009
1.74
0.033





4E family member 2


383
EIF4EBP1
1978
eukaryotic translation initiation factor
0.77
0.010
1.45
0.003





4E binding protein 1


384
EIF4G1
1981
eukaryotic translation initiation factor
0.82
0.043
1.70
0.033





4 gamma, 1


385
EIF6
3692
eukaryotic translation initiation factor
0.80
0.028
1.56
0.037





6


386
ELAC1
55520
elaC ribonuclease Z 1
0.81
0.035
0.73
0.028


387
ELMO2
63916
engulfment and cell motility 2
0.78
0.017
1.57
0.044


388
ELOVL5
60481
ELOVL fatty acid elongase 5
0.76
0.015
0.55
0.000


389
EMC2
9694
ER membrane protein complex subunit
0.80
0.035
2.07
0.000





2


390
EMC9
51016
ER membrane protein complex subunit
0.76
0.012
1.22
0.027





9


391
EMG1
10436
EMG1 N1-specific pseudouridine
0.79
0.020
1.72
0.024





methyltransferase


392
EML3
256364
echinoderm microtubule associated
0.79
0.019
0.51
0.002





protein like 3


393
ENOPH1
58478
enolase-phosphatase 1
0.79
0.014
2.12
0.000


394
ENOSF1
55556
enolase superfamily member 1
0.79
0.020
0.67
0.028


395
ENY2
56943
enhancer of yellow 2 homolog
0.75
0.008
1.91
0.000





(Drosophila)


396
EP400
57634
E1A binding protein p400
0.80
0.027
0.40
0.023


397
EPB41L2
2037
erythrocyte membrane protein band
0.81
0.047
0.57
0.016





4.1-like 2


398
EPRS
2058
glutamyl-prolyl-tRNA synthetase
0.77
0.006
1.87
0.002


399
ERBB2
2064
v-erb-b2 avian erythroblastic leukemia
0.81
0.048
1.30
0.024





viral oncogene homolog 2


400
ERBB4
2066
v-erb-b2 avian erythroblastic leukemia
0.77
0.013
0.75
0.016





viral oncogene homolog 4


401
ERCC6L
54821
excision repair cross-complementation
0.74
0.003
1.44
0.000





group 6-like


402
ERO1L
30001
ERO1-like (S. cerevisiae)
0.79
0.019
1.41
0.032


403
ESRP1
54845
epithelial splicing regulatory protein 1
0.80
0.024
1.77
0.002


404
ETFA
2108
electron-transfer-flavoprotein, alpha
0.77
0.011
1.50
0.041





polypeptide


405
ETV3
2117
ets variant 3
0.80
0.021
0.71
0.044


406
EXOC7
23265
exocyst complex component 7
0.80
0.025
0.52
0.023


407
EXOSC1
51013
exosome component 1
0.80
0.027
0.83
0.033


408
EXOSC4
54512
exosome component 4
0.80
0.037
1.47
0.023


409
EXT1
2131
exostosin glycosyltransferase 1
0.74
0.004
2.05
0.001


410
EZH1
2145
enhancer of zeste 1 polycomb
0.79
0.017
0.39
0.001





repressive complex 2 subunit


411
EZR
7430
ezrin
0.82
0.041
1.73
0.011


412
F8
2157
coagulation factor VIII, procoagulant
0.81
0.043
0.57
0.033





component


413
FABP1
2168
fatty acid binding protein 1, liver
0.81
0.037
1.34
0.036


414
FABP6
2172
fatty acid binding protein 6, ileal
0.79
0.017
1.18
0.016


415
FABP7
2173
fatty acid binding protein 7, brain
0.80
0.028
0.73
0.016


416
FADD
8772
Fas (TNFRSF6)-associated via death
0.78
0.021
1.57
0.005





domain


417
FAM120A
23196
family with sequence similarity 120A
0.79
0.018
0.53
0.019


418
FAM129A
116496
family with sequence similarity 129,
0.81
0.035
0.78
0.034





member A


419
FAM131B
9715
family with sequence similarity 131,
0.80
0.024
0.78
0.018





member B


420
FAM136A
84908
family with sequence similarity 136,
0.80
0.027
1.83
0.048





member A


421
FAM160B2
64760
family with sequence similarity 160,
0.78
0.014
0.58
0.032





member B2


422
FAM163A
148753
family with sequence similarity 163,
0.80
0.025
1.46
0.004





member A


423
FAM171A1
221061
family with sequence similarity 171,
0.80
0.040
0.55
0.005





member A1


424
FAM189A1
23359
family with sequence similarity 189,
0.80
0.020
0.74
0.015





member A1


425
FAM189A2
9413
family with sequence similarity 189,
0.79
0.019
0.63
0.016





member A2


426
FAM193B
54540
family with sequence similarity 193,
0.77
0.015
0.65
0.030





member B


427
FAM47E
8987
family with sequence similarity 47,
0.81
0.034
2.15
0.002





member E


428
FAM49B
51571
family with sequence similarity 49,
0.73
0.003
2.04
0.001





member B


429
FAM63A
55793
family with sequence similarity 63,
0.77
0.010
0.65
0.017





member A


430
FAM96B
51647
family with sequence similarity 96,
0.79
0.018
1.75
0.035





member B


431
FAM98A
25940
family with sequence similarity 98,
0.74
0.004
1.87
0.016





member A


432
FANCI
55215
Fanconi anemia, complementation
0.81
0.032
1.49
0.049





group I


433
FARP2
9855
FERM, RhoGEF and pleckstrin
0.76
0.007
0.46
0.005





domain protein 2


434
FAU
2197
Finkel-Biskis-Reilly murine sarcoma
0.82
0.045
0.59
0.035





virus (FBR-MuSV) ubiquitously





expressed


435
FBP2
8789
fructose-1,6-bisphosphatase 2
0.81
0.041
1.38
0.037


436
FBXO5
26271
F-box protein 5
0.74
0.005
1.49
0.012


437
FCGBP
8857
Fc fragment of IgG binding protein
0.79
0.016
0.82
0.006


438
FDPS
2224
famesyl diphosphate synthase
0.80
0.022
1.66
0.024


439
FEN1
2237
flap structure-specific endonuclease 1
0.77
0.012
1.77
0.003


440
FGB
2244
fibrinogen beta chain
0.78
0.020
1.27
0.023


441
FGF14
2259
fibroblast growth factor 14
0.81
0.036
0.77
0.012


442
FGF9
2254
fibroblast growth factor 9
0.80
0.025
0.81
0.048


443
FH
2271
fumarate hydratase
0.80
0.032
1.74
0.013


444
FIGF
2277
c-fos induced growth factor (vascular
0.80
0.028
0.83
0.044





endothelial growth factor D)


445
FJX1
24147
four jointed box 1 (Drosophila)
0.80
0.027
1.25
0.042


446
FKBP4
2288
FK506 binding protein 4, 59 kDa
0.80
0.036
2.11
0.000


447
FLJ42627
645644
uncharacterized LOC645644
0.78
0.011
0.63
0.030


448
FLNA
2316
filamin A, alpha
0.81
0.032
0.72
0.027


449
FLRT2
23768
fibronectin leucine rich transmembrane
0.81
0.043
0.67
0.022





protein 2


450
FLT3
2322
fms-related tyrosine kinase 3
0.79
0.019
0.78
0.039


451
FMO2
2327
flavin containing monooxygenase 2
0.79
0.031
0.54
0.000





(non-functional)


452
FMO5
2330
flavin containing monooxygenase 5
0.79
0.015
0.64
0.000


453
FOCAD
54914
focadhesin
0.82
0.044
0.59
0.032


454
FOXI1
2299
forkhead box I1
0.77
0.010
0.73
0.001


455
FOXM1
2305
forkhead box M1
0.78
0.015
1.42
0.002


456
FPGS
2356
folylpolyglutamate synthase
0.79
0.016
0.65
0.039


457
FPR3
2359
formyl peptide receptor 3
0.76
0.009
1.57
0.044


458
FST
10468
follistatin
0.80
0.029
0.68
0.024


459
FTH1P5
2509
ferritin, heavy polypeptide 1
0.81
0.036
0.62
0.023





pseudogene 5


460
FUBP3
8939
far upstream element (FUSE) binding
0.81
0.033
1.62
0.044





protein 3


461
FUT2
2524
fucosyltransferase 2 (secretor status
0.78
0.017
0.55
0.033





included)


462
FZD10
11211
frizzled class receptor 10
0.77
0.012
0.69
0.002


463
FZD4
8322
frizzled class receptor 4
0.77
0.010
0.54
0.013


464
G3BP2
9908
GTPase activating protein (SH3
0.80
0.032
2.37
0.005





domain) binding protein 2


465
GAA
2548
glucosidase, alpha; acid
0.81
0.027
0.56
0.015


466
GABRA1
2554
gamma-aminobutyric acid (GABA) A
0.81
0.046
1.49
0.022





receptor, alpha 1


467
GABRB2
2561
gamma-aminobutyric acid (GABA) A
0.80
0.026
0.77
0.041





receptor, beta 2


468
GABRB3
2562
gamma-aminobutyric acid (GABA) A
0.79
0.021
0.84
0.031





receptor, beta 3


469
GABRD
2563
gamma-aminobutyric acid (GABA) A
0.79
0.021
1.43
0.004





receptor, delta


470
GABRP
2568
gamma-aminobutyric acid (GABA) A
0.81
0.032
0.81
0.001





receptor, pi


471
GADD45GIP1
90480
growth arrest and DNA-damage-
0.77
0.008
1.64
0.002





inducible, gamma interacting protein 1


472
GAREM
64762
GRB2 associated, regulator of MAPK1
0.81
0.032
1.28
0.027


473
GARS
2617
glycyl-tRNA synthetase
0.81
0.025
2.24
0.005


474
GAS2
2620
growth arrest-specific 2
0.80
0.024
0.83
0.017


475
GATA3
2625
GATA binding protein 3
0.78
0.013
0.77
0.017


476
GATAD1
57798
GATA zinc finger domain containing 1
0.79
0.018
0.52
0.033


477
GCH1
2643
GTP cyclohydrolase 1
0.76
0.003
1.55
0.004


478
GDF10
2662
growth differentiation factor 10
0.78
0.015
0.79
0.019


479
GFPT1
2673
glutamine-fructose-6-phosphate
0.81
0.032
1.56
0.026





transaminase 1


480
GGA2
23062
golgi-associated, gamma adaptin ear
0.77
0.008
0.40
0.002





containing, ARF binding protein 2


481
GGCX
2677
gamma-glutamyl carboxylase
0.81
0.032
2.53
0.001


482
GHITM
27069
growth hormone inducible
0.82
0.044
2.52
0.005





transmembrane protein


483
GINS1
9837
GINS complex subunit 1 (Psf1
0.82
0.044
1.42
0.014





homolog)


484
GINS2
51659
GINS complex subunit 2 (Psf2
0.78
0.018
1.18
0.033





homolog)


485
GLB1L
79411
galactosidase, beta 1-like
0.80
0.023
0.57
0.004


486
GLI2
2736
GLI family zinc finger 2
0.81
0.044
0.56
0.006


487
GLI3
2737
GLI family zinc finger 3
0.77
0.012
0.70
0.047


488
GLO1
2739
glyoxalase I
0.81
0.040
1.84
0.003


489
GLRX2
51022
glutaredoxin 2
0.77
0.018
2.32
0.001


490
GLRX5
51218
glutaredoxin 5
0.77
0.017
1.30
0.042


491
GLUL
2752
glutamate-ammonia ligase
0.74
0.006
0.61
0.014


492
GLYR1
84656
glyoxylate reductase 1 homolog
0.79
0.010
0.55
0.017





(Arabidopsis)


493
GMFB
2764
glia maturation factor, beta
0.80
0.035
1.90
0.036


494
GMNN
51053
geminin, DNA replication inhibitor
0.77
0.010
1.52
0.013


495
GMPS
8833
guanine monphosphate synthase
0.79
0.020
1.64
0.009


496
GNA11
2767
guanine nucleotide binding protein (G
0.80
0.026
0.49
0.028





protein), alpha 11 (Gq class)


497
GNAZ
2781
guanine nucleotide binding protein (G
0.81
0.047
1.69
0.038





protein), alpha z polypeptide


498
GNB2L1
10399
guanine nucleotide binding protein (G
0.82
0.047
0.56
0.013





protein), beta polypeptide 2-like 1


499
GNG12
55970
guanine nucleotide binding protein (G
0.81
0.033
0.61
0.002





protein), gamma 12


500
GNL1
2794
guanine nucleotide binding protein-like
0.80
0.027
0.68
0.046





1


501
GNL2
29889
guanine nucleotide binding protein-like
0.80
0.022
1.60
0.042





2 (nucleolar)


502
GNPTAB
79158
N-acetylglucosamine-1-phosphate
0.79
0.014
1.66
0.033





transferase, alpha and beta subunits


503
GNRH1
2796
gonadotropin-releasing hormone 1
0.78
0.009
0.74
0.000





(luteinizing-releasing hormone)


504
GOLGA6L4
374650
golgin A6 family-like 4
0.78
0.017
0.73
0.018


505
GOLGA8A
23015
golgin A8 family, member A
0.79
0.018
0.79
0.017


506
GOLGA8A
23015
golgin A8 family, member A
0.79
0.015
0.66
0.002


507
GOLT1B
51026
golgi transport 1B
0.73
0.002
2.94
0.000


508
GON4L
54856
gon-4-like (C. elegans)
0.76
0.009
0.37
0.005


509
GORASP1
64689
golgi reassembly stacking protein 1,
0.80
0.030
0.46
0.018





65 kDa


510
GOT1
2805
glutamic-oxaloacetic transaminase 1,
0.76
0.010
1.48
0.017





soluble


511
GPN1
11321
GPN-loop GTPase 1
0.78
0.011
1.86
0.016


512
GPR153
387509
G protein-coupled receptor 153
0.75
0.007
0.66
0.029


513
GPR22
2845
G protein-coupled receptor 22
0.81
0.039
0.80
0.025


514
GPR35
2859
G protein-coupled receptor 35
0.80
0.023
0.75
0.029


515
GPRASP1
9737
G protein-coupled receptor associated
0.79
0.022
0.64
0.017





sorting protein 1


516
GPRC5C
55890
G protein-coupled receptor, class C,
0.79
0.020
1.41
0.034





group 5, member C


517
GPRIN2
9721
G protein regulated inducer of neurite
0.75
0.006
0.68
0.005





outgrowth 2


518
GPX4
2879
glutathione peroxidase 4
0.80
0.029
0.52
0.023


519
GREB1L
80000
growth regulation by estrogen in breast
0.80
0.023
0.82
0.049





cancer-like


520
GRK6
2870
G protein-coupled receptor kinase 6
0.80
0.021
1.75
0.020


521
GRSF1
2926
G-rich RNA sequence binding factor 1
0.78
0.016
2.08
0.009


522
GRWD1
83743
glutamate-rich WD repeat containing 1
0.80
0.033
1.65
0.026


523
GSK3B
2932
glycogen synthase kinase 3 beta
0.79
0.017
1.99
0.003


524
GSTM2
2946
glutathione S-transferase mu 2
0.79
0.016
0.75
0.047





(muscle)


525
GSTM4
2948
glutathione S-transferase mu 4
0.78
0.012
0.60
0.041


526
GTF2A2
2958
general transcription factor IIA, 2,
0.79
0.017
1.73
0.041





12 kDa


527
GTF2E1
2960
general transcription factor II E,
0.79
0.026
1.91
0.036





polypeptide 1, alpha 56 kDa


528
GTF2H5
404672
general transcription factor IIH,
0.80
0.028
2.02
0.011





polypeptide 5


529
GTF2IRD2B
389524
GTF2I repeat domain containing 2B
0.77
0.011
0.67
0.010


530
GTPBP4
23560
GTP binding protein 4
0.79
0.017
2.05
0.007


531
GUF1
60558
GUF1 GTPase homolog (S. cerevisiae)
0.82
0.046
2.03
0.009


532
GUK1
2987
guanylate kinase 1
0.79
0.017
1.77
0.016


533
GYS2
2998
glycogen synthase 2 (liver)
0.77
0.014
0.84
0.030


534
GZMB
3002
granzyme B (granzyme 2, cytotoxic T-
0.77
0.005
1.36
0.019





lymphocyte-associated serine esterase





1)


535
H2AFV
94239
H2A histone family, member V
0.80
0.015
3.04
0.000


536
H2AFZ
3015
H2A histone family, member Z
0.77
0.009
2.23
0.001


537
HAB1
55547
B1 for mucin
0.82
0.039
1.53
0.037


538
HAO1
54363
hydroxyacid oxidase (glycolate
0.76
0.008
0.81
0.015





oxidase) 1


539
HAUS2
55142
HAUS augmin-like complex, subunit 2
0.75
0.008
0.76
0.033


540
HCCS
3052
holocytochrome c synthase
0.80
0.034
2.95
0.003


541
HCFC2
29915
host cell factor C2
0.80
0.024
1.79
0.033


542
HCG18
414777
HLA complex group 18 (non-protein
0.79
0.018
0.82
0.022





coding)


543
HDAC2
3066
histone deacetylase 2
0.79
0.015
1.44
0.039


544
HDAC5
10014
histone deacetylase 5
0.80
0.021
0.72
0.043


545
HDAC7
51564
histone deacetylase 7
0.78
0.016
0.71
0.039


546
HDDC2
51020
HD domain containing 2
0.77
0.010
1.89
0.008


547
HEATR6
63897
HEAT repeat containing 6
0.78
0.015
1.79
0.001


548
HFE
3077
hemochromatosis
0.76
0.007
0.48
0.005


549
HGD
3081
homogentisate 1,2-dioxygenase
0.81
0.028
1.32
0.030


550
HIP1
3092
huntingtin interacting protein 1
0.81
0.038
0.49
0.005


551
HIST1H1C
3006
histone cluster 1, H1c
0.79
0.023
1.25
0.031


552
HIST1H2AG
8329
histone cluster 1, H2ag
0.77
0.011
1.36
0.017


553
HIST1H2AJ
8331
histone cluster 1, H2aj
0.79
0.024
1.47
0.046


554
HIST1H2BB
3018
histone cluster 1, H2bb
0.77
0.008
1.35
0.007


555
HIST1H3C
8352
histone cluster 1, H3c
0.80
0.023
1.61
0.001


556
HIST1H4H
8365
histone cluster 1, H4h
0.79
0.020
1.28
0.046


557
HIST3H2A
92815
histone cluster 3, H2a
0.81
0.034
1.28
0.027


558
HJURP
55355
Holliday junction recognition protein
0.79
0.023
1.36
0.049


559
HK2
3099
hexokinase 2
0.78
0.015
0.75
0.005


560
HLTF
6596
helicase-like transcription factor
0.82
0.038
1.79
0.005


561
HMBOX1
79618
homeobox containing 1
0.79
0.020
0.66
0.002


562
HMGB2
3148
high mobility group box 2
0.80
0.024
1.39
0.027


563
HMGN5
79366
high mobility group nucleosome
0.83
0.048
1.50
0.040





binding domain 5


564
HMHB1
57824
histocompatibility (minor) HB-1
0.82
0.047
1.71
0.010


565
HN1
51155
hematological and neurological
0.76
0.011
1.35
0.039





expressed 1


566
HNRNPA1
3178
heterogeneous nuclear
0.79
0.026
0.52
0.007





ribonucleoprotein A1


567
HNRNPA1
3178
heterogeneous nuclear
0.79
0.026
0.41
0.005





ribonucleoprotein A1


568
HNRNPDL
9987
heterogeneous nuclear
0.80
0.023
0.63
0.031





ribonucleoprotein D-like


569
HOXA10
3206
homeobox A10
0.81
0.046
0.75
0.026


570
HOXA4
3201
homeobox A4
0.78
0.015
0.74
0.011


571
HOXA5
3202
homeobox A5
0.80
0.032
0.65
0.000


572
HP
3240
haptoglobin
0.80
0.036
0.78
0.023


573
HPRT1
3251
hypoxanthine
0.77
0.009
1.83
0.003





phosphoribosyltransferase 1


574
HS2ST1
9653
heparan sulfate 2-O-sulfotransferase 1
0.77
0.012
1.48
0.042


575
HSBP1
3281
heat shock factor binding protein 1
0.77
0.011
1.65
0.021


576
HSD17B10
3028
hydroxysteroid (17-beta)
0.80
0.030
1.56
0.044





dehydrogenase 10


577
HSDL2
84263
hydroxysteroid dehydrogenase like 2
0.79
0.022
1.51
0.030


578
HSPA13
6782
heat shock protein 70 kDa family,
0.79
0.017
1.34
0.048





member 13


579
HSPH1
10808
heat shock 105 kDa/110 kDa protein 1
0.78
0.015
1.62
0.005


580
HTATIP2
10553
HIV-1 Tat interactive protein 2, 30 kDa
0.74
0.002
1.91
0.000


581
HTATSF1
27336
HIV-1 Tat specific factor 1
0.80
0.022
1.58
0.033


582
HTR1E
3354
5-hydroxytryptamine (serotonin)
0.77
0.016
0.77
0.007





receptor IE, G protein-coupled


583
HTR3B
9177
5-hydroxytryptamine (serotonin)
0.77
0.013
0.80
0.013





receptor 3B, ionotropic


584
HUWE1
10075
HECT, UBA and WWE domain
0.73
0.004
0.46
0.000





containing 1, E3 ubiquitin protein





ligase


585
HYPK
10169
huntingtin interacting protein K
0.78
0.019
1.93
0.003


586
IARS
3376
isoleucyl-tRNA synthetase
0.82
0.036
1.84
0.014


587
ICMT
23463
isoprenylcysteine carboxyl
0.80
0.025
2.03
0.043





methyltransferase


588
ICT1
3396
immature colon carcinoma transcript 1
0.77
0.015
1.83
0.003


589
ID4
3400
inhibitor of DNA binding 4, dominant
0.81
0.039
0.75
0.009





negative helix-loop-helix protein


590
IDH2
3418
isocitrate dehydrogenase 2 (NADP+),
0.75
0.008
1.45
0.017





mitochondrial


591
IDH3A
3419
isocitrate dehydrogenase 3 (NAD+)
0.79
0.022
1.82
0.019





alpha


592
IDI1
3422
isopentenyl-diphosphate delta
0.78
0.010
2.00
0.000





isomerase 1


593
IDO1
3620
indoleamine 2,3-dioxygenase 1
0.73
0.003
1.43
0.009


594
IDUA
3425
iduronidase, alpha-L-
0.77
0.011
0.66
0.044


595
IER2
9592
immediate early response 2
0.81
0.033
0.56
0.002


596
IFIH1
64135
interferon induced with helicase C
0.76
0.006
1.34
0.029





domain 1


597
IFIT5
24138
interferon-induced protein with
0.77
0.012
1.48
0.037





tetratricopeptide repeats 5


598
IFT46
56912
intraflagellar transport 46 homolog
0.81
0.015
0.35
0.000





(Chlamydomonas)


599
IGH
3492
immunoglobulin heavy locus
0.82
0.037
1.63
0.023


600
IKBKB
3551
inhibitor of kappa light polypeptide
0.76
0.007
0.63
0.010





gene enhancer in B-cells, kinase beta


601
IKZF1
10320
IKAROS family zinc finger 1 (Ikaros)
0.81
0.046
0.66
0.042


602
IKZF2
22807
IKAROS family zinc finger 2 (Helios)
0.79
0.016
0.83
0.030


603
IL11RA
3590
interleukin 11 receptor, alpha
0.80
0.025
0.77
0.023


604
IL1A
3552
interleukin 1, alpha
0.77
0.011
0.59
0.008


605
IL1RL2
8808
interleukin 1 receptor-like 2
0.78
0.023
1.40
0.006


606
IL5
3567
interleukin 5
0.82
0.047
1.25
0.019


607
IL6ST
3572
interleukin 6 signal transducer
0.78
0.013
0.69
0.003


608
ILF2
3608
interleukin enhancer binding factor 2
0.76
0.007
1.75
0.012


609
IMPA1
3612
inositol(myo)-1(or 4)-
0.78
0.013
1.58
0.008





monophosphatase 1


610
INADL
10207
InaD-like (Drosophila)
0.79
0.021
0.66
0.000


611
INPP5D
3635
inositol polyphosphate-5-phosphatase,
0.79
0.030
0.70
0.042





145 kDa


612
INPP5K
51763
inositol polyphosphate-5-phosphatase
0.83
0.046
0.40
0.019





K


613
INTS6
26512
integrator complex subunit 6
0.78
0.012
1.46
0.041


614
INTS7
25896
integrator complex subunit 7
0.75
0.007
1.61
0.014


615
INTS8
55656
integrator complex subunit 8
0.78
0.016
1.54
0.012


616
INTS9
55756
integrator complex subunit 9
0.78
0.018
0.47
0.007


617
IPO7
10527
importin 7
0.80
0.023
1.94
0.026


618
IQCK
124152
IQ motif containing K
0.82
0.038
0.51
0.008


619
IRAK3
11213
interleukin-1 receptor-associated
0.80
0.037
0.81
0.024





kinase 3


620
IRF2BP1
26145
interferon regulatory factor 2 binding
0.81
0.025
0.77
0.043





protein 1


621
ISG20
3669
interferon stimulated exonuclease gene
0.76
0.004
1.48
0.018





20 kDa


622
ISOC2
79763
isochorismatase domain containing 2
0.81
0.039
1.80
0.031


623
ITGA10
8515
integrin, alpha 10
0.77
0.011
0.67
0.047


624
ITGA2
3673
integrin, alpha 2 (CD49B, alpha 2
0.81
0.043
0.68
0.034





subunit of VLA-2 receptor)


625
ITGA7
3679
integrin, alpha 7
0.81
0.045
0.85
0.044


626
ITGA9
3680
integrin, alpha 9
0.81
0.031
0.73
0.028


627
ITIH4
3700
inter-alpha-trypsin inhibitor heavy
0.76
0.007
0.72
0.015





chain family, member 4


628
IVD
3712
isovaleryl-CoA dehydrogenase
0.83
0.049
0.56
0.030


629
IVNS1ABP
10625
influenza virus NS1A binding protein
0.79
0.015
1.79
0.017


630
JADE2
23338
jade family PHD finger 2
0.80
0.035
0.44
0.003


631
JAG2
3714
jagged 2
0.76
0.011
0.60
0.037


632
JAM2
58494
junctional adhesion molecule 2
0.82
0.041
0.62
0.022


633
JMJD6
23210
jumonji domain containing 6
0.77
0.010
2.24
0.005


634
JMJD7-PLA2G4B
8681
JMJD7-PLA2G4B readthrough
0.75
0.005
0.70
0.015


635
JUN
3725
jun proto-oncogene
0.82
0.042
0.61
0.002


636
JUP
3728
junction plakoglobin
0.78
0.024
0.78
0.011


637
KANK1
23189
KN motif and ankyrin repeat domains
0.79
0.027
0.51
0.003





1


638
KANK2
25959
KN motif and ankyrin repeat domains
0.80
0.028
0.63
0.044





2


639
KANSL1L
151050
KAT8 regulatory NSL complex
0.80
0.019
1.38
0.017





subunit 1-like


640
KANSL2
54934
KAT8 regulatory NSL complex
0.75
0.011
2.50
0.004





subunit 2


641
KCMF1
56888
potassium channel modulatory factor 1
0.77
0.005
3.20
0.002


642
KCNC4
3749
potassium voltage-gated channel,
0.78
0.013
0.79
0.029





Shaw-related subfamily, member 4


643
KCNG1
3755
potassium voltage-gated channel,
0.79
0.019
1.23
0.050





subfamily G, member 1


644
KCNK1
3775
potassium channel, subfamily K,
0.81
0.040
1.23
0.022





member 1


645
KCNMB1
3779
potassium large conductance calcium-
0.79
0.018
0.61
0.007





activated channel, subfamily M, beta





member 1


646
KCNS3
3790
potassium voltage-gated channel,
0.80
0.033
0.68
0.016





delayed-rectifier, subfamily S, member





3


647
KDELR2
11014
KDEL (Lys-Asp-Glu-Leu)
0.80
0.031
1.71
0.019





endoplasmic reticulum protein





retention receptor 2


648
KEAP1
9817
kelch-like ECH-associated protein 1
0.79
0.016
1.72
0.030


649
KHDRBS3
10656
KH domain containing, RNA binding,
0.80
0.023
1.35
0.043





signal transduction associated 3


650
KIAA0101
9768
KIAA0101
0.78
0.012
1.56
0.004


651
KIAA0391
5687
KIAA0391
0.76
0.008
1.67
0.027


652
KIAA0485
57235
uncharacterized LOC57235
0.80
0.022
0.73
0.002


653
KIAA0556
23247
KIAA0556
0.73
0.003
0.48
0.006


654
KIAA0753
9851
KIAA0753
0.76
0.004
0.47
0.001


655
KIAA0754
643314
KIAA0754
0.80
0.023
0.84
0.004


656
KIAA1024
23251
KIAA1024
0.77
0.008
1.31
0.007


657
KIF11
3832
kinesin family member 11
0.78
0.012
1.61
0.003


658
KIF13B
23303
kinesin family member 13B
0.77
0.012
0.59
0.002


659
KIF14
9928
kinesin family member 14
0.78
0.013
1.73
0.001


660
KIF20A
10112
kinesin family member 20A
0.76
0.011
1.46
0.005


661
KIF23
9493
kinesin family member 23
0.79
0.021
1.22
0.028


662
KIF2C
11004
kinesin family member 2C
0.78
0.009
1.90
0.003


663
KIF4A
24137
kinesin family member 4A
0.79
0.016
1.73
0.003


664
KIF5B
3799
kinesin family member 5B
0.80
0.022
1.77
0.033


665
KIFC1
3833
kinesin family member C1
0.78
0.015
1.35
0.003


666
KIT
3815
v-kit Hardy-Zuckerman 4 feline
0.79
0.023
0.77
0.010





sarcoma viral oncogene homolog


667
KLF13
51621
Kruppel-like factor 13
0.80
0.037
0.81
0.019


668
KLF2
10365
Kruppel-like factor 2
0.80
0.034
0.63
0.028


669
KLHL7
55975
kelch-like family member 7
0.78
0.010
1.57
0.036


670
KMT2A
4297
lysine (K)-specific methyltransferase
0.81
0.048
0.33
0.000





2A


671
KMT2A
4297
lysine (K)-specific methyltransferase
0.81
0.029
0.79
0.020





2A


672
KPNA2
3838
karyopherin alpha 2 (RAG cohort 1,
0.79
0.021
1.44
0.038





importin alpha 1)


673
KPTN
11133
kaptin (actin binding protein)
0.80
0.027
0.74
0.046


674
KRAS
3845
Kirsten rat sarcoma viral oncogene
0.82
0.047
1.87
0.016





homolog


675
KRR1
11103
KRR1, small subunit (SSU)
0.81
0.039
1.87
0.001





processome component, homolog





(yeast)


676
KRT14
3861
keratin 14
0.78
0.024
0.89
0.014


677
KRT15
3866
keratin 15
0.80
0.025
0.88
0.002


678
KRT23
25984
keratin 23 (histone deacetylase
0.79
0.014
0.85
0.015





inducible)


679
KRT5
3852
keratin 5
0.73
0.006
0.75
0.000


680
KRT6B
3854
keratin 6B
0.76
0.018
0.77
0.023


681
LAGE3
8270
L antigen family, member 3
0.79
0.022
1.47
0.010


682
LAMA1
284217
laminin, alpha 1
0.78
0.014
0.77
0.020


683
LAMA3
3909
laminin, alpha 3
0.81
0.027
0.81
0.002


684
LAMB2
3913
laminin, beta 2 (laminin S)
0.78
0.011
0.55
0.001


685
LAMP1
3916
lysosomal-associated membrane
0.78
0.018
0.59
0.031





protein 1


686
LANCL1
10314
LanC lantibiotic synthetase component
0.82
0.050
0.60
0.042





C-like 1 (bacterial)


687
LAPTM4A
9741
lysosomal protein transmembrane 4
0.77
0.010
0.35
0.001





alpha


688
LAPTM4B
55353
lysosomal protein transmembrane 4
0.80
0.036
1.34
0.012





beta


689
LDLR
3949
low density lipoprotein receptor
0.79
0.019
1.60
0.002


690
LDLRAP1
26119
low density lipoprotein receptor
0.80
0.044
0.50
0.002





adaptor protein 1


691
LEPR
3953
leptin receptor
0.81
0.036
0.54
0.008


692
LEPREL1
55214
leprecan-like 1
0.79
0.024
0.77
0.008


693
LEPREL4
10609
leprecan-like 4
0.79
0.029
1.30
0.046


694
LEPROT
54741
leptin receptor overlapping transcript
0.82
0.044
0.42
0.002


695
LETMD1
25875
LETM1 domain containing 1
0.77
0.006
0.44
0.001


696
LGR4
55366
leucine-rich repeat containing G
0.80
0.021
1.32
0.015





protein-coupled receptor 4


697
LGR5
8549
leucine-rich repeat containing G
0.81
0.034
0.77
0.027





protein-coupled receptor 5


698
LHFP
10186
lipoma HMGIC fusion partner
0.81
0.047
0.59
0.003


699
LHX1
3975
LIM homeobox 1
0.79
0.024
1.27
0.024


700
LINC00472
79940
long intergenic non-protein coding
0.79
0.014
0.73
0.000





RNA 472


701
LINC00965
349196
long intergenic non-protein coding
0.80
0.031
0.78
0.022





RNA 965


702
LMAN1
3998
lectin, mannose-binding, 1
0.78
0.014
1.41
0.012


703
LMBR1L
55716
limb development membrane protein
0.77
0.013
0.50
0.018





1-like


704
LMF1
64788
lipase maturation factor 1
0.77
0.009
0.58
0.013


705
LMO7
4008
LIM domain 7
0.78
0.015
0.75
0.000


706
LOC100129361
100129361
chromosome X open reading frame 69-
0.77
0.010
1.60
0.016





like


707
LOC100505498
100505498
uncharacterized LOC100505498
0.74
0.004
0.62
0.002


708
LOC100505915
100505915
uncharacterized LOC100505915
0.80
0.021
0.74
0.002


709
LOC100506282
100506282
uncharacterized LOC100506282
0.78
0.011
0.76
0.005


710
LOC100506469
100506469
uncharacterized LOC100506469
0.75
0.004
0.57
0.003


711
LONP2
100507577
Ion peptidase 2, peroxisomal
0.78
0.019
0.48
0.000


712
SIAH1
100507577
siah E3 ubiquitin protein ligase 1
0.78
0.017
0.71
0.004


713
SNORD14B
100508408
small nucleolar RNA, C/D box 14B
0.81
0.040
0.42
0.004


714
ZNF44
101060181
zinc finger protein 44
0.76
0.008
0.58
0.017


715
NPIPB5
101929910
nuclear pore complex interacting
0.77
0.007
0.76
0.003





protein family, member B5


716
POLR3E
101060521
polymerase (RNA) III (DNA directed)
0.81
0.022
0.43
0.001





polypeptide E (80 kD)


717
LOC101927792
101927792
uncharacterized LOC101927792
0.80
0.028
1.22
0.041


718
RSRP1
101928189
arginine/serine-rich protein 1
0.79
0.027
0.69
0.024


719
MFAP3L
101928198
microfibrillar-associated protein 3-like
0.82
0.045
1.30
0.043


720
SLC19A1
101928717
solute carrier family 19 (folate
0.79
0.023
1.46
0.033





transporter), member 1


721
TPT1
101928826
tumor protein, translationally-
0.74
0.005
0.46
0.001





controlled 1


722
SNX29
101929304
sorting nexin 29
0.82
0.031
0.72
0.019


723
RIF1
101929336
replication timing regulatory factor 1
0.79
0.014
1.59
0.011


724
SULT1A4
101929857
sulfotransferase family, cytosolic, 1A,
0.76
0.007
0.59
0.008





phenol-preferring, member 4


725
NPIPB5
101929910
nuclear pore complex interacting
0.80
0.020
0.73
0.005





protein family, member B5


726
PKD1P1
102724993
polycystic kidney disease 1 (autosomal
0.78
0.013
0.65
0.002





dominant) pseudogene 1


727
PKD1P1
101930075
polycystic kidney disease 1 (autosomal
0.79
0.018
0.70
0.016





dominant) pseudogene 1


728
PKD1
101930075
polycystic kidney disease 1 (autosomal
0.76
0.007
0.61
0.014





dominant)


729
PPP2R3B
102725016
protein phosphatase 2, regulatory
0.83
0.049
1.24
0.022





subunit B″, beta


730
LOC155060
155060
AI894139 pseudogene
0.79
0.012
0.65
0.019


731
LOC157562
157562
uncharacterized LOC157562
0.77
0.015
1.38
0.004


732
LOC202181
202181
SUMO-interacting motifs containing 1
0.79
0.018
0.83
0.015





pseudogene


733
PDE4C
729966
phosphodiesterase 4C, cAMP-specific
0.76
0.008
0.81
0.038


734
LONRF1
91694
LON peptidase N-terminal domain and
0.79
0.020
0.70
0.006





ring finger 1


735
LPCAT4
254531
lysophosphatidylcholine
0.80
0.027
0.43
0.002





acyltransferase 4


736
LPPR2
64748
lipid phosphate phosphatase-related
0.77
0.025
1.55
0.027





protein type 2


737
LRIG1
26018
leucine-rich repeats and
0.81
0.020
0.65
0.004





immunoglobulin-like domains 1


738
LRRC1
55227
leucine rich repeat containing 1
0.80
0.027
1.50
0.028


739
LRRC17
10234
leucine rich repeat containing 17
0.83
0.049
0.71
0.016


740
LRRC48
83450
leucine rich repeat containing 48
0.81
0.030
0.54
0.003


741
LRRC59
55379
leucine rich repeat containing 59
0.78
0.013
1.78
0.001


742
LRRFIP1
9208
leucine rich repeat (in FLII) interacting
0.79
0.019
0.73
0.048





protein 1


743
LRRN3
54674
leucine rich repeat neuronal 3
0.80
0.028
0.68
0.044


744
LRRTM2
26045
leucine rich repeat transmembrane
0.82
0.044
0.79
0.022





neuronal 2


745
LSM1
27257
LSM1, U6 small nuclear RNA
0.80
0.034
1.90
0.000





associated


746
LSM3
27258
LSM3 homolog, U6 small nuclear
0.75
0.007
1.68
0.019





RNA associated (S. cerevisiae)


747
LTBP3
4054
latent transforming growth factor beta
0.79
0.026
0.55
0.000





binding protein 3


748
LTBP4
8425
latent transforming growth factor beta
0.79
0.022
0.66
0.028





binding protein 4


749
LYRM4
57128
LYR motif containing 4
0.74
0.007
1.70
0.027


750
MAB21L1
4081
mab-21-like 1 (C. elegans)
0.82
0.046
0.79
0.010


751
MACF1
23499
microtubule-actin crosslinking factor 1
0.79
0.027
0.48
0.000


752
MAD2L1
4085
MAD2 mitotic arrest deficient-like 1
0.77
0.009
1.57
0.001





(yeast)


753
MAD2L1BP
9587
MAD2L1 binding protein
0.79
0.017
2.01
0.021


754
MAFK
7975
v-maf avian musculoaponeurotic
0.77
0.011
1.29
0.015





fibrosarcoma oncogene homolog K


755
MAGED1
9500
melanoma antigen family D, 1
0.78
0.012
1.56
0.014


756
MAGOHB
55110
mago-nashi homolog B (Drosophila)
0.78
0.016
1.23
0.031


757
MAK
4117
male germ cell-associated kinase
0.81
0.039
0.85
0.044


758
MAN1C1
57134
mannosidase, alpha, class 1C, member
0.80
0.022
0.56
0.018





1


759
MAN2C1
4123
mannosidase, alpha, class 2C, member
0.80
0.018
0.55
0.026





1


760
MAOA
4128
monoamine oxidase A
0.80
0.028
0.71
0.001


761
MAOB
4129
monoamine oxidase B
0.81
0.041
0.89
0.048


762
MAP2K1
5604
mitogen-activated protein kinase
0.77
0.014
1.99
0.016





kinase 1


763
MAP3K12
7786
mitogen-activated protein kinase
0.77
0.008
0.61
0.004





kinase kinase 12


764
MAP3K13
9175
mitogen-activated protein kinase
0.79
0.018
1.27
0.049





kinase kinase 13


765
MAP7D3
79649
MAP7 domain containing 3
0.76
0.010
0.72
0.027


766
MAPK1
5594
mitogen-activated protein kinase 1
0.77
0.016
1.75
0.015


767
MARCH2
51257
membrane-associated ring finger
0.83
0.039
0.51
0.005





(C3HC4) 2, E3 ubiquitin protein ligase


768
MARCH5
54708
membrane-associated ring finger
0.79
0.017
2.06
0.033





(C3HC4) 5


769
MARCH8
220972
membrane-associated ring finger
0.81
0.033
0.55
0.002





(C3HC4) 8, E3 ubiquitin protein ligase


770
MARS
4141
methionyl-tRNA synthetase
0.77
0.008
3.44
0.000


771
MAST4
375449
microtubule associated
0.81
0.031
0.74
0.043





serine/threonine kinase family member





4


772
MBD3
53615
methyl-CpG binding domain protein 3
0.79
0.020
0.58
0.049


773
MCAT
27349
malonyl CoA:ACP acyltransferase
0.79
0.021
1.84
0.015





(mitochondrial)


774
MCL1
4170
myeloid cell leukemia 1
0.81
0.045
0.59
0.021


775
MCM10
55388
minichromosome maintenance
0.79
0.016
1.39
0.001





complex component 10


776
MCM2
4171
minichromosome maintenance
0.81
0.028
1.53
0.012





complex component 2


777
MCM6
4175
minichromosome maintenance
0.77
0.013
1.63
0.020





complex component 6


778
MCUR1
63933
mitochondrial calcium uniporter
0.76
0.004
1.95
0.002





regulator 1


779
ME3
10873
malic enzyme 3, NADP(+)-dependent,
0.78
0.020
0.69
0.002





mitochondrial


780
MEA1
4201
male-enhanced antigen 1
0.80
0.030
1.66
0.044


781
MED1
5469
mediator complex subunit 1
0.78
0.012
1.40
0.003


782
MED31
51003
mediator complex subunit 31
0.80
0.020
0.66
0.022


783
MELK
9833
maternal embryonic leucine zipper
0.78
0.010
1.54
0.003





kinase


784
METTL2A
55798
methyltransferase like 2A
0.80
0.027
1.44
0.040


785
METTL5
29081
methyltransferase like 5
0.79
0.019
2.03
0.012


786
MGC12488
84786
uncharacterized protein MGC12488
0.78
0.010
0.78
0.002


787
MGP
4256
matrix Gla protein
0.81
0.027
0.81
0.007


788
MGRN1
23295
mahogunin ring finger 1, E3 ubiquitin
0.80
0.029
0.56
0.035





protein ligase


789
MIF
4282
macrophage migration inhibitory factor
0.78
0.026
1.67
0.003





(glycosylation-inhibiting factor)


790
MINK1
50488
misshapen-like kinase 1
0.80
0.035
0.53
0.005


791
NELFE
100302242
negative elongation factor complex
0.74
0.005
1.68
0.009





member E


792
UCK2
100500832
uridine-cytidine kinase 2
0.75
0.007
1.69
0.004


793
STMN1
100500808
stathmin 1
0.74
0.005
1.82
0.010


794
PDCD4
100616113
programmed cell death 4 (neoplastic
0.82
0.046
0.81
0.026





transformation inhibitor)


795
NDUFS8
102465669
NADH dehydrogenase (ubiquinone)
0.80
0.031
1.67
0.003





Fe—S protein 8, 23 kDa (NADH-





coenzyme Q reductase)


796
NEAT1
693197
nuclear paraspeckle assembly
0.77
0.010
0.81
0.040





transcript 1 (non-protein coding)


797
NEIL1
693216
nei endonuclease VIII-like 1 (E. coli)
0.80
0.030
0.72
0.043


798
PFDN6
102465501
prefoldin subunit 6
0.80
0.037
2.05
0.006


799
SEMA3B
102465526
sema domain, immunoglobulin domain
0.80
0.023
0.84
0.030





(Ig), short basic domain, secreted,





(semaphorin) 3B


800
QARS
102465536
glutaminyl-tRNA synthetase
0.77
0.013
0.36
0.002


801
MIS18A
54069
MIS18 kinetochore protein A
0.81
0.029
1.86
0.033


802
MKL2
57496
MKL/myocardin-like 2
0.80
0.020
0.57
0.025


803
MKNK2
2872
MAP kinase interacting
0.75
0.007
0.49
0.000





serine/threonine kinase 2


804
MLF1
4291
myeloid leukemia factor 1
0.80
0.027
1.39
0.041


805
MLLT10
8028
myeloid/lymphoid or mixed-lineage
0.75
0.010
0.52
0.049





leukemia; translocated to, 10


806
MLLT11
10962
myeloid/lymphoid or mixed-lineage
0.79
0.019
1.42
0.023





leukemia; translocated to, 11


807
MLLT3
4300
myeloid/lymphoid or mixed-lineage
0.78
0.018
0.55
0.006





leukemia; translocated to, 3


808
MLPH
79083
melanophilin
0.76
0.008
0.73
0.013


809
MMP1
4312
matrix metallopeptidase 1 (interstitial
0.74
0.004
1.22
0.000





collagenase)


810
MMP11
4320
matrix metallopeptidase 11
0.81
0.048
1.18
0.050





(stromelysin 3)


811
MNT
4335
MAX network transcriptional repressor
0.80
0.032
0.40
0.006


812
MOK
5891
MOK protein kinase
0.76
0.005
0.39
0.000


813
MOSPD1
56180
motile sperm domain containing 1
0.79
0.023
2.16
0.005


814
MPRIP
23164
myosin phosphatase Rho interacting
0.79
0.024
0.55
0.029





protein


815
MR1
3140
major histocompatibility complex,
0.79
0.029
0.53
0.033





class I-related


816
MRPL13
28998
mitochondrial ribosomal protein L13
0.78
0.017
1.72
0.000


817
MRPL15
29088
mitochondrial ribosomal protein L15
0.79
0.025
1.90
0.000


818
MRPL17
63875
mitochondrial ribosomal protein L17
0.76
0.008
2.05
0.002


819
MRPL18
29074
mitochondrial ribosomal protein LI8
0.77
0.011
2.21
0.001


820
MRPL22
29093
mitochondrial ribosomal protein L22
0.79
0.017
1.75
0.031


821
MRPL23
6150
mitochondrial ribosomal protein L23
0.79
0.022
1.51
0.025


822
MRPL3
11222
mitochondrial ribosomal protein L3
0.74
0.004
2.34
0.000


823
MRPL35
51318
mitochondrial ribosomal protein L35
0.73
0.005
1.21
0.022


824
MRPL40
64976
mitochondrial ribosomal protein L40
0.79
0.023
1.36
0.049


825
MRPL41
64975
mitochondrial ribosomal protein L41
0.83
0.050
1.28
0.049


826
MRPL48
51642
mitochondrial ribosomal protein L48
0.78
0.019
2.51
0.000


827
MRPS10
55173
mitochondrial ribosomal protein S10
0.81
0.038
1.96
0.010


828
MRPS11
64963
mitochondrial ribosomal protein S11
0.71
0.003
1.86
0.006


829
MRPS12
6183
mitochondrial ribosomal protein S12
0.77
0.015
1.48
0.028


830
MRPS14
63931
mitochondrial ribosomal protein S14
0.79
0.022
1.85
0.021


831
MRPS16
51021
mitochondrial ribosomal protein S16
0.75
0.006
1.65
0.007


832
MRPS17
51373
mitochondrial ribosomal protein S17
0.76
0.013
3.13
0.000


833
MRPS18A
55168
mitochondrial ribosomal protein S18A
0.81
0.035
1.87
0.019


834
MRPS18C
51023
mitochondrial ribosomal protein S18C
0.81
0.031
1.46
0.004


835
MRPS22
56945
mitochondrial ribosomal protein S22
0.79
0.015
5.21
0.000


836
MRPS28
28957
mitochondrial ribosomal protein S28
0.81
0.039
1.33
0.029


837
MRPS33
51650
mitochondrial ribosomal protein S33
0.79
0.017
1.65
0.027


838
MRPS7
51081
mitochondrial ribosomal protein S7
0.79
0.021
2.07
0.003


839
MRS2
57380
MRS2 magnesium transporter
0.78
0.011
2.22
0.004


840
MS4A5
64232
membrane-spanning 4-domains,
0.78
0.013
1.46
0.024





subfamily A, member 5


841
MSH6
2956
mutS homolog 6
0.76
0.008
2.86
0.001


842
MSMO1
6307
methylsterol monooxygenase 1
0.79
0.022
1.36
0.046


843
MSR1
4481
macrophage scavenger receptor 1
0.77
0.013
1.46
0.040


844
MSRA
4482
methionine sulfoxide reductase A
0.80
0.025
0.43
0.003


845
MST1
4485
macrophage stimulating 1 (hepatocyte
0.76
0.006
0.69
0.008





growth factor-like)


846
MTA1
9112
metastasis associated 1
0.79
0.015
0.50
0.001


847
MTA2
9219
metastasis associated 1 family, member
0.79
0.033
0.73
0.033





2


848
MTCH2
23788
mitochondrial carrier 2
0.75
0.005
2.43
0.002


849
MTERF3
51001
mitochondrial transcription termination
0.82
0.049
1.58
0.003





factor 3


850
MTFR1
9650
mitochondrial fission regulator 1
0.80
0.026
1.50
0.004


851
MTG1
92170
mitochondrial ribosome-associated
0.80
0.034
0.49
0.003





GTPase 1


852
MTHFD2
10797
methylenetetrahydrofolate
0.76
0.005
2.59
0.000





dehydrogenase 2,





methenyltetrahydrofolate





cyclohydrolase


853
MTHFS
10588
5,10-methenyltetrahydrofolate
0.78
0.015
1.61
0.048





synthetase


854
MTM1
4534
myotubularin 1
0.78
0.016
1.43
0.042


855
MTMR2
8898
myotubularin related protein 2
0.78
0.017
1.57
0.050


856
MTPAP
55149
mitochondrial poly(A) polymerase
0.80
0.028
2.25
0.022


857
MTSS1
9788
metastasis suppressor 1
0.82
0.032
1.86
0.001


858
MTX1
4580
metaxin 1
0.79
0.027
1.52
0.044


859
MTX2
10651
metaxin 2
0.79
0.022
1.82
0.049


860
MUC7
4589
mucin 7, secreted
0.78
0.016
0.82
0.044


861
MUM1
84939
melanoma associated antigen (mutated)
0.82
0.037
0.52
0.001





1


862
MUT
4594
methylmalonyl CoA mutase
0.78
0.011
2.20
0.003


863
MXD1
4084
MAX dimerization protein 1
0.81
0.036
1.28
0.043


864
MYBL2
4605
v-myb avian myeloblastosis viral
0.77
0.012
1.26
0.010





oncogene homolog-like 2


865
MYH6
4624
myosin, heavy chain 6, cardiac muscle,
0.79
0.019
1.24
0.001





alpha


866
MYL12B
103910
myosin, light chain 12B, regulatory
0.78
0.015
0.69
0.036


867
MYL6
4637
myosin, light chain 6, alkali, smooth
0.76
0.007
0.47
0.017





muscle and non-muscle


868
MYNN
55892
myoneurin
0.80
0.026
1.76
0.036


869
MYO15B
80022
myosin XVB pseudogene
0.76
0.009
0.55
0.010


870
MYO1C
4641
myosin IC
0.75
0.006
0.62
0.014


871
MYO5A
4644
myosin VA (heavy chain 12, myoxin)
0.79
0.017
2.02
0.012


872
MYO5C
55930
myosin VC
0.80
0.019
0.63
0.038


873
MYOF
26509
myoferlin
0.80
0.021
0.61
0.013


874
MYOZ2
51778
myozenin 2
0.82
0.040
1.30
0.006


875
MZF1
7593
myeloid zinc finger 1
0.76
0.010
0.48
0.007


876
NAA10
8260
N(alpha)-acetyltransferase 10, NatA
0.79
0.016
1.89
0.014





catalytic subunit


877
NAA15
80155
N(alpha)-acetyltransferase 15, NatA
0.81
0.027
1.68
0.009





auxiliary subunit


878
NAA40
79829
N(alpha)-acetyltransferase 40, NatD
0.77
0.007
0.57
0.018





catalytic subunit


879
NAA50
80218
N(alpha)-acetyltransferase 50, NatE
0.81
0.030
1.60
0.037





catalytic subunit


880
NAA60
79903
N(alpha)-acetyltransferase 60, NatF
0.78
0.014
0.55
0.036





catalytic subunit


881
NAALAD2
10003
N-acetylated alpha-linked acidic
0.80
0.024
0.72
0.000





dipeptidase 2


882
NAB1
4664
NGFI-A binding protein 1 (EGR1
0.81
0.047
0.49
0.009





binding protein 1)


883
NACA
4666
nascent polypeptide-associated
0.77
0.010
0.37
0.003





complex alpha subunit


884
NANS
54187
N-acetylneuraminic acid synthase
0.79
0.013
1.59
0.016


885
NAP1L2
4674
nucleosome assembly protein 1-like 2
0.80
0.029
0.81
0.038


886
NAPG
8774
N-ethylmaleimide-sensitive factor
0.79
0.015
2.48
0.004





attachment protein, gamma


887
NARF
26502
nuclear prelamin A recognition factor
0.79
0.018
1.48
0.041


888
NAV2
89797
neuron navigator 2
0.82
0.043
0.75
0.045


889
NBPF1
55672
neuroblastoma breakpoint family,
0.81
0.041
0.66
0.050





member 1


890
NBPF10
25832
neuroblastoma breakpoint family,
0.81
0.034
0.62
0.015





member 10


891
NBR2
10230
neighbor of BRCA1 gene 2 (non-
0.78
0.015
0.79
0.017





protein coding)


892
NCAPG
64151
non-SMC condensin I complex,
0.76
0.005
1.98
0.000





subunit G


893
NCOA1
8648
nuclear receptor coactivator 1
0.77
0.008
0.41
0.003


894
NCOR1
9611
nuclear receptor corepressor 1
0.82
0.043
0.48
0.007


895
NCSTN
23385
nicastrin
0.78
0.019
0.52
0.021


896
NDE1
54820
nudE neurodevelopment protein 1
0.78
0.013
0.62
0.011


897
NDP
4693
Norrie disease (pseudoglioma)
0.77
0.011
0.78
0.014


898
NDRG2
57447
NDRG family member 2
0.76
0.009
0.64
0.004


899
NDST2
8509
N-deacetylase/N-sulfotransferase
0.82
0.047
0.74
0.023





(heparan glucosaminyl) 2


900
NDUFA3
4696
NADH dehydrogenase (ubiquinone) 1
0.78
0.017
1.60
0.010





alpha subcomplex, 3, 9 kDa


901
NDUFA6
4700
NADH dehydrogenase (ubiquinone) 1
0.76
0.010
1.80
0.002





alpha subcomplex, 6, 14 kDa


902
NDUFA7
4701
NADH dehydrogenase (ubiquinone) 1
0.80
0.032
1.30
0.023





alpha subcomplex, 7, 14.5 kDa


903
NDUFA8
4702
NADH dehydrogenase (ubiquinone) 1
0.73
0.002
2.26
0.000





alpha subcomplex, 8, 19 kDa


904
NDUFB3
4709
NADH dehydrogenase (ubiquinone) 1
0.79
0.018
4.97
0.000





beta subcomplex, 3, 12 kDa


905
NDUFB5
4711
NADH dehydrogenase (ubiquinone) 1
0.74
0.004
2.54
0.001





beta subcomplex, 5, 16 kDa


906
NDUFS1
4719
NADH dehydrogenase (ubiquinone)
0.78
0.012
2.31
0.000





Fe—S protein 1, 75 kDa


907
NDUFS3
4722
NADH dehydrogenase (ubiquinone)
0.79
0.019
1.93
0.014





Fe—S protein 3, 30 kDa


908
NEK2
4751
NIMA-related kinase 2
0.80
0.026
1.45
0.003


909
NEK3
4752
NIMA-related kinase 3
0.80
0.035
0.61
0.043


910
NEK9
91754
NIMA-related kinase 9
0.83
0.043
0.42
0.001


911
NELL1
4745
NEL-like 1 (chicken)
0.80
0.034
1.36
0.003


912
NEU2
4759
sialidase 2 (cytosolic sialidase)
0.78
0.017
1.41
0.008


913
NFATC2IP
84901
nuclear factor of activated T-cells,
0.76
0.013
0.42
0.001





cytoplasmic, calcineurin-dependent 2





interacting protein


914
NFE2L1
4779
nuclear factor, erythroid 2-like 1
0.77
0.013
0.49
0.006


915
NFIB
4781
nuclear factor I/B
0.81
0.041
0.79
0.037


916
NFRKB
4798
nuclear factor related to kappaB
0.76
0.007
0.37
0.000





binding protein


917
NFX1
4799
nuclear transcription factor, X-box
0.79
0.016
0.30
0.001





binding 1


918
NHLH1
4807
nescient helix loop helix 1
0.82
0.048
1.42
0.041


919
NINL
22981
ninein-like
0.79
0.014
0.55
0.010


920
NIPSNAP3B
55335
nipsnap homolog 3B (C. elegans)
0.78
0.013
0.76
0.038


921
NISCH
11188
nischarin
0.77
0.016
0.52
0.002


922
NIT2
56954
nitrilase family, member 2
0.80
0.026
1.71
0.037


923
NKX2-5
1482
NK2 homeobox 5
0.82
0.044
1.35
0.016


924
NME1
4830
NME/NM23 nucleoside diphosphate
0.80
0.034
1.66
0.002





kinase 1


925
NME5
8382
NME/NM23 family member 5
0.79
0.018
0.73
0.022


926
NMT2
9397
N-myristoyltransferase 2
0.82
0.048
0.55
0.043


927
NOL11
25926
nucleolar protein 11
0.79
0.019
1.66
0.021


928
NOL7
51406
nucleolar protein 7, 27 kDa
0.81
0.030
2.23
0.003


929
NOL9
79707
nucleolar protein 9
0.82
0.047
0.44
0.037


930
NOP10
55505
NOP10 ribonucleoprotein
0.72
0.002
2.63
0.001


931
NOP16
51491
NOP16 nucleolar protein
0.77
0.011
1.64
0.005


932
NOS1
4842
nitric oxide synthase 1 (neuronal)
0.77
0.011
0.69
0.038


933
NOTCH2
4853
notch 2
0.81
0.040
0.56
0.029


934
NOTCH2NL
388677
notch 2 N-terminal like
0.82
0.044
0.79
0.040


935
NOV
4856
nephroblastoma overexpressed
0.77
0.011
0.63
0.015


936
NPAS2
4862
neuronal PAS domain protein 2
0.81
0.032
0.70
0.035


937
NPC1L1
29881
NPC1-like 1
0.78
0.024
1.48
0.001


938
NPEPPS
9520
aminopeptidase puromycin sensitive
0.81
0.027
0.57
0.022


939
NPIPA1
9284
nuclear pore complex interacting
0.78
0.015
0.59
0.004





protein family, member A1


940
NPIPA1
9284
nuclear pore complex interacting
0.78
0.011
0.79
0.005





protein family, member A1


941
NPL
80896
N-acetylneuraminate pyruvate lyase
0.74
0.001
1.32
0.003





(dihydrodipicolinate synthase)


942
NPR3
4883
natriuretic peptide receptor 3
0.82
0.045
1.41
0.038


943
NPY1R
4886
neuropeptide Y receptor Y1
0.82
0.034
0.88
0.011


944
NPY5R
4889
neuropeptide Y receptor Y5
0.77
0.009
0.80
0.011


945
NQO1
1728
NAD(P)H dehydrogenase, quinone 1
0.79
0.020
1.35
0.009


946
NRAS
4893
neuroblastoma RAS viral (v-ras)
0.78
0.012
1.58
0.050





oncogene homolog


947
NRDE2
55051
NRDE-2, necessary for RNA
0.81
0.019
0.50
0.000





interference, domain containing


948
NRN1
51299
neuritin 1
0.80
0.027
0.56
0.011


949
NRXN1
9378
neurexin 1
0.77
0.009
0.68
0.012


950
NSDHL
50814
NAD(P) dependent steroid
0.78
0.016
1.59
0.015





dehydrogenase -like


951
NSFL1C
55968
NSFL1 (p97) cofactor (p47)
0.79
0.018
1.81
0.024


952
NSL1
25936
NSL1, MIS12 kinetochore complex
0.78
0.015
1.96
0.020





component


953
NT5DC3
51559
5′-nucleotidase domain containing 3
0.76
0.007
0.57
0.013


954
NT5E
4907
5′-nucleotidase, ecto (CD73)
0.76
0.011
1.30
0.045


955
NTRK2
4915
neurotrophic tyrosine kinase, receptor,
0.80
0.022
0.48
0.000





type 2


956
NUDT21
11051
nudix (nucleoside diphosphate linked
0.75
0.005
1.66
0.016





moiety X)-type motif 21


957
NUMA1
4926
nuclear mitotic apparatus protein 1
0.78
0.022
0.56
0.002


958
NUP155
9631
nucleoporin 155 kDa
0.80
0.031
1.50
0.048


959
NUP93
9688
nucleoporin 93 kDa
0.75
0.005
1.17
0.028


960
NUS1
11049
nuclear undecaprenyl pyrophosphate
0.79
0.019
1.59
0.028





synthase 1 homolog (S. cerevisiae)


961
NUS1P3
11049
nuclear undecaprenyl pyrophosphate
0.74
0.010
1.35
0.034





synthase 1 homolog (S. cerevisiae)





pseudogene 3


962
NUSAP1
51203
nucleolar and spindle associated
0.79
0.016
1.73
0.000





protein 1


963
NVL
4931
nuclear VCP-like
0.76
0.009
1.20
0.038


964
NXF1
10482
nuclear RNA export factor 1
0.79
0.025
0.43
0.003


965
NXF3
56000
nuclear RNA export factor 3
0.81
0.038
1.26
0.033


966
NXT2
55916
nuclear transport factor 2-like export
0.79
0.019
1.80
0.013





factor 2


967
OGT
8473
O-linked N-acetylglucosamine
0.78
0.017
0.56
0.002





(GlcNAc) transferase


968
OMD
4958
osteomodulin
0.80
0.024
0.74
0.038


969
OR5I1
10798
olfactory receptor, family 5, subfamily
0.78
0.014
0.79
0.035





I, member 1


970
OR7E47P
26628
olfactory receptor, family 7, subfamily
0.79
0.025
1.27
0.030





E, member 47 pseudogene


971
ORAI3
93129
ORAI calcium release-activated
0.77
0.006
0.46
0.002





calcium modulator 3


972
ORC4
5000
origin recognition complex, subunit 4
0.78
0.013
1.55
0.036


973
ORC5
5001
origin recognition complex, subunit 5
0.77
0.011
1.86
0.021


974
ORMDL2
29095
ORMDL sphingolipid biosynthesis
0.77
0.010
2.27
0.000





regulator 2


975
OS9
10956
osteosarcoma amplified 9, endoplasmic
0.80
0.032
0.53
0.037





reticulum lectin


976
OSBPL10
114884
oxysterol binding protein-like 10
0.81
0.046
0.57
0.022


977
OXR1
55074
oxidation resistance 1
0.82
0.044
1.50
0.012


978
P2RY2
5029
purinergic receptor P2Y, G-protein
0.80
0.031
1.19
0.049





coupled, 2


979
PA2G4
5036
proliferation-associated 2G4, 38 kDa
0.82
0.043
2.15
0.007


980
PAFAH1B3
5050
platelet-activating factor
0.77
0.012
1.98
0.000





acetylhydrolase 1b, catalytic subunit 3





(29 kDa)


981
PALM
5064
paralemmin
0.79
0.013
0.55
0.031


982
PALMD
54873
palmdelphin
0.81
0.045
0.74
0.041


983
PANX1
24145
pannexin 1
0.78
0.011
2.19
0.021


984
PARL
55486
presenilin associated, rhomboid-like
0.78
0.014
2.17
0.016


985
PARPBP
55010
PARP1 binding protein
0.79
0.022
1.45
0.017


986
PAX7
5081
paired box 7
0.77
0.014
1.60
0.004


987
PBK
55872
PDZ binding kinase
0.78
0.012
1.55
0.001


988
PCDHB17
54661
protocadherin beta 17 pseudogene
0.80
0.027
1.32
0.027


989
PCNXL2
80003
pecanex-like 2 (Drosophila)
0.81
0.049
0.53
0.006


990
PCSK1N
27344
proprotein convertase subtilisin/kexin
0.80
0.026
1.22
0.032





type 1 inhibitor


991
PCSK5
5125
proprotein convertase subtilisin/kexin
0.81
0.038
0.66
0.037





type 5


992
PCTP
58488
phosphatidylcholine transfer protein
0.77
0.011
1.17
0.046


993
PDAP1
11333
PDGFA associated protein 1
0.82
0.040
1.77
0.036


994
PDCD10
11235
programmed cell death 10
0.81
0.033
1.80
0.029


995
PDCD5
9141
programmed cell death 5
0.78
0.010
2.16
0.001


996
PDCL
5082
phosducin-like
0.77
0.014
2.29
0.008


997
PDE2A
5138
phosphodiesterase 2A, cGMP-
0.79
0.025
0.77
0.012





stimulated


998
PDE6D
5147
phosphodiesterase 6D, cGMP-specific,
0.78
0.013
1.38
0.039





rod, delta


999
PDGFD
80310
platelet derived growth factor D
0.81
0.031
0.60
0.003


1000
PDHA1
5160
pyruvate dehydrogenase (lipoamide)
0.77
0.009
3.34
0.001





alpha 1


1001
PDHX
8050
pyruvate dehydrogenase complex,
0.80
0.025
1.60
0.032





component X


1002
PDLIM1
9124
PDZ and LIM domain 1
0.75
0.010
0.69
0.005


1003
PDZK1
5174
PDZ domain containing 1
0.80
0.027
0.86
0.018


1004
PEX13
5194
peroxisomal biogenesis factor 13
0.77
0.015
1.33
0.039


1005
PEX5L
51555
peroxisomal biogenesis factor 5-like
0.80
0.028
0.80
0.020


1006
PFDN1
5201
prefoldin subunit 1
0.76
0.013
2.20
0.007


1007
PFDN2
5202
prefoldin subunit 2
0.76
0.008
1.94
0.002


1008
PFDN4
5203
prefoldin subunit 4
0.80
0.029
1.48
0.025


1009
PGAM1
5223
phosphoglycerate mutase 1 (brain)
0.80
0.032
1.74
0.048


1010
PGK1
5230
phosphoglycerate kinase 1
0.79
0.020
2.35
0.001


1011
PGM3
5238
phosphoglucomutase 3
0.79
0.016
1.50
0.031


1012
PHB
5245
prohibitin
0.78
0.015
1.53
0.016


1013
PHF20L1
51105
PHD finger protein 20-like 1
0.81
0.034
1.84
0.011


1014
PHKA2
5256
phosphorylase kinase, alpha 2 (liver)
0.80
0.022
0.48
0.021


1015
PHKG2
5261
phosphorylase kinase, gamma 2 (testis)
0.78
0.009
0.56
0.009


1016
PHTF2
57157
putative homeodomain transcription
0.79
0.014
1.68
0.011





factor 2


1017
PI15
51050
peptidase inhibitor 15
0.79
0.015
0.76
0.006


1018
PIGL
9487
phosphatidylinositol glycan anchor
0.77
0.009
0.62
0.042





biosynthesis, class L


1019
PIK3CA
5290
phosphatidylinositol-4,5-bisphosphate
0.79
0.015
1.48
0.027





3-kinase, catalytic subunit alpha


1020
PIK3CB
5291
phosphatidylinositol-4,5-bisphosphate
0.79
0.019
1.80
0.009





3-kinase, catalytic subunit beta


1021
PIK3R4
30849
phosphoinositide-3-kinase, regulatory
0.78
0.015
1.90
0.019





subunit 4


1022
PIN1
5300
peptidylprolyl cis/trans isomerase,
0.75
0.006
1.26
0.010





NIMA-interacting 1


1023
PIN1P1
5301
peptidylprolyl cis/trans isomerase,
0.78
0.013
1.31
0.013





NIMA-interacting 1 pseudogene 1


1024
PIN4
5303
protein (peptidylprolyl cis/trans
0.79
0.025
2.08
0.012





isomerase) NIMA-interacting, 4





(parvulin)


1025
PIP
5304
prolactin-induced protein
0.80
0.035
0.87
0.003


1026
PIP4K2C
79837
phosphatidylinositol-5-phosphate 4-
0.79
0.023
1.79
0.011





kinase, type II, gamma


1027
PIPOX
51268
pipecolic acid oxidase
0.81
0.036
1.45
0.030


1028
PIR
8544
pirin (iron-binding nuclear protein)
0.78
0.012
1.47
0.004


1029
PITRM1
10531
pitrilysin metallopeptidase 1
0.80
0.019
1.93
0.030


1030
PITX1
5307
paired-like homeodomain 1
0.80
0.031
1.15
0.046


1031
PKP4
8502
plakophilin 4
0.80
0.028
0.54
0.020


1032
PLA2G12A
81579
phospholipase A2, group XIIA
0.81
0.049
2.68
0.000


1033
PLA2G7
7941
phospholipase A2, group VII (platelet-
0.76
0.004
1.39
0.012





activating factor acetylhydrolase,





plasma)


1034
PLAA
9373
phospholipase A2-activating protein
0.77
0.012
1.61
0.028


1035
PLAT
5327
plasminogen activator, tissue
0.82
0.045
0.81
0.050


1036
PLAUR
5329
plasminogen activator, urokinase
0.79
0.022
1.53
0.031





receptor


1037
PLEKHF2
79666
pleckstrin homology domain
0.80
0.035
1.57
0.003





containing, family F (with FYVE





domain) member 2


1038
PLEKHG3
26030
pleckstrin homology domain
0.82
0.036
0.60
0.018





containing, family G (with RhoGef





domain) member 3


1039
PLIN2
123
perilipin 2
0.75
0.005
1.43
0.006


1040
PLK1
5347
polo-like kinase 1
0.81
0.037
1.53
0.020


1041
PLK4
10733
polo-like kinase 4
0.78
0.021
1.34
0.048


1042
PLLP
51090
plasmolipin
0.79
0.021
0.60
0.041


1043
PLSCR3
57048
phospholipid scramblase 3
0.77
0.012
0.55
0.009


1044
PLSCR4
57088
phospholipid scramblase 4
0.82
0.047
0.52
0.000


1045
PMEL
6490
premelanosome protein
0.77
0.010
0.71
0.008


1046
PMP22
5376
peripheral myelin protein 22
0.82
0.043
0.71
0.011


1047
PMS2P8
729299
postmeiotic segregation increased 2
0.77
0.011
0.62
0.017





pseudogene 8


1048
PNISR
25957
PNN-interacting serine/arginine-rich
0.80
0.036
0.69
0.021





protein


1049
PNLIPRP1
5407
pancreatic lipase-related protein 1
0.77
0.013
0.74
0.022


1050
PNMA2
10687
paraneoplastic Ma antigen 2
0.78
0.014
0.67
0.010


1051
PODNL1
79883
podocan-like 1
0.77
0.009
0.74
0.002


1052
PODXL2
50512
podocalyxin-like 2
0.77
0.007
1.38
0.000


1053
POLA1
5422
polymerase (DNA directed), alpha 1,
0.79
0.017
1.91
0.018





catalytic subunit


1054
POLA2
23649
polymerase (DNA directed), alpha 2,
0.74
0.006
1.41
0.010





accessory subunit


1055
POLD4
57804
polymerase (DNA-directed), delta 4,
0.76
0.008
1.37
0.002





accessory subunit


1056
POLR2A
5430
polymerase (RNA) II (DNA directed)
0.80
0.028
0.55
0.009





polypeptide A, 220 kDa


1057
POLR2B
5431
polymerase (RNA) II (DNA directed)
0.79
0.021
2.50
0.011





polypeptide B, 140 kDa


1058
POLR2J4
84820
polymerase (RNA) II (DNA directed)
0.80
0.019
0.61
0.038





polypeptide J4, pseudogene


1059
POLR2K
5440
polymerase (RNA) II (DNA directed)
0.78
0.017
1.85
0.000





polypeptide K, 7.0 kDa


1060
POM121L9P
29774
POM121 transmembrane nucleoporin-
0.81
0.034
0.79
0.047





like 9, pseudogene


1061
POMP
51371
proteasome maturation protein
0.81
0.033
2.13
0.022


1062
PON2
5445
paraoxonase 2
0.77
0.017
0.66
0.045


1063
POP1
10940
processing of precursor 1, ribonuclease
0.81
0.036
1.54
0.012





P/MRP subunit (S. cerevisiae)


1064
POP7
10248
processing of precursor 7, ribonuclease
0.81
0.030
2.40
0.000





P/MRP subunit (S. cerevisiae)


1065
POU2F3
25833
POU class 2 homeobox 3
0.78
0.014
0.79
0.010


1066
PP14571
100130449
uncharacterized LOC100130449
0.81
0.045
1.18
0.033


1067
PPA2
27068
pyrophosphatase (inorganic) 2
0.78
0.017
1.58
0.046


1068
PPEF1
5475
protein phosphatase, EF-hand calcium
0.82
0.050
1.55
0.035





binding domain 1


1069
PPIEL
728448
peptidylprolyl isomerase E-like
0.81
0.030
0.71
0.011





pseudogene


1070
PPL
5493
periplakin
0.76
0.006
0.64
0.001


1071
PPP1CA
5499
protein phosphatase 1, catalytic
0.76
0.010
1.45
0.008





subunit, alpha isozyme


1072
PPP1R12B
4660
protein phosphatase 1, regulatory
0.78
0.015
0.42
0.004





subunit 12B


1073
PPP1R14B
26472
protein phosphatase 1, regulatory
0.79
0.022
1.73
0.009





(inhibitor) subunit 14B


1074
PPP1R2
5504
protein phosphatase 1, regulatory
0.79
0.021
1.60
0.044





(inhibitor) subunit 2


1075
PPP2R3C
55012
protein phosphatase 2, regulatory
0.78
0.014
1.91
0.033





subunit B″, gamma


1076
PPP3CA
5530
protein phosphatase 3, catalytic
0.82
0.039
1.52
0.020





subunit, alpha isozyme


1077
PPP3CB
5532
protein phosphatase 3, catalytic
0.80
0.024
1.69
0.048





subunit, beta isozyme


1078
PPRC1
23082
peroxisome proliferator-activated
0.81
0.031
1.95
0.020





receptor gamma, coactivator-related 1


1079
PQLC1
80148
PQ loop repeat containing 1
0.79
0.016
0.45
0.002


1080
PRAME
23532
preferentially expressed antigen in
0.82
0.049
1.21
0.032





melanoma


1081
PRC1
9055
protein regulator of cytokinesis 1
0.79
0.019
1.82
0.000


1082
PRDM10
56980
PR domain containing 10
0.79
0.011
0.42
0.020


1083
PRDX4
10549
peroxiredoxin 4
0.78
0.010
2.00
0.000


1084
PREP
5550
prolyl endopeptidase
0.79
0.016
2.04
0.005


1085
PRH1
5554
proline-rich protein HaeIII subfamily 1
0.76
0.011
1.79
0.003


1086
PRICKLE4
29964
prickle homolog 4 (Drosophila)
0.79
0.024
1.91
0.019


1087
PRIM1
5557
primase, DNA, polypeptide 1 (49 kDa)
0.80
0.035
2.44
0.000


1088
PRKCD
5580
protein kinase C, delta
0.79
0.024
1.38
0.022


1089
PRKCI
5584
protein kinase C, iota
0.79
0.014
2.45
0.003


1090
PRMT1
3276
protein arginine methyltransferase 1
0.80
0.029
1.64
0.044


1091
PRO2012
55478
uncharacterized protein PRO2012
0.77
0.011
0.86
0.045


1092
PROC
5624
protein C (inactivator of coagulation
0.79
0.019
0.73
0.042





factors Va and VIIIa)


1093
PROL1
58503
proline rich, lacrimal 1
0.81
0.032
0.74
0.010


1094
PRPF18
8559
pre-mRNA processing factor 18
0.82
0.037
2.09
0.017


1095
PRPF4
9128
pre-mRNA processing factor 4
0.74
0.008
1.81
0.025


1096
PRR14
78994
proline rich 14
0.80
0.021
0.51
0.029


1097
PSMA1
5682
proteasome (prosome, macropain)
0.77
0.009
2.33
0.005





subunit, alpha type, 1


1098
PSMA2
5683
proteasome (prosome, macropain)
0.78
0.010
1.94
0.010





subunit, alpha type, 2


1099
PSMA3
5684
proteasome (prosome, macropain)
0.75
0.005
1.74
0.010





subunit, alpha type, 3


1100
PSMA4
5685
proteasome (prosome, macropain)
0.78
0.007
2.27
0.009





subunit, alpha type, 4


1101
PSMA7
5688
proteasome (prosome, macropain)
0.79
0.020
1.69
0.006





subunit, alpha type, 7


1102
PSMB3
5691
proteasome (prosome, macropain)
0.80
0.033
1.70
0.003





subunit, beta type, 3


1103
PSMB5
5693
proteasome (prosome, macropain)
0.77
0.016
2.13
0.002





subunit, beta type, 5


1104
PSMB7
5695
proteasome (prosome, macropain)
0.78
0.010
1.93
0.011





subunit, beta type, 7


1105
PSMC3
5702
proteasome (prosome, macropain) 26S
0.82
0.032
1.56
0.022





subunit, ATPase, 3


1106
PSMC3IP
29893
PSMC3 interacting protein
0.78
0.018
0.69
0.039


1107
PSMC6
5706
proteasome (prosome, macropain) 26S
0.78
0.016
1.58
0.031





subunit, ATPase, 6


1108
PSMD10
5716
proteasome (prosome, macropain) 26S
0.77
0.007
2.27
0.001





subunit, non-ATPase, 10


1109
PSMD12
5718
proteasome (prosome, macropain) 26S
0.79
0.018
1.93
0.003





subunit, non-ATPase, 12


1110
PSMD14
10213
proteasome (prosome, macropain) 26S
0.78
0.012
2.24
0.005





subunit, non-ATPase, 14


1111
PSMD2
5708
proteasome (prosome, macropain) 26S
0.80
0.025
2.42
0.001





subunit, non-ATPase, 2


1112
PSMD3
5709
proteasome (prosome, macropain) 26S
0.78
0.015
1.34
0.012





subunit, non-ATPase, 3


1113
PSMD4
5710
proteasome (prosome, macropain) 26S
0.80
0.025
1.79
0.032





subunit, non-ATPase, 4


1114
PSMD6
9861
proteasome (prosome, macropain) 26S
0.80
0.019
2.09
0.013





subunit, non-ATPase, 6


1115
PSMD7
5713
proteasome (prosome, macropain) 26S
0.78
0.010
2.22
0.003





subunit, non-ATPase, 7


1116
PTBP3
9991
polypyrimidine tract binding protein 3
0.81
0.033
1.59
0.035


1117
PTCH1
5727
patched 1
0.81
0.035
0.58
0.013


1118
PTGER3
5733
prostaglandin E receptor 3 (subtype
0.81
0.026
0.73
0.020





EP3)


1119
PTK2
5747
protein tyrosine kinase 2
0.81
0.043
1.50
0.021


1120
PTN
5764
pleiotrophin
0.78
0.012
0.64
0.002


1121
PTPN18
26469
protein tyrosine phosphatase, non-
0.82
0.042
0.55
0.050





receptor type 18 (brain-derived)


1122
PTPRD
5789
protein tyrosine phosphatase, receptor
0.80
0.033
0.62
0.013





type, D


1123
PTPRF
5792
protein tyrosine phosphatase, receptor
0.78
0.013
0.62
0.040





type, F


1124
PTPRT
11122
protein tyrosine phosphatase, receptor
0.82
0.047
0.85
0.041





type, T


1125
PTPRZ1
5803
protein tyrosine phosphatase, receptor-
0.79
0.021
0.83
0.014





type, Z polypeptide 1


1126
PTRF
284119
polymerase I and transcript release
0.81
0.030
0.73
0.043





factor


1127
PTRH2
51651
peptidyl-tRNA hydrolase 2
0.77
0.015
1.66
0.001


1128
PTS
5805
6-pyruvoyltetrahydropterin synthase
0.80
0.026
1.89
0.022


1129
PTTG1
9232
pituitary tumor-transforming 1
0.80
0.017
2.08
0.000


1130
PTTG3P
26255
pituitary tumor-transforming 3,
0.76
0.012
1.50
0.002





pseudogene


1131
PURA
5813
purine-rich element binding protein A
0.81
0.045
0.59
0.041


1132
PURG
29942
purine-rich element binding protein G
0.79
0.023
0.80
0.013


1133
PWP1
11137
PWP1 homolog (S. cerevisiae)
0.79
0.020
2.10
0.022


1134
PYCRL
65263
pyrroline-5-carboxylate reductase-like
0.80
0.031
1.19
0.032


1135
QPCT
25797
glutaminyl-peptide cyclotransferase
0.82
0.029
1.36
0.002


1136
RAB11A
8766
RAB11A, member RAS oncogene
0.80
0.022
1.79
0.046





family


1137
RAB11FIP1
80223
RAB11 family interacting protein 1
0.81
0.042
1.27
0.018





(class I)


1138
RAB26
25837
RAB26, member RAS oncogene
0.77
0.009
0.65
0.043





family


1139
RABIF
5877
RAB interacting factor
0.79
0.030
2.38
0.002


1140
RACGAP1
29127
Rac GTPase activating protein 1
0.77
0.013
1.75
0.000


1141
RAD23A
5886
RAD23 homolog A (S. cerevisiae)
0.79
0.014
1.96
0.002


1142
RAD23B
5887
RAD23 homolog B (S. cerevisiae)
0.76
0.009
1.84
0.016


1143
RAD51AP1
10635
RAD51 associated protein 1
0.80
0.015
1.46
0.007


1144
RAD51C
5889
RAD51 paralog C
0.77
0.015
1.42
0.047


1145
RAD54B
25788
RAD54 homolog B (S. cerevisiae)
0.78
0.021
1.63
0.001


1146
RAG2
5897
recombination activating gene 2
0.81
0.034
0.88
0.027


1147
RAI2
10742
retinoic acid induced 2
0.82
0.043
0.82
0.037


1148
RALA
5898
v-ral simian leukemia viral oncogene
0.77
0.007
2.50
0.000





homolog A (ras related)


1149
RAMP3
10268
receptor (G protein-coupled) activity
0.82
0.044
0.72
0.049





modifying protein 3


1150
RANBP1
5902
RAN binding protein 1
0.79
0.021
1.53
0.033


1151
RANBP9
10048
RAN binding protein 9
0.82
0.043
1.59
0.033


1152
RAP1GDS1
5910
RAP1, GTP-GDP dissociation
0.78
0.018
1.57
0.009





stimulator 1


1153
RAP2A
5911
RAP2A, member of RAS oncogene
0.78
0.011
1.65
0.020





family


1154
RASL12
51285
RAS-like, family 12
0.80
0.032
0.58
0.003


1155
RASSF9
9182
Ras association (RalGDS/AF-6)
0.81
0.040
0.78
0.017





domain family (N-terminal) member 9


1156
RBM3
5935
RNA binding motif (RNP1, RRM)
0.78
0.016
0.48
0.012





protein 3


1157
RBM5
10181
RNA binding motif protein 5
0.80
0.029
0.47
0.001


1158
RBPMS
11030
RNA binding protein with multiple
0.77
0.008
0.62
0.002





splicing


1159
RBX1
9978
ring-box 1, E3 ubiquitin protein ligase
0.76
0.012
1.57
0.018


1160
RC3H2
54542
ring finger and CCCH-type domains 2
0.76
0.003
2.46
0.000


1161
RECQL4
9401
RecQ protein-like 4
0.81
0.040
1.28
0.003


1162
RELN
5649
reelin
0.78
0.017
0.72
0.003


1163
REPS1
85021
RALBP1 associated Eps domain
0.82
0.036
0.79
0.020





containing 1


1164
RERE
473
arginine-glutamic acid dipeptide (RE)
0.78
0.016
0.47
0.008





repeats


1165
RFC2
5982
replication factor C (activator 1) 2,
0.78
0.012
1.74
0.014





40 kDa


1166
RFC4
5984
replication factor C (activator 1) 4,
0.78
0.014
1.83
0.019





37 kDa


1167
RFC5
5985
replication factor C (activator 1) 5,
0.80
0.027
1.91
0.012





36.5 kDa


1168
RFNG
5986
RFNG O-fucosylpeptide 3-beta-N-
0.78
0.014
0.47
0.008





acetylglucosaminyltransferase


1169
RGL1
23179
ral guanine nucleotide dissociation
0.79
0.032
0.56
0.010





stimulator-like 1


1170
RGPD3
84220
RANBP2-like and GRIP domain
0.80
0.029
0.54
0.008





containing 3


1171
RGS5
8490
regulator of G-protein signaling 5
0.81
0.046
0.75
0.021


1172
RHBDD3
25807
rhomboid domain containing 3
0.82
0.043
1.75
0.045


1173
RHBDF1
64285
rhomboid 5 homolog 1 (Drosophila)
0.77
0.005
0.59
0.010


1174
RHOA
387
ras homolog family member A
0.81
0.042
0.48
0.003


1175
RIC3
79608
RIC3 acetylcholine receptor chaperone
0.78
0.014
0.75
0.006


1176
RIOK3
8780
RIO kinase 3
0.79
0.019
0.67
0.029


1177
RIPK2
8767
receptor-interacting serine-threonine
0.80
0.027
1.78
0.022





kinase 2


1178
RIT1
6016
Ras-like without CAAX 1
0.75
0.006
1.91
0.005


1179
RITA1
84934
RBPJ interacting and tubulin
0.76
0.015
1.47
0.037





associated 1


1180
RLN2
6019
relaxin 2
0.77
0.008
0.60
0.000


1181
RMDN1
51115
regulator of microtubule dynamics 1
0.82
0.050
1.61
0.006


1182
RMI1
80010
RecQ mediated genome instability 1
0.81
0.030
1.70
0.009


1183
RNASEH2A
10535
ribonuclease H2, subunit A
0.79
0.022
1.69
0.012


1184
RNASEL
6041
ribonuclease L (2′,5′-oligoisoadenylate
0.78
0.019
0.70
0.015





synthetase-dependent)


1185
RNF13
11342
ring finger protein 13
0.75
0.006
2.76
0.001


1186
RNF139
11236
ring finger protein 139
0.77
0.016
2.62
0.000


1187
RNF40
9810
ring finger protein 40, E3 ubiquitin
0.79
0.015
0.52
0.014





protein ligase


1188
RNF41
10193
ring finger protein 41, E3 ubiquitin
0.78
0.015
1.24
0.041





protein ligase


1189
RNF6
6049
ring finger protein (C3H2C3 type) 6
0.80
0.020
1.49
0.050


1190
RNF7
9616
ring finger protein 7
0.78
0.025
2.00
0.014


1191
RNU86
6122
RNA, U86 small nucleolar
0.79
0.020
0.55
0.014


1192
ROBO1
6091
roundabout, axon guidance receptor,
0.79
0.014
1.28
0.048





homolog 1 (Drosophila)


1193
ROPN1B
152015
rhophilin associated tail protein 1B
0.80
0.026
0.80
0.035


1194
RORC
6097
RAR-related orphan receptor C
0.80
0.020
0.67
0.033


1195
RPA2
6118
replication protein A2, 32 kDa
0.76
0.013
0.47
0.009


1196
RPAP3
79657
RNA polymerase II associated protein
0.78
0.015
1.68
0.016





3


1197
SNORA70
26778
small nucleolar RNA, H/ACA box 70
0.80
0.026
0.49
0.006


1198
RPL12
6136
ribosomal protein L12
0.81
0.043
0.58
0.018


1199
SNORD68
606500
small nucleolar RNA, C/D box 68
0.80
0.034
0.61
0.023


1200
RPL13AP5
26816
ribosomal protein L13a pseudogene 5
0.77
0.014
0.41
0.001


1201
RPL13A
23521
ribosomal protein L13a
0.81
0.046
0.62
0.008


1202
RPL17
6139
ribosomal protein L17
0.81
0.036
0.76
0.010


1203
RPL22
6146
ribosomal protein L22
0.78
0.018
0.59
0.033


1204
RPL23A
6147
ribosomal protein L23a
0.76
0.007
0.52
0.006


1205
SNORD42A
26809
small nucleolar RNA, C/D box 42A
0.75
0.006
0.36
0.001


1206
RPL27
6155
ribosomal protein L27
0.70
0.001
0.27
0.000


1207
SNORA45A
619562
small nucleolar RNA, H/ACA box 45A
0.78
0.016
0.36
0.001


1208
RPL29
6159
ribosomal protein L29
0.79
0.020
0.48
0.005


1209
RPL3
6122
ribosomal protein L3
0.75
0.008
0.42
0.000


1210
RPL31
6160
ribosomal protein L31
0.78
0.021
0.39
0.000


1211
RPL32
6161
ribosomal protein L32
0.80
0.027
0.40
0.002


1212
RPL34
6164
ribosomal protein L34
0.82
0.043
0.56
0.003


1213
RPL36
25873
ribosomal protein L36
0.82
0.047
0.59
0.010


1214
RPL37A
6168
ribosomal protein L37a
0.72
0.003
0.39
0.001


1215
RPL38
6169
ribosomal protein L38
0.77
0.008
0.45
0.006


1216
RPL39
6170
ribosomal protein L39
0.74
0.005
0.26
0.000


1217
RPL41
6171
ribosomal protein L41
0.72
0.003
0.29
0.000


1218
SNORD21
6125
small nucleolar RNA, C/D box 21
0.78
0.018
0.53
0.004


1219
RPL6
6128
ribosomal protein L6
0.76
0.009
0.42
0.004


1220
RPL7
6129
ribosomal protein L7
0.78
0.012
0.55
0.013


1221
SNORD36B
26820
small nucleolar RNA, C/D box 36B
0.80
0.029
0.49
0.005


1222
RPL9
6133
ribosomal protein L9
0.75
0.006
0.46
0.004


1223
RPLP0
6175
ribosomal protein, large, P0
0.78
0.014
0.55
0.028


1224
RPLP1
6176
ribosomal protein, large, P1
0.76
0.010
0.32
0.000


1225
SNORA52
619565
small nucleolar RNA, H/ACA box 52
0.81
0.034
0.42
0.000


1226
RPP21
56658
ribonuclease P/MRP 21 kDa subunit
0.78
0.017
2.31
0.004


1227
RPP40
10799
ribonuclease P/MRP 40 kDa subunit
0.79
0.022
2.20
0.012


1228
RPS10
6204
ribosomal protein S10
0.79
0.020
0.56
0.012


1229
RPS11
6205
ribosomal protein S11
0.81
0.040
0.34
0.001


1230
RPS14
6208
ribosomal protein S14
0.77
0.016
0.29
0.000


1231
RPS15
6209
ribosomal protein S15
0.80
0.028
0.39
0.001


1232
RPS16
6217
ribosomal protein S16
0.81
0.038
0.45
0.004


1233
RPS18
6222
ribosomal protein S18
0.75
0.007
0.40
0.001


1234
RPS2
6187
ribosomal protein S2
0.76
0.007
0.34
0.000


1235
RPS20
6224
ribosomal protein S20
0.77
0.012
0.56
0.019


1236
RPS21
6227
ribosomal protein S21
0.79
0.019
0.54
0.014


1237
RPS23
6228
ribosomal protein S23
0.72
0.002
0.24
0.000


1238
RPS24
6229
ribosomal protein S24
0.79
0.022
0.56
0.037


1239
RPS25
6230
ribosomal protein S25
0.80
0.042
0.30
0.000


1240
RPS27
6232
ribosomal protein S27
0.78
0.014
0.34
0.000


1241
RPS27A
6233
ribosomal protein S27a
0.80
0.035
0.47
0.000


1242
RPS28
6234
ribosomal protein S28
0.74
0.004
0.51
0.001


1243
RPS29
6235
ribosomal protein S29
0.78
0.023
0.31
0.000


1244
RPS2P45
80052
ribosomal protein S2 pseudogene 45
0.78
0.009
0.83
0.008


1245
RPS3
6188
ribosomal protein S3
0.79
0.017
0.61
0.006


1246
RPS3A
6189
ribosomal protein S3A
0.74
0.006
0.38
0.000


1247
RPS4X
6191
ribosomal protein S4, X-linked
0.75
0.007
0.42
0.001


1248
RPS6
6194
ribosomal protein S6
0.75
0.009
0.43
0.000


1249
RPS6KA2
6196
ribosomal protein S6 kinase, 90 kDa,
0.79
0.030
0.59
0.044





polypeptide 2


1250
RPS6KB1
6198
ribosomal protein S6 kinase, 70 kDa,
0.81
0.029
1.45
0.048





polypeptide 1


1251
RPS6KC1
26750
ribosomal protein S6 kinase, 52 kDa,
0.78
0.008
3.11
0.000





polypeptide 1


1252
RPS
6202
ribosomal protein S8
0.74
0.006
0.58
0.004


1253
RRM2
6241
ribonucleotide reductase M2
0.77
0.010
1.50
0.004


1254
RRN3P1
730092
RNA polymerase I transcription factor
0.79
0.014
0.60
0.005





homolog (S. cerevisiae) pseudogene 1


1255
RRP15
51018
ribosomal RNA processing 15
0.76
0.010
1.61
0.009





homolog (S. cerevisiae)


1256
RRP7A
27341
ribosomal RNA processing 7 homolog
0.77
0.015
1.43
0.034





A (S. cerevisiae)


1257
RRP7B
91695
ribosomal RNA processing 7 homolog
0.76
0.008
0.53
0.002





B (S. cerevisiae)


1258
RSAD2
91543
radical S-adenosyl methionine domain
0.74
0.003
1.40
0.007





containing 2


1259
RSF1
51773
remodeling and spacing factor 1
0.81
0.035
1.35
0.032


1260
RTCA
8634
RNA 3′-terminal phosphate cyclase
0.77
0.010
1.50
0.036


1261
RUFY1
80230
RUN and FYVE domain containing 1
0.78
0.016
0.40
0.028


1262
RUFY2
55680
RUN and FYVE domain containing 2
0.80
0.024
0.73
0.005


1263
RUNX1T1
862
runt-related transcription factor 1;
0.77
0.014
0.61
0.019





translocated to, 1 (cyclin D-related)


1264
S100P
6286
S100 calcium binding protein P
0.81
0.028
1.24
0.000


1265
SAFB2
9667
scaffold attachment factor B2
0.80
0.031
0.55
0.034


1266
SALL2
6297
spalt-like transcription factor 2
0.79
0.017
0.63
0.029


1267
SAMHD1
25939
SAM domain and HD domain 1
0.74
0.003
1.46
0.022


1268
SAR1B
51128
secretion associated, Ras related
0.78
0.011
1.46
0.024





GTPase 1B


1269
SCFD1
23256
sec1 family domain containing 1
0.75
0.004
3.31
0.000


1270
SCN4A
6329
sodium channel, voltage-gated, type
0.81
0.029
1.34
0.036





IV, alpha subunit


1271
SCUBE2
57758
signal peptide, CUB domain, EGF-like
0.82
0.033
0.87
0.043





2


1272
SDF2L1
23753
stromal cell-derived factor 2-like 1
0.77
0.013
1.49
0.033


1273
SDS
10993
serine dehydratase
0.81
0.038
1.59
0.026


1274
SEC24D
9871
SEC24 family member D
0.75
0.007
1.95
0.005


1275
SEC31B
25956
SEC31 homolog B (S. cerevisiae)
0.78
0.013
0.72
0.027


1276
SEC61G
23480
Sec61 gamma subunit
0.78
0.019
1.94
0.009


1277
SEC62
7095
SEC62 homolog (S. cerevisiae)
0.77
0.008
2.22
0.001


1278
SELP
6403
selectin P (granule membrane protein
0.75
0.009
0.42
0.000





140 kDa, antigen CD62)


1279
SEMA3G
56920
sema domain, immunoglobulin domain
0.77
0.010
0.41
0.000





(Ig), short basic domain, secreted,





(semaphorin) 3G


1280
SEMA6D
80031
sema domain, transmembrane domain
0.76
0.006
0.63
0.001





(TM), and cytoplasmic domain,





(semaphorin) 6D


1281
SERPINB8
5271
serpin peptidase inhibitor, clade B
0.80
0.022
1.51
0.023





(ovalbumin), member 8


1282
SESN1
27244
sestrin 1
0.81
0.035
0.38
0.000


1283
SF3B1
23451
splicing factor 3b, subunit 1, 155 kDa
0.79
0.026
0.50
0.023


1284
SFI1
9814
Sfi1 homolog, spindle assembly
0.80
0.018
0.57
0.014





associated (yeast)


1285
SFRP1
6422
secreted frizzled-related protein 1
0.81
0.041
0.64
0.000


1286
SGCB
6443
sarcoglycan, beta (43 kDa dystrophin-
0.78
0.012
1.77
0.006





associated glycoprotein)


1287
SGK2
10110
serum/glucocorticoid regulated kinase
0.73
0.003
0.55
0.003





2


1288
SGSM2
9905
small G protein signaling modulator 2
0.76
0.006
0.54
0.007


1289
SH2B1
25970
SH2B adaptor protein 1
0.72
0.002
0.50
0.000


1290
SH2D3A
10045
SH2 domain containing 3A
0.75
0.007
0.58
0.029


1291
SHC2
25759
SHC (Src homology 2 domain
0.78
0.012
0.61
0.012





containing) transforming protein 2


1292
SHH
6469
sonic hedgehog
0.82
0.048
0.76
0.013


1293
SHMT2
6472
serine hydroxymethyltransferase 2
0.81
0.038
1.63
0.038





(mitochondrial)


1294
SIK3
23387
SIK family kinase 3
0.82
0.049
0.53
0.004


1295
SIN3B
23309
SIN3 transcription regulator family
0.78
0.016
0.48
0.003





member B


1296
SIX1
6495
SIX homeobox 1
0.80
0.028
1.26
0.008


1297
SKA1
220134
spindle and kinetochore associated
0.79
0.025
1.35
0.012





complex subunit 1


1298
SKP1
6500
S-phase kinase-associated protein 1
0.76
0.007
0.46
0.014


1299
SLBP
7884
stem-loop binding protein
0.80
0.036
2.56
0.001


1300
SLC10A3
8273
solute carrier family 10, member 3
0.77
0.012
1.19
0.044


1301
SLC13A1
6561
solute carrier family 13 (sodium/sulfate
0.81
0.038
0.82
0.019





symporter), member 1


1302
SLC19A3
80704
solute carrier family 19 (thiamine
0.79
0.025
0.77
0.015





transporter), member 3


1303
SLC1A5
6510
solute carrier family 1 (neutral amino
0.80
0.024
1.48
0.030





acid transporter), member 5


1304
SLC22A18
5002
solute carrier family 22, member 18
0.80
0.024
0.66
0.012


1305
SLC22A18AS
5003
solute carrier family 22 (organic cation
0.81
0.040
0.71
0.020





transporter), member 18 antisense


1306
SLC22A5
6584
solute carrier family 22 (organic
0.75
0.007
0.51
0.021





cation/camitine transporter), member 5


1307
SLC25A1
6576
solute carrier family 25 (mitochondrial
0.81
0.033
1.57
0.028





carrier; citrate transporter), member 1


1308
SLC25A12
8604
solute carrier family 25
0.81
0.043
0.48
0.011





(aspartate/glutamate carrier), member





12


1309
SLC25A23
79085
solute carrier family 25 (mitochondrial
0.78
0.021
1.54
0.016





carrier; phosphate carrier), member 23


1310
SLC25A32
81034
solute carrier family 25 (mitochondrial
0.81
0.033
1.81
0.004





folate carrier), member 32


1311
SLC25A37
51312
solute carrier family 25 (mitochondrial
0.79
0.016
0.47
0.000





iron transporter), member 37


1312
SLC26A2
1836
solute carrier family 26 (anion
0.80
0.024
1.36
0.045





exchanger), member 2


1313
SLC2A10
81031
solute carrier family 2 (facilitated
0.81
0.036
1.37
0.023





glucose transporter), member 10


1314
SLC2A6
11182
solute carrier family 2 (facilitated
0.77
0.010
1.24
0.035





glucose transporter), member 6


1315
SLC30A9
10463
solute carrier family 30 (zinc
0.78
0.023
2.24
0.005





transporter), member 9


1316
SLC33A1
9197
solute carrier family 33 (acetyl-CoA
0.79
0.020
1.97
0.008





transporter), member 1


1317
SLC35A5
55032
solute carrier family 35, member A5
0.78
0.016
1.55
0.036


1318
SLC35B1
10237
solute carrier family 35, member B1
0.80
0.032
1.85
0.003


1319
SLC35E1
79939
solute carrier family 35, member E1
0.77
0.010
0.78
0.041


1320
SLC35E2
9906
solute carrier family 35, member E2
0.79
0.019
0.53
0.004


1321
SLC35E3
55508
solute carrier family 35, member E3
0.81
0.042
1.37
0.026


1322
SLC35F2
54733
solute carrier family 35, member F2
0.78
0.016
0.60
0.021


1323
SLC37A4
2542
solute carrier family 37 (glucose-6-
0.81
0.035
0.49
0.011





phosphate transporter), member 4


1324
SLC44A1
23446
solute carrier family 44 (choline
0.81
0.034
0.83
0.035





transporter), member 1


1325
SLC4A1AP
22950
solute carrier family 4 (anion
0.80
0.026
2.41
0.034





exchanger), member 1, adaptor protein


1326
SLC6A10P
6535
solute carrier family 6
0.79
0.021
1.25
0.039





(neurotransmitter transporter), member





10, pseudogene


1327
SLC6A5
9152
solute carrier family 6
0.76
0.011
0.78
0.017





(neurotransmitter transporter), member





5


1328
SLC7A10
56301
solute carrier family 7 (neutral amino
0.78
0.018
0.79
0.047





acid transporter light chain, asc





system), member 10


1329
SLC7A2
6542
solute carrier family 7 (cationic amino
0.81
0.030
0.85
0.040





acid transporter, y+ system), member 2


1330
SLC7A5
8140
solute carrier family 7 (amino acid
0.81
0.028
1.30
0.015





transporter light chain, L system),





member 5


1331
SLC8B1
80024
solute carrier family 8
0.80
0.026
0.59
0.001





(sodium/lithium/calcium exchanger),





member B1


1332
SLC9A3
6550
solute carrier family 9, subfamily A
0.77
0.019
1.77
0.002





(NHE3, cation proton antiporter 3),





member 3


1333
SLCO2A1
6578
solute carrier organic anion transporter
0.80
0.031
0.57
0.036





family, member 2A1


1334
SMARCA2
6595
SWI/SNF related, matrix associated,
0.79
0.028
0.45
0.002





actin dependent regulator of chromatin,





subfamily a, member 2


1335
SMARCE1
6605
SWI/SNF related, matrix associated,
0.81
0.046
1.59
0.032





actin dependent regulator of chromatin,





subfamily e, member 1


1336
SMC2
10592
structural maintenance of
0.78
0.011
1.95
0.004





chromosomes 2


1337
SMC4
10051
structural maintenance of
0.80
0.019
1.67
0.007





chromosomes 4


1338
SMC5
23137
structural maintenance of
0.83
0.046
1.77
0.015





chromosomes 5


1339
SMG7-AS1
284649
SMG7 antisense RNA 1
0.77
0.018
1.48
0.009


1340
SMG8
55181
SMG8 nonsense mediated mRNA
0.81
0.037
1.66
0.041





decay factor


1341
SMS
6611
spermine synthase
0.78
0.019
2.13
0.000


1342
SNAI1
6615
snail family zinc finger 1
0.81
0.036
1.34
0.015


1343
SNAP29
9342
synaptosomal-associated protein,
0.80
0.028
1.66
0.020





29 kDa


1344
SNAPC5
10302
small nuclear RNA activating complex,
0.75
0.007
2.12
0.011





polypeptide 5, 19 kDa


1345
TCP1
677812
t-complex 1
0.78
0.014
1.93
0.002


1346
TBRG4
677795
transforming growth factor beta
0.75
0.003
1.37
0.003





regulator 4


1347
SNRNP70
6625
small nuclear ribonucleoprotein 70 kDa
0.76
0.007
0.57
0.005





(U1)


1348
SNRPB2
6629
small nuclear ribonucleoprotein
0.78
0.019
2.17
0.003





polypeptide B


1349
SNRPC
6631
small nuclear ribonucleoprotein
0.78
0.017
2.21
0.007





polypeptide C


1350
SNRPD3
6634
small nuclear ribonucleoprotein D3
0.79
0.024
1.45
0.027





polypeptide 18 kDa


1351
SNX16
64089
sorting nexin 16
0.80
0.029
1.36
0.042


1352
SNX29P2
440352
sorting nexin 29 pseudogene 2
0.79
0.018
0.80
0.000


1353
SON
6651
SON DNA binding protein
0.81
0.045
0.48
0.022


1354
SORBS1
10580
sorbin and SH3 domain containing 1
0.78
0.010
0.54
0.004


1355
SORBS3
10174
sorbin and SH3 domain containing 3
0.80
0.025
0.63
0.048


1356
SORL1
6653
sortilin-related receptor, L(DLR class)
0.80
0.032
0.56
0.005





A repeats containing


1357
SOSTDC1
25928
sclerostin domain containing 1
0.76
0.012
0.76
0.001


1358
SPAG5
10615
sperm associated antigen 5
0.80
0.028
1.34
0.031


1359
SPAG9
9043
sperm associated antigen 9
0.82
0.047
1.85
0.048


1360
SPANXB1
728695
SPANX family, member B1
0.79
0.023
1.15
0.046


1361
SPATA6
54558
spermatogenesis associated 6
0.82
0.037
0.55
0.019


1362
SPEF1
25876
sperm flagellar 1
0.78
0.012
0.61
0.021


1363
SPEN
23013
spen family transcriptional repressor
0.78
0.017
0.29
0.000


1364
SPG11
80208
spastic paraplegia 11 (autosomal
0.81
0.031
0.55
0.020





recessive)


1365
SPG20
23111
spastic paraplegia 20 (Troyer
0.80
0.037
0.57
0.012





syndrome)


1366
SPPL2B
56928
signal peptide peptidase like 2B
0.78
0.012
0.74
0.037


1367
SPSB3
90864
splA/ryanodine receptor domain and
0.74
0.005
0.55
0.012





SOCS box containing 3


1368
SPTA1
6708
spectrin, alpha, erythrocytic 1
0.77
0.010
0.75
0.017


1369
SPTSSA
171546
serine palmitoyltransferase, small
0.80
0.029
1.65
0.027





subunit A


1370
SRP14
6727
signal recognition particle 14 kDa
0.75
0.003
0.24
0.000





(homologous Alu RNA binding





protein)


1371
SRP19
6728
signal recognition particle 19 kDa
0.80
0.027
1.97
0.030


1372
SRP54
6729
signal recognition particle 54 kDa
0.79
0.018
1.70
0.029


1373
SRP72
6731
signal recognition particle 72 kDa
0.80
0.036
2.79
0.002


1374
SRPK2
6733
SRSF protein kinase 2
0.82
0.049
1.64
0.029


1375
SRPRB
58477
signal recognition particle receptor, B
0.82
0.046
1.75
0.040





subunit


1376
SRRD
402055
SRR1 domain containing
0.78
0.017
1.57
0.047


1377
SRRM1
10250
serine/arginine repetitive matrix 1
0.82
0.046
0.40
0.004


1378
SRRM2
23524
serine/arginine repetitive matrix 2
0.74
0.004
0.72
0.014


1379
SRSF11
9295
serine/arginine-rich splicing factor 11
0.78
0.017
0.64
0.020


1380
SRSF5
6430
serine/arginine-rich splicing factor 5
0.77
0.008
0.48
0.000


1381
SRSF8
10929
serine/arginine-rich splicing factor 8
0.77
0.011
0.37
0.001


1382
SSB
6741
Sjogren syndrome antigen B
0.79
0.019
2.10
0.009





(autoantigen La)


1383
SSBP1
6742
single-stranded DNA binding protein
0.77
0.013
1.78
0.041





1, mitochondrial


1384
SSNA1
8636
Sjogren syndrome nuclear autoantigen
0.82
0.050
1.86
0.016





1


1385
SSSCA1
10534
Sjogren syndrome/scleroderma
0.76
0.012
1.43
0.005





autoantigen 1


1386
STAG3L3
442578
stromal antigen 3-like 3
0.78
0.011
0.72
0.003


1387
STAMBP
10617
STAM binding protein
0.81
0.035
3.38
0.001


1388
STARD13
90627
StAR-related lipid transfer (START)
0.81
0.034
0.63
0.026





domain containing 13


1389
STARD3
10948
StAR-related lipid transfer (START)
0.77
0.013
1.30
0.035





domain containing 3


1390
STAT5B
6777
signal transducer and activator of
0.76
0.008
0.32
0.001





transcription 5B


1391
STAT6
6778
signal transducer and activator of
0.77
0.016
0.53
0.006





transcription 6, interleukin-4 induced


1392
STAU2
27067
staufen double-stranded RNA binding
0.82
0.048
1.43
0.039





protein 2


1393
STC2
8614
stanniocalcin 2
0.77
0.006
0.81
0.003


1394
STIP1
10963
stress-induced phosphoprotein 1
0.75
0.005
1.52
0.004


1395
STK25
10494
serine/threonine kinase 25
0.80
0.025
0.57
0.039


1396
STK3
6788
serine/threonine kinase 3
0.76
0.012
1.33
0.021


1397
STMN1
3925
stathmin 1
0.78
0.013
1.51
0.041


1398
STRAP
11171
serine/threonine kinase receptor
0.80
0.027
2.50
0.009





associated protein


1399
STXBP1
6812
syntaxin binding protein 1
0.80
0.027
0.66
0.007


1400
SUGCT
79783
succinyl-CoA:glutarate-CoA
0.79
0.022
1.37
0.005





transferase


1401
SULT1A2
6799
sulfotransferase family, cytosolic, 1A,
0.76
0.008
0.63
0.042





phenol-preferring, member 2


1402
SULT4A1
25830
sulfotransferase family 4A, member 1
0.79
0.020
0.80
0.045


1403
SUMO3
6612
small ubiquitin-like modifier 3
0.77
0.009
2.15
0.006


1404
SUPT4H1
6827
suppressor of Ty 4 homolog 1 (S. cerevisiae)
0.79
0.026
1.44
0.042


1405
SUV420H1
51111
suppressor of variegation 4-20
0.82
0.047
1.91
0.008





homolog 1 (Drosophila)


1406
SYNM
23336
synemin, intermediate filament protein
0.81
0.039
0.78
0.032


1407
SYNPO
11346
synaptopodin
0.80
0.030
0.65
0.003


1408
SYT17
51760
synaptotagmin XVII
0.82
0.042
0.85
0.035


1409
SZT2
23334
seizure threshold 2 homolog (mouse)
0.76
0.011
0.55
0.019


1410
TAC1
6863
tachykinin, precursor 1
0.81
0.030
0.84
0.026


1411
TACC3
10460
transforming, acidic coiled-coil
0.75
0.005
1.25
0.013





containing protein 3


1412
TAF1B
9014
TATA box binding protein (TBP)-
0.81
0.036
1.77
0.015





associated factor, RNA polymerase I,





B, 63 kDa


1413
TAF1C
9013
TATA box binding protein (TBP)-
0.79
0.026
0.70
0.000





associated factor, RNA polymerase I,





C, 110 kDa


1414
TAF2
6873
TAF2 RNA polymerase II, TATA box
0.80
0.027
1.70
0.007





binding protein (TBP)-associated





factor, 150 kDa


1415
TAGLN2
8407
transgelin 2
0.82
0.037
1.51
0.019


1416
TALDO1
6888
transaldolase 1
0.77
0.010
1.53
0.024


1417
TARS
6897
threonyl-tRNA synthetase
0.76
0.006
1.77
0.006


1418
TAT
6898
tyrosine aminotransferase
0.77
0.017
0.85
0.032


1419
TAZ
6901
tafazzin
0.78
0.012
0.60
0.042


1420
TBC1D30
23329
TBC1 domain family, member 30
0.81
0.047
1.93
0.011


1421
TBC1D31
93594
TBC1 domain family, member 31
0.77
0.009
1.64
0.006


1422
TBC1D9B
23061
TBC1 domain family, member 9B
0.78
0.014
0.47
0.038





(with GRAM domain)


1423
TBCC
6903
tubulin folding cofactor C
0.76
0.007
1.59
0.022


1424
TBCE
6905
tubulin folding cofactor E
0.82
0.036
1.89
0.005


1425
TBK1
29110
TANK-binding kinase 1
0.81
0.041
2.02
0.043


1426
TBL2
26608
transducin (beta)-like 2
0.80
0.026
1.61
0.049


1427
TBX5
6910
T-box 5
0.81
0.028
0.75
0.040


1428
TCEA1
6917
transcription elongation factor A (SII),
0.81
0.046
1.52
0.025





1


1429
TCEAL2
140597
transcription elongation factor A (SII)-
0.80
0.022
0.81
0.043





like 2


1430
TCEB1
6921
transcription elongation factor B (SIII),
0.80
0.030
1.80
0.000





polypeptide 1 (15 kDa, elongin C)


1431
TCF7
6932
transcription factor 7 (T-cell specific,
0.79
0.020
0.66
0.015





HMG-box)


1432
TCF7L1
83439
transcription factor 7-like 1 (T-cell
0.80
0.023
0.57
0.005





specific, HMG-box)


1433
TCN1
6947
transcobalamin I (vitamin B12 binding
0.80
0.026
0.84
0.037





protein, R binder family)


1434
TCTN1
79600
tectonic family member 1
0.82
0.028
0.59
0.014


1435
TDP2
51567
tyrosyl-DNA phosphodiesterase 2
0.76
0.007
1.61
0.035


1436
TELO2
9894
telomere maintenance 2
0.77
0.009
0.55
0.015


1437
TENC1
23371
tensin like C1 domain containing
0.72
0.002
0.44
0.000





phosphatase (tensin 2)


1438
TERF1
7013
telomeric repeat binding factor
0.82
0.038
1.59
0.030





(NIMA-interacting) 1


1439
TFB2M
64216
transcription factor B2, mitochondrial
0.79
0.016
1.83
0.009


1440
TFDP2
7029
transcription factor Dp-2 (E2F
0.78
0.019
1.51
0.034





dimerization partner 2)


1441
TFEC
22797
transcription factor EC
0.76
0.004
1.43
0.038


1442
TFIP11
24144
tuftelin interacting protein 11
0.76
0.012
1.67
0.042


1443
TFPT
29844
TCF3 (E2A) fusion partner (in
0.75
0.007
1.56
0.011





childhood Leukemia)


1444
TGFB3
7043
transforming growth factor, beta 3
0.80
0.024
0.59
0.011


1445
TGFBR3
7049
transforming growth factor, beta
0.81
0.032
0.63
0.000





receptor III


1446
TGFBRAP1
9392
transforming growth factor, beta
0.81
0.039
0.44
0.030





receptor associated protein 1


1447
TGOLN2
10618
trans-golgi network protein 2
0.78
0.018
0.40
0.007


1448
THNSL2
55258
threonine synthase-like 2 (S.
0.79
0.016
0.56
0.002






cerevisiae)



1449
THSD4
79875
thrombospondin, type I, domain
0.77
0.006
0.72
0.002





containing 4


1450
TIMM17A
10440
translocase of inner mitochondrial
0.77
0.008
2.17
0.001





membrane 17 homolog A (yeast)


1451
TIMM23
100287932
translocase of inner mitochondrial
0.76
0.008
1.78
0.018





membrane 23 homolog (yeast)


1452
TIMM44
10469
translocase of inner mitochondrial
0.78
0.014
1.82
0.026





membrane 44 homolog (yeast)


1453
TIMP2
7077
TIMP metallopeptidase inhibitor 2
0.72
0.007
1.33
0.044


1454
TIPIN
54962
TIMELESS interacting protein
0.79
0.015
1.32
0.010


1455
TIPRL
261726
TOR signaling pathway regulator
0.77
0.010
2.03
0.000


1456
TK1
7083
thymidine kinase 1, soluble
0.80
0.034
1.41
0.030


1457
TLE2
7089
transducin-like enhancer of split 2
0.78
0.012
0.67
0.034


1458
TLE6
79816
transducin-like enhancer of split 6
0.81
0.031
0.64
0.000


1459
TLN2
83660
talin 2
0.82
0.048
0.59
0.044


1460
TLR6
10333
toll-like receptor 6
0.81
0.029
0.79
0.035


1461
TM2D1
83941
TM2 domain containing 1
0.80
0.025
0.53
0.011


1462
TM9SF1
10548
transmembrane 9 superfamily member
0.76
0.012
2.13
0.007





1


1463
TM9SF3
56889
transmembrane 9 superfamily member
0.80
0.023
2.44
0.003





3


1464
TMEM100
55273
transmembrane protein 100
0.81
0.033
0.74
0.003


1465
TMEM106B
54664
transmembrane protein 106B
0.79
0.025
1.53
0.026


1466
TMEM132A
54972
transmembrane protein 132A
0.79
0.033
1.44
0.028


1467
TMEM14A
28978
transmembrane protein 14A
0.78
0.012
1.61
0.022


1468
TMEM165
55858
transmembrane protein 165
0.80
0.028
1.55
0.037


1469
TMEM183A
92703
transmembrane protein 183A
0.77
0.013
2.12
0.002


1470
TMEM184C
55751
transmembrane protein 184C
0.78
0.016
1.79
0.040


1471
TMEM187
8269
transmembrane protein 187
0.80
0.028
1.78
0.009


1472
TMEM194A
23306
transmembrane protein 194A
0.80
0.022
1.59
0.036


1473
TMEM259
91304
transmembrane protein 259
0.80
0.027
0.64
0.038


1474
TMEM33
55161
transmembrane protein 33
0.78
0.014
2.04
0.005


1475
TMEM70
54968
transmembrane protein 70
0.79
0.024
1.72
0.003


1476
TMEM97
27346
transmembrane protein 97
0.80
0.040
1.51
0.004


1477
TMPO
7112
thymopoietin
0.78
0.013
1.62
0.006


1478
TMPRSS3
64699
transmembrane protease, serine 3
0.80
0.036
0.82
0.020


1479
TMPRSS6
164656
transmembrane protease, serine 6
0.78
0.014
0.63
0.009


1480
TMUB2
79089
transmembrane and ubiquitin-like
0.80
0.019
0.52
0.015





domain containing 2


1481
TNFAIP2
7127
tumor necrosis factor, alpha-induced
0.78
0.017
0.70
0.034





protein 2


1482
TNFRSF25
8718
tumor necrosis factor receptor
0.78
0.016
0.66
0.044





superfamily, member 25


1483
TNIK
23043
TRAF2 and NCK interacting kinase
0.80
0.033
0.63
0.040


1484
TNS1
7145
tensin 1
0.77
0.015
0.57
0.001


1485
TNXA
7146
tenascin XA (pseudogene)
0.80
0.028
0.76
0.005


1486
TOMM70A
9868
translocase of outer mitochondrial
0.79
0.036
3.41
0.000





membrane 70 homolog A (S.






cerevisiae)



1487
TOP2A
7153
topoisomerase (DNA) II alpha 170 kDa
0.80
0.029
1.51
0.000


1488
TOR1B
27348
torsin family 1, member B (torsin B)
0.79
0.016
3.10
0.001


1489
TP53
7157
tumor protein p53
0.82
0.043
0.58
0.025


1490
TP63
8626
tumor protein p63
0.74
0.006
0.56
0.001


1491
TP73-AS1
57212
TP73 antisense RNA 1
0.79
0.026
0.39
0.003


1492
TPCN1
53373
two pore segment channel 1
0.76
0.009
0.64
0.024


1493
TPRKB
51002
TP53RK binding protein
0.78
0.011
1.77
0.027


1494
TRA2A
29896
transformer 2 alpha homolog
0.78
0.013
0.61
0.035





(Drosophila)


1495
TRAF2
7186
TNF receptor-associated factor 2
0.81
0.032
1.29
0.035


1496
TRAK1
22906
trafficking protein, kinesin binding 1
0.80
0.028
0.57
0.023


1497
TRAM1
23471
translocation associated membrane
0.80
0.035
1.61
0.045





protein 1


1498
TRAPPC2
6399
trafficking protein particle complex 2
0.77
0.012
1.67
0.005


1499
TRIAP1
51499
TP53 regulated inhibitor of apoptosis 1
0.81
0.040
2.38
0.019


1500
TRIM13
10206
tripartite motif containing 13
0.80
0.028
0.44
0.025


1501
TRIM29
23650
tripartite motif containing 29
0.75
0.012
0.70
0.002


1502
TRIP13
9319
thyroid hormone receptor interactor 13
0.80
0.026
1.79
0.003


1503
TRMT12
55039
tRNA methyltransferase 12 homolog
0.80
0.037
2.76
0.000





(S. cerevisiae)


1504
TROVE2
6738
TROVE domain family, member 2
0.80
0.016
2.24
0.001


1505
TRPM2
7226
transient receptor potential cation
0.81
0.042
1.81
0.014





channel, subfamily M, member 2


1506
TSC2
7249
tuberous sclerosis 2
0.79
0.024
0.52
0.000


1507
TSFM
10102
Ts translation elongation factor,
0.82
0.042
1.78
0.029





mitochondrial


1508
TSG101
7251
tumor susceptibility 101
0.78
0.015
1.84
0.021


1509
TSKU
25987
tsukushi, small leucine rich
0.78
0.027
1.24
0.040





proteoglycan


1510
TSPAN7
7102
tetraspanin 7
0.80
0.028
0.73
0.007


1511
TSPYL2
64061
TSPY-like 2
0.78
0.019
0.70
0.018


1512
TSR1
55720
TSR1, 20S rRNA accumulation,
0.78
0.015
0.61
0.013





homolog (S. cerevisiae)


1513
TSSK2
23617
testis-specific serine kinase 2
0.82
0.048
1.66
0.030


1514
TTC12
54970
tetratricopeptide repeat domain 12
0.81
0.041
0.45
0.001


1515
TTC19
54902
tetratricopeptide repeat domain 19
0.78
0.017
0.52
0.022


1516
TTC26
79989
tetratricopeptide repeat domain 26
0.80
0.022
1.47
0.008


1517
TTC28
23331
tetratricopeptide repeat domain 28
0.77
0.009
0.39
0.002


1518
TTI1
9675
TELO2 interacting protein 1
0.80
0.044
1.50
0.035


1519
TTK
7272
TTK protein kinase
0.77
0.006
1.59
0.000


1520
TTLL4
9654
tubulin tyrosine ligase-like family,
0.78
0.016
0.69
0.040





member 4


1521
TUBA4B
80086
tubulin, alpha 4b (pseudogene)
0.76
0.008
0.59
0.001


1522
TUBB
203068
tubulin, beta class I
0.78
0.018
1.75
0.047


1523
TUBB3
10381
tubulin, beta 3 class III
0.81
0.038
1.71
0.022


1524
TUBB4B
10383
tubulin, beta 4B class IVb
0.80
0.026
1.87
0.013


1525
TUBD1
51174
tubulin, delta 1
0.77
0.009
1.64
0.018


1526
TUBG2
27175
tubulin, gamma 2
0.79
0.012
0.59
0.007


1527
TXN
7295
thioredoxin
0.79
0.015
2.82
0.000


1528
TXNDC9
10190
thioredoxin domain containing 9
0.79
0.016
2.97
0.002


1529
TXNL4A
10907
thioredoxin-like 4A
0.79
0.019
1.67
0.033


1530
TXNRD1
7296
thioredoxin reductase 1
0.78
0.013
1.75
0.002


1531
UAP1
6675
UDP-N-acteylglucosamine
0.80
0.030
1.46
0.044





pyrophosphorylase 1


1532
UBA2
10054
ubiquitin-like modifier activating
0.78
0.012
1.93
0.018





enzyme 2


1533
UBB
7314
ubiquitin B
0.82
0.049
0.48
0.033


1534
UBC
7316
ubiquitin C
0.79
0.018
0.42
0.016


1535
UBE2A
7319
ubiquitin-conjugating enzyme E2A
0.82
0.050
1.69
0.048


1536
UBE2C
11065
ubiquitin-conjugating enzyme E2C
0.81
0.041
1.99
0.000


1537
UBE2D3
7323
ubiquitin-conjugating enzyme E2D 3
0.79
0.021
2.52
0.020


1538
UBE2J1
51465
ubiquitin-conjugating enzyme E2, J1
0.77
0.007
1.71
0.050


1539
UBE2K
3093
ubiquitin-conjugating enzyme E2K
0.76
0.006
1.80
0.006


1540
UBE2L3
7332
ubiquitin-conjugating enzyme E2L 3
0.75
0.007
2.67
0.000


1541
UBE2N
7334
ubiquitin-conjugating enzyme E2N
0.79
0.022
2.21
0.001


1542
UBE2S
27338
ubiquitin-conjugating enzyme E2S
0.76
0.008
1.44
0.002


1543
UBE2V2
7336
ubiquitin-conjugating enzyme E2
0.81
0.035
2.11
0.000





variant 2


1544
UBE4B
10277
ubiquitination factor E4B
0.79
0.025
0.48
0.032


1545
UBFD1
56061
ubiquitin family domain containing 1
0.77
0.015
1.71
0.038


1546
UBL4A
8266
ubiquitin-like 4A
0.80
0.024
1.69
0.015


1547
UBL5
59286
ubiquitin-like 5
0.78
0.017
1.86
0.004


1548
UBP1
7342
upstream binding protein 1 (LBP-1a)
0.79
0.017
0.46
0.007


1549
UBQLN2
29978
ubiquilin 2
0.80
0.023
1.83
0.045


1550
UBQLN4
56893
ubiquilin 4
0.74
0.003
0.60
0.002


1551
UBR4
23352
ubiquitin protein ligase E3 component
0.79
0.031
0.44
0.010





n-recognin 4


1552
UCHL1
7345
ubiquitin carboxyl-terminal esterase L1
0.82
0.049
1.38
0.038





(ubiquitin thiolesterase)


1553
UCHL3
7347
ubiquitin carboxyl-terminal esterase L3
0.77
0.007
1.47
0.014





(ubiquitin thiolesterase)


1554
UFD1L
7353
ubiquitin fusion degradation 1 like
0.75
0.005
1.75
0.004





(yeast)


1555
UIMC1
51720
ubiquitin interaction motif containing 1
0.79
0.016
0.34
0.012


1556
UMPS
7372
uridine monophosphate synthetase
0.79
0.021
2.60
0.004


1557
UQCRC2
7385
ubiquinol-cytochrome c reductase core
0.81
0.025
0.47
0.012





protein II


1558
URB2
9816
URB2 ribosome biogenesis 2 homolog
0.81
0.041
1.66
0.024





(S. cerevisiae)


1559
USO1
8615
USO1 vesicle transport factor
0.80
0.023
1.61
0.049


1560
USP18
11274
ubiquitin specific peptidase 18
0.76
0.008
1.29
0.045


1561
USP32
84669
ubiquitin specific peptidase 32
0.77
0.010
1.83
0.002


1562
USP34
9736
ubiquitin specific peptidase 34
0.78
0.013
0.70
0.032


1563
USP46
64854
ubiquitin specific peptidase 46
0.80
0.036
1.75
0.026


1564
USP9X
8239
ubiquitin specific peptidase 9, X-linked
0.79
0.012
2.71
0.007


1565
UTP18
51096
UTP18 small subunit (SSU)
0.79
0.017
1.49
0.034





processome component homolog





(yeast)


1566
UTP3
57050
UTP3, small subunit (SSU)
0.79
0.018
1.47
0.033





processome component, homolog (S.






cerevisiae)



1567
VAMP1
6843
vesicle-associated membrane protein 1
0.78
0.015
0.58
0.003





(synaptobrevin 1)


1568
VAMP2
6844
vesicle-associated membrane protein 2
0.78
0.015
0.52
0.006





(synaptobrevin 2)


1569
VAMP7
6845
vesicle-associated membrane protein 7
0.80
0.023
1.98
0.011


1570
VBP1
7411
von Hippel-Lindau binding protein 1
0.76
0.006
1.83
0.001


1571
VDAC1
7416
voltage-dependent anion channel 1
0.80
0.033
1.79
0.021


1572
VDAC2
7417
voltage-dependent anion channel 2
0.79
0.016
1.81
0.010


1573
VIM
7431
vimentin
0.81
0.039
0.70
0.018


1574
VIP
7432
vasoactive intestinal peptide
0.80
0.026
0.84
0.035


1575
VIPAS39
63894
VPS33B interacting protein, apical-
0.77
0.010
0.30
0.005





basolateral polarity regulator, spe-39





homolog


1576
VIPR1
7433
vasoactive intestinal peptide receptor 1
0.78
0.010
0.67
0.003


1577
VPS11
55823
vacuolar protein sorting 11 homolog
0.81
0.024
0.46
0.025





(S. cerevisiae)


1578
VPS51
738
vacuolar protein sorting 51 homolog
0.81
0.036
0.64
0.047





(S. cerevisiae)


1579
VPS53
55275
vacuolar protein sorting 53 homolog
0.79
0.017
0.71
0.028





(S. cerevisiae)


1580
VRK1
7443
vaccinia related kinase 1
0.80
0.020
1.47
0.011


1581
VWA1
64856
von Willebrand factor A domain
0.80
0.027
1.32
0.040





containing 1


1582
WASF2
10163
WAS protein family, member 2
0.80
0.038
0.50
0.002


1583
WBSCR22
114049
Williams Beuren syndrome
0.81
0.032
1.84
0.042





chromosome region 22


1584
WDR19
57728
WD repeat domain 19
0.82
0.039
0.60
0.013


1585
WDR26
80232
WD repeat domain 26
0.81
0.030
1.97
0.012


1586
WDR3
10885
WD repeat domain 3
0.80
0.022
1.84
0.029


1587
WDR73
84942
WD repeat domain 73
0.80
0.023
0.53
0.048


1588
WDR78
79819
WD repeat domain 78
0.78
0.014
0.65
0.033


1589
WDTC1
23038
WD and tetratricopeptide repeats 1
0.75
0.005
0.54
0.006


1590
WDYHV1
55093
WDYHV motif containing 1
0.80
0.027
1.81
0.004


1591
WISP3
8838
WNT1 inducible signaling pathway
0.80
0.021
0.84
0.021





protein 3


1592
WSB2
55884
WD repeat and SOCS box containing 2
0.77
0.007
2.73
0.000


1593
XDH
7498
xanthine dehydrogenase
0.76
0.009
0.63
0.009


1594
XPC
7508
xeroderma pigmentosum,
0.78
0.020
0.48
0.010





complementation group C


1595
XPOT
11260
exportin, tRNA
0.76
0.009
1.97
0.001


1596
YIF1A
10897
Yip1 interacting factor homolog A (S.
0.81
0.031
1.63
0.016






cerevisiae)



1597
YIPF6
286451
Yip1 domain family, member 6
0.78
0.018
1.54
0.010


1598
YKT6
10652
YKT6 v-SNARE homolog (S.
0.75
0.009
1.34
0.048






cerevisiae)



1599
YLPM1
56252
YLP motif containing 1
0.80
0.038
0.40
0.016


1600
YTHDF3
253943
YTH domain family, member 3
0.81
0.037
1.91
0.003


1601
ZBTB11
27107
zinc finger and BTB domain
0.78
0.015
1.96
0.017





containing 11


1602
ZBTB16
7704
zinc finger and BTB domain
0.78
0.018
0.73
0.001





containing 16


1603
ZBTB17
7709
zinc finger and BTB domain
0.82
0.046
0.44
0.027





containing 17


1604
ZBTB25
7597
zinc finger and BTB domain
0.78
0.015
0.66
0.048





containing 25


1605
ZC3H15
55854
zinc finger CCCH-type containing 15
0.79
0.010
2.67
0.000


1606
ZFHX3
463
zinc finger homeobox 3
0.79
0.026
1.26
0.028


1607
ZFHX4
79776
zinc finger homeobox 4
0.80
0.031
0.60
0.027


1608
ZFP2
80108
ZFP2 zinc finger protein
0.81
0.029
0.81
0.011


1609
ZFP36L1
677
ZFP36 ring finger protein-like 1
0.77
0.010
0.63
0.001


1610
ZFYVE9
9372
zinc finger, FYVE domain containing
0.79
0.016
0.52
0.009





9


1611
ZNF141
7700
zinc finger protein 141
0.79
0.021
0.85
0.044


1612
ZNF148
7707
zinc finger protein 148
0.80
0.027
1.73
0.022


1613
ZNF160
90338
zinc finger protein 160
0.77
0.008
0.76
0.015


1614
ZNF184
7738
zinc finger protein 184
0.76
0.009
1.44
0.020


1615
ZNF202
7753
zinc finger protein 202
0.81
0.035
0.48
0.042


1616
ZNF204P
7754
zinc finger protein 204, pseudogene
0.80
0.027
0.77
0.006


1617
ZNF213-AS1
100507458
ZNF213 antisense RNA 1 (head to
0.82
0.046
0.79
0.008





head)


1618
ZNF236
7776
zinc finger protein 236
0.73
0.004
0.57
0.018


1619
ZNF286A
57335
zinc finger protein 286A
0.77
0.011
0.79
0.022


1620
ZNF322
79692
zinc finger protein 322
0.75
0.009
1.68
0.031


1621
ZNF385D
79750
zinc finger protein 385D
0.82
0.043
0.82
0.011


1622
ZNF423
23090
zinc finger protein 423
0.82
0.048
0.71
0.017


1623
ZNF446
55663
zinc finger protein 446
0.78
0.012
0.73
0.014


1624
ZNF468
90333
zinc finger protein 468
0.80
0.026
1.54
0.046


1625
ZNF480
147657
zinc finger protein 480
0.80
0.034
1.45
0.047


1626
ZNF500
26048
zinc finger protein 500
0.77
0.005
0.54
0.006


1627
ZNF562
54811
zinc finger protein 562
0.76
0.007
0.52
0.013


1628
ZNF606
80095
zinc finger protein 606
0.80
0.024
1.93
0.005


1629
ZNF611
81856
zinc finger protein 611
0.76
0.008
0.71
0.019


1630
ZNF623
9831
zinc finger protein 623
0.80
0.035
1.94
0.000


1631
ZNF629
23361
zinc finger protein 629
0.76
0.008
0.37
0.010


1632
ZNF706
51123
zinc finger protein 706
0.80
0.030
1.72
0.001


1633
ZNF767P
79970
zinc finger family member 767,
0.77
0.010
0.59
0.031





pseudogene


1634
ZWILCH
55055
zwilch kinetochore protein
0.75
0.006
1.43
0.010


1635
ZWINT
11130
ZW10 interacting kinetochore protein
0.82
0.042
1.46
0.033









Evaluation on Biological Functions of Prognostic Genes with Increased Prediction Accuracy Through Gene Combinations


Gene Ontology (GO) analysis was used to investigate any association of the above selected 1,635 genes, which were found to improve such prediction accuracy through their combinatory use with BTN3A2 gene, with biological processes. As a result of GO analysis using relevant GO terms (such as mitotic cell cycle, cell cycle process, mitotic cell cycle phase transition, and so on), it was verified that those 1,635 genes are particularly involved in cell proliferation, as shown in the following Table 4.









TABLE 4







Gene Ontology (GO) analysis









GO Terms—Biological process
Nos. of Genes
Adjusted p-value












mitotic cell cycle
170
8.80E−17


cell cycle process
213
3.90E−16


establishment of protein localization
129
5.70E−16


to organelle




ribosome biogenesis
82
2.00E−15


mitotic cell cycle phase transition
106
7.10E−15









Further, among the 213 cell-proliferation-related genes, it was confirmed via bivariate Cox analysis that a total of 159 genes (159/213, 74.6%), which are named p-genes as described above, were associated with a poor prognosis of breast as shown in the following Table 5, suggesting that each of these 159 p-genes may be used in combination with BTN3A2 gene to increase the accuracy of prognosis prediction of a cancer, particularly a breast cancer.









TABLE 5







Bivariate Cox analysis on p-genes and BTN3A2










BTN3A2
p-gene















Gene



p-

p-


No
Symbol
Gene ID
Gene Name
HR
value
HR
value

















1
APITD1
378708
apoptosis-inducing, TAF9-like
0.81
0.041
1.86
0.027





domain 1


2
BID
637
BH3 interacting domain death agonist
0.77
0.007
1.72
0.011


3
BUB1B
701
BUB1 mitotic checkpoint
0.80
0.031
1.71
0.002





serine/threonine kinase B


4
BUB1
699
BUB1 mitotic checkpoint
0.78
0.014
1.73
0.005





serine/threonine kinase


5
CKS1B
1163
CDC28 protein kinase regulatory
0.78
0.013
1.52
0.027





subunit 1B


6
CKS2
1164
CDC28 protein kinase regulatory
0.79
0.023
1.55
0.002





subunit 2


7
DLGAP5
9787
discs, large (Drosophila) homolog-
0.75
0.004
1.76
0.000





associated protein 5


8
POLA1
5422
polymerase (DNA directed), alpha 1,
0.79
0.017
1.91
0.018





catalytic subunit


9
POLA2
23649
polymerase (DNA directed), alpha 2,
0.74
0.006
1.41
0.010





accessory subunit


10
DSCC1
79075
DNA replication and sister chromatid
0.80
0.021
1.93
0.000





cohesion 1


11
DNA2
1763
DNA replication helicase/nuclease 2
0.81
0.034
1.49
0.047


12
E2F8
79733
E2F transcription factor 8
0.81
0.025
1.46
0.004


13
ERCC6L
54821
excision repair cross-
0.74
0.003
1.44
0.000





complementation group 6-like


14
FBXO5
26271
F-box protein 5
0.74
0.005
1.49
0.012


15
FANCI
55215
Fanconi anemia, complementation
0.81
0.032
1.49
0.049





group I


16
GADD45GIP1
90480
growth arrest and DNA-damage-
0.77
0.008
1.64
0.002





inducible, gamma interacting protein 1


17
GINS1
9837
GINS complex subunit 1 (Psf1
0.82
0.044
1.42
0.014





homolog)


18
GINS2
51659
GINS complex subunit 2 (Psf2
0.78
0.018
1.18
0.033





homolog)


19
MAD2L1
4085
MAD2 mitotic arrest deficient-like 1
0.77
0.009
1.57
0.001





(yeast)


20
MAD2L1BP
9587
MAD2L1 binding protein
0.79
0.017
2.01
0.021


21
MIS18A
54069
MIS18 kinetochore protein A
0.81
0.029
1.86
0.033


22
MYBL2
4605
v-myb avian myeloblastosis viral
0.77
0.012
1.26
0.010





oncogene homolog-like 2


23
NAA50
80218
N(alpha)-acetyltransferase 50, NatE
0.81
0.030
1.60
0.037





catalytic subunit


24
NEK2
4751
NIMA-related kinase 2
0.80
0.026
1.45
0.003


25
NSL1
25936
NSL1, MIS12 kinetochore complex
0.78
0.015
1.96
0.020





component


26
PBK
55872
PDZ binding kinase
0.78
0.012
1.55
0.001


27
RAB11A
8766
RAB11A, member RAS oncogene
0.80
0.022
1.79
0.046





family


28
RAD51C
5889
RAD51 paralog C
0.77
0.015
1.42
0.047


29
RAD54B
25788
RAD54 homolog B (S. cerevisiae)
0.78
0.021
1.63
0.001


30
RANBP1
5902
RAN binding protein 1
0.79
0.021
1.53
0.033


31
RALA
5898
v-ral simian leukemia viral oncogene
0.77
0.007
2.50
0.000





homolog A (ras related)


32
RACGAP1
29127
Rac GTPase activating protein 1
0.77
0.013
1.75
0.000


33
SSNA1
8636
Sjogren syndrome nuclear
0.82
0.050
1.86
0.016





autoantigen 1


34
STAMBP
10617
STAM binding protein
0.81
0.035
3.38
0.001


35
SSSCA1
10534
Sjogren syndrome/scleroderma
0.76
0.012
1.43
0.005





autoantigen 1


36
TAF2
6873
TAF2 RNA polymerase II, TATA
0.80
0.027
1.70
0.007





box binding protein (TBP)-associated





factor, 150 kDa


37
TIPIN
54962
TIMELESS interacting protein
0.79
0.015
1.32
0.010


38
TIPRL
261726
TOR signaling pathway regulator
0.77
0.010
2.03
0.000


39
TRIAP1
51499
TP53 regulated inhibitor of apoptosis 1
0.81
0.040
2.38
0.019


40
TTK
7272
TTK protein kinase
0.77
0.006
1.59
0.000


41
ZWINT
11130
ZW10 interacting kinetochore protein
0.82
0.042
1.46
0.033


42
ASPM
259266
asp (abnormal spindle) homolog,
0.79
0.011
1.32
0.002





microcephaly associated (Drosophila)


43
AURKA
6790
aurora kinase A
0.80
0.024
1.59
0.002


44
AURKB
9212
aurora kinase B
0.81
0.029
1.26
0.044


45
BRD7
29117
bromodomain containing 7
0.76
0.007
1.54
0.021


46
CSNK2A1
1457
casein kinase 2, alpha 1 polypeptide
0.75
0.004
2.13
0.002


47
CDC20
991
cell division cycle 20
0.78
0.011
1.32
0.019


48
CDC25C
995
cell division cycle 25C
0.80
0.038
1.70
0.011


49
CENPA
1058
centromere protein A
0.73
0.002
2.00
0.000


50
CENPE
1062
centromere protein E, 312 kDa
0.78
0.012
1.33
0.001


51
CENPF
1063
centromere protein F, 350/400 kDa
0.79
0.016
1.64
0.002


52
CENPI
2491
centromere protein I
0.81
0.030
1.50
0.040


53
CENPM
79019
centromere protein M
0.75
0.005
1.39
0.001


54
CENPN
55839
centromere protein N
0.78
0.006
1.50
0.001


55
CENPU
79682
centromere protein U
0.79
0.025
1.77
0.000


56
CEP55
55165
centrosomal protein 55 kDa
0.75
0.007
1.48
0.001


57
CHEK1
1111
checkpoint kinase 1
0.80
0.024
1.75
0.016


58
CDT1
81620
chromatin licensing and DNA
0.78
0.015
1.32
0.040





replication factor 1


59
C11orf80
79703
chromosome 11 open reading frame
0.82
0.039
1.31
0.004





80


60
CCNA2
890
cyclin A2
0.82
0.043
1.70
0.006


61
CCNB1
891
cyclin B1
0.79
0.021
1.58
0.004


62
CCNB2
9133
cyclin B2
0.77
0.007
1.67
0.001


63
CCNE2
9134
cyclin E2
0.80
0.027
2.07
0.000


64
CDK1
983
cyclin-dependent kinase 1
0.76
0.005
1.56
0.001


65
CDKN3
1033
cyclin-dependent kinase inhibitor 3
0.81
0.037
2.26
0.000


66
CKAP5
9793
cytoskeleton associated protein 5
0.78
0.020
2.12
0.022


67
DTL
51514
d.enticleless E3 ubiquitin protein
0.80
0.023
1.47
0.003





ligase homolog (Drosophila)


68
DCTN2
10540
dynactin 2 (p50)
0.77
0.012
1.85
0.015


69
DYNLT1
6993
dynein, light chain, Tctex-type 1
0.77
0.010
2.21
0.003


70
ECD
11319
ecdysoneless homolog (Drosophila)
0.75
0.007
1.70
0.036


71
ECT2
1894
epithelial cell transforming 2
0.79
0.022
1.69
0.001


72
EIF4G1
1981
eukaryotic translation initiation factor
0.82
0.043
1.70
0.033





4 gamma, 1


73
EIF4EBP1
1978
eukaryotic translation initiation factor
0.77
0.010
1.45
0.003





4E binding protein 1


74
EZR
7430
ezrin
0.82
0.041
1.73
0.011


75
FEN1
2237
flap structure-specific endonuclease 1
0.77
0.012
1.77
0.003


76
FOXM1
2305
forkhead box M1
0.78
0.015
1.42
0.002


77
GSK3B
2932
glycogen synthase kinase 3 beta
0.79
0.017
1.99
0.003


78
HMGN5
79366
high mobility group nucleosome
0.83
0.048
1.50
0.040





binding domain 5


79
INTS7
25896
integrator complex subunit 7
0.75
0.007
1.61
0.014


80
KIF11
3832
kinesin family member 11
0.78
0.012
1.61
0.003


81
KIF14
9928
kinesin family member 14
0.78
0.013
1.73
0.001


82
KIF20A
10112
kinesin family member 20A
0.76
0.011
1.46
0.005


83
KIF23
9493
kinesin family member 23
0.79
0.021
1.22
0.028


84
KIF2C
11004
kinesin family member 2C
0.78
0.009
1.90
0.003


85
KIF4A
24137
kinesin family member 4A
0.79
0.016
1.73
0.003


86
KIFC1
3833
kinesin family member C1
0.78
0.015
1.35
0.003


87
MIF
4282
macrophage migration inhibitory
0.78
0.026
1.67
0.003





factor (glycosylation-inhibiting





factor)


88
MELK
9833
maternal embryonic leucine zipper
0.78
0.010
1.54
0.003





kinase


89
MED1
5469
mediator complex subunit 1
0.78
0.012
1.40
0.003


90
MCM10
55388
minichromosome maintenance
0.79
0.016
1.39
0.001





complex component 10


91
MCM2
4171
minichromosome maintenance
0.81
0.028
1.53
0.012





complex component 2


92
MCM6
4175
minichromosome maintenance
0.77
0.013
1.63
0.020





complex component 6


93
MAP2K1
5604
mitogen-activated protein kinase
0.77
0.014
1.99
0.016





kinase 1


94
MSH6
2956
mutS homolog 6
0.76
0.008
2.86
0.001


95
MLF1
4291
myeloid leukemia factor 1
0.80
0.027
1.39
0.041


96
NCAPG
64151
non-SMC condensin I complex,
0.76
0.005
1.98
0.000





subunit G


97
NUSAP1
51203
nucleolar and spindle associated
0.79
0.016
1.73
0.000





protein 1


98
NUP155
9631
nucleoporin 155 kDa
0.80
0.031
1.50
0.048


99
NUP93
9688
nucleoporin 93 kDa
0.75
0.005
1.17
0.028


100
ORC4
5000
origin recognition complex, subunit 4
0.78
0.013
1.55
0.036


101
ORC5
5001
origin recognition complex, subunit 5
0.77
0.011
1.86
0.021


102
PIN1
5300
peptidylprolyl cis/trans isomerase,
0.75
0.006
1.26
0.010





NIMA-interacting 1


103
PIK3R4
30849
phosphoinositide-3-kinase, regulatory
0.78
0.015
1.90
0.019





subunit 4


104
PTTG1
9232
pituitary tumor-transforming 1
0.80
0.017
2.08
0.000


105
PTTG3P
26255
pituitary tumor-transforming 3,
0.76
0.012
1.50
0.002





pseudogene


106
PLK1
5347
polo-like kinase 1
0.81
0.037
1.53
0.020


107
PLK4
10733
polo-like kinase 4
0.78
0.021
1.34
0.048


108
PRIM1
5557
primase, DNA, polypeptide 1
0.80
0.035
2.44
0.000





(49 kDa)


109
PA2G4
5036
proliferation-associated 2G4, 38 kDa
0.82
0.043
2.15
0.007


110
LEPREL4
10609
leprecan-like 4
0.79
0.029
1.30
0.046


111
PSMC3
5702
proteasome (prosome, macropain)
0.82
0.032
1.56
0.022





26S subunit, ATPase, 3


112
PSMC6
5706
proteasome (prosome, macropain)
0.78
0.016
1.58
0.031





26S subunit, ATPase, 6


113
PSMD10
5716
proteasome (prosome, macropain)
0.77
0.007
2.27
0.001





26S subunit, non-ATPase, 10


114
PSMD12
5718
proteasome (prosome, macropain)
0.79
0.018
1.93
0.003





26S subunit, non-ATPase, 12


115
PSMD14
10213
proteasome (prosome, macropain)
0.78
0.012
2.24
0.005





26S subunit, non-ATPase, 14


116
PSMD2
5708
proteasome (prosome, macropain)
0.80
0.025
2.42
0.001





26S subunit, non-ATPase, 2


117
PSMD3
5709
proteasome (prosome, macropain)
0.78
0.015
1.34
0.012





26S subunit, non-ATPase, 3


118
PSMD4
5710
proteasome (prosome, macropain)
0.80
0.025
1.79
0.032





26S subunit, non-ATPase, 4


119
PSMD6
9861
proteasome (prosome, macropain)
0.80
0.019
2.09
0.013





26S subunit, non-ATPase, 6


120
PSMD7
5713
proteasome (prosome, macropain)
0.78
0.010
2.22
0.003





26S subunit, non-ATPase, 7


121
PSMA1
5682
proteasome (prosome, macropain)
0.77
0.009
2.33
0.005





subunit, alpha type, 1


122
PSMA2
5683
proteasome (prosome, macropain)
0.78
0.010
1.94
0.010





subunit, alpha type, 2


123
PSMA3
5684
proteasome (prosome, macropain)
0.75
0.005
1.74
0.010





subunit, alpha type, 3


124
PSMA4
5685
proteasome (prosome, macropain)
0.78
0.007
2.27
0.009





subunit, alpha type, 4


125
PSMA6
9692
proteasome (prosome, macropain)
0.76
0.008
1.67
0.027





subunit, alpha type, 6


126
PSMA7
5688
proteasome (prosome, macropain)
0.79
0.020
1.69
0.006





subunit, alpha type, 7


127
PSMB3
5691
proteasome (prosome, macropain)
0.80
0.033
1.70
0.003





subunit, beta type, 3


128
PSMB5
5693
proteasome (prosome, macropain)
0.77
0.016
2.13
0.002





subunit, beta type, 5


129
PSMB7
5695
proteasome (prosome, macropain)
0.78
0.010
1.93
0.011





subunit, beta type, 7


130
PRMT1
3276
protein arginine methyltransferase 1
0.80
0.029
1.64
0.044


131
PPP2R3B
102725016
protein phosphatase 2, regulatory
0.83
0.049
1.24
0.022





subunit B″, beta


132
PPP3CA
5530
protein phosphatase 3, catalytic
0.82
0.039
1.52
0.020





subunit, alpha isozyme


133
PRC1
9055
protein regulator of cytokinesis 1
0.79
0.019
1.82
0.000


134
RRM2
6241
ribonucleotide reductase M2
0.77
0.010
1.50
0.004


135
RPS6KB1
6198
ribosomal protein S6 kinase, 70 kDa,
0.81
0.029
1.45
0.048





polypeptide 1


136
SPAG5
10615
sperm associated antigen 5
0.80
0.028
1.34
0.031


137
SKA1
220134
spindle and kinetochore associated
0.79
0.025
1.35
0.012





complex subunit 1


138
STMN1
3925
stathmin 1
0.78
0.013
1.51
0.041


139
SLBP
7884
stem-loop binding protein
0.80
0.036
2.56
0.001


140
SMC2
10592
structural maintenance of
0.78
0.011
1.95
0.004





chromosomes 2


141
SMC4
10051
structural maintenance of
0.80
0.019
1.67
0.007





chromosomes 4


142
SMC5
23137
structural maintenance of
0.83
0.046
1.77
0.015





chromosomes 5


143
TERF1
7013
telomeric repeat binding factor
0.82
0.038
1.59
0.030





(NIMA-interacting) 1


144
TXNL4A
10907
thioredoxin-like 4A
0.79
0.019
1.67
0.033


145
TRIP13
9319
thyroid hormone receptor interactor
0.80
0.026
1.79
0.003





13


146
TOP2A
7153
topoisomerase (DNA) II alpha
0.80
0.029
1.51
0.000





170 kDa


147
TFDP2
7029
transcription factor Dp-2 (E2F
0.78
0.019
1.51
0.034





dimerization partner 2)


148
TACC3
10460
transforming, acidic coiled-coil
0.75
0.005
1.25
0.013





containing protein 3


149
TUBB3
10381
tubulin, beta 3 class III
0.81
0.038
1.71
0.022


150
TUBB4B
10383
tubulin, beta 4B class IVb
0.80
0.026
1.87
0.013


151
TUBB
203068
tubulin, beta class I
0.78
0.018
1.75
0.047


152
TSG101
7251
tumor susceptibility 101
0.78
0.015
1.84
0.021


153
UBE2C
11065
ubiquitin-conjugating enzyme E2C
0.81
0.041
1.99
0.000


154
UBE2L3
7332
ubiquitin-conjugating enzyme E2L 3
0.75
0.007
2.67
0.000


155
UBE2S
27338
ubiquitin-conjugating enzyme E2S
0.76
0.008
1.44
0.002


156
USP9X
8239
ubiquitin specific peptidase 9, X-
0.79
0.012
2.71
0.007





linked


157
VRK1
7443
vaccinia related kinase 1
0.80
0.020
1.47
0.011


158
ZFHX3
463
zinc finger homeobox 3
0.79
0.026
1.26
0.028


159
ZWILCH
55055
zwilch kinetochore protein
0.75
0.006
1.43
0.010


160
MMP11
4320
matrix metallopeptidase 11
0.81
0.048
1.18
0.050





(stromelysin 3)









Combinatory Use of BTN3A2 and RRM2 gene for Prognosis Prediction


Following is the description of the combinatory use of one of the p-genes, i.e. RRM2 (Ribonucleotide Reductase M2) and BTN3A2 for the prognosis prediction of breast cancer. As shown in Table 6 below, bivariate Cox analysis indicates that BTN3A2 and RRM2 are an independent prognostic factor associated with the prognosis of breast cancer, respectively. In particular, it was confirmed that BTN3A2 is correlated with a good prognosis of breast cancer (HR<1), whereas RRM2 with a poor prognosis (HR>1).









TABLE 6







Results of Bivariate Cox analysis on BTN3A2 and RRM2 genes











HR





(Hazard




Gene
Ratio)
95% C.I.
p-value





BTN3A2
0.77
0.64 to 0.94
0.010


RRM2
1.50
1.14 to 1.98
0.004









Further, as shown in FIG. 9, it was found that C-index (0.63) of the combination of BTN3A2 and RRM2 was larger than that of either BTN3A2 (0.55) or RRM2 (0.61) alone, suggesting that the combinatory use of the i-gene BTN3A2 and at least one p-gene would lead to an improved accuracy of the prognosis prediction.


In conclusion, these results suggest that the combinatory use of the i-genes (such as BTN3A2) with various genes may improve its prognostic performance. Particularly, such a combinatory use of the i-gene and the proliferation-related p-gene (such as RRM2) may significantly increase the prediction accuracy of survival probabilities in a breast cancer patient.


As set forth above, the present invention provides a genetic marker for early-stage breast cancer prognosis prediction and diagnosis. The genetic marker of the present invention can enable the prediction or diagnosis of the prognosis of a breast cancer patient, and thus can be favorably used to present clues for the future direction of breast cancer treatment, including the determination on whether anticancer treatment is needed.


It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the spirit or scope of the invention. Thus, it is intended that the present invention cover the modifications and variations of this invention provided they come within the scope of the appended claims and their equivalents.

Claims
  • 1. A method for treating a breast cancer in a breast cancer patient, the method comprising the steps of: collecting a sample from the breast cancer patient;isolating mRNA from the sample from the breast cancer patient;measuring a first mRNA expression level for the mRNA of an i-gene BTN3A2 (butyrophilin, subfamily 3, member A2) and a second mRNA expression level for the mRNA of RRM2 (Ribonucleotide Reductase M2);normalizing the first and second mRNA expression levels to determine a normalized value;detecting in the breast cancer patient sample a decrease in normalized value of BTN3A2 compared to a reference breast tumor or an increased normalized value of RRM2 compared to a reference breast tumor;diagnosing the breast cancer patient who has a decrease in normalized value of BTN3A2 compared to a reference breast tumor sample or has an increased normalized value of RRM2 compared to a reference breast tumor sample as requiring treatment; andtreating the diagnosed breast cancer patient by administering at least one of an anti-cancer agent, a surgery, and a radiation therapy,wherein the method comprises a step of using a plurality of primer pairs, wherein the plurality of the primer pairs comprises a primer pair for the i-gene BTN3A2 (butyrophilin, subfamily 3, member A2), and a primer pair for the p-gene RRM2, wherein the primer pairs are selected to amplify the i-gene and the p-gene through PCR amplification.
  • 2. The method of claim 1, wherein the sample is a formalin-fixed paraffin-embedded (FFPE) sample of tissue containing cancer cells of the breast cancer patient.
  • 3. The method of claim 1, wherein the step of normalizing is conducted by calculating a ratio of a mean expression level of the gene with a mean expression level of at least one standard gene selected from the group consisting of CTBP1 (C-terminal-binding protein 1), TBP (TATA-binding protein), HMBS (hydroxymethylbilane synthase), CUL1 (cullin 1), and UBQLN1 (ubiquilin-1).
Priority Claims (1)
Number Date Country Kind
10-2013-0043160 Apr 2013 KR national
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation-in-part of U.S. application Ser. No. 14/886,948, filed on Oct. 19, 2015, which is a continuation of International Application PCT/KR2014/003384, filed on Apr. 18, 2014, which claims priority from and the benefit of Korean Patent Application No. 10-2013-0043160, filed on Apr. 18, 2013, which is hereby incorporated by reference for all purposes as if fully set forth herein.

US Referenced Citations (3)
Number Name Date Kind
8557525 Wang Oct 2013 B1
20060246470 Fuqua Nov 2006 A1
20100113297 Lidereau May 2010 A1
Foreign Referenced Citations (3)
Number Date Country
10-2012-0079295 Jul 2012 KR
10-1183522 Sep 2012 KR
2012093821 Jul 2012 WO
Non-Patent Literature Citations (24)
Entry
Pawitan et al. (Breast Cancer Research, vol. 7, R953-964, 2005) (Year: 2005).
Lee et al. (J. of Biochemistry and Molecular Biology, vol. 40, No. 2, pp. 226-231, Mar. 2007). (Year: 2007).
Chang, H.Y., et al., “Gene Expression Signature of Fibroblast Serum Response Predicts Human Cancer Progression: Similarities between Tumors and Wounds”, PLoS Biology, Feb. 2004, pp. 0206-0214, vol. 2, Issue 2.
Marc J. Van De Vijver, M.D., Ph.D, et al., “A Gene-Expression Signature As a Predictor of Survival in Breast Cancer”, The New England Journal of Medicine, Dec. 19, 2002, pp. 1999-2009, vol. 347, No. 25.
Laura J. Van'T Veer, et al., “Gene Expression Profiling Predicts Clinical Outcome of Breast Cancer”, Macmillan Magazines Ltd, Jan. 31, 2002, pp. 530-536, vol. 415.
Yixin Wang, et al., “Gene-Expression Profiles to Predict Distant Metastasis of Lymph-Node-Negative Primary Breast Cancer”, The Lancet, Feb. 19, 2005, pp. 671-679, vol. 365.
Marc Buyse, et al., “Validation and Clinical Utility of a 70-Gene Prognostic Signature for Women Wth Node-Negative Breast Cancer”, Journal of the National Cancer Institute, Sep. 6, 2006, pp. 1183-1192, vol. 98, No. 17.
Soonmyung Paik, et al., “Development and Clinical Utility of a 21-Gene Recurrence Score Prognostic Assay in Patients with Early Breast Cancer Treated with Tamoxifen”, The Oncologist, 2007, pp. 631-635, vol. 12.
Soonmyung Paik, M.D., et al., “A Multigene Assay to Predict Recurrence of Tamoxifen-Treated, Node-Negative Breast Cancer”, The New England Journal of Medicine, Dec. 30, 2004, pp. 2817-2826.
Christos Sotiriou, et al., “Gene Expression Profiling in Breast Cancer: Understanding the Molecular Basis of Histologic Grade to Improve Prognosis”, Journal of the National Cancer Institute, Feb. 15, 2006, pp. 261-272, vol. 98.
Yudi Pawitan, et al., “Gene Expression Profiling Spares Early Breast Cancer Patients from Adjuvant Therapy: Derived and Validated in Two Population-based Chohorts”, Breast Cancer Research, 2005, pp. R953-R964, vol. 7, No. 6.
Lance D. Miller, et al., “An Expression Signature for p53 Status in Human Breast Cancer Predicts Mutation Status, Transcriptional Effects, and Patient Survival”, PNAS, Sep. 20, 2005, pp. 13550 13555, vol. 102, No. 38.
Andrea H. Bild, et al. “Oncogenic Pathway Signatures in Human Cancers as a Guide to Targeted Therapies”, Nature, Jan. 19, 2006, pp. 353-357, vol. 439.
Andrew E. Teschedorff, et al. “A Consensus Prognostic Gene Expression Classifier for ER Positive Breast Cancer”, Genome Biology, Oct. 31, 2006, vol. 7.
Christine Desmedt, et al. “Strong Time Dependence of the 76-Gene Prognostic Signature for Node-Negative Breast Cancer Patients in the TRANSBIG Multicenter Independent Validation Series”, American Association for Cancer Research, Sep. 30, 2015, pp. 3207-3214.
Tony E. Godfrey, et al. “Quantitative mRNA Expression Analysis from Formalin-Fixed, Paraffin-Embedded Tissues Using 5′ Nuclease Quantitative Reverse Transcription-Polymerase Chain Reaction”, Journal of Molecular Diagnostics, May 2000, pp. 84-91, vol. 2, No. 2.
Katja Specht, et al. “Quantitative Gene Expression Analysis in Microdissected Archival Formalin-Fixed and Paraffin-Embedded Tumor Tissue”, American Journal of Patbology, Feb. 2001, vol. 158, No. 2.
Christian A. Heid, et al. “Real Time Quantitative PCR”, Genome Methods, Sep. 30, 2015, Published by Cold Spring Harbor Laboratory Press, Jun. 3, 1996, pp. 986-994.
Cecile Le Page, et al. “BTN3A2 Expression in Epithelial Ovarian Cancer is Associated with Higher Tumor Infiltrating T Cells and a Better Prognosis” PLoS One, Jun. 2012, pp. 1-12, vol. 7.
Stavropoulos et al. (PNAS, vol. 98, No. 18, pp. 10232-10237, Aug. 2001). (Year: 2001).
LePage et al, (Cancer Epidemiol Biomarkers Prev, vol. 17, No. 4, pp. 913-920, Apr. 2008).
Non-Final Office Action dated May 19, 2017, issued in U.S. Appl. No. 14/886,948.
Non-Final Office Action dated Oct. 12, 2017, issued in U.S. Appl. No. 14/886,948.
Final Office Action dated Apr. 10, 2018, issued in U.S. Appl. No. 14/886,948.
Related Publications (1)
Number Date Country
20190040473 A1 Feb 2019 US
Continuations (1)
Number Date Country
Parent PCT/KR2014/003384 Apr 2014 US
Child 14886948 US
Continuation in Parts (1)
Number Date Country
Parent 14886948 Oct 2015 US
Child 16155220 US